WO2019149178A1 - 吡喃葡萄糖基衍生物及其用途 - Google Patents

吡喃葡萄糖基衍生物及其用途 Download PDF

Info

Publication number
WO2019149178A1
WO2019149178A1 PCT/CN2019/073550 CN2019073550W WO2019149178A1 WO 2019149178 A1 WO2019149178 A1 WO 2019149178A1 CN 2019073550 W CN2019073550 W CN 2019073550W WO 2019149178 A1 WO2019149178 A1 WO 2019149178A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
atoms
alkylene
methyl
alkyl
Prior art date
Application number
PCT/CN2019/073550
Other languages
English (en)
French (fr)
Inventor
顾峥
伍武勇
康盼盼
曲桐
张宗远
黄伟明
吴天云
张英俊
Original Assignee
广东东阳光药业有限公司
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 广东东阳光药业有限公司 filed Critical 广东东阳光药业有限公司
Priority to US16/965,438 priority Critical patent/US11186602B2/en
Priority to CN201980008449.3A priority patent/CN111630058B/zh
Priority to EP19748264.9A priority patent/EP3747892A4/en
Publication of WO2019149178A1 publication Critical patent/WO2019149178A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H9/00Compounds containing a hetero ring sharing at least two hetero atoms with a saccharide radical
    • C07H9/02Compounds containing a hetero ring sharing at least two hetero atoms with a saccharide radical the hetero ring containing only oxygen as ring hetero atoms
    • C07H9/04Cyclic acetals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H3/00Compounds containing only hydrogen atoms and saccharide radicals having only carbon, hydrogen, and oxygen atoms
    • C07H3/10Anhydrosugars, e.g. epoxides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/08Bridged systems

Definitions

  • the present invention belongs to the field of medicine, and particularly relates to a glucopyranosyl derivative which is an inhibitor of sodium-dependent glucose transporters (SGLTs), particularly as an inhibitor of sodium-dependent glucose transporter 1 (SGLT1), a method for preparing the same, Pharmaceutical compositions comprising the derivatives and uses of the derivatives and compositions thereof. More specifically, it is a compound represented by the formula (I) or a pharmaceutically acceptable salt thereof or a stereoisomer thereof or a pharmaceutical composition containing the same, and the compound and the pharmaceutical composition for preparation Use of drugs to treat diabetes and diabetes related diseases.
  • SGLTs sodium-dependent glucose transporters
  • SGLT1 sodium-dependent glucose transporter 1
  • Diabetes is a common chronic disease characterized by hyperglycemia, which is accompanied by insulin resistance in peripheral tissues, decreased insulin secretion in the body, and an increase in hepatic gluconeogenesis.
  • additional insulin or oral hypoglycemic agents are needed for treatment.
  • Current hypoglycemic agents include biguanides, sulfonylureas, insulin sensitizers, glinides, alpha-glucosidase inhibitors, and DPP-IV (dipeptidyl peptidase IV) inhibitors.
  • hypoglycemic drugs are lacking, the biguanides cause lactic acidosis, the sulfonylureas can cause severe hypoglycemia, and the inappropriate use of the glinide can also cause hypoglycemia.
  • Insulin sensitizers can cause edema and heart. Insufficiency and weight gain, ⁇ -glucosidase inhibitors cause abdominal flatulence and diarrhea, and DPP-IV inhibitors need to be combined with metformin to achieve the desired hypoglycemic effect. Therefore, there is an urgent need to develop new safer and more effective hypoglycemic agents.
  • SGLT2 is mainly distributed in the kidney, mainly expressed in the S1 segment of the renal proximal convoluted tubule, and transports glucose at a ratio of 1:1 to sodium-glucose.
  • SGLTs actively transport glucose in an inverse concentration gradient while consuming energy, while GLUTs transport glucose in a concentration-prone manner in a diffusion-prone manner, and the transport process does not consume energy.
  • SGLT2 is the first level to regulate cellular glucose metabolism, and are also an ideal target for effective treatment of diabetes.
  • SGLTs inhibitors Inhibition of SGLTs does not affect the normal glucose counter-regulatory mechanism and causes hypoglycemia; at the same time, lowering blood glucose by increasing renal glucose excretion can cause weight loss in obese patients.
  • the study also found that the mechanism of action of SGLTs inhibitors does not depend on the degree of islet ⁇ -cell dysfunction or insulin resistance, and therefore, its effect does not decrease with ⁇ -cell function failure or severe insulin resistance. It can be used alone or in combination with other hypoglycemic agents to better exert hypoglycemic effects through complementary mechanisms. Therefore, SGLTs inhibitors are ideal new hypoglycemic agents.
  • SGLTs inhibitors can be used in the treatment of diabetes-related complications. Such as retinopathy, neuropathy, kidney disease, insulin resistance caused by glucose metabolism disorders, hyperinsulinemia, hyperlipidemia, obesity and so on.
  • SGLTs inhibitors can also be used in combination with existing therapeutic drugs, such as sulfonamide, thiazolidinedione, metformin and insulin, to reduce the dose without affecting the efficacy, thereby avoiding or reducing adverse reactions. Occurrence, improving patient compliance with treatment.
  • SGLT2 inhibitors are selective SGLT2 inhibitors.
  • SGLT1 inhibitors may exhibit greater benefits from inhibiting glucose reabsorption (U.S. Patent Application Publication No. US20110218159). It has been reported that in patients with congenital SGLT1 abnormalities, there is insufficient absorption of glucose and galactose, which provides a basis for reducing the absorption of carbohydrates by inhibiting SGLT1 activity.
  • the present invention provides a class of compounds having significant/excellent SGLTs inhibitory activity, particularly SGLT1 inhibitory activity, for improving intestinal environment; or for treating diabetes, insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipemia Disease, obesity, X syndrome, atherosclerosis, cardiovascular disease, congestive heart failure, hypomagnesemia, hyponatremia, renal failure, disorders associated with blood concentration, constipation or hypertension, and their complication.
  • the invention also provides methods of preparing such compounds, pharmaceutical compositions comprising the same, and methods of using the compounds and compositions to prepare a medicament for treating the above-described diseases in mammals, particularly humans, as compared to existing analogous compounds
  • the compound of the present invention not only has better pharmacological activity, but also has more excellent in vivo metabolic kinetic properties and in vivo pharmacodynamic properties.
  • the compound of the present invention has excellent SGLT1 inhibitory activity and thus has an excellent hypoglycemic and urinary excretion effect. Therefore, the compound provided by the present invention has more excellent drug-yielding properties than the currently-prepared compound.
  • the invention relates to a compound which is a stereoisomer, geometric isomer, tautomer, oxynitride, solvate of a compound of formula (I) or a compound of formula (I). , metabolites, pharmaceutically acceptable salts, dimers, trimers or prodrugs thereof,
  • q 1, 2, 3, 4, 5 or 6;
  • R 2 and R 3 are each independently H, D, CN, OH, NH 2 , -SH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy a heterocyclic group consisting of a C 1-6 alkylthio group, a C 1-6 alkylamino group, a C 3-6 cycloalkyl group or a 3-6 atom, wherein the C 1-6 alkyl group, C 2 ⁇ 6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, C 1-6 alkylthio, C 1-6 alkylamino, C 3-6 cycloalkyl and 3-6 atomic hetero
  • R 2 , R 3 together with the carbon atom to which they are attached form a carbonyl group
  • R 2 , R 3 together with the carbon atom to which they are attached form a C 3-6 carbocyclic ring or a heterocyclic ring composed of 3 to 6 atoms, wherein the C 3-6 carbocyclic ring and 3 to 6 atoms are composed of
  • R 10 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , C 1-6 alkyl or C 1-6 alkoxy;
  • R 6 and R 7 are each independently H, D, F, Cl, Br, I, CN, NO 2 , OH, NH 2 , C 1-6 alkyl, C 1-6 alkoxy, C 1-6 Alkylamino, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 3-8 cycloalkyl, heterocyclic group consisting of 3-8 atoms, C 6-10 aryl or 5-8 atoms Heteroaryl, wherein the C 1-6 alkyl group, C 1-6 alkoxy group, C 1-6 alkylamino group, C 1-6 haloalkyl group, C 1-6 haloalkoxy group, C 3-8 A cycloalkyl group, a heterocyclic group of 3-8 atoms, a C 6-10 aryl group, and a heteroaryl group of 5-8 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents.
  • heterocyclic ring composed of 3-8 atoms and the heteroaryl ring composed of 5-8 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents, the substitution
  • R 8 and R 9 are each independently H, D, R e OC 1-4 alkylene, R d R c NC 1-4 alkylene, C 1-8 alkyl, C 1-6 haloalkyl, C 3 -8 cycloalkyl, C 3-8 cycloalkyl-C 1-4 alkylene, heterocyclic group consisting of 3-8 atoms, (heterocyclic group consisting of 3-8 atoms) -C 1-4 Alkylene group, C 6-10 aryl group, C 6-10 aryl-C 1-4 alkylene group, heteroaryl group composed of 5-8 atoms or (heteroaryl group of 5-8 atoms)- a C 1-4 alkylene group, wherein the C 1-8 alkyl group, the C 1-6 haloalkyl group, the C 3-8 cycloalkyl group, the C 3-8 cycloalkyl-C 1-4 alkylene group, 3 Heterocyclic group consisting of -8 atoms, (hetero
  • R 8 , R 9 together with the nitrogen atom to which they are attached form a heterocyclic ring of 3-8 atoms or a heteroaromatic ring of 5-8 atoms, wherein the heterocyclic ring consisting of 3-8 atoms
  • the heteroaryl rings of 5-8 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents independently selected from D, F, Cl, Br, I, CN.
  • R c , R d , R e and R f are each independently H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-4 alkylene group, heterocyclic group composed of 3-8 atoms, (heterocyclic group of 3-8 atoms)-C 1-4 alkylene group, C 6-10 aryl group, C 6-10 An aryl-C 1-4 alkylene group, a heteroaryl group of 5-8 atoms or a (heteroaryl group of 5-8 atoms)-C 1-4 alkylene group, wherein the C 1- 6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-4 alkylene, heterocyclic group consisting of 3-8 atoms, (3-8 Heterocyclic group consisting of a hetero atom)-C 1-4 alkylene group, C 6
  • n 0, 1, 2 or 3;
  • t is 0, 1, 2, 3, 4, 5 or 6; with the proviso that when R 5 and R 8 are both H and R 9 is When t is not 0.
  • the present invention relates to a compound of the formula (II) or a stereoisomer, geometric isomer, tautomer, oxynitride, solvate, metabolite, pharmacy thereof.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , n and t have the definitions described in the present invention.
  • R 1 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , methyl, ethyl, n-propyl, isopropyl, propenyl, propyl Alkynyl, methoxy, ethoxy, hydroxymethyl, trifluoromethyl, trifluoroethyl, monofluoromethyl, trifluoromethoxy, difluoromethoxy, methylamino, cyclopropyl, ring Butyl, cyclopentyl, cyclohexyl or cyclopropyl-methylene, wherein the methyl, ethyl, n-propyl, isopropyl, propenyl, propynyl, methoxy, ethoxy groups , hydroxymethyl, trifluoroethyl, monofluoromethyl, difluoromethoxy, methylamino, cyclopropyl, cyclobutyl
  • R 2 and R 3 are each independently H, D, CN, OH, NH 2 , —SH, methyl, ethyl, n-propyl, isopropyl, vinyl, propynyl, a methoxy group, an ethoxy group, a methylthio group, a methylamino group, a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, a cyclohexyl group or a heterocyclic group consisting of 5-6 atoms, wherein the methyl group and the ethyl group are , n-propyl, isopropyl, vinyl, propynyl, methoxy, ethoxy, methylthio, methylamino, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and 5-6
  • the heterocyclic groups consisting of atoms are each independently unsubstidi
  • R 2 , R 3 together with the carbon atom to which they are attached form a carbonyl group
  • R 4 is H, D, -OR 4a or -SR 4b ;
  • R 4a and R 4b are each independently H, D, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 3-6 cycloalkyl- C 1-2 alkylene, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, (heterocyclic ring of 5-6 atoms) -C 1-2 alkylene, phenyl, phenyl-C 1-2 alkylene, furyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, cacao Azolyl, isoxazolyl, oxadiazolyl, 1,3,5-triazinyl,
  • R 10 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , methyl, ethyl, n-propyl, isopropyl, methoxy or Ethoxy.
  • R 6 and R 7 are each independently H, D, F, Cl, Br, I, CN, NO 2 , OH, NH 2 , C 1-4 alkyl, C 1-4 alkoxy, C 1-4 Alkylamino, C 1-4 haloalkyl, C 1-4 haloalkoxy, C 3-6 cycloalkyl, heterocyclic group consisting of 5-6 atoms, C 6-10 aryl or 5-6 atoms Heteroaryl, wherein the C 1-4 alkyl group, C 1-4 alkoxy group, C 1-4 alkylamino group, C 1-4 haloalkyl group, C 1-4 haloalkoxy group, C 3-6 A cycloalkyl group, a heterocyclic group of 5-6 atoms, a C 6-10 aryl group, and a heteroaryl group of 5-6 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents.
  • R 5 , R 6 together with the atoms to which they are attached, or R 5 , R 7 and the atoms to which they are attached, form a heterocyclic ring of 3-6 atoms or a heteroaryl composed of 5-6 atoms.
  • the heterocyclic ring composed of 3-6 atoms and the heteroaryl ring composed of 5-6 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents, the substitution
  • R 5 is H, D, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, trifluoromethyl, cyclopropyl, cyclo Butyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazine Base, phenyl, furanyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, oxadiazolyl, 1,3,5-triazinyl, thiazolyl , thienyl, pyr
  • R 6 , R 7 together with the carbon atom to which they are attached form cyclopropane, cyclobutane, cyclopentane, cyclohexane, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyran, tetrahydrothiopyran, piperazine Pyridinium, morpholine, thiomorpholine, piperazine, benzene ring, furan, pyrrole, pyridine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3,5-triazine, thiazole , thiophene, pyrazine, pyridazine or pyrimidine, wherein the cyclopropane, cyclobutane, cyclopentane, cyclohexane, pyrrolidine, tetrahydrofuran
  • R 5 , R 6 together with the atoms to which they are attached, or R 5 , R 7 and the atoms to which they are attached, form aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine , oxazolidine, thiazolidine, piperidine, morpholine, thiomorpholine, piperazine, pyrrole, pyridine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3,5- a triazine, a thiazole, a pyrazine, a pyridazine or a pyrimidine, wherein the aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, Thiomorpholine, piperazine, pyrrole, pyridine
  • R 8 and R 9 are each independently H, D, R e OC 1-4 alkylene, R d R c NC 1-4 alkylene, C 1-6 alkyl, C 1-4 haloalkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyl-C 1-2 alkylene, heterocyclic group consisting of 5-6 atoms, (5-6 atomic hetero Cycloalkyl)-C 1-2 alkylene, C 6-10 aryl, C 6-10 aryl-C 1-2 alkylene, heteroaryl consisting of 5-6 atoms or (5-6 Atomic composition of heteroaryl)-C 1-2 alkylene, wherein said C 1-6 alkyl, C 1-4 haloalkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyl-C 1-2 alkylene group, heterocyclic group composed of 5-6 atoms, (heterocyclic group of 5-6 atoms)-C 1-2 alkylene group, C 6-10
  • R 8 , R 9 together with the nitrogen atom to which they are attached form a heterocyclic ring composed of 3-6 atoms or a heteroaryl ring composed of 5-6 atoms, wherein the heterocyclic ring composed of the 3-6 atoms
  • the heteroaryl rings of 5-6 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents independently selected from D, F, Cl, Br, I, CN.
  • R 8 , R 9 together with the nitrogen atom to which they are attached form aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, thio Morpholine, piperazine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3,5-triazine, thiazole, pyrazine, pyridazine or pyrimidine, wherein the aziridine , azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, thiomorpholine, piperazine, pyrazole, imidazole, triazole, tetrazole, evil An azole, an oxadiazole, a 1,3,5-triazine
  • R c , R d , R e , and R f are each independently H, D, C 1-4 alkyl, C 1-4 haloalkyl, C 3-6 cycloalkyl, C 3 - 6 -cycloalkyl-C 1-2 alkylene group, heterocyclic group composed of 5-6 atoms, (heterocyclic group of 5-6 atoms)-C 1-2 alkylene group, C 6-10 aryl group a C 6-10 aryl-C 1-2 alkylene group, a heteroaryl group of 5-6 atoms or a heteroaryl group of 5-6 atoms, a C 1-2 alkylene group, wherein , the C 1-4 alkyl group, the C 1-4 haloalkyl group, the C 3-6 cycloalkyl group, the C 3-6 cycloalkyl-C 1-2 alkylene group, the heterocyclic ring composed of 5-6 atoms Base, (heterocyclic group of 5-6
  • R c , R d , R e , and R f are each independently H, D, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl , trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 3-6 cycloalkyl-C 1-2 alkylene, pyrrolidinyl, pyrazolidinyl, imidazolidinyl , oxazolidinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, (5 Heterocyclic group consisting of -6 atoms
  • R c , R d together with the nitrogen atom to which they are attached form aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, thio Morpholine, piperazine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3,5-triazine, thiazole, pyrazine, pyridazine or pyrimidine, wherein the aziridine , azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, thiomorpholine, piperazine, pyrazole, imidazole, triazole, tetrazole, evil The azole, oxadiazole, 1,3,5-triazine,
  • the invention in another aspect, relates to a pharmaceutical composition comprising a compound of the invention.
  • compositions of the present invention further comprise a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle, or combination thereof.
  • the pharmaceutical composition of the present invention further comprises one or more additional additional therapeutic agents, wherein the additional additional therapeutic agent is selected from the group consisting of an anti-diabetic drug, an anti-hyperglycemic drug, an anti-obesity drug, An antihypertensive drug, an appetite suppressing drug, a lipid lowering drug, or a combination thereof.
  • the additional additional therapeutic agent is selected from the group consisting of an anti-diabetic drug, an anti-hyperglycemic drug, an anti-obesity drug, An antihypertensive drug, an appetite suppressing drug, a lipid lowering drug, or a combination thereof.
  • the antidiabetic and antihyperglycemic agents of the invention are each independently selected from the group consisting of an SGLT2 inhibitor, a biguanide, a sulfonylurea, a glucosidase inhibitor, a PPAR agonist.
  • Oxide proliferator-activated receptor agonist Oxide proliferator-activated receptor agonist
  • ⁇ P2 inhibitor adipocyte fatty acid binding protein inhibitor
  • PPAR ⁇ / ⁇ dual activator peroxisome proliferator-activated receptor ⁇ / ⁇ dual activator
  • dipeptidyl peptidase IV inhibitor dipeptidyl peptidase IV inhibitor
  • glinide class insulin
  • glucagon-like peptide-1 inhibitor PTP1B inhibitor
  • PTP1B inhibitor protein tyrosine phosphatase 1B inhibitor
  • glycogen phosphorylase inhibition Agent glucose-6-phosphatase inhibitor or a combination thereof.
  • the anti-obesity agent of the present invention is selected from the group consisting of central anti-obesity agents, MCH (anthraquinone-concentrating hormone) receptor antagonists, neuropeptide Y receptor antagonists, and cannabinoids.
  • MCH anthraquinone-concentrating hormone
  • neuropeptide Y receptor antagonists neuropeptide Y receptor antagonists
  • cannabinoids cannabinoids.
  • Body antagonist brain gut peptide antagonist, lipase inhibitor, ⁇ 3 agonist, 11 ⁇ -HSD1 (11 ⁇ hydroxysteroid dehydrogenase 1) inhibitor, DGAT-1 (diacylglycerol acyltransferase 1) inhibitor, peptide A sexual appetite suppressant, a Cholecystokinin agonist, a feeding inhibitor, or a combination thereof.
  • the lipid-lowering drug of the present invention is selected from the group consisting of an MTP inhibitor (microsomal triglyceride transfer protein inhibitor) and an HMGCoA reductase inhibitor (hydroxymethylglutaryl coenzyme A reductase inhibitor) , squalene synthetase inhibitor, betabutyric acid hypolipidemic drug (also known as fibrate lipid-lowering drug), ACAT inhibitor (acetylcholesteryl acetyltransferase inhibitor), lipoxygenase inhibitor, cholesterol absorption inhibition Agent, ileal sodium ion/bile acid cotransporter inhibitor, upregulator of LDL receptor activity, niacin hypolipidemic drug, bile acid chelate or a combination thereof.
  • MTP inhibitor microsomal triglyceride transfer protein inhibitor
  • HMGCoA reductase inhibitor hydroxymethylglutaryl coenzyme A reductase inhibitor
  • squalene synthetase inhibitor hydroxymethylg
  • the lipid lowering drug of the present invention is selected from the group consisting of pravastatin, simvastatin, atorvastatin, fluvastatin, cerivastatin, etavatatin, russatin, or a combination thereof.
  • 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, wherein the medicament is for inhibiting SGLT1.
  • 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, wherein the medicament is for improving the intestinal environment.
  • the present invention relates to the use of a compound of the present invention or the pharmaceutical composition for the preparation of a medicament, wherein the medicament is for preventing or treating a disease, alleviating the symptoms of the disease or delaying the disease.
  • Development or seizure wherein the disease is diabetes, diabetic complications, insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipidemia, obesity, syndrome X, atherosclerosis, cardiovascular disease, congestive heart Failure, hypomagnesemia, hyponatremia, renal failure, disorders associated with blood concentration, constipation or high blood pressure.
  • the invention in another aspect, relates to a method of inhibiting SGLT1 comprising administering to a patient an effective amount of a compound of the invention or a pharmaceutical composition of the invention.
  • the invention in another aspect, relates to a method of improving intestinal environment comprising administering to a patient an effective amount of a compound of the invention or a pharmaceutical composition of the invention.
  • the invention in another aspect, relates to a method of preventing or treating a disease, alleviating the symptoms of the disease, or delaying the progression or onset of the disease, comprising administering to the patient an effective amount of a compound of the invention or a method of the invention a pharmaceutical composition, wherein the disease is diabetes, diabetic complications, insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipidemia, obesity, X syndrome, atherosclerosis, cardiovascular disease Congestive heart failure, hypomagnesemia, hyponatremia, renal failure, disorders associated with blood concentration, constipation or high blood pressure.
  • the invention relates to the use of a compound of the invention or a pharmaceutical composition of the invention for inhibiting SGLT1.
  • the invention relates to the use of a compound of the invention or a pharmaceutical composition of the invention for improving the intestinal environment.
  • the invention relates to the use of a compound of the invention or a pharmaceutical composition according to the invention for preventing or treating a disease, alleviating the symptoms of the disease or delaying the development or onset of the disease, wherein the disease
  • a disease For diabetes, diabetic complications, insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipidemia, obesity, syndrome X, atherosclerosis, cardiovascular disease, congestive heart failure, hypomagnesemia, low Natriuresis, renal failure, disorders associated with blood concentration, constipation or high blood pressure.
  • the diabetic complication of the invention is diabetic retinopathy, diabetic neuropathy or diabetic nephropathy.
  • the hyperlipidemia of the invention is hypertriglyceridemia.
  • the invention provides a glucopyranosyl derivative, a preparation method thereof and application thereof in medicine, and those skilled in the art can learn from the contents of the paper and appropriately improve the process parameters. It is to be noted that all such alternatives and modifications are obvious to those skilled in the art and are considered to be included within the scope of the invention.
  • the articles used herein are used to refer to the articles of one or more than one (ie, at least one).
  • a component refers to one or more components, that is, there may be more than one component contemplated for use or use in embodiments of the embodiments.
  • the compounds of the present invention may be optionally substituted with one or more substituents, such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • substituents such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • a class of compounds such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • substituents such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • a class of compounds A class of compounds.
  • substituents such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • a class of compounds such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • substituents such as the compounds of the above formula, or specific examples, subclasses, and inclusions of the present invention.
  • a class of compounds such as the
  • the term “optionally” whether or not preceded by the term “substituted” means that one or more hydrogen atoms in the given structure are unsubstituted or substituted with a particular substituent. Unless otherwise indicated, an optional substituent group can be substituted at each substitutable position of the group. When more than one position in the given formula can be substituted by one or more substituents selected from a particular group, the substituents may be substituted at the various positions, either identically or differently.
  • C 1-8 alkyl specifically refers to independently disclosed C 1 alkyl (methyl), C 2 alkyl (ethyl), C 3 alkyl, C 4 alkyl, C 5 alkyl, C 6 alkyl, C 7 alkyl and C 8 alkyl
  • C 3-8 cycloalkyl refers to independently disclosed C 3 cycloalkyl (cyclopropyl), C 4 cycloalkyl (cyclobutyl), C 5 -cycloalkyl (cyclopentyl), C 6 cycloalkyl (cyclohexyl), C 7 cycloalkyl (cycloheptyl) and C 8 cycloalkyl (cyclooctyl);
  • 3-8 atoms The heterocyclic ring refers to
  • linking substituents are described.
  • the Markush variable recited for that group is understood to be a linking group.
  • the definition of the Markush group for the variable is "alkyl” or "aryl”
  • the “alkyl” or “aryl” respectively represent the attached An alkylene group or an arylene group.
  • alkyl or "alkyl group” as used herein, denotes a saturated straight or branched monovalent hydrocarbon radical containing from 1 to 20 carbon atoms. Unless otherwise specified, an alkyl group contains from 1 to 20 carbon atoms; in some embodiments, the alkyl group contains from 1 to 12 carbon atoms; in some embodiments, the alkyl group contains from 1 to 10 a carbon atom; in other embodiments, the alkyl group contains from 1 to 9 carbon atoms; in other embodiments, the alkyl group contains from 1 to 8 carbon atoms; in other embodiments, the alkyl group contains 1 to 6 carbon atoms, i.e., C 1-6 alkyl; in other embodiments, the alkyl group contains 1 to 4 carbon atoms, i.e., C 1-4 alkyl; and other examples are alkyl groups.
  • the group contains 1-3 carbon atoms, i.e., C 1-3 alkyl; in other embodiments, the alkyl group contains a
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), n-propyl (n-Pr, -CH 2 CH 2 CH 3 ), isopropyl (i-Pr, -CH(CH 3 ) 2 ), n-butyl (n-Bu, -CH 2 CH 2 CH 2 CH 3 ), isobutyl (i-Bu, -CH 2 CH) (CH 3 ) 2 ), sec-butyl (s-Bu, -CH(CH 3 )CH 2 CH 3 ), tert-butyl (t-Bu, -C(CH 3 ) 3 ), n-pentyl (-CH) 2 CH 2 CH 2 CH 2 CH 3 ), 2-pentyl (-CH(CH 3 )CH 2 CH 2 CH 3 ), 3-pentyl (-CH(CH 2 CH 3 ) 2 ), 2-methyl -2-butyl (-C(CHCH
  • alkyl and its prefix “alk”, as used herein, encompass both straight-chain and branched saturated carbon chains.
  • alkylene means a saturated divalent hydrocarbon group derived by removing two hydrogen atoms from a saturated linear or branched hydrocarbon group. Unless otherwise specified, an alkylene group contains from 1 to 12 carbon atoms. In some embodiments, the alkylene group contains from 1 to 8 carbon atoms; in other embodiments, the alkylene group contains from 1 to 6 carbon atoms; in other embodiments, the alkylene group The group contains 1-4 carbon atoms, i.e., C 1-4 alkylene; in still other embodiments, the alkylene group contains 1-3 carbon atoms, i.e., C 1-3 alkylene; In an embodiment, the alkylene group contains 1-2 carbon atoms, i.e., C 1-2 alkylene.
  • Such examples include methylene (-CH 2 -), ethylene (including -CH 2 CH 2 - or -CH(CH 3 )-), isopropylidene (including -CH(CH 3 )CH 2 - Or -C(CH 3 ) 2 -), n-propylene (including -CH 2 CH 2 CH 2 -, -CH(CH 2 CH 3 )- or -CH 2 CH(CH 3 )-), Base (including -CH 2 (CH 2 ) 2 CH 2 -, -CH(CH 2 CH 2 CH 3 )-, -CH 2 CH(CH 2 CH 3 )-, -CH 2 CH(CH 3 )- Or -CH(CH 3 )CH(CH 3 )-), tert-butyl (including -CH(CH(CH 3 ) 2 )-, -CH 2 CH(CH 3 )CH 2 - or -CH 2 C ( CH 3 ) 2 -), pentylene (e.g., -CH 2
  • alkenyl denotes a straight or branched chain monovalent hydrocarbon radical containing from 2 to 12 carbon atoms, wherein at least one of the sites of unsaturation is a carbon-carbon sp 2 double bond, wherein the alkenyl group is capable Optionally, it is replaced by one or more substituents described herein, including the positioning of "cis” and “trans”, or the positioning of "E” and "Z”.
  • the alkenyl group contains 2-8 carbon atoms; in other embodiments, the alkenyl group contains 2-6 carbon atoms, ie, C 2-6 alkenyl; in yet other embodiments In the alkenyl group, the alkenyl group contains 2 to 4 carbon atoms, that is, a C 2-4 alkenyl group.
  • alkynyl means a straight or branched chain monovalent hydrocarbon radical containing from 2 to 12 carbon atoms, wherein at least one of the sites of unsaturation is a carbon-carbon sp triple bond.
  • an alkynyl group contains 2-8 carbon atoms; in other embodiments, an alkynyl group contains 2-6 carbon atoms, ie, a C 2-6 alkynyl group; in still other embodiments In the above, the alkynyl group contains 2 to 4 carbon atoms, that is, a C 2-4 alkynyl group.
  • alkynyl groups include, but are not limited to, ethynyl (-C ⁇ CH), propynyl (including 1-propynyl (-C ⁇ C-CH 3 ) and propargyl (-CH 2 C ⁇ CH) )), 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1-butynyl, 1-hexynyl, 1-heptynyl, 1-octynyl, and the like.
  • the alkynyl group can be independently and optionally substituted with one or more substituents described herein.
  • alkoxy refers to an alkyl group attached to the remainder of the molecule through an oxygen atom, i.e., alkyl-O-, wherein the alkyl group has the meaning as described herein.
  • the alkoxy group contains from 1 to 20 carbon atoms; in other embodiments, the alkoxy group contains from 1 to 10 carbon atoms; in still other embodiments, the alkoxy group The group contains from 1 to 8 carbon atoms; in still other embodiments, the alkoxy group contains from 1 to 6 carbon atoms, i.e., a C1-6 alkoxy group; in still other embodiments, the alkoxy group Containing from 1 to 4 carbon atoms, i.e., C 1-4 alkoxy; in still other embodiments, the alkoxy group contains from 1 to 3 carbon atoms, i.e., C 1-3 alkoxy; In the scheme, the alkoxy group contains 1-2 carbon atoms, that is, a C 1-2 alkoxy group.
  • alkoxy groups include, but are not limited to, methoxy (MeO, -OCH 3 ), ethoxy (EtO, -OCH 2 CH 3 ), 1-propoxy (n-PrO, n- Propyloxy, -OCH 2 CH 2 CH 3 ), 2-propoxy (i-PrO, i-propoxy, -OCH(CH 3 ) 2 ), 1-butoxy (n-BuO, n- Butoxy, -OCH 2 CH 2 CH 2 CH 3 ), 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH 2 CH(CH 3 ) 2 ), 2-butyl Oxygen (s-BuO, s-butoxy, -OCH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH) 3 ) 3 ), 1-pentyloxy (n-pentyloxy, -OCH 2 CH 2 CH 2 CH 2 CH
  • alkylthio means that the alkyl group is attached to the remainder of the molecule through a sulfur atom, i.e., alkyl-S-, wherein the alkyl group has the meaning as described herein.
  • the alkylthio group contains 1-6 carbon atoms, ie, a C1-6 alkylthio group; in other embodiments, the alkylthio group contains 1-4 carbon atoms, ie, C.
  • alkylthio group contains 1-3 carbon atoms, ie, a C 1-3 alkylthio group; in still other embodiments, the alkylthio group contains 1 - 2 carbon atoms, ie C 1-2 alkylthio groups.
  • alkylthio groups include, but are not limited to, methylthio, ethylthio, and the like.
  • the alkylthio group can be optionally substituted with one or more substituents described herein.
  • alkylamino or “alkylamino” includes “N-alkylamino” and "N,N-dialkylamino” wherein the amino groups are each independently substituted with one or two alkyl groups.
  • the alkylamino group is a lower alkylamino group having one or two C1-6 alkyl groups attached to the nitrogen atom.
  • the alkylamino group is a lower alkylamino group of C1-4 .
  • the alkylamino group is a lower alkylamino group of C1-3 .
  • the alkylamino group is a lower alkylamino group of C 1-2 .
  • Suitable alkylamino groups may be monoalkylamino or dialkylamino, examples of which include, but are not limited to, methylamino (also known as N-methylamino), ethylamino (also known as N-ethylamino), N, N-dimethylamino, N,N-diethylamino and the like. Wherein the alkylamino group can be independently unsubstituted or substituted with one or more substituents described herein.
  • haloalkyl refers to an alkyl group having one or more halo substituents.
  • the haloalkyl group contains 1-10 carbon atoms, and in other embodiments, the haloalkyl group contains 1-8 carbon atoms, and in still other embodiments, the haloalkyl group contains 1 -6 carbon atoms, i.e., C 1-6 haloalkyl; in still other embodiments, the haloalkyl group contains 1-4 carbon atoms, i.e., C 1-4 haloalkyl; and in some embodiments, haloalkyl The group contains 1-3 carbon atoms, i.e., C 1-3 haloalkyl; in some embodiments, the haloalkyl group contains 1-2 carbon atoms, i.e., C 1-2 haloalkyl.
  • haloalkyl groups include, but are not limited to, monofluoromethyl (-CH 2 F), difluoromethyl (-CHF 2 ), trifluoromethyl (-CF 3 ), fluoroethyl (-CHFCH 3 ,- CH 2 CH 2 F), difluoroethyl (-CF 2 CH 3 , -CFHCFH 2 , -CH 2 CHF 2 ), perfluoroethyl, fluoropropyl (-CHFCH 2 CH 3 , -CH 2 CHFCH 3 , -CH 2 CH 2 CH 2 F), difluoropropyl (-CF 2 CH 2 CH 3 , -CFHCFHCH 3 , -CH 2 CH 2 CHF 2 , -CH 2 CF 2 CH 3 , -CH 2 CHFCH 2 F) , trifluoropropyl, 1,1-dichloroethyl, 1,2-dichloropropyl, and the like.
  • haloalkoxy means that the alkoxy group is substituted by one or more halogen substituents.
  • a haloalkoxy group contains from 1 to 10 carbon atoms, and in still other embodiments, a haloalkoxy group contains from 1 to 8 carbon atoms; in still other embodiments, a haloalkoxy group The group contains from 1 to 6 carbon atoms, i.e., a C1-6 haloalkoxy group; in still other embodiments, the haloalkoxy group contains from 1 to 4 carbon atoms, i.e., a C1-4 haloalkoxy group; embodiment, haloalkoxy groups contain 1-3 carbon atoms, i.e.
  • haloalkoxy a halogenated alkyl group containing 1-2 carbon atoms, i.e. C 1-2 Haloalkoxy.
  • haloalkoxy groups include, but are not limited to, trifluoromethoxy, difluoromethoxy, and the like.
  • the haloalkoxy group can be optionally substituted with one or more substituents described herein.
  • hydroxyalkyl refers to an alkyl group having one or more hydroxy substituents, wherein the alkyl group has the meaning as described herein.
  • the hydroxyalkyl group contains 1-6 carbon atoms, ie, C 1-6 hydroxyalkyl; in other embodiments, the hydroxyalkyl group contains 1-4 carbon atoms, ie, C 1-4 hydroxyalkyl; in still other embodiments, the hydroxyalkyl group contains 1-3 carbon atoms, ie, C 1-3 hydroxyalkyl; in still other embodiments, the hydroxyalkyl group contains 1 - 2 carbon atoms, ie C 1-2 hydroxyalkyl.
  • hydroxyalkyl groups include, but are not limited to, hydroxymethyl, 2-hydroxyethyl (-CH 2 CH 2 OH), 1-hydroxyethyl (-CHOHCH 3 ), 1,2-dihydroxyethyl (- CHOHCH 2 OH), 2,3-dihydroxypropyl (-CH 2 CHOHCH 2 OH), 1-hydroxypropyl (-CH 2 CH 2 CH 2 OH), 2-hydroxypropyl, 3-hydroxypropyl, Hydroxybutyl, and so on.
  • the hydroxyalkyl group can be optionally substituted with one or more substituents described herein.
  • cycloalkyl refers to a monocyclic, bicyclic or tricyclic ring system having one or more attachment points attached to the remainder of the molecule, saturated, containing from 3 to 12 ring carbon atoms.
  • a cycloalkyl group is a ring system containing from 3 to 10 ring carbon atoms; in other embodiments, a cycloalkyl group is a ring system containing from 3 to 8 ring carbon atoms, ie, C 3 -8 cycloalkyl; in other embodiments, the cycloalkyl is a ring system containing from 5 to 8 ring carbon atoms; in other embodiments, the cycloalkyl group is a ring containing from 3 to 6 ring carbon atoms The system, i.e., C 3-6 cycloalkyl; in other embodiments, the cycloalkyl is a ring system containing from 5 to 6 ring carbon atoms.
  • cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • the cycloalkyl group can be independently unsubstituted or substituted with one or more substituents described herein.
  • Carbocyclyl may be used alone or as a large part of “carbocyclylalkyl” or “carbocyclylalkoxy”, meaning saturated or containing one or more units of unsaturation, containing from 3 to 14 A non-aromatic carbocyclic ring system of ring carbon atoms and free of any aromatic rings.
  • carbocyclic “carbocyclic” or “carbocyclic” are used interchangeably herein.
  • the number of ring carbon atoms of the carbocyclic ring is from 3 to 12; in other embodiments, the number of ring carbon atoms of the carbocyclic ring is from 3 to 10; in other embodiments, the carbocyclic ring The number of ring carbon atoms is 3-8, that is, C 3-8 carbocyclic ring; in other embodiments, the number of ring carbon atoms of the carbocyclic ring is 3-6, that is, C 3-6 carbon Ring (base); In other embodiments, the number of ring carbon atoms of the carbocyclic ring is 5-6; in other embodiments, the number of ring carbon atoms of the carbocyclic ring is 5-8.
  • the number of ring carbon atoms of the carbocyclic ring is from 6 to 8.
  • Carbocyclyl includes monocyclic, bicyclic or polycyclic fused, spiro or bridged carbocyclic ring systems, and also includes wherein the carbocyclic ring may be bonded to one or more non-aromatic carbocyclic rings or one or more aromatic rings Or a combination thereof, a fused polycyclic ring system in which the attached atomic groups or points are on the carbocyclic ring.
  • Bicyclic carbocyclyl groups include bridged bicyclic carbocyclyl, fused bicyclic carbocyclyl and spiro bicyclic carbocyclyl, and "fused" bicyclic ring systems contain two rings sharing two adjacent ring atoms.
  • a bridged bicyclic group includes two rings that share 2, 3 or 4 adjacent ring atoms.
  • the spiro ring system shares one ring atom.
  • Suitable carbocyclic groups include, but are not limited to, cycloalkyl, cycloalkenyl and cycloalkynyl.
  • carbocyclic groups further include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-enyl, 1-cyclopentyl-2-alkenyl, 1- Cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-1-alkenyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexadienyl, cycloheptane Base, cyclooctyl, cyclodecyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
  • Bridged carbocyclyl groups include, but are not limited to, bicyclo [2.2.2] octyl, bicyclo [2.2.1] heptyl, bicyclo [3.3.1] fluorenyl, bicyclo [3.2.3] fluorene Base, and so on.
  • the carbocyclyl group can be optionally substituted with one or more substituents described herein.
  • heterocyclyl may be used alone or as a large part of “heterocyclylalkyl” or “heterocyclylalkoxy”, meaning saturated or partially unsaturated, containing from 3 to 12 ring atoms, An aromatic monocyclic, bicyclic or tricyclic ring system in which 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, and the ring system There are one or more connection points connected to the rest of the molecule.
  • heterocyclyl includes monocyclic, bicyclic or polycyclic fused, spiro or bridged heterocyclic ring systems.
  • Bicyclic heterocyclic groups include bridged bicyclic heterocyclic groups, fused bicyclic heterocyclic groups, and spirobicyclic heterocyclic groups.
  • heterocyclyl and “heterocycle” are used interchangeably herein.
  • the sulfur atom of the ring can be optionally oxidized to an S-oxide.
  • the nitrogen atom of the ring can be optionally oxidized to an N-oxygen compound.
  • a heterocyclic group is a ring system of from 3 to 8 ring atoms; in other embodiments, a heterocyclic group is a ring system of from 3 to 6 ring atoms; in other embodiments, The heterocyclic group is a ring system of 5 to 7 ring atoms; in other embodiments, the heterocyclic group is a ring system of 5 to 10 ring atoms; in other embodiments, the heterocyclic group is 5- a ring system of 8 ring atoms; in other embodiments, the heterocyclic group is a ring system of 6-8 ring atoms; in other embodiments, the heterocyclic group is composed of 5-6 ring atoms.
  • the heterocyclic group is a ring system of 4 ring atoms; in other embodiments, the heterocyclic group is a ring system of 5 ring atoms; in other embodiments, The heterocyclic group is a ring system composed of 6 ring atoms; in other embodiments, the heterocyclic group is a ring system composed of 7 ring atoms; in other embodiments, the heterocyclic group is composed of 8 ring atoms. Ring system.
  • heterocyclic groups include, but are not limited to, oxiranyl, aziridine, azetidinyl, oxetanyl, thioheterobutyl, pyrrolidinyl, 2-pyrroline, 3-pyrroline, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, oxazolidine, thiazolidine, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothienyl, 1 , 3-dioxocyclopentyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, Polinyl, thiomorpholinyl, piperazinyl, dioxoalkyl, dithiaal
  • Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, a sulfolane group and a 1,1-dioxothiomorpholinyl group.
  • the bridged heterocyclyl group includes, but is not limited to, 2-oxabicyclo[2.2.2]octyl, 1-azabicyclo[2.2.2]octyl, 3-azabicyclo[3.2. 1] ⁇ , and so on.
  • the heterocyclyl group can be optionally substituted with one or more substituents described herein.
  • m atoms typically describes the number of ring atoms in the molecule in which the number of ring atoms is m.
  • piperidinyl is a heterocyclic group consisting of 6 ring atoms
  • 1,2,3,4-tetrahydronaphthyl is a carbocyclic group consisting of 10 ring atoms.
  • aryl may be used alone or as a large part of “arylalkyl” or “arylalkoxy”, meaning 6-14 ring atoms, or 6-12 ring atoms, or 6-10. Monocyclic, bicyclic, and tricyclic aromatic carbocyclic ring systems of ring atoms wherein each ring contains from 3 to 7 ring atoms and one or more attachment points are attached to the remainder of the molecule.
  • aryl may be used interchangeably with the terms "aromatic ring” or “aromatic ring”, as aryl may include phenyl, naphthyl and anthracenyl.
  • the aryl group may be independently unsubstituted or substituted with one or more substituents described herein.
  • heteroaryl may be used alone or as a large part of “heteroarylalkyl” or “heteroarylalkoxy”, meaning a monocyclic, bicyclic and tricyclic aromaticity of a 5-16 ring atom. a system wherein at least one ring comprises one or more heteroatoms, wherein each ring comprises 5-7 ring atoms, wherein at least one ring system is aromatic, and at the same time, the heteroaryl has one or more attachments The point is connected to the rest of the molecule. Unless otherwise stated, the heteroaryl group can be attached to the remainder of the molecule (e.g., the host structure in the formula) by any reasonable site (which can be C in CH, or N in NH).
  • heteroaryl can be used interchangeably with the terms “heteroaryl ring” or “heteroaromatic compound”.
  • a heteroaryl is a heteroaryl group of 5 to 14 atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • the heteroaryl is a heteroaryl group comprising from 5 to 12 atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N; in other embodiments, A heteroaryl group is a heteroaryl group comprising from 5 to 10 atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N; in other embodiments, the heteroaryl group comprises 1, 2, 3 or 4 heteroaryl groups of 5-8 atoms independently selected from the heteroatoms of O, S and N; in other embodiments, the heteroaryl group comprises 1, 2, 3 or 4 independent a heteroaryl group consisting of 5-7 atoms of a hetero atom selected from O, S and N; in other embodiments, the heteroaryl group comprises 1, 2, 3 or 4 independently selected from O, S and N a heteroaryl group of 5-6 atoms of a heteroatom; in other embodiments, the heteroaryl group is 5 atoms comprising 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • the heteroaryl group is
  • the heteroaryl group includes the following monocyclic groups, but is not limited to these monocyclic groups: 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazole , 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl , 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (eg 3-pyridazine) Base, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (eg 5H-tetrazolyl, 2H-tetrazolyl), triazolyl (eg 2-triazolyl, 2-tri
  • heteroatom means O, S, N, P and Si, including the form of any oxidation state of S, N and P; the form of primary, secondary, tertiary and quaternary ammonium salts; or the nitrogen atom of a heterocyclic ring.
  • a form in which hydrogen is substituted for example, N (like N in 3,4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR (like in N-substituted pyrrolidinyl) NR, R are the substituents described in the present invention).
  • halogen means F, Cl, Br, I.
  • nitro refers to -NO 2 .
  • mercapto refers to -SH.
  • hydroxy refers to -OH.
  • amino refers to -NH 2 .
  • cyano refers to -CN.
  • D refers to deuterated, ie 2 H.
  • drawing a bond to form a ring system with a substituent attached to the center of the ring means that the substituent can be substituted at any substitutable position on the ring system.
  • the formula a represents that any position on the A ring that may be substituted may be optionally substituted by n R; when the A ring is a bicyclic structure, R may be substituted at any substitutable position on any of the rings;
  • b represents that the substituent R may be substituted at any position on the phenyl ring which may be substituted, as shown by the formulae b-1 to b-3:
  • protecting group refers to a substituent group used to block or protect a particular functionality when other functional groups in the compound react.
  • protecting group of an amino group refers to a substituent attached to an amino group to block or protect the functionality of an amino group in a compound.
  • Suitable amino protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl. (BOC, Boc), benzyloxycarbonyl (CBZ, Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc).
  • a "hydroxy protecting group” refers to a substituent of a hydroxy group used to block or protect the functionality of a hydroxy group.
  • Suitable protecting groups include, but are not limited to, acetyl, benzoyl, benzyl, p-methoxy A benzyl group, a silane group and the like.
  • Carboxy protecting group means a substituent of a carboxy group used to block or protect the functionality of a carboxy group.
  • Typical carboxy protecting groups include -CH 2 CH 2 SO 2 Ph, cyanoethyl, 2-(trimethylsilane Ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl)ethyl, 2-(p-nitrophenylsulfonyl)ethyl, 2-(diphenyl Phosphine) ethyl, nitroethyl, and the like.
  • a general description of protecting groups can be found in the literature: T W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991; and PJ Kocienski, Protecting Groups, Thieme, Stuttgart, 2005.
  • LG refers to an atom or functional group that is liberated from a larger molecule in a chemical reaction and is a term used in the nucleophilic substitution reaction and elimination reaction.
  • a reactant attacked by a nucleophile is referred to as a substrate, and an atom or group of atoms that are cleaved from a substrate molecule with a pair of electrons is referred to as a leaving group.
  • Common leaving groups are, for example but not limited to, halogen atoms, ester groups, sulfonate groups, nitro groups, azide groups or hydroxyl groups, and the like.
  • pharmaceutically acceptable means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal treated therewith.
  • pharmaceutically acceptable refers to the use in animals, particularly humans, as approved by a federal or national government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia.
  • carrier includes any solvent, dispersion medium, coating, surfactant, antioxidant, preservative (eg antibacterial, antifungal), isotonic, salt, drug stabilizer, binder, Forming agents, dispersing agents, lubricants, sweeteners, flavoring agents, coloring agents, or combinations thereof, are known to those skilled in the art (e.g., Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289-1329). The use thereof in a therapeutic or pharmaceutical composition is encompassed except where any conventional carrier is incompatible with the active ingredient.
  • 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 Excipients such as carriers, excipients, diluents, binders, fillers, and the like, as well as anti-diabetic agents, anti-hyperglycemic agents, anti-obesity agents, antihypertensive agents, anti-platelet agents, anti-atherosclerotic agents or An additional therapeutic agent such as a lipid lowering agent.
  • the purpose of the pharmaceutical composition is to facilitate the administration of the compound to the organism.
  • prodrug denotes a compound which is converted in vivo to a compound of formula (I). Such transformation is affected by the hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue.
  • the prodrug-like compound of the present invention may be an ester.
  • the ester may be used as a prodrug such as a phenyl ester, an aliphatic (C 1-24 ) ester, an acyloxymethyl ester, or a carbonate. , carbamates and amino acid esters.
  • a compound of the invention comprises a hydroxyl group, i.e., it can be acylated to give a compound in the form of a prodrug.
  • Other prodrug forms include phosphates, such as those obtained by phosphorylation of a hydroxy group on the parent.
  • metabolite refers to a product obtained by metabolism of a particular compound or salt thereof in vivo. Metabolites of a compound can be identified by techniques well known in the art, and the activity can be characterized by experimental methods as described herein. Such a product may be obtained by administering a compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage and the like. Accordingly, the invention includes metabolites of a compound, including metabolites produced by intimate contact of a compound of the invention with a mammal for a period of time.
  • pharmaceutically acceptable salt refers to organic and inorganic salts of the compounds of the present invention.
  • Pharmaceutically acceptable salts are well known in the art, as described in the literature: Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmacol Sci, 1997, 66, 1-19.
  • Examples of pharmaceutically acceptable non-limiting salts include inorganic acid salts formed by reaction with amino groups, hydrochlorides, hydrobromides, phosphates, metaphosphates, sulfates, nitrates, perchlorates, And organic acid salts such as methanesulfonate, ethanesulfonate, acetate, trifluoroacetate, glycolate, isethionate, oxalate, maleate, tartrate, lemon Acid salts, succinates, malonates, besylate, p-toluenesulfonate, malate, fumarate, lactate, lactobionate, or other methods described in the literature These salts are obtained by ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, besylate, benzoate, disulfate, borate, butyrate, camphoric acid Salt, camphorsulfonate, cyclopentylpropionate, digluconate, lauryl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphoric acid Salt, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, laurate, lauryl sulfate, malonate , 2-naphthalene sulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectate, persulphate, 3-phenylpropionate, picrate, pentane Acid salts, propionates
  • Salts obtained by appropriate bases include the alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • the present invention also contemplates quaternary ammonium salts formed from any of the compounds comprising a group of N. Water soluble or oil soluble or dispersed products can be obtained by quaternization.
  • Alkali metal or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Pharmaceutically acceptable salts further comprise suitable amine cation nontoxic ammonium, quaternary ammonium, and the counterion, such as halide, hydroxide, carboxylate, sulfated, phosphorylated compounds, nitrate compounds, C 1 -8 sulfonate and aromatic sulfonate.
  • suitable amine cation nontoxic ammonium, quaternary ammonium, and the counterion such as halide, hydroxide, carboxylate, sulfated, phosphorylated compounds, nitrate compounds, C 1 -8 sulfonate and aromatic sulfonate.
  • solvate refers to an association of one or more solvent molecules with a compound of the invention.
  • Solvent-forming solvents include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, aminoethanol.
  • hydrate means that the solvent molecule is an association formed by water.
  • nitrogen oxide means that when a compound contains several amine functional groups, one or more than one nitrogen atom can be oxidized to form an N-oxide.
  • N-oxides are N-oxides of tertiary amines or N-oxides of nitrogen-containing heterocyclic nitrogen atoms.
  • the corresponding amine can be treated with an oxidizing agent such as hydrogen peroxide or a peracid such as peroxycarboxylic acid to form an N-oxide (see Advanced Organic Chemistry, Wiley Interscience, 4th edition, Jerry March, pages).
  • the N-oxide can be prepared by the method of LWDeady (Syn. Comm. 1977, 7, 509-514), wherein the amine compound and 3-chloroperoxybenzoic acid (m) are, for example, in an inert solvent such as dichloromethane. -CPBA) reaction.
  • any asymmetric atom (e.g., carbon, etc.) of the compounds of the invention may exist in racemic or enantiomerically enriched form, such as (R)-, (S)-, (R, S)- or (S, The S)-configuration form exists.
  • each asymmetric atom has at least 50% enantiomeric excess in the (R)- or (S)-configuration, 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.
  • the substituent on the atom having an unsaturated double bond may exist in the form -(Z)- or -(E)-.
  • the compounds of the invention may exist in the form of one of the possible isomers, rotamers, atropisomers, tautomers, or mixtures thereof.
  • it is a substantially pure geometric (cis or trans) isomer, a diastereomer, an optical isomer (enantiomer), a racemate or a mixture thereof.
  • the resulting mixture of any isomers can be separated into pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example by chromatography and/or fractionation, depending on the physicochemical differences of the components.
  • the crystallization is carried out to carry out the separation.
  • racemate of any of the resulting end products or intermediates can be resolved into the optical antipodes by methods known to those skilled in the art by known methods, for example, by obtaining the diastereomeric salts thereof. Separation.
  • the racemic product can also be separated by chiral chromatography, such as high pressure liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high pressure liquid chromatography
  • enantiomers can be prepared by asymmetric synthesis (e.g., Jacques, et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Principles of Asymmetric Synthesis (2 nd Ed. Robert E.
  • the invention also includes isotopically-labeled compounds of the invention which are identical to those described herein except that the one or more atoms are replaced by an atom having an atomic mass or mass number different from the natural common atomic mass or mass number.
  • Exemplary isotopes which may also be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 16 O, 17 O, 31 P, 32 P, 36 S, 18 F and 37 Cl.
  • Isotopically labeled compounds of the invention such as radioisotopes, such as 3 H and 14 C
  • Isotope, i.e., 3 H, and carbon-14, i.e., 14 C is particularly preferred because of ease of preparation and detection.
  • substitution with heavy isotopes such as deuterium, i.e., 2 H may provide some therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements. Therefore, it may be preferable in some cases.
  • stereochemical definitions and conventions used in the present invention are generally in accordance with SP Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., "Stereochemistry Definitions and conventions as documented by The Organic Compounds, John Wiley & Sons, Inc., New York, 1994.
  • the compounds of the invention may contain asymmetric centers or chiral centers and therefore exist in different stereoisomeric forms. It is contemplated that all stereoisomeric forms of the compounds of the invention include, but are not limited to, diastereomers, enantiomers and atropisomers, and mixtures thereof such as racemic mixtures, It is also included in the scope of the invention.
  • optically active forms i.e., they have the ability to rotate a plane of plane polarized light.
  • the prefixes D and L or R and S are used to indicate the absolute configuration of the molecule with respect to the chiral center (or multiple chiral centers) in the molecule.
  • the prefixes d and l or (+) and (–) are symbols used to specify the rotation of the plane polarized light caused by the compound, where (–) or l indicates that the compound is left-handed.
  • Compounds prefixed with (+) or d are dextrorotatory. For a given chemical structure, these stereoisomers are identical except that these stereoisomers are mirror images of one another.
  • stereoisomers may also be referred to as enantiomers, and mixtures of such isomers are often referred to as mixtures of enantiomers.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur when there is no stereoselectivity or stereospecificity in a chemical reaction or process. Spin body.
  • the compounds of the invention may exist as one of the possible isomers or as a mixture thereof, for example as a pure optical isomer, or as a mixture of isomers, such as as racemic and non-
  • the corresponding isomer mixture depends on the number of asymmetric carbon atoms.
  • the optically active (R)- or (S)-isomers can be prepared using chiral synthons or chiral preparations or resolved using conventional techniques. If the compound contains a double bond, the substituent may be in the E or Z configuration; if the compound contains a disubstituted cycloalkyl group, the substituent of the cycloalkyl group may be cis or trans (cis- or trans-) structure.
  • the compounds of the invention may contain asymmetric centers or chiral centers and therefore exist in different stereoisomeric forms. It is contemplated that all stereoisomeric forms of the compounds of the invention include, but are not limited to, diastereomers, enantiomers and atropisomers and geometric (or conformational) isomers. And mixtures thereof, such as racemic mixtures, are within the scope of the invention.
  • structures depicted in the present invention are also meant to include all isomers (e.g., enantiomers, diastereomeric atropisomers, and geometric (or conformation)) forms of this structure; for example, each The R and S configurations of the asymmetric center, the (Z) and (E) double bond isomers, and the (Z) and (E) conformers.
  • isomers e.g., enantiomers, diastereomeric atropisomers, and geometric (or conformation)
  • tautomer or "tautomeric form” refers to structural isomers having different energies that are interconvertible by a low energy barrier. If tautomerism is possible (as in solution), the chemical equilibrium of the tautomers can be achieved.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by recombination of some bonding electrons.
  • keto-enol tautomerization is the interconversion of a pentane-2,4-dione and a 4-hydroxypent-3-en-2-one tautomer.
  • Another example of tautomerization is phenol-keto tautomerization.
  • a specific example of phenol-keto tautomerization is the interconversion of pyridin-4-ol and pyridine-4(1H)-one tautomers. All tautomeric forms of the compounds of the invention are within the scope of the invention unless otherwise indicated.
  • dimer refers to a substance of the same or the same species that appears in a double form and may have properties or functions that are not found in a single state. Common examples include dicyclopentadiene, dimerized cuprous chloride, sucrose, and the like.
  • trimer refers to three identical or identical species that polymerize into a new molecule, a new molecule that is a trimer and a low molecular weight polymer.
  • subject refers to an animal. Typically the animal is a mammal. Subjects also refer to primates (eg, humans), cattle, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds, and the like. In certain embodiments, the subject is a primate. In still other embodiments, the subject is a human.
  • subject and patient refer to animals (eg, birds or mammals such as chickens, quails, or turkeys), particularly “mammals” including non-primates (eg, cows, pigs). , horses, sheep, rabbits, guinea pigs, rats, cats, dogs, and mice) and primates (eg, monkeys, chimpanzees, and humans), and more particularly humans.
  • the subject is a non-human animal, such as a domestic animal (eg, a horse, cow, pig, or sheep) or a pet (eg, a dog, cat, guinea pig, or rabbit).
  • "patient” refers to a human.
  • X Syndrome also known as the disease, disease of the metabolic syndrome, is described in detail in Johannsson et al., J. Clin. Endocrinol. Metab., 1997, 82, 727-734.
  • intestinal improvement means an increase in beneficial bacteria such as Bifidobacterium, Lactobacillus or the like, an increase in organic acids in the intestine, and a reduction in spoilage products in the intestine.
  • cancer and “cancerous” refer to or describe a physiological condition in a patient that is typically characterized by uncontrolled cell growth.
  • a “tumor” contains one or more cancer cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia, or lymphoid malignancies.
  • squamous cell carcinoma such as squamous cell carcinoma
  • lung cancer including small cell lung cancer, non-small cell lung cancer (NSCLC), lung adenocarcinoma, and lung squamous cell carcinoma
  • peritoneal cancer Hepatocellular cancer
  • gastric or stomach cancer including gastrointestinal cancer
  • pancreatic cancer malignant glioma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma (hepatoma) ), breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial cancer or uterine cancer, salivary gland cancer, kidney or renal cancer, prostate cancer, vulvar cancer, thyroid cancer, liver cancer ( Hepatic carcinoma), anal cancer, penile cancer, and head and neck cancer.
  • the structural formulae of the compounds described herein include enriched isotopes of one or more different atoms.
  • any disease or condition as used in the present invention refers to ameliorating a disease or condition (ie, slowing or preventing or alleviating the progression of a disease or at least one of its clinical symptoms).
  • “treating” refers to alleviating or ameliorating at least one physical parameter, including physical parameters that may not be perceived by the patient.
  • “treating” refers to modulating a disease or condition from the body (eg, stabilizing a detectable symptom) or physiologically (eg, stabilizing the body's parameters) or both.
  • “treating” refers to preventing or delaying the onset, onset, or exacerbation of a disease or condition.
  • the present invention provides a class of compounds having excellent SGLTs inhibitory activity, especially those having excellent SGLT1 inhibitory activity, for preparing a drug for improving intestinal environment; or for preparing for treating diabetes, diabetic complications, insulin resistance, and high Blood sugar, hyperinsulinemia, hyperlipidemia, obesity, X syndrome, atherosclerosis, cardiovascular disease, congestive heart failure, hypomagnesemia, hyponatremia, renal failure, and blood concentration Disorders, constipation or drugs for high blood pressure.
  • the invention also provides methods of preparing these compounds, pharmaceutical compositions comprising these compounds, and methods of using the compounds and compositions to prepare a medicament for treating the above-described diseases in mammals, particularly humans.
  • the compounds of the present invention Compared with the existing analogous compounds, the compounds of the present invention not only have good pharmacological activities, but also have excellent in vivo metabolic kinetic properties and in vivo pharmacodynamic properties. At the same time, the preparation method is simple and easy, the process method is stable, and is suitable for industrial production. Therefore, the compound provided by the present invention has more excellent drug-yielding properties than the currently-prepared compound.
  • the invention relates to a compound which is a stereoisomer, geometric isomer, tautomer, oxynitride, solvate of a compound of formula (I) or a compound of formula (I). , metabolites, pharmaceutically acceptable salts, dimers, trimers or prodrugs thereof,
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , L, n and t have the definitions as defined in the present invention, provided that R 5 and R 8 are both H and R 9 is When t is not 0.
  • q 1, 2, 3, 4, 5 or 6;
  • p 0, 1, 2, 3, 4, 5 or 6;
  • R 1 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 Alkoxy, C 1-6 hydroxyalkyl, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 1-6 alkylamino, C 1-6 alkylthio, C 3-6 cycloalkyl or a C 3-6 cycloalkyl-C 1-4 alkylene group, wherein the C 1-6 alkyl group, C 2-6 alkenyl group, C 2-6 alkynyl group, C 1-6 alkoxy group, C 1-6 hydroxyalkyl, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 1-6 alkylamino, C 1-6 alkylthio, C 3-6 cycloalkyl and C 3-6 ring
  • the alkyl-C 1-4 alkylene groups are each
  • R 2 and R 3 are each independently H, D, CN, OH, NH 2 , -SH, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy a heterocyclic group consisting of a C 1-6 alkylthio group, a C 1-6 alkylamino group, a C 3-6 cycloalkyl group or a 3-6 atom, wherein the C 1-6 alkyl group, C 2 ⁇ 6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, C 1-6 alkylthio, C 1-6 alkylamino, C 3-6 cycloalkyl and 3-6 atomic hetero
  • R 2 , R 3 together with the carbon atom to which they are attached form a carbonyl group
  • R 2 , R 3 together with the carbon atom to which they are attached form a C 3-6 carbocyclic ring or a heterocyclic ring composed of 3 to 6 atoms, wherein the C 3-6 carbocyclic ring and 3 to 6 atoms are composed of
  • R 4 is H, D, -OR 4a or -SR 4b ;
  • R 4a and R 4b are each independently H, D, C 1-6 alkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyl-C 1-4 alkylene, 5-6 atoms Heterocyclic group, (heterocyclic group of 5-6 atoms)-C 1-4 alkylene group, C 6-10 aryl group, C 6-10 aryl-C 1-4 alkylene group, 5- a 6-atom heteroaryl group or a (5-6 atom heteroaryl group)-C 1-4 alkylene group, wherein the C 1-6 alkyl group, C 3-6 cycloalkyl group, C 3-6 cycloalkyl-C 1-4 alkylene group, heterocyclic group composed of 5-6 atoms, (heterocyclic group of 5-6 atoms)-C 1-4 alkylene group, C 6- 10 aryl, C 6-10 aryl-C 1-4 alkylene, heteroaryl group of 5-6 atoms and (heteroaryl group of 5-6
  • R 10 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , C 1-6 alkyl or C 1-6 alkoxy;
  • R 6 and R 7 are each independently H, D, F, Cl, Br, I, CN, NO 2 , OH, NH 2 , C 1-6 alkyl, C 1-6 alkoxy, C 1-6 Alkylamino, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 3-8 cycloalkyl, heterocyclic group consisting of 3-8 atoms, C 6-10 aryl or 5-8 atoms Heteroaryl, wherein the C 1-6 alkyl group, C 1-6 alkoxy group, C 1-6 alkylamino group, C 1-6 haloalkyl group, C 1-6 haloalkoxy group, C 3-8 A cycloalkyl group, a heterocyclic group of 3-8 atoms, a C 6-10 aryl group, and a heteroaryl group of 5-8 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents.
  • R 6 , R 7 together with the carbon atom to which they are attached form a C 3-8 carbocyclic ring, a heterocyclic ring of 3-8 atoms, a C 6-10 aromatic ring or a heteroaryl ring of 5-8 atoms,
  • R 5 , R 6 together with the atoms to which they are attached, or R 5 , R 7 and the atoms to which they are attached, form a heterocyclic ring of 3-8 atoms or a heteroaryl composed of 5-8 atoms.
  • heterocyclic ring composed of 3-8 atoms and the heteroaryl ring composed of 5-8 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents, the substitution
  • R 8 and R 9 are each independently H, D, R e OC 1-4 alkylene, R d R c NC 1-4 alkylene, C 1-8 alkyl, C 1-6 haloalkyl, C 3 -8 cycloalkyl, C 3-8 cycloalkyl-C 1-4 alkylene, heterocyclic group consisting of 3-8 atoms, (heterocyclic group consisting of 3-8 atoms) -C 1-4 Alkylene group, C 6-10 aryl group, C 6-10 aryl-C 1-4 alkylene group, heteroaryl group composed of 5-8 atoms or (heteroaryl group of 5-8 atoms)- a C 1-4 alkylene group, wherein the C 1-8 alkyl group, the C 1-6 haloalkyl group, the C 3-8 cycloalkyl group, the C 3-8 cycloalkyl-C 1-4 alkylene group, 3 Heterocyclic group consisting of -8 atoms, (hetero
  • R 8 , R 9 together with the nitrogen atom to which they are attached form a heterocyclic ring of 3-8 atoms or a heteroaromatic ring of 5-8 atoms, wherein the heterocyclic ring consisting of 3-8 atoms
  • the heteroaryl rings of 5-8 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents independently selected from D, F, Cl, Br, I, CN.
  • R c , R d , R e and R f are each independently H, D, C 1-6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-4 alkylene group, heterocyclic group composed of 3-8 atoms, (heterocyclic group of 3-8 atoms)-C 1-4 alkylene group, C 6-10 aryl group, C 6-10 An aryl-C 1-4 alkylene group, a heteroaryl group of 5-8 atoms or a (heteroaryl group of 5-8 atoms)-C 1-4 alkylene group, wherein the C 1- 6 alkyl, C 1-6 haloalkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-4 alkylene, heterocyclic group consisting of 3-8 atoms, (3-8 Heterocyclic group consisting of a hetero atom)-C 1-4 alkylene group, C 6
  • R c , R d together with the nitrogen atom to which they are attached form a heterocyclic ring of 3-8 atoms or a heteroaromatic ring of 5-8 atoms, wherein the heterocyclic ring consisting of 3-8 atoms
  • the heteroaryl rings of 5-8 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents independently selected from D, F, Cl, Br, I, CN.
  • n 0, 1, 2 or 3;
  • t is 0, 1, 2, 3, 4, 5 or 6; with the proviso that when R 5 and R 8 are both H and R 9 is When t is not 0.
  • the present invention relates to a compound of the formula (Ia) or a stereoisomer, geometric isomer, tautomer, oxynitride, solvate, metabolite, pharmacy thereof.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , L, n and t have the definitions as defined in the present invention.
  • the present invention relates to a compound of the formula (II) or a stereoisomer, geometric isomer, tautomer, oxynitride, solvate, metabolite, pharmacy thereof.
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , n and t have the definitions as defined in the present invention.
  • R 1 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , C 1-4 alkyl, C 2-4 alkenyl, C 2-4 Alkynyl, C 1-4 alkoxy, C 1-4 hydroxyalkyl, C 1-4 haloalkyl, C 1-4 haloalkoxy, C 1-4 alkylamino, C 1-4 alkylthio, C a 3-6 cycloalkyl group or a C 3-6 cycloalkyl-C 1-2 alkylene group, wherein the C 1-4 alkyl group, the C 2-4 alkenyl group, the C 2-4 alkynyl group, C 1 -4 alkoxy, C 1-4 hydroxyalkyl, C 1-4 haloalkyl, C 1-4 haloalkoxy, C 1-4 alkylamino, C 1-4 alkylthio, C 3-6 naphthenic And the C 3-6 cycloalkyl-C 1-2
  • R 1 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , methyl, ethyl, n-propyl, isopropyl, propenyl, propyl Alkynyl, methoxy, ethoxy, hydroxymethyl, trifluoromethyl, trifluoroethyl, monofluoromethyl, trifluoromethoxy, difluoromethoxy, methylamino, cyclopropyl, ring Butyl, cyclopentyl, cyclohexyl or cyclopropyl-methylene, wherein the methyl, ethyl, n-propyl, isopropyl, propenyl, propynyl, methoxy, ethoxy groups , hydroxymethyl, trifluoroethyl, monofluoromethyl, difluoromethoxy, methylamino, cyclopropyl, cyclobuty
  • R 2 and R 3 are each independently H, D, CN, OH, NH 2 , -SH, methyl, ethyl, n-propyl, isopropyl, vinyl, propynyl a methoxy group, an ethoxy group, a methylthio group, a methylamino group, a cyclopropyl group, a cyclobutyl group, a cyclopentyl group, a cyclohexyl group or a heterocyclic group consisting of 5-6 atoms, wherein the methyl group and the ethyl group Base, n-propyl, isopropyl, vinyl, propynyl, methoxy, ethoxy, methylthio, methylamino, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and 5-6
  • the heterocyclic groups consisting of one atom are each independently unsubstit
  • R 2 , R 3 and the carbon atom to which they are attached form a carbonyl group.
  • R 2 , R 3 and the carbon atom to which they are attached form a C 3-6 carbocyclic ring or a heterocyclic ring of 5-6 atoms, wherein the C 3-6 carbocyclic ring and 5
  • R 4 is H, D, -OR 4a or -SR 4b ;
  • R 4a and R 4b are each independently H, D, C 1-4 alkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyl-C 1-2 alkylene, 5-6 atoms Heterocyclic group, (heterocyclic group of 5-6 atoms)-C 1-2 alkylene group, C 6-10 aryl group, C 6-10 aryl-C 1-2 alkylene group, 5- a 6-atom heteroaryl group or a (5-6 atom heteroaryl group)-C 1-2 alkylene group, wherein the C 1-4 alkyl group, C 3-6 cycloalkyl group, C 3-6 cycloalkyl-C 1-2 alkylene group, heterocyclic group composed of 5-6 atoms, (heterocyclic group of 5-6 atoms)-C 1-2 alkylene group, C 6- 10 aryl, C 6-10 aryl-C 1-2 alkylene, heteroaryl group of 5-6 atoms and (heteroaryl group of 5-6
  • R 4 is H, D, -OR 4a or -SR 4b ;
  • R 4a and R 4b are each independently H, D, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 3-6 cycloalkyl- C 1-2 alkylene, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, (heterocyclic ring of 5-6 atoms) -C 1-2 alkylene, phenyl, phenyl-C 1-2 alkylene, furyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, cacao Azolyl, isoxazolyl, oxadiazolyl, 1,3,5-triazinyl,
  • R 10 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , C 1-4 alkyl or C 1-4 alkoxy.
  • R 10 is H, D, F, Cl, Br, I, OH, CN, NO 2 , NH 2 , methyl, ethyl, n-propyl, isopropyl, methoxy or Ethoxy.
  • R 5 is H, D, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, trifluoromethyl, cyclopropyl, cyclo Butyl, cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazine Base, phenyl, furanyl, pyrrolyl, pyridyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, oxadiazolyl, 1,3,5-triazinyl, thiazolyl , thienyl, pyr
  • R 6 and R 7 are each independently H, D, F, Cl, Br, I, CN, NO 2 , OH, NH 2 , C 1-4 alkyl, C 1-4 alkane Oxyl group, C 1-4 alkylamino group, C 1-4 haloalkyl group, C 1-4 haloalkoxy group, C 3-6 cycloalkyl group, heterocyclic group composed of 5-6 atoms, C 6-10 aryl group Or a heteroaryl group of 5-6 atoms, wherein the C 1-4 alkyl group, the C 1-4 alkoxy group, the C 1-4 alkylamino group, the C 1-4 haloalkyl group, the C 1-4 halogenated alkane An oxy group, a C 3-6 cycloalkyl group, a heterocyclic group of 5-6 atoms, a C 6-10 aryl group, and a heteroaryl group of 5-6 atoms are each independently unsubstituted or 1, 2 Sub
  • R 6 and R 7 are each independently H, D, F, Cl, Br, I, CN, NO 2 , OH, NH 2 , methyl, ethyl, n-propyl, isopropyl Base, n-butyl, isobutyl, tert-butyl, methoxy, ethoxy, methylamino, trifluoromethyl, trifluoromethoxy, difluoromethoxy, cyclopropyl, cyclobutyl, Cyclopentyl, cyclohexyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, benzene , furyl, pyrrolyl, pyridyl, pyrazolyl,
  • R 6 , R 7 and the carbon atom to which they are attached form a C 3-6 carbocyclic ring, a heterocyclic ring of 5-6 atoms, a C 6-10 aromatic ring or 5-6 atoms.
  • R 6 , R 7 and the carbon atom to which they are attached form a cyclopropane, cyclobutane, cyclopentane, cyclohexane, pyrrolidine, tetrahydrofuran, tetrahydrothiophene, tetrahydropyran, Tetrahydrothiopyran, piperidine, morpholine, thiomorpholine, piperazine, benzene ring, furan, pyrrole, pyridine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3, 5-triazine, thiazole, thiophene, pyrazine, pyridazine or pyrimidine, wherein the cyclopropane, cyclobutane, cyclopentane, cyclohexane, pyrrolidine, tetrahydrofuran,
  • R 5 , R 6 together with the atoms to which they are attached, or R 5 , R 7 and the atoms to which they are attached form aziridine, azetidine, pyrrolidine, Pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, thiomorpholine, piperazine, pyrrole, pyridine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3,5-triazine, thiazole, pyrazine, pyridazine or pyrimidine, wherein the aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, Piperidine, morpholine, thiomorpholine, piperazine, pyrrole, pyridine, pyrazole
  • R 8 and R 9 are each independently H, D, R e OC 1-4 alkylene, R d R c NC 1-4 alkylene, C 1-6 alkyl, C 1 -4 haloalkyl, C 3-6 cycloalkyl, C 3-6 cycloalkyl-C 1-2 alkylene, heterocyclic group consisting of 5-6 atoms, (heterocyclic ring of 5-6 atoms) —C 1-2 alkylene, C 6-10 aryl, C 6-10 aryl-C 1-2 alkylene, heteroaryl consisting of 5-6 atoms or (5-6 atoms) a heteroaryl)-C 1-2 alkylene group, wherein the C 1-6 alkyl group, C 1-4 haloalkyl group, C 3-6 cycloalkyl group, C 3-6 cycloalkyl-C 1 -2 alkylene group, heterocyclic group consisting of 5-6 atoms, (heterocyclic group of 5-6 atoms
  • R 8 and R 9 are each independently H, D, R d R c NC 1-4 alkylene, methyl, ethyl, n-propyl, isopropyl, n-butyl, iso Butyl, tert-butyl, n-pentyl, n-hexyl, trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 3-6 cycloalkyl-C 1-2 alkylene, Pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, Thiomorpholyl, piperazinyl, (heter
  • R 8 , R 9 and the nitrogen atom to which they are attached form a heterocyclic ring of 3-6 atoms or a heteroaryl ring of 5-6 atoms, wherein said 3-6
  • the heterocyclic ring composed of an atom and the heteroaryl ring composed of 5-6 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents independently selected from D, F, Cl.
  • R 8 , R 9 and the nitrogen atom to which they are attached form aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine , morpholine, thiomorpholine, piperazine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3,5-triazine, thiazole, pyrazine, pyridazine or pyrimidine, wherein , aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, thiomorpholine, piperazine, pyrazole, imidazole, three Oxazole, tetrazole, oxazole, oxadiazole, 1,3,5-triazine,
  • R c , R d , R e and R f are each independently H, D, C 1-4 alkyl, C 1-4 haloalkyl, C 3-6 cycloalkyl, C 3 -6 cycloalkyl-C 1-2 alkylene group, heterocyclic group composed of 5-6 atoms, (heterocyclic group of 5-6 atoms)-C 1-2 alkylene group, C 6-10 An aryl group, a C 6-10 aryl-C 1-2 alkylene group, a heteroaryl group of 5-6 atoms or a heteroaryl group of 5-6 atoms, a C 1-2 alkylene group, Wherein the C 1-4 alkyl group, the C 1-4 haloalkyl group, the C 3-6 cycloalkyl group, the C 3-6 cycloalkyl-C 1-2 alkylene group, the heterocyclic ring composed of 5-6 atoms Cyclic group, (heterocyclic group of 5-6 atoms Cyc
  • R c , R d , R e , and R f are each independently H, D, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl , trifluoromethyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, C 3-6 cycloalkyl-C 1-2 alkylene, pyrrolidinyl, pyrazolidinyl, imidazolidinyl , oxazolidinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, (5 Heterocyclic group consisting of -6 atoms
  • R c , R d and the nitrogen atom to which they are attached form a heterocyclic ring of 3-6 atoms or a heteroaryl ring of 5-6 atoms, wherein said 3-6
  • the heterocyclic ring composed of an atom and the heteroaryl ring composed of 5-6 atoms are each independently unsubstituted or substituted by 1, 2, 3 or 4 substituents independently selected from D, F, Cl.
  • R c , R d and the nitrogen atom to which they are attached form an aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine , morpholine, thiomorpholine, piperazine, pyrazole, imidazole, triazole, tetrazole, oxazole, oxadiazole, 1,3,5-triazine, thiazole, pyrazine, pyridazine or pyrimidine, wherein , aziridine, azetidine, pyrrolidine, pyrazolidine, imidazolidine, oxazolidine, thiazolidine, piperidine, morpholine, thiomorpholine, piperazine, pyrazole, imidazole, three Oxazole, tetrazole, oxazole, oxadiazole, 1,3,
  • the invention relates to a structure of one of the following, or a stereoisomer, geometric isomer, tautomer, oxynitride, solvate, metabolite, pharmaceutically acceptable Salts, dimers, trimers or their prodrugs,
  • the invention in another aspect, relates to a pharmaceutical composition comprising a compound of the invention.
  • compositions of the present invention further comprise a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle, or combination thereof.
  • the pharmaceutical composition of the present invention further comprises one or more additional additional therapeutic agents, wherein the additional additional therapeutic agent is selected from the group consisting of an anti-diabetic drug, an anti-hyperglycemic drug, an anti-obesity drug, An antihypertensive drug, an appetite suppressing drug, a lipid lowering drug, or a combination thereof.
  • the additional additional therapeutic agent is selected from the group consisting of an anti-diabetic drug, an anti-hyperglycemic drug, an anti-obesity drug, An antihypertensive drug, an appetite suppressing drug, a lipid lowering drug, or a combination thereof.
  • compositions of the invention may be in the form of a liquid, solid, semi-solid, gel or spray.
  • the antidiabetic and antihyperglycemic agents of the invention are each independently selected from the group consisting of an SGLT2 inhibitor, a biguanide, a sulfonylurea, a glucosidase inhibitor, a PPAR agonist.
  • Oxide proliferator-activated receptor agonist Oxide proliferator-activated receptor agonist
  • ⁇ P2 inhibitor adipocyte fatty acid binding protein inhibitor
  • PPAR ⁇ / ⁇ dual activator peroxisome proliferator-activated receptor ⁇ / ⁇ dual activator
  • dipeptidyl peptidase IV inhibitor dipeptidyl peptidase IV inhibitor
  • glinide class insulin
  • glucagon-like peptide-1 inhibitor PTP1B inhibitor
  • PTP1B inhibitor protein tyrosine phosphatase 1B inhibitor
  • glycogen phosphorylase inhibition Agent glucose-6-phosphatase inhibitor or a combination thereof.
  • the anti-obesity agent of the present invention is selected from the group consisting of central anti-obesity agents, MCH (anthraquinone-concentrating hormone) receptor antagonists, neuropeptide Y receptor antagonists, and cannabinoids.
  • MCH anthraquinone-concentrating hormone
  • neuropeptide Y receptor antagonists neuropeptide Y receptor antagonists
  • cannabinoids cannabinoids.
  • Body antagonist brain gut peptide antagonist, lipase inhibitor, ⁇ 3 agonist, 11 ⁇ -HSD1 (11 ⁇ hydroxysteroid dehydrogenase 1) inhibitor, DGAT-1 (diacylglycerol acyltransferase 1) inhibitor, peptide A sexual appetite suppressant, a Cholecystokinin agonist, a feeding inhibitor, or a combination thereof.
  • the antihypertensive agent of the present invention is selected from the group consisting of an angiotensin converting enzyme inhibitor, an angiotensin II receptor antagonist, a calcium channel antagonist, a potassium channel opener, a diuretic, or a combination thereof. .
  • the lipid-lowering drug of the present invention is selected from the group consisting of an MTP inhibitor (microsomal triglyceride transfer protein inhibitor) and an HMGCoA reductase inhibitor (hydroxymethylglutaryl coenzyme A reductase inhibitor) , squalene synthetase inhibitor, betabutyric acid hypolipidemic drug (also known as fibrate lipid-lowering drug), ACAT inhibitor (acetylcholesteryl acetyltransferase inhibitor), lipoxygenase inhibitor, cholesterol absorption inhibition Agent, ileal sodium ion/bile acid cotransporter inhibitor, upregulator of LDL receptor activity, niacin hypolipidemic drug, bile acid chelate or a combination thereof.
  • MTP inhibitor microsomal triglyceride transfer protein inhibitor
  • HMGCoA reductase inhibitor hydroxymethylglutaryl coenzyme A reductase inhibitor
  • squalene synthetase inhibitor hydroxymethylg
  • the lipid lowering drug of the present invention is selected from the group consisting of pravastatin, simvastatin, atorvastatin, fluvastatin, cerivastatin, etavatatin, russatin, or a combination thereof.
  • 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, wherein the medicament is for inhibiting SGLT1 in a human or animal body.
  • 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, wherein the medicament is for improving the intestinal environment.
  • the invention relates to the use of a compound or pharmaceutical composition according to the invention for the preparation of a medicament, wherein the medicament is for preventing or treating a disease, alleviating the symptoms of the disease or delaying the progression of the disease or Attack, wherein the disease is diabetes, diabetic complications, insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipidemia, obesity, syndrome X, atherosclerosis, cardiovascular disease, congestive heart failure, Hypomagnesemia, hyponatremia, renal failure, disorders associated with blood concentration, constipation or high blood pressure.
  • the diabetic complication is diabetic retinopathy, diabetic neuropathy, or diabetic nephropathy.
  • the hyperlipidemia is hypertriglyceridemia.
  • the invention in another aspect, relates to a method of inhibiting SGLT1 activity using a compound or pharmaceutical composition of the invention, the method comprising administering to a patient a therapeutically effective amount of the compound or the pharmaceutical composition.
  • the invention in another aspect, relates to a method of using the compounds or pharmaceutical compositions of the invention to improve the intestinal environment, the method comprising administering to the patient a therapeutically effective amount of the compound or the pharmaceutical composition.
  • the present invention relates to a method of using the compound or pharmaceutical composition of the present invention for preventing or treating a disease comprising administering to a patient an effective treatment of a compound or a pharmaceutical composition of the present invention Amount, wherein the disease is diabetes, diabetic complications, insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipidemia, obesity, syndrome X, atherosclerosis, cardiovascular disease, congestive heart failure , hypomagnesemia, hyponatremia, renal failure, disorders associated with blood concentration, constipation or high blood pressure.
  • the above compounds or pharmaceutical compositions thereof provided by the present invention may be co-administered with other therapies or therapeutic agents.
  • the mode of administration can be simultaneous, sequential or at regular intervals.
  • the dosage of the compound or pharmaceutical composition required to effect a therapeutic, prophylactic or prolonged effect will generally depend on the particular compound being administered, the patient, the particular disease or condition and its severity, the route and frequency of administration, and the like, and The specific situation is judged. For example, when the compound or pharmaceutical composition provided by the present invention is administered by the intravenous route, it can be administered once a week or even at a longer interval.
  • the invention relates to the use of a compound or pharmaceutical composition of the invention for inhibiting the activity of SGLT1.
  • the invention relates to the use of a compound or pharmaceutical composition of the invention for improving the intestinal environment.
  • the present invention relates to the use of a compound or a pharmaceutical composition according to the present invention for the prevention or treatment of a disease which alleviates or delays the development or onset of a disease in which diabetes or diabetic complications , insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipidemia, obesity, syndrome X, atherosclerosis, cardiovascular disease, congestive heart failure, hypomagnesemia, hyponatremia, renal failure , disorders associated with blood concentration, constipation or high blood pressure.
  • the salt refers to a pharmaceutically acceptable salt.
  • pharmaceutically acceptable means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal treated therewith.
  • the compounds of the invention also include other salts of such compounds which are not necessarily pharmaceutically acceptable salts and which are useful as compounds for the preparation and/or purification of the invention and/or for the isolation of the compounds of the invention.
  • the intermediate of the enantiomer is not necessarily pharmaceutically acceptable salts and which are useful as compounds for the preparation and/or purification of the invention and/or for the isolation of the compounds of the invention.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic and organic acids, such as acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/ Carbonate, hydrogen sulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorophylline, citrate, ethanedisulfonate, fumarate, glucoheptonate, Portuguese Saccharate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, Malay Acid salt, malonate, mandelic acid salt, methanesulfonate, methyl sulfate, naphthoate, naphthalene sulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalic acid Salt, palmitate
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid. , ethanesulfonic acid, p-toluenesulfonic acid, sulfosalicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic bases and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of Groups I to XII of the Periodic Table.
  • the salt is derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include the ammonium, potassium, sodium, calcium, and magnesium salts.
  • Organic bases from which salts can be derived include primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins and the like.
  • Certain organic amines include, for example, isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine, and tromethamine. .
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid form of these compounds with a stoichiometric amount of a suitable base such as Na, Ca, Mg or K hydroxide, carbonate, bicarbonate, and the like, or by The free base form of these compounds is prepared by reaction with a stoichiometric amount of a suitable acid. This type of reaction is usually carried out in water or an organic solvent or a mixture of the two.
  • a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • the compounds of the present invention may also be obtained in the form of their hydrates or include other solvents for their crystallization.
  • the compounds of the invention may be formed intrinsically or by design to form solvates having a pharmaceutically acceptable solvent, including water; thus, the invention is intended to include both solvated and unsolvated forms.
  • isotopically labeled compounds have the structure depicted by the general formula given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that may be incorporated into the 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, 18 F , 31 P, 32 P, 36 S, 37 Cl or 125 I.
  • the compounds of the invention include compounds defined by the invention, labeled with various isotopes, for example, those in which a radioisotope such as 3 H, 14 C and 18 F is present, or in which a non-radioactive isotope is present, Such as 2 H and 13 C.
  • a radioisotope such as 3 H, 14 C and 18 F
  • a non-radioactive isotope such as 2 H and 13 C.
  • isotopically labeled compounds can be used for metabolic studies (using 14 C), reaction kinetic studies (using, for example, 2 H or 3 H), detection or imaging techniques such as positron emission tomography (PET) or including drugs or primers.
  • PET positron emission tomography
  • SPECT Single photon emission computed tomography
  • 18 F-labeled compounds are particularly desirable for PET or SPECT studies.
  • Isotopically labeled compounds of formula (I) can be prepared by conventional techniques familiar to those skilled in the art or by the use of suitable isotopically labeled reagents in place of the previously used unlabeled reagents as described in the Examples and Preparations of the present invention.
  • substitution of heavier isotopes may provide certain therapeutic advantages resulting from higher metabolic stability. For example, increased in vivo half-life or reduced dose requirements or improved therapeutic index.
  • hydrazine in this context is considered to be a substituent of the compound of formula (I).
  • Isotopic enrichment factors can be used to define the concentration of such heavier isotopes, particularly ruthenium.
  • isotopic enrichment factor refers to the ratio between the isotope abundance and the natural abundance of a given isotope.
  • a substituent of a compound of the invention is designated as hydrazine
  • the compound has at least 3500 for each of the specified hydrazine atoms (52.5% of ruthenium incorporation at each of the specified ruthenium atoms), at least 4,000 (60% of ruthenium incorporation), At least 4,500 (67.5% of cerium incorporation), at least 5,000 (75% of cerium incorporation), at least 5,500 (82.5% of cerium incorporation), at least 6,000 (90% of cerium incorporation), at least 6333.3 (95%) Iridium enrichment factor with at least 6466.7 (97% cerium incorporation), at least 6600 (99% cerium incorporation) or at least 6633.3 (99.5% cerium incorporation).
  • the present invention can include pharmaceutically acceptable solvates wherein the solvent of crystallization may be isotopically substituted, for example D 2 O, acetone -d 6, or DMSO-d 6 solvate of those.
  • compositions, formulations and administration of the compounds of the invention are provided.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention or a compound of the structure shown in the examples, or a stereoisomer, geometric isomer, tautomer, oxynitride or solvate thereof. , metabolites, dimers, trimers, and pharmaceutically acceptable salts or prodrugs thereof.
  • the pharmaceutical composition further comprises at least one pharmaceutically acceptable carrier, excipient, adjuvant, vehicle or combination thereof, and optionally, other therapeutic and/or prophylactic ingredients.
  • the amount of the compound in the pharmaceutical composition of the present invention is effective to detectably inhibit the activity of sodium-dependent glucose transporters (SGLTs), particularly SGLT1.
  • SGLTs sodium-dependent glucose transporters
  • pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable prodrugs, salts, esters, ester salts, or any other agent which can be administered, directly or indirectly, depending on the needs of the patient.
  • a pharmaceutically acceptable carrier may contain inert ingredients which do not unduly inhibit the biological activity of the compound.
  • the pharmaceutically acceptable carrier should be biocompatible, for example non-toxic, non-inflammatory, non-immunogenic or have no other adverse effects or side effects once administered to the patient. Standard pharmaceutical technology can be used.
  • the pharmaceutical compositions or pharmaceutically acceptable compositions of the present invention further comprise a pharmaceutically acceptable carrier, adjuvant or excipient, as applied herein, including suitable for specific Target dosage form, any solvent, diluent, liquid excipient, dispersant, suspending agent, surfactant, isotonic agent, thickener, emulsifier, preservative, solid binder or lubricant, etc. .
  • a pharmaceutically acceptable carrier including suitable for specific Target dosage form, any solvent, diluent, liquid excipient, dispersant, suspending agent, surfactant, isotonic agent, thickener, emulsifier, preservative, solid binder or lubricant, etc.
  • Remington The Science and Practice of Pharmacy, 21st edition, 2005, ed. DB Trooy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J.
  • materials that can be used as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (eg, human serum albumin), buffer substances (eg, Tween 80) , phosphate, glycine, sorbic acid or potassium sorbate), a mixture of partial glycerides of saturated plant fatty acids, water, salts or electrolytes (eg, protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride or zinc salts) , silica gel, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylate, wax, polyethylene-polyoxypropylene-block copolymer, methyl cellulose, hydroxypropyl methylcellulose, lanolin, sugar (for example) Lactose, glucose and sucrose), starch (such as corn starch and potato starch), cellulose and its derivatives (such as sodium carboxymethylcellulose, e
  • the compounds or compositions of this invention may be administered by any suitable means, orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments or drops) depending on the severity of the infection being treated.
  • the oral cavity is administered to a human or other animal as a mouth or nasal spray or the like and the pharmaceutically acceptable composition.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage form may contain inert diluents conventional in the art such as water or other solvents, solubilizers and emulsifiers such as ethanol, isopropanol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate.
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening agents, flavoring agents, and flavoring agents.
  • sterile injectable preparations such as sterile injectable aqueous or oily suspensions
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P., and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as a solvent or suspending medium.
  • any odorless, fixed oil may be employed including synthetic monoglycerides or diglycerides.
  • fatty acids such as oleic acid, are used in the preparation of injectables.
  • the injectable preparation can be sterilized by filtration through a bacterial retention filter or by the addition of a bactericidal agent which is previously dissolved or dispersible in sterile water or other sterile injectable medium.
  • Injectable depot forms are made by forming a microcapsule matrix of the compound in a biodegradable polymer such as polylactide-polyglycolic acid. The rate of compound release can be controlled based on the ratio of compound to polymer and the nature of the particular polymer employed. Examples of other biodegradable polymers include polyorthoesters and polyanhydrides. Injectable depot formulations are also prepared by entrapping the compound in liposomes or microemulsions which are compatible with body tissues.
  • Composition for rectal or vaginal administration is especially a suppository which can be prepared by admixing a compound according to the invention and a suitable non- irritating excipient or carrier, such as cocoa butter, polyethylene glycol or suppository wax, the excipient Or the carrier is solid at ambient temperature but liquid at body temperature and thus melts in the rectum or vaginal cavity and releases the active compound.
  • a suitable non- irritating excipient or carrier such as cocoa butter, polyethylene glycol or suppository wax
  • Oral solid dosage forms include capsules, tablets, pills, powders, and granules.
  • the active compound is admixed with at least one inert pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) filler or bulking agent, for example starch, lactose, sucrose , glucose, mannitol and silicic acid, b) binders such as carboxymethylcellulose, alginates, gels, polyvinylpyrrolidone, sucrose and acacia, c) humectants, such as glycerin, d) disintegrants For example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate, e) solution blockers, such as paraffin, f) absorption accelerators, such as quaternary ammonium compounds, g) Wetting agents, such as cetyl alcohol and gly
  • Solid compositions of a similar type may also be employed as fillers in soft and hard gel capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols.
  • Solid dosage forms of tablets, lozenges, capsules, pills and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical art. They may optionally contain opacifying agents and may also be of a nature such that the active ingredient is optionally released in a delayed manner or, preferably, in a portion of the intestinal tract. Examples of embedding compositions that can be used include polymers and waxes.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard gel capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols.
  • the active compounds may also be presented in microencapsulated form with one or more of the above-mentioned excipients.
  • Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, controlled release coatings, and other coatings well known in the pharmaceutical arts.
  • the active compound may be mixed with at least one inert diluent such as sucrose, lactose or starch.
  • inert diluent such as sucrose, lactose or starch.
  • such dosage forms may also contain additional materials other than inert diluents, such as tableting lubricants and other tableting aids such as magnesium stearate and microcrystalline cellulose.
  • the dosage form may also contain a buffer.
  • opacifying agents may optionally contain opacifying agents and may also be of a nature such that the active ingredient is optionally released in a delayed manner or, preferably, in a portion of the intestinal tract.
  • embedding compositions include polymers and waxes.
  • Topical or transdermal administration forms of the compounds of the invention include ointments, ointments, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active compound is admixed under sterile conditions with a pharmaceutically acceptable carrier and any required preservatives or buffers which may be required.
  • Ophthalmic formulations, ear drops, and eye drops are also contemplated as being within the scope of the invention.
  • the present invention contemplates the use of a dermal patch that provides the added benefit of controlling the delivery of the compound to the body.
  • This dosage form can be made by dissolving or dispersing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound through the skin. The rate can be controlled by providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • compositions of the present invention may also be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or by implantation of a kit.
  • parenteral as used in the present invention includes, but is not limited to, subcutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional, and intracranial injection or infusion techniques.
  • the composition is administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of the invention may be aqueous or oily suspensions. These suspensions can be prepared following techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent such as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any odorless, fixed oil may be employed including synthetic monoglycerides or diglycerides.
  • oils especially in the form of polyoxyethylenated, such as olive oil or castor oil
  • fatty acids such as oleic acid and its glyceride derivatives
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersing agent, such as carboxymethylcellulose or similar dispersing agents which are conventionally employed in the formulation of pharmaceutically acceptable dosage forms, including emulsions and suspensions.
  • a long-chain alcohol diluent or dispersing agent such as carboxymethylcellulose or similar dispersing agents which are conventionally employed in the formulation of pharmaceutically acceptable dosage forms, including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans, and other emulsifiers or bioavailability enhancers commonly used in the manufacture of pharmaceutically acceptable solid, liquid or other dosage forms may also be used for formulation purposes.
  • compositions of the present invention can be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • conventional carriers include, but are not limited to, lactose and starch.
  • a lubricant such as magnesium stearate is also usually added.
  • useful diluents include lactose and dried cornstarch.
  • compositions of the invention may be administered in the form of a suppository for rectal use.
  • These pharmaceutical compositions can be prepared by mixing agents and non-irritating excipients which are solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the drug.
  • agents and non-irritating excipients include, but are not limited to, cocoa butter, beeswax, and polyethylene glycol.
  • compositions of the present invention may also be administered topically, especially when the therapeutic target includes areas or organs that are readily accessible by topical application, including ocular, cutaneous or low intestinal disease. It is easy to prepare suitable topical formulations for each of these areas or organs.
  • Topical application to the lower intestinal tract can be achieved in a rectal suppository formulation (see above) or a suitable enema formulation.
  • a topical skin patch can also be used.
  • the pharmaceutical compositions may be formulated as a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers suitable for topical application to the compounds of the present invention include, but are not limited to, mineral oil, petroleum jelly, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical compositions may be formulated as a suitable lotion or cream containing the active component in suspension or in apharmaceutically acceptable carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water. .
  • the pharmaceutical compositions may be formulated as micronized suspensions in isotonic pH adjusted sterile saline, or especially in isotonic pH adjusted sterile saline, with or without a preservative such as benzalkonium chloride.
  • the pharmaceutical composition can be formulated as a salve, such as petrolatum.
  • the pharmaceutical composition can also be administered by nasal aerosolized spray or by inhalation.
  • This composition is prepared according to techniques well known in the pharmaceutical arts and prepared in saline using benzyl alcohol and other suitable preservatives, bioavailability absorption enhancers, fluorocarbons and/or other conventional solubilizers or dispersants. Solution.
  • the compounds used in the methods of the invention can be formulated in unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unitary dosages of the subject, each unit containing a predetermined amount of active ingredient calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier.
  • the unit dosage form can be one of a single daily dose or multiple daily doses (e.g., about 1-4 or more times per day). When multiple daily doses are used, the unit dosage form for each dose may be the same or different.
  • the compounds of the invention may be administered as the sole pharmaceutical agent or in combination with one or more additional therapeutic (pharmaceutical) agents, wherein the combination causes an acceptable adverse reaction for diabetes, diabetic complications, and other related diseases.
  • the treatment has special significance, including, but not limited to, type I diabetes, type 2 diabetes, diabetic retinopathy, diabetic neuropathy, diabetic nephropathy, insulin resistance, hyperglycemia, hyperinsulinemia , elevated levels of fatty acids or glycerol in the blood, hyperlipidemia, obesity, hypertriglyceridemia, X syndrome, diabetic complications, atherosclerosis, cardiovascular disease, congestive heart failure, low magnesium Hypertension, hyponatremia, renal failure, disorders associated with blood concentration, constipation or high blood pressure.
  • the "additional therapeutic agent" used in the present invention includes an antidiabetic drug, an antihyperglycemic drug, an antiobesity drug, an antihypertensive drug, an appetite suppressing drug, a lipid lowering drug, or a combination thereof.
  • the anti-diabetic agents of the present invention include, but are not limited to, SGLT2 inhibitors (for example, dapagliflozin, canagliflozin, tofogliflozin, iglitavir ( Ipragliflozin), luseogliflozin, empagliflozin, biguanide drugs (eg phenformin, metformin), sulfonylureas (eg acesulfame) (acetohexamide), chlorpropamide, glibenclamide, glipizide (glipizide), gliclazide (glimezide) Glemepiride, glipentide, gliquidone, tolazamide, tolbutamide, and meglitinide Lina drugs (such as repaglinide and nateglinide), alpha-glucoside hydrolase inhibitors (such as acarbose), alpha-glucosidase inhibitors
  • the antihyperglycemic agent of the present invention includes, but is not limited to, an SGLT2 inhibitor (eg, dapagliflozin, canagliflozin, tofogliflozin, igrelin) (ipragliflozin), luseogliflozin, empagliflozin, biguanide drugs (such as phenformin, metformin), sulfonylureas (such as vinegar ring) Urea (acetohexamide), chlorpropamide, glibenclamide, glipizide (glipizide), gliclazide (glimezide) , glimepiride, glipentide, gliquidone, tolazamide, tolbutamide, and meglitinide, Glinide drugs (such as repaglinide and nateglinide), alpha-glucoside hydrolase inhibitors (such as acarbose), alpha-glucosidase inhibitor
  • the anti-obesity drugs of the present invention include, but are not limited to, central anti-obesity drugs (for example, dexfenfluramine, fenfluramine, phentermine, xibuchamin). (sibutramine), amfepramone, d-amphetamine, mazidol, phenylpropanolamine, clobenzorex, MCH receptor antagonism Agents (for example, compounds described in WO06035967, SB-568849; SNAP-7941, T-226296), neuropeptide Y receptor antagonists (eg CP-422935), cannabinoid receptor antagonists (eg rimonabant) , SR-147778), brain gut peptide antagonists, lipase inhibitors (eg, orlistat, ATL-962), ⁇ 3 agonists (eg AJ-9677, AZ40140), 11 ⁇ -HSD1 inhibitors (eg BVT-3498
  • the lipid-lowering reagents of the present invention include, but are not limited to, MTP inhibitors, HMG CoA reductase inhibitors, squalene synthetase inhibitors, and fibrate lipid-lowering drugs (beanbutyric lipid-lowering drugs) , ACAT inhibitor, lipoxygenase inhibitor, cholesterol absorption inhibitor, ileal sodium ion/bile acid cotransporter inhibitor, upregulator of LDL receptor activity, bile acid chelate or niacin hypolipidemic drug .
  • the lipid lowering agent is selected from the group consisting of pravastatin, simvastatin, atorvastatin, fluvastatin, cerivastatin, etatastatin or rosuvastatin.
  • the anti-obesity agent is selected from the group consisting of CB-1 antagonists (eg, rimonabant, taranabant, surinabant, otenabant, SLV319 and AVE1625), intestinal-selective MTP inhibitors (eg, dirlotapide, mitratapide, and implitapide), CCKa agonists, 5HT2c agonists (eg, chlorine) Lorcaserin, MCR4 agonist, lipase inhibitor (eg Cetilistat), PYY 3-36 , opioid antagonist (eg naltrexone), oleoyl-female Ketone, obinepitide, pramlintide, tesofensine, leptin,
  • the appropriate anti-inflammatory agents according to the present invention include genital/urinary tract infection prevention and treatment drugs, such as Vaccinium macrocarpon and cranberry derivatives, such as cranberry juice, cranberry extract or cranberry. Vine flavonols.
  • genital/urinary tract infection prevention and treatment drugs such as Vaccinium macrocarpon and cranberry derivatives, such as cranberry juice, cranberry extract or cranberry. Vine flavonols.
  • other suitable anti-inflammatory agents include, but are not limited to, aspirin, non-steroidal anti-inflammatory drugs, glucocorticosteroids, sulfasalazine, and cyclooxygenase II selective inhibitors.
  • the amount of the compound in the compound or pharmaceutical composition of the present invention can effectively and detectably inhibit the activity of sodium-dependent glucose transporters (SGLTs), particularly the inhibition of SGLT1 activity. Therefore, the compound of the present invention will be applied to the prevention, treatment or improvement of the symptoms of these diseases of diabetes and related diseases.
  • SGLTs sodium-dependent glucose transporters
  • the compounds of the present invention will be, but are not limited to, administered to a patient using an effective amount of a compound or pharmaceutical composition of the present invention to prevent or treat diabetes and related diseases in a patient, or to alleviate symptoms of diabetes and related diseases, or delay diabetes And the development or onset of related diseases or for increasing the level of high-density lipoprotein.
  • diseases include, but are not limited to, diabetes, especially type II diabetes, as well as insulin resistance, hyperglycemia, hyperinsulinemia, hyperlipidemia such as hypertriglyceridemia, diabetic complications, such as diabetic retinopathy. , diabetic neuropathy or diabetic nephropathy, obesity, X syndrome, atherosclerosis, cardiovascular disease, congestive heart failure, hypomagnesemia, hyponatremia, renal failure, disorders associated with blood concentration, Constipation or high blood pressure.
  • the compound of the present invention has an excellent intestinal environment-improving effect, and can increase the number of beneficial bacteria such as bifidobacteria and lactobacilli, increase the organic acid in the intestine, and reduce the spoilage products in the intestine.
  • beneficial bacteria such as bifidobacteria and lactobacilli
  • By covering the intestinal environment diseases associated with changes in the intestinal environment can be improved.
  • the "disease associated with changes in the intestinal environment” includes, but is not limited to, chronic kidney disease, pseudomembranous colitis/hemorrhagic enteritis, infectious enteritis, ulcerative colitis, Crohn's disease, irritable bowel Syndrome, obesity, arteriosclerosis, hypertension, Guillain-Barre syndrome, allergic disease, diabetes, multiple sclerosis, autoimmune disease, alcoholic liver dysfunction, nonalcoholic fatty liver disease, nonalcoholic Fatty hepatitis, non-steroidal anti-inflammatory drugs causing enteritis, stress, depression, influenza, periodontal disease, cancer, hay fever, functional dyspepsia, pruritus, etc.
  • the compounds or pharmaceutical compositions of the invention are also suitable for the prevention and treatment of late diabetic lesions such as kidney disease, retinopathy, neuropathy, and myocardial infarction, peripheral arterial occlusive disease, thrombosis, arteriosclerosis, inflammation, immune disease, autoimmunity Sexual diseases such as AIDS, asthma, osteoporosis, cancer, psoriasis, Alzheimer's disease, schizophrenia and infectious diseases.
  • late diabetic lesions such as kidney disease, retinopathy, neuropathy, and myocardial infarction, peripheral arterial occlusive disease, thrombosis, arteriosclerosis, inflammation, immune disease, autoimmunity Sexual diseases such as AIDS, asthma, osteoporosis, cancer, psoriasis, Alzheimer's disease, schizophrenia and infectious diseases.
  • an “effective amount”, “effective therapeutic amount” or “effective amount” of a compound of the invention or a pharmaceutically acceptable pharmaceutical composition refers to an effective amount to treat or ameliorate the severity of one or more of the conditions mentioned herein. .
  • the compounds of the invention or pharmaceutically acceptable pharmaceutical compositions are effective over a relatively wide dosage range. For example, a daily dose of about 0.1 mg to 1000 mg per person is divided into one or several administrations.
  • the methods, compounds and pharmaceutical compositions according to the present invention can be administered in any amount and in any route of administration to effectively treat or alleviate the severity of the disease. The exact amount required will vary depending on the patient's condition, depending on race, age, general condition of the patient, severity of infection, specific factors, mode of administration, and the like.
  • the compounds or pharmaceutical compositions of this invention may be administered in combination with one or more other therapeutic agents, as discussed herein.
  • the compounds of the present invention can be prepared by the methods described herein, unless otherwise stated, wherein the substituents are as defined for formula (I), formula (I-a) or formula (II).
  • the following reaction schemes and examples are provided to further illustrate the contents of the present invention.
  • the structure of the compound was determined by nuclear magnetic resonance ( 1 H-NMR, 13 C-NMR or/and 19 F-NMR).
  • 1 H-NMR, 13 C-NMR, 19 F-NMR chemical shifts ( ⁇ ) are given in parts per million (ppm).
  • 1 H-NMR, 13 C-NMR, and 19 F-NMR were measured using a Bruker Ultrashield-400 nuclear magnetic resonance spectrometer and a Bruker Avance III HD 600 nuclear magnetic resonance spectrometer.
  • the solvent was deuterated chloroform (CDCl 3 ), deuterated. Methanol (CD 3 OD or MeOH-d 4 ) or deuterated dimethyl sulfoxide (DMSO-d 6 ).
  • Preparative purification or preparative resolution is generally carried out using a Novasep pump 250 high performance liquid chromatograph.
  • the LC-MS was measured using an Agilen-6120 Quadrupole LC/MS mass spectrometer.
  • the starting materials of the present invention are known and commercially available from Shanghai Accela Company, Energy Company, J&K, Tianjin Alpha Companies such as the Alfa Company can be synthesized or synthesized according to methods known in the art.
  • Nitrogen atmosphere means that the reaction flask is connected to a nitrogen balloon or steel kettle of about 1L volume;
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume or a stainless steel high pressure reactor having a volume of about 1 L;
  • the solution means an aqueous solution
  • reaction temperature is room temperature
  • room temperature is 20 ° C to 30 ° C unless otherwise specified.
  • the progress of the reaction in the examples was monitored by thin layer chromatography (TLC).
  • TLC thin layer chromatography
  • the systems used for the reaction were: dichloromethane and methanol systems, dichloromethane and ethyl acetate systems, petroleum ether and ethyl acetate systems.
  • the volume ratio of the solvent is adjusted depending on the polarity of the compound.
  • Column chromatography eluent systems include: A: petroleum ether and ethyl acetate systems, B: dichloromethane and ethyl acetate systems, C: dichloromethane and methanol systems.
  • the volume ratio of the solvent is adjusted depending on the polarity of the compound, and may be adjusted by adding a small amount of ammonia water, acetic acid or the like.
  • DMSO-d 6 deuterated dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • Mg magnesium; Cbz: benzyloxycarbonyl; Ac: acetyl;
  • ⁇ L microliter
  • mmol millimolar
  • MPa MPa; atm: standard atmospheric pressure; DCM: dichloromethane;
  • HBTU O-benzotriazole-tetramethylurea hexafluorophosphate
  • HATU O-(7-azabenzotriazol-1-yl)-N,N,N,N-tetramethyluronium hexafluorophosphate
  • DBU 1,8-diazabicyclo[5.4.0]undec-7-ene.
  • the intermediate represented by the formula (6) can be synthesized by the method disclosed in Synthesis Scheme 1.
  • the compound (1) is reacted with benzyl chloroformate to give a compound (2) .
  • the compound (2) is condensed with the compound (3) to form the compound (4) .
  • the compound (4) is subjected to catalytic hydrogenation to remove the Cbz protecting group on the amino group to give the compound (5) .
  • the compound (5) is salted with hydrogen chloride to give the compound (6) .
  • the intermediate represented by the formula (10) can be synthesized by the method disclosed in Synthesis Scheme 2. First, the compound (7) is reduced with a reducing agent to give a compound (8) . Then, the compound (8) is substituted with allyl bromide under basic conditions to give a compound (9) . Finally, compound (9) is reacted with metal Mg to give compound (10) .
  • the intermediate represented by the formula (15) can be synthesized by the method disclosed in Synthesis Scheme 3. First, the compound (11) is reacted with oxalyl chloride to give a compound (12) . Then, the compound (12) is reacted with dimethylhydroxylamine hydrochloride under basic conditions to give a compound (13) . Next, the compound (13) is reacted with the compound (10) to form a compound (14) . Finally, the compound (14) is subjected to a reduction reaction under the action of an acid and triethylsilane to give a compound (15) .
  • the intermediate represented by the formula (29) or the intermediate represented by the formula (30) can be synthesized by the method disclosed in Synthesis Scheme 4.
  • the compound (16) is catalytically oxidized in the presence of 2,2,6,6-tetramethylpiperidine oxide to give a compound (17) .
  • Compound (17) is reacted with acetic anhydride to give compound (18) .
  • Compound (15) reacts with isopropylmagnesium chloride and reacts with compound (18) to form compound (19) .
  • Compound (19) is reacted with methanol under acidic conditions to give compound (20) .
  • Compound (20) is oxidized by an oxidizing agent to form compound (21) .
  • Compound (21) reacts with formaldehyde under the action of DBU to form compound (22) . Then, the compound (22) is reduced with sodium borohydride to form a compound (23) . Compound (23) is subjected to ring closure under acidic conditions to give compound (24) . Compound (24) is oxidized with a Des Martin oxidant to give compound (25) . Compound (25) is reacted with methanol under acidic conditions to give compound (26) . Next, the compound (26) is reacted with a Grignard reagent at a low temperature to form a compound (27) . Compound (27) deprotects the hydroxy protecting group allylation compound (28) under the action of a catalyst.
  • the compound (28) is oxidized with iodobenzenediacetic acid in the presence of the catalyst 2,2,6,6-tetramethylpiperidine oxide to give the compound (29) .
  • Compound (29) is subjected to catalytic hydrogenation to remove the hydroxy protecting group Bn to give compound (30) .
  • the compound of the formula (32) can be synthesized by the method disclosed in Synthesis Scheme 5. First, the compound (29) is condensed with the compound (6) to give a compound (31) . Then, the compound (31) is subjected to catalytic hydrogenation to remove the hydroxy protecting group Bn to give the compound (32) .
  • the compound of the formula (32) can also be synthesized by the method disclosed in Synthesis Scheme 6. First, the compound (30) is reacted with acetic anhydride under basic conditions to give a compound (33) . Next, the compound (33) is condensed with the compound (6) to form a compound (34) . Finally, the compound (34) is deprotected under basic conditions to give the compound (32) .
  • Step 1 N-[1,1-Dimethyl-2-oxo-2-(2-pyrrolidin-1-ylethylamino)ethyl]carbamate
  • Step 11 (3R, 4S, 5R, 6R)-3,4,5-tribenzyloxy-6-(benzyloxymethyl)tetrahydropyran-2-one
  • Step 12 [(2R,3R,4S,5R)-3,4,5-Tribenzyloxy-6-oxo-tetrahydropyran-2-yl]methyl acetate
  • Glacial acetic acid 350 mL was added to (3R,4S,5R,6R)-3,4,5-tribenzyloxy-6-(benzyloxymethyl)tetrahydropyran-2-one at room temperature (
  • Step 13 [(2R,3R,4S,5R,6S)-6-[3-[[4-(4-Allyloxybutyl)phenyl]methyl]-4-methyl-phenyl] -3,4,5-tribenzyloxy-6-hydroxy-tetrahydro Pyr-2-yl]methyl acetate
  • Step 14 [(2R,3R,4S,5R,6S)-6-[3-[[4-(4-Allyloxybutyl)phenyl]methyl]-4-methyl-phenyl] -3,4,5-tribenzyloxy-6-methoxy-tetra Hydropyran-2-yl]methanol
  • Step 15 [(2S,3S,4S,5R,6S)-6-[3-[[4-(4-allyloxybutyl)phenyl]methyl]-4-methyl-phenyl] -3,4,5-tribenzyloxy-6-methoxy-tetrahydro Pyran-2-yl]formaldehyde
  • Step 16 (2R, 3S, 4S, 5R, 6S)-6-[3-[[4-(4-allyloxybutyl)phenyl]methyl]-4-methyl-phenyl]- 3,4,5-tribenzyloxy-2-(hydroxymethyl)-6- Methoxy-tetrahydropyran-2-carbaldehyde
  • Step 17 [(3S,4S,5R,6S)-6-[3-[[4-(4-allyloxybutyl)phenyl]methyl]-4-methyl-phenyl]-3 ,4,5-tribenzyloxy-2-(hydroxymethyl)-6-A Oxy-tetrahydropyran-2-yl]methanol
  • Step 18 [(1S, 2S, 3S, 4R, 5S)-5-[3-[[4-(4-allyloxybutyl)phenyl]methyl]-4-methyl-phenyl] -2,3,4-tribenzyloxy-6,8-dioxa double Cyclo [3.2.1] octane-1-yl]methanol
  • Step 20 (1S, 2S, 3S, 4R, 5S)-5-[3-[[4-(4-allyloxybutyl)phenyl]methyl]-4-methyl-phenyl]- 2,3,4-tribenzyloxy-6,8-dioxa double Ring [3.2.1] octane-1-carboxylic acid methyl ester
  • Step 21 2-[(1S,2S,3S,4R,5S)-5-[3-[[4-(4-allyloxybutyl)phenyl]methyl]-4-methyl-benzene 2,3,4-tribenzyloxy-6,8-dioxa Bicyclo[3.2.1]octane-1-yl]propan-2-ol
  • Step 22 4-[4-[[2-Methyl-5-[(1S,2S,3S,4R,5S)-2,3,4-tribenzyloxy-1-(1-hydroxy-1-) Methyl-ethyl)-6,8-dioxabicyclo[3.2.1] octane Alkan-5-yl]phenyl]methyl]phenyl]butan-1-ol
  • Step 23 4-[4-[[2-Methyl-5-[(1S,2S,3S,4R,5S)-2,3,4-tribenzyloxy-1-(1-hydroxy-1-) Methyl-ethyl)-6,8-dioxabicyclo[3.2.1] octane Alkan-5-yl]phenyl]methyl]phenyl]butyric acid
  • Step 24 2-Methyl-2-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S)-2,3,4-tribenzyloxy-1 -(1-hydroxy-1-methyl-ethyl)-6,8-dioxbicyclo [3.2.1] Octane-5-yl]phenyl]methyl]phenyl]butyrylamino]-N-(2-pyrrolidin-1-ylethyl)propanamide
  • Step 25 2-Methyl-2-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-( 1-hydroxy-1-methyl-ethyl)-6,8-dioxbicyclo [3.2.1] Octane-5-yl]phenyl]methyl]phenyl]butyrylamino]-N-(2-pyrrolidin-1-ylethyl)propanamide
  • 3-Aminopropionic acid (3.0 g, 34 mmol) was dissolved in dioxane (20 mL) at room temperature, sodium carbonate (11.0 g, 104 mmol) and water (60 mL) were added and cooled to 0 ° C.
  • Benzyl ester (6.0 mL, 41 mmol) was stirred at rt overnight. After washing with n-hexane (100 mL), the aqueous phase was adjusted to pH 1 with dilute hydrochloric acid (1M), ethyl acetate (100 mL ⁇ 2), and the combined organic phase was washed with saturated brine (100 mL)
  • the title compound (6.9 g, m.
  • Step 2 N-[3-(2-Dimethylaminoethylamino)-3-oxo-propyl]carbamic acid benzyl ester
  • Step 5 4-[4-[[2-Methyl-5-[(1S,2S,3S,4R,5S)-2,3,4-trihydroxy-1-(1-hydroxy-1-methyl) -ethyl)-6,8-dioxabicyclo[3.2.1]octane-5- Phenyl]methyl]phenyl]butyric acid
  • Step 6 4-[4-[[2-Methyl-5-[(1S,2S,3S,4R,5S)-2,3,4-triacetoxy-1-(1-hydroxy-1-) Methyl-ethyl)-6,8-dioxabicyclo[3.2.1] octane Alkan-5-yl]phenyl]methyl]phenyl]butyric acid
  • Step 7 [(1S,2S,3S,4R,5S)-2,4-Diacetoxy-5-[3-[[4-[4-[[3-(2-methylaminoethylamino) )-3-oxo-propyl]amino]-butyl]benzene Methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxabicyclo[3.2.1]octane-3-yl Acetate
  • Step 8 N-[3-(2-Methylaminoethylamino)-3-oxo-propyl]-4-[4-[[2-methyl-5-[(1S,2S,3S, 4R,5S)-2,3,4-trihydroxy-1-(1-hydroxyl -1-methyl-ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butanamide
  • Step 1 N-[2-(2-Diethylaminoethylamino)-1,1-dimethyl-2-oxo-ethyl]carbamic acid benzyl ester
  • N-[2-(2-Diethylaminoethylamino)-1,1-dimethyl-2-oxo-ethyl]carbamic acid benzyl ester (1.0 g, 3.0 mmol) at room temperature It was dissolved in anhydrous methanol (20 mL), and a 10% palladium/carbon catalyst (0.20 g, 0.18 mmol) was added and stirred under a hydrogen atmosphere for 5 hours. Filtration and concentration gave the title compound (m.
  • Step 4 [(1S,2S,3S,4R,5S)-2,4-Diacetoxy-5-[3-[[4-[4-[[2-(2-diethylaminoethylamino)) -1,1-dimethyl-2-oxoethyl]ammonium 4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxa Bicyclo[3.2.1]octane-3-yl]acetate
  • Step 5 N-(2-Diethylaminoethyl)-2-methyl-2-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S)) -2,3,4-trihydroxy-1-(1-hydroxy-1-methyl- Ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyrylamino]propanamide
  • 3-Amino-3-methyl-butyric acid (3.0 g, 26 mmol) was dissolved in dioxane (20 mL), EtOAc (EtOAc) Benzyl chloroformate (4.5 mL, 30 mmol) was added at 0<0>C. After washing with n-hexane (100 mL), the aqueous phase was adjusted to pH 1 with dilute hydrochloric acid (1M), ethyl acetate (100 mL ⁇ 2), and the combined organic phase was washed with saturated brine (100 mL) The title compound (5.4 g, m.
  • Step 2 N-[3-(2-Dimethylaminoethylamino)-1,1-dimethyl-3-oxo-propyl]carbamic acid benzyl ester
  • Step 5 [(1S,2S,3S,4R,5S)-2,4-Diacetoxy-5-[3-[[4-[4-[[3-(2-methylaminoethylamino) )-1,1-dimethyl-3-oxo-propyl]ammonia 4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxa Bicyclo[3.2.1]octane-3-yl]acetate
  • Step 6 N-(2-Dimethylaminoethyl)-3-methyl-3-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S)- 2,3,4-trihydroxy-1-(1-hydroxy-1-methyl-B 6,8-Dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyrylamino]butanamide
  • Step 1 N-[2-[3-(Dimethylamino)propylamino]-1,1-dimethyl-2-oxo-ethyl]carbamic acid benzyl ester
  • Step 4 [(1S, 2S, 3S, 4R, 5S)-2,4-diacetoxy-5-[3-[[4-[4-[[2-[3-(dimethylamino)) 1,1,1-dimethyl-2-oxo-ethyl]ammonia 4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxa Bicyclo[3.2.1]octane-3-yl]acetate
  • EtOAc EtOAc
  • EtOAc 2-Amino-N-[3-(dimethylamino)propyl]-2-methyl-propanamide dihydrochloride
  • v/v 8/1].
  • Step 5 N-[3-(Dimethylamino)propyl]-2-methyl-2-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S) )-2,3,4-trihydroxy-1-(1-hydroxy-1-methyl-B ,6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyrylamino]propanamide
  • Step 1 N-[2-[(3R)-3-fluoropyrrolidin-1-yl]ethyl]carbamic acid tert-butyl ester
  • N-[2-[(3R)-3-fluoropyrrolidin-1-yl]ethyl]carbamic acid tert-butyl ester (3.5 g, 15 mmol) was dissolved in ethyl acetate (30 mL) Ethyl HCl solution (10 mL, 4 M) was stirred for 2 h. The residue was dissolved in EtOAc (EtOAc)EtOAc.
  • Step 3 N-[2-[(3R)-3-fluoropyrrolidin-1-yl]ethylamino]-1,1-dimethyl-2-oxo-ethyl]carbamate
  • Step 6 [(1S, 2S, 3S, 4R, 5S)-2,4-diacetoxy-5-[3-[[4-[4-[[2-[2-[(3))) -fluoropyrrolidin-1-yl]ethylamino]-1,1-dimethyl-2- Oxo-ethyl]amino]-4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6 , 8-dioxabicyclo[3.2.1]octane -3-yl]acetate
  • EtOAc EtOAc, EtOAc (EtOAc) 2-Amino-N-[2-[(3R)-3-fluoropyrrolidin-1-yl]ethyl]-2-methyl-propanamide dihydrochloride (93 mg, 0.32 mmol).
  • EtOAc EtOAc
  • Step 7 N-[2-[(3R)-3-fluoropyrrolidin-1-yl]ethyl]-2-methyl-2-[4-[4-[[2-methyl-5-[ (1S, 2S, 3S, 4R, 5S)-2,3,4-trihydroxy-1-(1-hydroxyl -1-methyl-ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyrylamino]propanamide
  • Step 2 N-[1-[2-[(3R)-3-fluoropyrrolidin-1-yl]ethylcarbamoyl]cyclohexyl]carbamic acid benzyl ester
  • Step 5 [(1S, 2S, 3S, 4R, 5S)-2,4-diacetoxy-5-[3-[[4-[4-[[1-[2-[(3[ -fluoropyrrolidin-1-yl]ethylcarbamoyl]cyclohexyl] Amino]-4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxa Bicyclo[3.2.1]octane-3-yl]acetic acid ester
  • Step 6 N-[2-[(3R)-3-fluoropyrrolidin-1-yl]ethyl]-1-[4-[4-[[2-methyl-5-[(1S,2S, 3S,4R,5S)-2,3,4-trihydroxy-1-(1-hydroxy-1-methyl -ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyrylamino]cyclohexylcarboxamide
  • Step 1 N-[2-(2-Dimethylaminoethylamino)-1,1,-dimethyl-2-oxo-ethyl]-N-methyl-carboxylic acid tert-butyl ester
  • Step 3 [(1S,2S,3S,4R,5S)-2,4-Diacetoxy-5-[3-[[4-[4-[[2-(2-[ Amino)-1,1-dimethyl-2-oxo-ethyl] Methyl-amino]-4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8- Dioxabicyclo[3.2.1]octane-3-yl] Acetate
  • EtOAc EtOAc
  • Step 4 N-(2-Dimethylaminoethyl)-2-methyl-2-[methyl-[4-[4-[[2-methyl-5-[(1S,2S,3S, 4R,5S)-2,3,4-trihydroxy-1-(1-hydroxy-1- Methyl-ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyryl]amino]propanamide
  • Step 1 N-[2-[2-Dimethylaminoethyl(methyl)amino]-1,1,-dimethyl-2-oxo-ethyl]-carboxylic acid tert-butyl ester
  • Step 3 [(1S, 2S, 3S, 4R, 5S)-2,4-Diacetoxy-5-[3-[[4-[4-[[2-[2-dimethylaminoethyl] (methyl)amino]-1,1-dimethyl-2-oxo -ethyl]-amino]-4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6, 8-Dioxabicyclo[3.2.1]octane-3-yl] Acetate
  • EtOAc EtOAc
  • Step 4 N-(2-Dimethylaminoethyl)-N,2-dimethyl-2-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R, 5S)-2,3,4-trihydroxy-1-(1-hydroxy-1-methyl -ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyryl]propanamide
  • Step 1 (2S)-2-(2-Dimethylaminoethylcarbamoyl)azetidin-1-carboxylic acid tert-butyl ester
  • Step 3 [(1S, 2S, 3S, 4R, 5S)-2,4-Diacetoxy-5-[3-[[4-[(2S)-2-(2-dimethylaminoethyl) Carbamoyl)azetidin-1- 4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxa Bicyclo[3.2.1]octane-3-yl]acetate
  • Step 4 (2S)-N-(2-Dimethylaminoethyl)-1-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S) -2,3,4-trihydroxy-1-(1-hydroxy-1-methyl-B 6,6-Dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butanoyl]azetidin-2-carboxamide
  • Step 1 (2S)-2-(2-Dimethylaminoethylcarbamoyl)pyrrolidine-1-carboxylic acid benzyl ester
  • Step 4 [(1S,2S,3S,4R,5S)-2,4-Diacetoxy-5-[3-[[4-[(2S)-2-(2-dimethylaminoethyl) Carbamoyl)pyrrolidin-1-yl]-4- Oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxabicyclo[3.2.1 Octane-3-yl]acetate
  • Step 5 (2S)-N-(2-Dimethylaminoethyl)-1-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S)) -2,3,4-trihydroxy-1-(1-hydroxy-1-methyl-B 6,6-Dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyryl]pyrrolidine-2-carboxamide
  • Step 1 (2S,4S)-2-(2-Dimethylaminoethylcarbamoyl)-4-hydroxy-pyrrolidine-1-carboxylic acid benzyl ester
  • Step 4 [(1S, 2S, 3S, 4R, 5S)-2,4-diacetoxy-5-[3-[[4-[4-[(2S,4S)-2-(2-) Methylaminoethylcarbamoyl)-4-hydroxy-pyrrole Alkyl-1-yl]-4-oxo-butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8 -dioxabicyclo[3.2.1]octane-3-yl]] Acetate
  • Step 5 (2S,4S)-N-(2-Dimethylaminoethyl)-4-hydroxy-1-[4-[4-[[2-methyl-5-[(1S,2S,3S) ,4R,5S)-2,3,4-trihydroxy-1-(1-hydroxy-1- Methyl-ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyryl]pyrrolidine-2-carboxamide
  • Step 2 (2S,4R)-2-(2-Dimethylaminoethylcarbamoyl)-4-fluoro-pyrrolidine-1-carboxylic acid benzyl ester
  • Step 6 (2S,4R)-N-(2-Dimethylaminoethyl)-4-fluoro-1-[4-[4-[[2-methyl-5-[(1S,2S,3S) ,4R,5S)-2,3,4-trihydroxy-1-(1-hydroxy-1- Methyl-ethyl)-6,8-dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyryl]pyrrolidine-2-carboxamide
  • Step 1 (2S)-2-(2-Dimethylaminoethylcarbamoyl)piperidine-1-carboxylic acid tert-butyl ester
  • Step 3 [(1S, 2S, 3S, 4R, 5S)-2,4-Diacetoxy-5-[3-[[4-[(2S)-2-(2-dimethylaminoethyl) Carbamoyl)piperidin-1-yl]-4-oxo ⁇ -butyl]phenyl]methyl]-4-methyl-phenyl]-1-(1-hydroxy-1-methyl-ethyl)-6,8-dioxabicyclo[3.2.1] Octane-3-yl]acetate
  • Step 4 (2S)-N-(2-Dimethylaminoethyl)-1-[4-[4-[[2-methyl-5-[(1S,2S,3S,4R,5S) -2,3,4-trihydroxy-1-(1-hydroxy-1-methyl-B 6,6-Dioxabicyclo[3.2.1]octane-5-yl]phenyl]methyl]phenyl]butyryl]piperidine-2-carboxamide
  • ⁇ -methyl glucoside was purchased from Sigma, Cat. No. M9376-100G;
  • N-methyl-D-glucosamine was purchased from Sigma, Cat. No. M2004-100G;
  • Root bark was purchased from Sigma, Cat. No. P3449-1G;
  • a 96-well cell culture plate was purchased from Corning, Cat. No. 3903.
  • test results show that the compound of the present invention has a significant inhibitory effect on SGLT1.
  • the following method is used to determine the effect of the compounds of the invention on improving oral glucose tolerance and promoting urinary glucose excretion.
  • mice C57BL/6 mice were fasted for 15 hours overnight, and the fasting blood glucose concentration was measured.
  • the blood glucose concentration was determined according to body weight and fasting blood glucose.
  • the respective test groups were given a single intragastric administration of the test compound at a dose of 1 mg/kg.
  • the blank control group was given a vehicle, and the blood glucose level (ie, 0 hour blood glucose) was detected after 15 minutes of administration. After the blood glucose was detected at 0, the mice in each group were immediately given a single gavage of glucose (2.5 g/kg), 15 min after the sugar administration.
  • each group of animals was placed in a metabolic cage, and urine was collected in the metabolic cage for 2.25-6 hours and 6-24 hours after administration, and the urine volume at each time point was recorded. During the collection process, free diet and drinking water were collected. The urine was collected and centrifuged to obtain the supernatant. The Roche automatic biochemical analyzer was used to detect the urine sugar concentration of C57BL/6 mice at each time point.
  • mice in each group were immediately given a single gavage of glucose (4.0 g/kg), 15 min after the sugar administration. Blood was collected from the tail vein at 30 min and 60 min. The blood glucose meter continuously detected the blood glucose concentration of SD rats, and the decrease rate of the area under the blood glucose curve (AUC Glu 0-60 min) within 60 min after the sugar load was calculated.
  • each group of animals was placed in a metabolic cage, and urine was collected in the metabolic cage for 1.5-24 hours and 24-48 hours after administration, and the urine volume at each time point was recorded. During the collection process, free diet and drinking water were collected. The urine was collected and centrifuged to obtain the supernatant. The Roche automatic biochemical analyzer was used to detect the urine glucose concentration of SD rats at each time point.
  • test results show that the compound of the present invention is effective in lowering blood sugar levels.
  • test results show that the compound of the present invention has a remarkable effect in promoting urinary glucose excretion.
  • test rats were weighed overnight for 15 hours, weighed, and randomized according to body weight, and the test compound was dissolved in 5% DMSO + 5% Koliphor HS 15 + 90% Saline vehicle.
  • test animals were administered at a dose of 1 mg/kg, 2 mg/kg or 5 mg/kg; for the test group administered orally, the test animals were given a dose of 5 mg/kg.
  • venous blood about 0.2 mL was taken at 0.083 hours before administration and at a time point after administration of 0.083 (intravenous group only), 0.25, 0.5, 1.0, 2.0, 5.0, 7.0, and 24 hours, and placed in EDTAK.
  • test results show that the compound provided by the present invention exhibits excellent pharmacokinetic properties when administered intravenously or orally.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Diabetes (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Obesity (AREA)
  • Genetics & Genomics (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明涉及吡喃葡萄糖基衍生物及其用途。具体地,本发明涉及一种作为钠依赖性葡萄糖转运蛋白(SGLTs)抑制剂,尤其作为钠依赖性葡萄糖转运蛋白-1(SGLT1)抑制剂的吡喃葡萄糖基衍生物及其药学上可接受的盐或立体异构体,进一步涉及含有该衍生物的药物组合物。本发明还涉及所述化合物及其药物组合物用于制备治疗糖尿病和糖尿病相关疾病的药物的用途。

Description

吡喃葡萄糖基衍生物及其用途
相关申请的交叉引用
本申请要求在2018年01月31日提交的中国专利申请号201810093154.8享有优先权,该专利通过引用被全部合并于此。
技术领域
本发明属于医药领域,具体涉及一种作为钠依赖性葡萄糖转运蛋白(SGLTs)抑制剂,尤其作为钠依赖性葡萄糖转运蛋白1(SGLT1)抑制剂的吡喃葡萄糖基衍生物、制备他们的方法、包含所述衍生物的药物组合物及所述衍生物和其组合物的用途。更具体地说,是通式(I)所示的化合物或者其在药学上可接受的盐或其立体异构体或含有该化合物的药物组合物和所述的化合物及药物组合物用于制备治疗糖尿病和糖尿病相关疾病的药物的用途。
背景技术
糖尿病是一种常见的以高血糖为特征的慢性疾病,糖尿病的发生伴随着外周组织的胰岛素抵抗、体内胰岛素分泌减少以及肝脏糖异生作用的增加。当无法通过饮食和运动的方法来有效地控制病症时,需要另外使用胰岛素或者口服降血糖药来治疗。目前的降血糖药包括双胍类、磺酰脲类、胰岛素增敏剂、格列奈类、α-葡萄糖苷酶抑制剂以及DPP-IV(二肽基肽酶IV)抑制剂等。然而,目前这些降血糖药都存在欠缺,双胍类会引起乳酸中毒,磺酰脲类会引起严重的低血糖,格列奈类使用不当也会引起低血糖,胰岛素增敏剂会造成水肿、心脏衰竭和体重增加,α-葡萄糖苷酶抑制剂会造成腹部胀气和下痢,DPP-IV抑制剂需要和二甲双胍联合用药才能达到理想的降糖效果。因此,迫切需要开发更安全有效的新型降血糖药。
研究发现,葡萄糖转运蛋白是一类镶嵌在细胞膜上转运葡萄糖的载体蛋白质,葡萄糖必须借助葡萄糖转运蛋白才能通过细胞膜的脂质双层结构。葡萄糖转运蛋白分两大类,一类是钠依赖性葡萄糖转运蛋白(sodium-dependent glucose transporters,SGLTs);另一类是葡萄糖转运蛋白(glucose transporters,GLUTs)。SGLTs的两个主要家族成员为SGLT1和SGLT2。SGLT1主要分布在小肠、肾脏、心脏和气管中,主要表达于小肠刷状缘和肾近曲小管的S3阶段中,少量表达于心脏和气管,以钠-葡萄糖2:1的比率转运葡萄糖和半乳糖。而SGLT2主要分布在肾脏中,主要表达于肾近曲小管的S1节段中,以钠-葡萄糖1:1的比率转运葡萄糖。在生物体里,SGLTs以主动方式逆浓度梯度转运葡萄糖,同时消耗能量,而GLUTs以易化扩散的方式顺浓度梯度转运葡萄糖,其转运过程不消耗能量。研究表明,血浆葡萄糖通常在肾脏的肾小球中过滤并有90%的葡萄糖在肾小管近端S1段被SGLT2主动转运至上皮细胞中,10%的葡萄糖在肾小管远端S3段被SGLT1主动转运至上皮细胞中,又被上皮细胞基底膜侧的GLUT转运至周围毛细管网中,完成了肾小管对葡萄糖的重吸收。因此,SGLTs是调控细胞糖代谢的第一道关卡,也是能有效治疗糖尿病的理想靶点。抑制SGLTs不会影响正常葡萄糖反调节机制而造成低血糖风险;同时通过增加肾脏葡萄糖的***来降低血糖,能促使肥胖症患者的体重下降。研究还发现,SGLTs抑制剂作用机制不依赖于胰岛β-细胞功能异常或者胰岛素抵抗的程度,因此,其效果不会随着β-细胞的功能衰竭或者严重胰岛素抵抗而下降。它可以单独使用,也可以和其他的降血糖药联合治疗,通过机制互补而更好的发挥降糖作用。因此,SGLTs抑制剂是理想的新型降血糖药。
此外,研究还发现SGLTs抑制剂可以用于糖尿病相关并发症的治疗。如视网膜病变、神经病、肾病,葡萄糖代谢紊乱造成的胰岛素耐受、高胰岛素血症、高血脂、肥胖等。同时SGLTs抑制剂亦可与现有的治疗药物联合使用,如磺酰胺、噻唑烷二酮、二甲双胍和胰岛素等,在不影响药效的情况下,降低用药剂量,从而避免或减轻了不良反应的发生,提高了患者对治疗的顺应性。
目前,研究主要聚焦于发现选择性的SGLT2抑制剂。目前在临床试验中的大多数SGLTs抑制剂,如Dapagliflozin(达格列净)、canagliflozin(卡格列净)和empagliflozin(恩格列净),均是选择性的SGLT2抑制剂。然而,最近的临床试验结果表明,SGLT1抑制剂可表现出由抑制葡萄糖重吸收所提供更大的益处(美国专利申请公开号US20110218159)。据报道,先天性SGLT1异常的病人中,存在葡萄糖和半乳糖的吸收不足,这为通过抑制SGLT1活性减少碳水化合物的吸收提供事实依据。另外,在OLETF大鼠和患有链脲 酶诱导的糖尿病症状的大鼠中,SGLT1的mRNA和蛋白增加,并且葡萄糖吸收加快。因此,阻断SGLT1活性可抑制碳水化合物如葡萄糖在小肠的吸收,随后可防止血糖水平的升高。尤其是,基于上述机理来延迟葡萄糖的吸收,可有效得使餐后高血糖正常化。此外,SGLT1抑制剂还具有提高胰高血糖素样肽-1(GLP-1)的水平(Moriya,R.等,Am JPhysiol Endorinol Metab,297:E1358-E1365(2009))。
综上所述,SGLT1抑制剂,作为新型的糖尿病治疗药物有着良好的开发前景。
发明摘要
本发明提供了一类具有显著/优异的SGLTs抑制活性,尤其是SGLT1抑制活性的化合物,用于改善肠内环境;或用于治疗糖尿病、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症,肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或高血压以及它们的并发症。本发明也提供了制备这些化合物的方法、包含这些化合物的药物组合物,以及使用这些化合物和组合物制备治疗哺乳动物,尤其是人类的上述疾病的药物的方法,与已有的同类化合物相比,本发明的化合物不仅具有更好的药理活性,还具有更优良的体内代谢动力学性质和体内药效学性质。具体而言,本发明化合物具有优良的SGLT1抑制活性,因此具有优良的降血糖及促尿糖***效果。因此,本发明提供的化合物相对于目前已有的同类化合物而言,具有更优良的成药性。
一方面,本发明涉及一种化合物,其为式(I)所示化合物或式(I)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药,
Figure PCTCN2019073550-appb-000001
其中,
L为-(CR aR b) q-、-CH=CH-(CR aR b) p-、-O-(CR aR b) p-、-NH-(CR aR b) p-、-S-(CR aR b) p-、-S(=O)-(CR aR b) p-或-S(=O) 2-(CR aR b) p-;
q为1、2、3、4、5或6;
p为0、1、2、3、4、5或6;
R a和R b各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基或3-6个原子组成的杂环基;或R a、R b和与它们相连的碳原子一起形成C 3-6碳环或3-6个原子组成的杂环;
R 1为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6羟基烷基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 1-6烷氨基、C 1-6烷硫基、C 3-6环烷基或C 3-6环烷基-C 1-4亚烷基,其中,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6羟基烷基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 1-6烷氨基、C 1-6烷硫基、C 3-6环烷基和C 3-6环烷基-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 2和R 3各自独立地为H、D、CN、OH、NH 2、-SH、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6烷硫基、C 1-6烷氨基、C 3-6环烷基或3-6个原子组成的杂环基,其中,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6烷硫基、C 1-6烷氨基、C 3-6环烷基和3-6个原子组成的杂环基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R 2、R 3和与它们相连的碳原子一起形成羰基;
或R 2、R 3和与它们相连的碳原子一起形成C 3-6碳环或3-6个原子组成的杂环,其中,所述C 3-6碳环和3-6个原子组成的杂环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、 F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 4为H、D、-OR 4a或-SR 4b
R 4a和R 4b各自独立地为H、D、C 1-6烷基、C 3-6环烷基、C 3-6环烷基-C 1-4亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-4亚烷基,其中,所述C 1-6烷基、C 3-6环烷基、C 3-6环烷基-C 1-4亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 10为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-6烷基或C 1-6烷氧基;
R 5为H、D、C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基或5-8个原子组成的杂芳基,其中,所述C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基和5-8个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基或5-8个原子组成的杂芳基,其中,所述C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基和5-8个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
或R 6、R 7和与它们相连的碳原子一起形成C 3-8碳环、3-8个原子组成的杂环、C 6-10芳环或5-8个原子组成的杂芳环,其中,所述C 3-8碳环、3-8个原子组成的杂环、C 6-10芳环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 8和R 9各自独立为H、D、R eO-C 1-4亚烷基、R dR cN-C 1-4亚烷基、C 1-8烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基或(5-8个原子组成的杂芳基)-C 1-4亚烷基,其中所述C 1-8烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基和(5-8个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R 8、R 9和与它们相连的氮原子一起形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所 取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R c、R d、R e和R f各自独立地为H、D、C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基或(5-8个原子组成的杂芳基)-C 1-4亚烷基,其中,所述C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基和(5-8个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R c、R d和与它们相连的氮原子一起形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、OH、-NH 2、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
n为0、1、2或3;
t为0、1、2、3、4、5或6;条件是,当R 5和R 8同时为H,且R 9
Figure PCTCN2019073550-appb-000002
时,t不为0。
在另一些实施方案中,本发明涉及一种式(II)所示结构的化合物或其立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药,
Figure PCTCN2019073550-appb-000003
其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、n和t具有本发明所述的定义。
在另一些实施方案中,R 1为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、甲基、乙基、正丙基、异丙基、丙烯基、丙炔基、甲氧基、乙氧基、羟基甲基、三氟甲基、三氟乙基、一氟甲基、三氟甲氧基、二氟甲氧基、甲氨基、环丙基、环丁基、环戊基、环己基或环丙基-亚甲基,其中,所述甲基、乙基、正丙基、异丙基、丙烯基、丙炔基、甲氧基、乙氧基、羟基甲基、三氟乙基、一氟甲基、二氟甲氧基、甲氨基、环丙基、环丁基、环戊基、环己基和环丙基-亚甲基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在一些实施方案中,R 2和R 3各自独立地为H、D、CN、OH、NH 2、-SH、甲基、乙基、正丙基、异丙基、乙烯基、丙炔基、甲氧基、乙氧基、甲硫基、甲氨基、环丙基、环丁基、环戊基、环己基或5-6个原子组成的杂环基,其中,所述甲基、乙基、正丙基、异丙基、乙烯基、丙炔基、甲氧基、乙氧基、甲硫基、甲氨基、环丙基、环丁基、环戊基、环己基和5-6个原子组成的杂环基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
或R 2、R 3和与它们相连的碳原子一起形成羰基;
或R 2、R 3和与它们相连的碳原子一起形成环丙烷、环丁烷、环戊烷、环己烷或5-6个原子组成的杂环,其中,所述环丙烷、环丁烷、环戊烷、环己烷和5-6个原子组成的杂环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在一些实施方案中,R 4为H、D、-OR 4a或-SR 4b
R 4a和R 4b各自独立地为H、D、甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 10为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、甲基、乙基、正丙基、异丙基、甲氧基或乙氧基。
在另一些实施方案中,R 5为H、D、C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基,其中,所述C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基和5-6个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基,其中,所述C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基和5-6个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
或R 6、R 7和与它们相连的碳原子一起形成C 3-6碳环、5-6个原子组成的杂环、C 6-10芳环或5-6个原子组成的杂芳环,其中,所述C 3-6碳环、5-6个原子组成的杂环、C 6-10芳环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基;
或R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 5为H、D、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、三氟甲基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基或嘧啶基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、 吡嗪基、哒嗪基和嘧啶基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基或苯基;
R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、甲氧基、乙氧基、甲氨基、三氟甲基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基或嘧啶基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、甲氧基、乙氧基、甲氨基、二氟甲氧基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基和嘧啶基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基或苯基;
或R 6、R 7和与它们相连的碳原子一起形成环丙烷、环丁烷、环戊烷、环己烷、吡咯烷、四氢呋喃、四氢噻吩、四氢吡喃、四氢噻喃、哌啶、吗啉、硫代吗啉、哌嗪、苯环、呋喃、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、噻吩、吡嗪、哒嗪或嘧啶,其中,所述环丙烷、环丁烷、环戊烷、环己烷、吡咯烷、四氢呋喃、四氢噻吩、四氢吡喃、四氢噻喃、哌啶、吗啉、硫代吗啉、哌嗪、苯环、呋喃、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、噻吩、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
或R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 8和R 9各自独立为H、D、R eO-C 1-4亚烷基、R dR cN-C 1-4亚烷基、C 1-6烷基、、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中所述C 1-6烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基;
或R 8、R 9和与它们相连的氮原子一起形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 8和R 9各自独立为H、D、R dR cN-C 1-4亚烷基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、正戊基、正己基、三氟甲基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、C 6-10芳基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、正戊基、正己基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、C 6-10芳基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OH、-NR cR d、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
或R 8、R 9和与它们相连的氮原子一起形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶,各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OH、-NR cR d、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在一些实施方案中,R c、R d、R e和R f各自独立地为H、D、C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基;
或R c、R d和与它们相连的氮原子一起形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、OH、-NH 2、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R c、R d、R e和R f各自独立地为H、D、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、三氟甲基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3 或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
或R c、R d和与它们相连的氮原子一起形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
另一方面,本发明涉及一种药物组合物,其包含本发明所述的化合物。
在一些实施方案中,本发明所述的药物组合物,其进一步包含药学上可接受的载体、赋形剂、辅剂、媒介物或它们的组合。
在一些实施方案中,本发明所述的药物组合物,进一步包含一种或多种其他附加治疗剂,其中所述其他附加治疗剂选自抗糖尿病药物、抗高血糖药物、抗肥胖症药物、抗高血压药物、食欲抑制药物、降脂药物或其组合。
在另一些实施方案中,本发明所述的抗糖尿病药物和抗高血糖药物分别独立地选自SGLT2抑制剂、双胍类药物、磺酰脲类药物、葡萄糖苷酶抑制剂、PPAR激动剂(过氧化物酶体增殖物激活受体激动剂)、αP2抑制剂(脂肪细胞脂肪酸结合蛋白抑制剂)、PPARα/γ双激活剂(过氧化物酶体增殖物激活受体α/γ双激活剂)、二肽酰肽酶IV抑制剂、格列奈类药物、胰岛素、胰高血糖素样肽-1抑制剂、PTP1B抑制剂(蛋白酪氨酸磷酸酶1B抑制剂)、糖原磷酸化酶抑制剂、葡萄糖-6-磷酸酶抑制剂或其组合。
在另一些实施方案中,本发明所述的抗肥胖症药物选自中枢抗肥胖药、MCH(黒色素浓集激素)受体拮抗剂、神经肽Y受体拮抗剂、***素(cannabinoid)受体拮抗剂、脑肠肽拮抗剂、脂肪酶抑制剂、β3激动剂、11β-HSD1(11β羟基类固醇脱氢酶1)抑制剂、DGAT-1(二酰甘油酰基转移酶1)抑制剂、肽性食欲抑制剂、缩胆囊肽(Cholecystokinin)激动剂、摄食抑制剂或其组合。
在另一些实施例中,本发明所述降脂药物选自MTP抑制剂(微粒体甘油三酯转移蛋白抑制剂)、HMGCoA还原酶抑制剂(羟甲基戊二酰辅酶A还原酶抑制剂)、角鲨烯合成酶抑制剂、贝丁酸类降血脂药物(又称贝特类降血脂药物)、ACAT抑制剂(乙酰胆固醇乙酰转移酶抑制剂)、脂加氧酶抑制剂、胆固醇吸收抑制剂、回肠钠离子/胆汁酸协同转运蛋白抑制剂、LDL受体活性的向上调节物、烟酸类降血脂药物、胆汁酸螯合物或其组合。
在又一些实施例中,本发明所述降脂药物选自普伐他汀、辛伐他汀、阿伐他汀、氟伐他汀、西立伐他汀、埃塔伐他汀、罗素他汀或其组合。
另一方面,本发明涉及本发明所述的化合物或本发明所述的药物组合物在制备药物中的用途,其中,所述药物用于抑制SGLT1。
另一方面,本发明涉及本发明所述的化合物或本发明所述的药物组合物在制备药物中的用途,其中,所述药物用于改善肠内环境。
另一方面,本发明涉及本发明所述的化合物或所述的药物组合物在制备药物中的用途,其中,所述药物用于预防或治疗疾病,减轻所述疾病症状或者延缓所述疾病的发展或发作,其中所述疾病是糖尿病,糖尿病并发症,胰岛素抵抗,高血糖,高胰岛素血症,高脂血症,肥胖症,X综合症,动脉粥样硬化,心血管疾病,充血性心力衰竭,低镁血症,低钠血症,肾衰竭,与血浓缩相关的障碍、便秘或者高血压。
另一方面,本发明涉及一种抑制SGLT1的方法,包括给患者使用有效量的本发明所述的化合物或本发明所述的药物组合物。
另一方面,本发明涉及一种改善肠内环境方法,包括给患者使用有效量的本发明所述的化合物或本发明所述的药物组合物。
另一方面,本发明涉及一种预防或治疗疾病、减轻所述疾病症状或者延缓所述疾病的发展或发作的方法,包括给患者使用有效量的本发明所述的化合物或本发明所述的药物组合物,其中所述疾病为所述疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压。
另一方面,本发明涉及本发明所述化合物或或本发明所述的药物组合物用于抑制SGLT1的用途。
另一方面,本发明涉及本发明所述化合物或或本发明所述的药物组合物用于改善肠内环境的用途。
另一方面,本发明涉及本发明所述化合物或或本发明所述的药物组合物用于预防或治疗疾病、减轻所述疾病症状或者延缓所述疾病的发展或发作的用途,其中所述疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压。
在一些实施方案中,本发明所述糖尿病并发症为糖尿病性视网膜病、糖尿病性神经病或糖尿病性肾病。
在一些实施方案中,本发明所述高脂血症为高甘油三酯血症。
前面所述内容只概述了本发明的某些方面,但不限于这些方面。这些方面及其他的方面的内容将在下面更加具体完整的描述。
本发明详细说明书
本发明提供了一类吡喃葡萄糖基衍生物、其制备方法及其在医药上的应用,本领域技术人员可以借鉴本文内容,适当改进工艺参数实现。特别需要指出的是,所有类似的替换和改动对本领域技术人员来说是显而易见的,它们都被视为包括在本发明的范围中。
定义和一般术语
现在详细描述本发明的某些实施方案,其实例由随附的结构式和化学式说明。本发明意图涵盖所有的替代、修改和等同技术方案,它们均包括在本发明范围内。本领域技术人员应认识到,许多与本文所述类似或等同的方法和材料能够用于实践本发明。本发明绝不限于本文所述的方法和材料,在所结合的文献、专利和类似材料的一篇或多篇与本申请不同或相矛盾的情况下(包括但不限于所定义的术语、术语应用、所描述的技术,等等),以本申请为准。
应进一步认识到,本发明的某些特征,为清楚可见,在多个独立的实施方案中进行了描述,但也可以在单个实施例中以组合形式提供。反之,本发明的各种特征,为简洁起见,在单个实施方案中进行了描述,但也可以单独或以任意适合的子组合提供。
除非另外说明,本发明所使用的所有科技术语具有与本发明所属领域技术人员的通常理解相同的含义。本发明涉及的所有专利和公开出版物通过引用的方式整体并入本发明。除非另外说明,应当应用本文所使用的下列定义。出于本发明的目的,化学元素与元素周期表CAS版,和《化学和物理手册》,第75版,1994一致。此外,有机化学一般原理可参考"Organic Chemistry",Thomas Sorrell,University Science Books,Sausalito:1999和"March's Advanced Organic Chemistry"by Michael B.Smith and Jerry March,John Wiley & Sons,New York:2007中的描述,其全部内容通过引用并入本文。
除非另有说明或者上下文中有明显的冲突,本文所使用的冠词“一”、“一个(种)”和“所述”旨在包括“至少一个”或“一个或多个”。因此,本文所使用的这些冠词是指一个或多于一个(即至少一个)宾语的冠词。例如,“一组分”指一个或多个组分,即可能有多于一个的组分被考虑在所述实施方案的实施方式中采用或使用。
除非另有说明,本发明所用在说明书和权利要求书中的术语具有下述定义。
术语“包含”为开放式表达,即包括本发明所指明的内容,但并不排除其他方面的内容。
像本发明所描述的,本发明的化合物可以任选地被一个或多个取代基所取代,如上面的通式化合物,或者像实施例里面特殊的例子,子类,和本发明所包含的一类化合物。应了解术语“任选取代的”、“未被取代或被……取代基所取代”与“取代或非取代的”这个术语可以交换使用。术语“任选地”,“任选的”或“任选”是指随后所述的事件或状况可以但未必发生,并且该描述包括其中发生该事件或状况的情况,以及其中未 发生该事件或状况的情况。一般而言,术语“任选地”不论是否位于术语“取代的”之前,都表示所给结构中的一个或多个氢原子未被取代或被具体取代基所取代。除非其他方面表明,一个任选的取代基团可以在基团各个可取代的位置进行取代。当所给出的结构式中不只一个位置能被选自具体基团的一个或多个取代基所取代,那么取代基可以相同或不同地在各个位置取代。其中所述的取代基可以是,但并不限于,D、F、Cl、Br、I、CN、NO 2、OH、NH 2、氧代(=O)、-C(=O)OH、-C(=O)NH 2、-SH、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、烷基、卤代烷基、烷氧基、烷氨基、烷硫基、卤代烷氧基、烯基、炔基、羟基烷基、环烷基、环烷基-亚烷基、碳环基、碳环基-亚烷基、杂环基、杂环基-亚烷基、芳基、芳基-亚烷基、杂芳基或杂芳基-亚烷基,其中所述的R c、R d、R e和R f具有本发明所述的定义。
另外,需要说明的是,除非以其他方式明确指出,在本发明中所采用的描述方式“各……独立(地)为”与“……各自独立(地)为”和“……独立(地)为”可以互换,均应做广义理解,其既可以是指在不同基团中,相同符号之间所表达的具体选项之间互相不影响,也可以表示在相同的基团中,相同符号之间所表达的具体选项之间互相不影响。
在本说明书的各部分,本发明公开化合物的取代基按照基团种类或范围公开。特别指出,本发明包括这些基团种类和范围的各个成员的每一个独立的次级组合。例如,术语“C 1-8烷基”特别指独立公开的C 1烷基(甲基)、C 2烷基(乙基)、C 3烷基、C 4烷基、C 5烷基、C 6烷基、C 7烷基和C 8烷基;“C 3-8环烷基”指独立公开的C 3环烷基(环丙基)、C 4环烷基(环丁基)、C 5环烷基(环戊基)、C 6环烷基(环己基)、C 7环烷基(环庚基)和C 8环烷基(环辛基);“3-8个原子组成的杂环(基)”指独立公开的3个原子组成的杂环、4个原子组成的杂环、5个原子组成的杂环、6个原子组成的杂环、7个原子组成的杂环和8个原子组成的杂环。
在本发明书的各部分,描述了连接取代基。当该结构清楚地需要连接基团时,针对该基团所列举的马库什变量应理解为连接基团。例如,如果该结构需要连接基团并且针对该变量的马库什基团定义列举了“烷基”或“芳基”,则应当理解,该“烷基”或“芳基”分别代表连接的亚烷基基团或亚芳基基团。
本发明使用的术语“烷基”或“烷基基团”,表示含1-20个碳原子的饱和直链或支链的一价碳氢化合物原子团。除非另外详细说明,烷基基团含有1-20个碳原子;其中一些实施例是,烷基基团含有1-12个碳原子;其中一些实施例是,烷基基团含有1-10个碳原子;另外一些实施例是,烷基基团含有1-9个碳原子;另外一些实施例是,烷基基团含有1-8个碳原子;另外一些实施例是,烷基基团含有1-6个碳原子,即C 1-6烷基;另外一些实施例是,烷基基团含有1-4个碳原子,即C 1-4烷基;另外一些实施例是,烷基基团含有1-3个碳原子,即C 1-3烷基;另外一些实施例是,烷基基团含有1-2个碳原子,即C 1-2烷基。
烷基基团的实例包含,但并不限于,甲基(Me、-CH 3),乙基(Et、-CH 2CH 3),正丙基(n-Pr、-CH 2CH 2CH 3),异丙基(i-Pr、-CH(CH 3) 2),正丁基(n-Bu、-CH 2CH 2CH 2CH 3),异丁基(i-Bu、-CH 2CH(CH 3) 2),仲丁基(s-Bu、-CH(CH 3)CH 2CH 3),叔丁基(t-Bu、-C(CH 3) 3),正戊基(-CH 2CH 2CH 2CH 2CH 3),2-戊基(-CH(CH 3)CH 2CH 2CH 3),3-戊基(-CH(CH 2CH 3) 2),2-甲基-2-丁基(-C(CH 3) 2CH 2CH 3),3-甲基-2-丁基(-CH(CH 3)CH(CH 3) 2),3-甲基-1-丁基(-CH 2CH 2CH(CH 3) 2),2-甲基-1-丁基(-CH 2CH(CH 3)CH 2CH 3),正己基(-CH 2CH 2CH 2CH 2CH 2CH 3),2-己基(-CH(CH 3)CH 2CH 2CH 2CH 3),3-己基(-CH(CH 2CH 3)(CH 2CH 2CH 3)),2-甲基-2-戊基(-C(CH 3) 2CH 2CH 2CH 3),3-甲基-2-戊基(-CH(CH 3)CH(CH 3)CH 2CH 3),4-甲基-2-戊基(-CH(CH 3)CH 2CH(CH 3) 2),3-甲基-3-戊基(-C(CH 3)(CH 2CH 3) 2),2-甲基-3-戊基(-CH(CH 2CH 3)CH(CH 3) 2),2,3-二甲基-2-丁基(-C(CH 3) 2CH(CH 3) 2),3,3-二甲基-2-丁基(-CH(CH 3)C(CH 3) 3),正庚基,正辛基,等等。其中所述烷基基团可以任选地被一个或多个本发明所述的取代基所取代。
本发明所使用的术语“烷基”和其前缀“烷”,都包含直链和支链的饱和碳链。
术语“亚烷基”表示从饱和的直链或支链烃基中去掉两个氢原子所得到的饱和的二价烃基基团。除非另外详细说明,亚烷基基团含有1-12个碳原子。在一些实施方案中,亚烷基基团含1-8个碳原子;在另一些实施方案中,亚烷基基团含1-6个碳原子;在另一些实施方案中,亚烷基基团含有1-4个碳原子,即C 1-4亚烷基;在又一些实施方案中,亚烷基基团含有1-3个碳原子,即C 1-3亚烷基;还在一些实施方案中,亚烷基基团含有1-2个碳原子,即C 1-2亚烷基。这样的实例包括亚甲基(-CH 2-),亚乙基(包括-CH 2CH 2-或-CH(CH 3)-),亚异丙基(包括-CH(CH 3)CH 2-或-C(CH 3) 2-)、亚正丙基(包括-CH 2CH 2CH 2-、-CH(CH 2CH 3)- 或-CH 2CH(CH 3)-)、亚正丁基(包括-CH 2(CH 2) 2CH 2-、-CH(CH 2CH 2CH 3)-、-CH 2CH(CH 2CH 3)-、-CH 2CH 2CH(CH 3)-或-CH(CH 3)CH(CH 3)-)、亚叔丁基(包括-CH(CH(CH 3) 2)-、-CH 2CH(CH 3)CH 2-或-CH 2C(CH 3) 2-)、亚戊基(例如-CH 2(CH 2) 3CH 2-)、亚己基(例如-CH 2(CH 2) 4CH 2-)等等。其中,所述亚烷基可以任选地被一个或多个本发明所描述的取代基所取代。
术语“烯基”表示含有2-12个碳原子的直链或支链一价烃基,其中至少有一个不饱和位点为碳-碳sp 2双键,其中,所述烯基基团可以任选地被一个或多个本发明所描述的取代基所取代,其包括“cis”和“trans”的定位,或者“E”和“Z”的定位。在一些实施方案中,烯基基团包含2-8个碳原子;在另一些实施方案中,烯基基团包含2-6个碳原子,即C 2-6烯基;在又一些实施方案中,烯基基团包含2-4个碳原子,即C 2-4烯基。烯基基团的实例包括,但并不限于,乙烯基(-CH=CH 2)、丙烯基(-CH 2CH=CH 2、-CH=CHCH 3)、丁烯基(-CH=CHCH 2CH 3、-CH 2CH=CHCH 3、-CH 2CH 2CH=CH 2、-CH=C(CH 3) 2、-CH=C(CH 3) 2、-CH 2C(CH 3)=CH 2)、戊烯基(-CH 2CH 2CH 2CH=CH 2、-CH 2CH 2CH=CHCH 3、-CH 2CH 2CH=CHCH 3、-CH 2CH=CHCH 2CH 3、-CH=CHCH 2CH 2CH 3、-CH 2CH 2C(CH 3)=CH 2、-CH 2CH=C(CH 3) 2、-CH=CHCH(CH 3) 2、-C(CH 2CH 3)=CHCH 3、-CH(CH 2CH 3)CH=CH 2)等等。
术语“炔基”表示含有2-12个碳原子的直链或支链一价烃基,其中至少有一个不饱和位点为碳-碳sp三键。在一些实施方案中,炔基基团包含2-8个碳原子;在另一些实施方案中,炔基基团包含2-6个碳原子,即C 2-6炔基;在又一些实施方案中,炔基基团包含2-4个碳原子,即C 2-4炔基。炔基的实例包括,但并不限于,乙炔基(-C≡CH)、丙炔基(包括1-丙炔基(-C≡C-CH 3)和炔丙基(-CH 2C≡CH))、1-丁炔基、2-丁炔基、1-戊炔基、2-戊炔基、3-甲基-1-丁炔基、1-己炔基、1-庚炔基、1-辛炔基,等等。所述炔基基团可以独立任选地被一个或多个本发明所描述的取代基所取代。
术语“烷氧基”或“烷基氧基”是指烷基基团通过氧原子与分子其余部分相连,即烷基-O-,其中烷基基团具有如本发明所述的含义。在一些实施方案中,烷氧基基团含有1-20个碳原子;在另一些实施方案中,烷氧基基团含有1-10个碳原子;在又一些实施方案中,烷氧基基团含有1-8个碳原子;在又一些实施方案中,烷氧基基团含有1-6个碳原子,即C 1-6烷氧基;在又一些实施方案中,烷氧基基团含有1-4个碳原子,即C 1-4烷氧基;在又一些实施方案中,烷氧基基团含有1-3个碳原子,即C 1-3烷氧基;在又一些实施方案中,烷氧基基团含有1-2个碳原子,即C 1-2烷氧基。
烷氧基基团的实例包含,但并不限于,甲氧基(MeO,-OCH 3),乙氧基(EtO,-OCH 2CH 3),1-丙氧基(n-PrO,n-丙氧基,-OCH 2CH 2CH 3),2-丙氧基(i-PrO,i-丙氧基,-OCH(CH 3) 2),1-丁氧基(n-BuO,n-丁氧基,-OCH 2CH 2CH 2CH 3),2-甲基-l-丙氧基(i-BuO,i-丁氧基,-OCH 2CH(CH 3) 2),2-丁氧基(s-BuO,s-丁氧基,-OCH(CH 3)CH 2CH 3),2-甲基-2-丙氧基(t-BuO,t-丁氧基,-OC(CH 3) 3),1-戊氧基(n-戊氧基,-OCH 2CH 2CH 2CH 2CH 3),2-戊氧基(-OCH(CH 3)CH 2CH 2CH 3),3-戊氧基(-OCH(CH 2CH 3) 2),2-甲基-2-丁氧基(-OC(CH 3) 2CH 2CH 3),3-甲基-2-丁氧基(-OCH(CH 3)CH(CH 3) 2),3-甲基-l-丁氧基(-OCH 2CH 2CH(CH 3) 2),2-甲基-l-丁氧基(-OCH 2CH(CH 3)CH 2CH 3),等等,其中所述烷氧基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“烷硫基”是指烷基基团通过硫原子与分子其余部分相连,即烷基-S-,其中烷基基团具有如本发明所述的含义。在一些实施方案中,烷硫基基团含有1-6个碳原子,即C 1-6烷硫基;在另一些实施方案中,烷硫基基团含有1-4个碳原子,即C 1-4烷硫基;在又一些实施方案中,烷硫基基团含有1-3个碳原子,即C 1-3烷硫基;在又一些实施方案中,烷硫基基团含有1-2个碳原子,即C 1-2烷硫基。烷硫基的实例包括,但并不限于甲硫基、乙硫基,等等。所述烷硫基基团可以任选地被一个或多个本发明描述的取代基所取代。
术语“烷氨基”或“烷基氨基”包括“N-烷氨基”和“N,N-二烷氨基”,其中氨基基团分别独立地被一个或两个烷基基团所取代。其中一些实施例是,烷氨基是一个或两个C 1-6烷基连接到氮原子上的较低级的烷基氨基基团。另外一些实施例是,烷氨基是C 1-4的较低级的烷基氨基基团。另外一些实施例是,烷氨基是C 1-3的较低级的烷基氨基基团。另外一些实施例是,烷氨基是C 1-2的较低级的烷基氨基基团。合适的烷氨基基团可以是单烷氨基或二烷氨基,这样的实例包括,但并不限于,甲氨基(也称N-甲氨基),乙氨基(也称N-乙氨基),N,N-二甲氨基,N,N-二乙氨基等等。其中所述烷氨基基团可以独立地未被取代或被一个或多个 本发明所描述的取代基所取代。
术语“卤代烷基”是指具有一个或者多个卤素取代基的烷基。在一些实施方案中,卤代烷基基团含有1-10个碳原子,在另一些实施方案中,卤代烷基基团含有1-8个碳原子,在又一些实施方案中,卤代烷基基团含有1-6个碳原子,即C 1-6卤代烷基;在又一些实施方案中,卤代烷基基团含有1-4个碳原子,即C 1-4卤代烷基;还在一些实施方案中,卤代烷基基团含有1-3个碳原子,即C 1-3卤代烷基;还在一些实施方案中,卤代烷基基团含有1-2个碳原子,即C 1-2卤代烷基。卤代烷基的实例包括,但并不限于一氟甲基(-CH 2F)、二氟甲基(-CHF 2)、三氟甲基(-CF 3)、氟乙基(-CHFCH 3,-CH 2CH 2F)、二氟乙基(-CF 2CH 3,-CFHCFH 2,-CH 2CHF 2)、全氟乙基、氟丙基(-CHFCH 2CH 3,-CH 2CHFCH 3,-CH 2CH 2CH 2F)、二氟丙基(-CF 2CH 2CH 3,-CFHCFHCH 3,-CH 2CH 2CHF 2,-CH 2CF 2CH 3,-CH 2CHFCH 2F)、三氟丙基、1,1-二氯乙基、1,2-二氯丙基等。所述卤代烷基基团可以任选地被一个或多个本发明描述的取代基所取代。
术语“卤代烷氧基”是指烷氧基基团被一个或多个卤素取代基所取代。在一些实施方案中,卤代烷氧基基团含有1-10个碳原子,在另一些实施方案中,卤代烷氧基基团含有1-8个碳原子;在又一些实施方案中,卤代烷氧基基团含有1-6个碳原子,即C 1-6卤代烷氧基;在又一些实施方案中,卤代烷氧基基团含有1-4个碳原子,即C 1-4卤代烷氧基;还在一些实施方案中,卤代烷氧基基团含有1-3个碳原子,即C 1-6卤代烷氧基;还在一些实施方案中,卤代烷基基团含有1-2个碳原子,即C 1-2卤代烷氧基。卤代烷氧基的实例包括,但并不限于三氟甲氧基、二氟甲氧基等。所述卤代烷氧基基团可以任选地被一个或多个本发明描述的取代基所取代。
术语“羟基烷基”或“羟烷基”是指具有一个或多个羟基取代基的烷基,其中烷基基团具有如本发明所述的含义。在一些实施方案中,羟基烷基基团含有1-6个碳原子,即C 1-6羟基烷基;在另一些实施方案中,羟基烷基基团含有1-4个碳原子,即C 1-4羟基烷基;在又一些实施方案中,羟基烷基基团含有1-3个碳原子,即C 1-3羟基烷基;在又一些实施方案中,羟基烷基基团含有1-2个碳原子,即C 1-2羟基烷基。羟基烷基的实例包括,但并不限于羟基甲基、2-羟基乙基(-CH 2CH 2OH)、1-羟基乙基(-CHOHCH 3)、1,2-二羟基乙基(-CHOHCH 2OH)、2,3-二羟基丙基(-CH 2CHOHCH 2OH)、1-羟基丙基(-CH 2CH 2CH 2OH)、2-羟基丙基、3-羟基丙基、羟丁基,等等。所述羟基烷基基团可以任选地被一个或多个本发明描述的取代基所取代。
术语“环烷基”是指有一个或多个连接点连接到分子的其余部分,饱和的,含有3-12个环碳原子的单环,双环或三环体系。其中,在一些实施方案中,环烷基是含3-10个环碳原子的环体系;在另一些实施方案中,环烷基是含3-8个环碳原子的环体系,即C 3-8环烷基;在另一些实施方案中,环烷基是含5-8个环碳原子的环体系;在另一些实施方案中,环烷基是含3-6个环碳原子的环体系,即C 3-6环烷基;在另一些实施例,环烷基是含5-6个环碳原子的环体系。环烷基的实例包括,但不限于:环丙基、环丁基、环戊基和环己基。所述环烷基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“碳环基”可以单独使用或作为“碳环基烷基”或“碳环基烷氧基”的一大部分,是指饱和或含有一个或多个不饱和单元、含有3-14个环碳原子的非芳香族碳环体系,且不含任何芳香环。术语“碳环”、“碳环基”或“碳环的”在此处可交换使用。在一些实施方案中,碳环的环碳原子的数量为3-12个;在另一些实施方案中,碳环的环碳原子的数量为3-10个;在其它一些实施方案中,碳环的环碳原子的数量为3-8个,即C 3-8碳环(基);在其它一些实施方案中,碳环的环碳原子的数量为3-6个,即C 3-6碳环(基);在其它一些实施方案中,碳环的环碳原子的数量为5-6个;在其它一些实施方案中,碳环的环碳原子的数量为5-8个。在其它一些实施方案中,碳环的环碳原子的数量为6-8个。此“碳环基”包括单环、双环或多环稠合、螺式或桥连碳环环系,还包括其中碳环可与一个或多个非芳香族碳环或一个或多个芳环或其组合稠合的多环环系,其中连接的原子团或点在碳环上。双环碳环基包括桥连双环碳环基、稠合双环碳环基和螺双环碳环基,“稠合”双环环系包含两个共用2个相邻环原子的环。桥连双环基团包括两个共用2、3或4个相邻环原子的环。螺环环系共用1个环原子。合适的碳环基团包括,但并不限于,环烷基,环烯基和环炔基。碳环基团的实例进一步包括,但绝不限于,环丙基,环丁基,环戊基,1-环戊基-1-烯基,1-环戊基-2-烯基,1-环戊基-3-烯基,环己基,1-环己基-1-烯基,1-环己基-2-烯基,1-环己基-3-烯基,环己二烯基,环庚基,环辛基,环壬基,环癸基,环十一烷基,环十二烷基,等等。桥连碳环基基团包括但不限于,二环[2.2.2]辛基,二 环[2.2.1]庚基,二环[3.3.1]壬基,二环[3.2.3]壬基,等等。所述碳环基基团可以任选地被一个或多个本发明描述的取代基所取代。
术语“杂环基”可以单独使用或作为“杂环基烷基”或“杂环基烷氧基”的一大部分,是指包含3-12个环原子的饱和或部分不饱和的,非芳香性的单环、双环或三环体系,其中至少一个环原子选自氮、硫和氧原子,其中,所述杂环基是非芳香性的,且不含任何的芳香环,且此环体系有一个或多个连接点与分子的其余部分相连。术语“杂环基”包括单环、双环或多环稠合、螺式或桥连杂环环系。双环杂环基包括桥连双环杂环基、稠合双环杂环基和螺双环杂环基。术语“杂环基”和“杂环”在此处可交换使用。除非另有说明,杂环基可以是碳基或氮基,且-CH 2-基团可以任选被-C(=O)-替代。环的硫原子可以任选被氧化成S-氧化物。环的氮原子可以任选地被氧化成N-氧化合物。在一些实施方案中,杂环基为3-8个环原子组成的环体系;在另一些实施方案中,杂环基为3-6个环原子组成的环体系;在其他一些实施方案中,杂环基为5-7个环原子组成的环体系;在另一些实施方案中,杂环基为5-10个环原子组成的环体系;在其他一些实施方案中,杂环基为5-8个环原子组成的环体系;在其他一些实施方案中,杂环基为6-8个环原子组成的环体系;在其他一些实施方案中,杂环基为5-6个环原子组成的环体系;在其他一些实施方案中,杂环基为4个环原子组成的环体系;在其他一些实施方案中,杂环基为5个环原子组成的环体系;在其他一些实施方案中,杂环基为6个环原子组成的环体系;在其他一些实施方案中,杂环基为7个环原子组成的环体系;在其他一些实施方案中,杂环基为8个环原子组成的环体系。
杂环基的实例包括,但不限于:环氧乙烷基,氮丙啶基,氮杂环丁基,氧杂环丁基,硫杂环丁基,吡咯烷基,2-吡咯啉基,3-吡咯啉基,吡唑啉基,吡唑烷基,咪唑啉基,咪唑烷基,噁唑烷,噻唑烷,四氢呋喃基,二氢呋喃基,四氢噻吩基,二氢噻吩基,1,3-二氧环戊基,二硫环戊基,四氢吡喃基,二氢吡喃基,2H-吡喃基,4H-吡喃基,四氢噻喃基,哌啶基,吗啉基,硫代吗啉基,哌嗪基,二噁烷基,二噻烷基,噻噁烷基,高哌嗪基,高哌啶基,氧杂环庚烷基,硫杂环庚烷基,四氢吡咯基,二氢吡咯基,四氢吡啶基,四氢嘧啶基,四氢吡嗪基,四氢哒嗪基。杂环基中-CH 2-基团被-C(=O)-取代的实例包括,但不限于,2-氧代吡咯烷基,氧代-1,3-噻唑烷基,2-哌啶酮基,3,5-二氧代哌啶基,嘧啶二酮基。杂环基中硫原子被氧化的实例包括,但不限于,环丁砜基和1,1-二氧代硫代吗啉基。桥连杂环基基团包括,但不限于,2-氧杂二环[2.2.2]辛基、1-氮杂二环[2.2.2]辛基、3-氮杂二环[3.2.1]辛基,等等。所述的杂环基基团可以任选地被一个或多个本发明所描述的取代基所取代。
术语“m个原子组成的”,其中m是整数,典型地描述分子中成环原子的数目,在所述分子中成环原子的数目是m。例如,哌啶基是6个环原子组成的杂环基,而1,2,3,4-四氢萘基是10个环原子组成的碳环基基团。
术语“芳基”可以单独使用或作为“芳基烷基”或“芳基烷氧基”的一大部分,表示含有6-14个环原子,或6-12个环原子,或6-10个环原子的单环,双环,和三环的芳香性碳环体系,其中,每一个环包含3-7个环原子,且有一个或多个附着点与分子的其余部分相连。术语“芳基”可以和术语“芳环”或“芳香环”交换使用,如芳基可以包括苯基,萘基和蒽基。所述芳基基团可以独立地未被取代或被一个或多个本发明所描述的取代基所取代。
术语“杂芳基”可以单独使用或作为“杂芳基烷基”或“杂芳基烷氧基”的一大部分,表示含有5-16环原子的单环、双环和三环的芳香性体系,其中,至少一个环包含一个或多个杂原子,其中每一个环包含5-7个环原子,其中至少一个环体系是芳香族的,同时,所述杂芳基有一个或多个附着点与分子其余部分相连。除非另外说明,所述的杂芳基基团可以通过任何合理的位点(可以为CH中的C,或者NH中N)连接到分子其余部分(例如通式中的主体结构)上。当杂芳基基团存在-CH 2-基团时,所述-CH 2-基团可以任选地被-C(=O)-替代。术语“杂芳基”可以与术语“杂芳环”或“杂芳族化合物”交换使用。在一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-14个原子组成的杂芳基。在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-12个原子组成的杂芳基;在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-10个原子组成的杂芳基;在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-8个原子组成的杂芳基;在 另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-7个原子组成的杂芳基;在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5-6个原子组成的杂芳基;在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的5个原子组成的杂芳基;在另一些实施方案中,杂芳基为包含1,2,3或4个独立选自O,S和N的杂原子的6个原子组成的杂芳基。
另外一些实施例是,杂芳基包括以下的单环基团,但并不限于这些单环基团:2-呋喃基,3-呋喃基,N-咪唑基,2-咪唑基,4-咪唑基,5-咪唑基,3-异噁唑基,4-异噁唑基,5-异噁唑基,2-噁唑基,4-噁唑基,5-噁唑基,N-吡咯基,2-吡咯基,3-吡咯基,2-吡啶基,3-吡啶基,4-吡啶基,2-嘧啶基,4-嘧啶基,5-嘧啶基,哒嗪基(如3-哒嗪基),2-噻唑基,4-噻唑基,5-噻唑基,四唑基(如5H-四唑基,2H-四唑基),***基(如2-***基,5-***基,4H-1,2,4-***基,1H-1,2,4-***基,1,2,3-***基),2-噻吩基,3-噻吩基,吡唑基(如2-吡唑基和3-吡唑基),异噻唑基,1,2,3-噁二唑基,1,2,5-噁二唑基,1,2,4-噁二唑基,1,3,4-噁二唑基,1,2,3-硫代二唑基,1,3,4-硫代二唑基,1,2,5-硫代二唑基,吡嗪基,1,3,5-三嗪基;也包括以下的双或者三环基团,但绝不限于这些基团:吲哚啉基、1,2,3,4-四氢异喹啉基、苯并咪唑基,苯并呋喃基,苯并噻吩基,吲哚基(如2-吲哚基),嘌呤基,喹啉基(如2-喹啉基,3-喹啉基,4-喹啉基),异喹啉基(如1-异喹啉基,3-异喹啉基或4-异喹啉基),吩噁噻基,二苯并咪唑基,二苯并呋喃基,二苯并噻吩基。所述杂芳基基团任选地被一个或多个本发明所描述的取代基所取代。
术语“杂原子”是指O、S、N、P和Si,包括S,N和P任何氧化态的形式;伯、仲、叔胺和季铵盐的形式;或者杂环中氮原子上的氢被取代的形式,例如,N(像3,4-二氢-2H-吡咯基中的N),NH(像吡咯烷基中的NH)或NR(像N-取代的吡咯烷基中的NR,R为本发明所述的取代基)。
术语“卤素”是指F、Cl、Br、I。
术语“硝基”是指-NO 2
术语“巯基”是指-SH。
术语“羟基”是指-OH。
术语“氨基”是指-NH 2
术语“氰基”是指-CN。
术语“羧酸”或“羧基”是指-C(=O)OH。
术语“羰基”,无论是单独使用还是和其他术语连用,表示-(C=O)-。
术语“D”是指氘代,即 2H。
像本发明所描述的,画一个键将取代基连接到环的中心形成的环体系代表取代基在该环体系上任何可取代的位置都可以取代。例如,式a代表A环上任何可能被取代的位置可以任选地被n个R取代;当A环为双环结构时,R可以在任意一个环上任何可取代的位置进行取代;又例如式b代表取代基R可以在苯环上任何可能被取代的位置取代,如式b-1至b-3所示:
Figure PCTCN2019073550-appb-000004
术语“保护基团”或“PG”是指当化合物中其他官能团发生反应的时候,用来阻断或保护特定的功能性的取代基团。例如,“氨基的保护基团”是指一个取代基与氨基基团相连来阻断或保护化合物中氨基的功能性,合适的氨基保护基团包括乙酰基,三氟乙酰基,叔丁氧羰基(BOC,Boc),苄氧羰基(CBZ,Cbz)和9-芴甲 氧羰基(Fmoc)。相似地,“羟基保护基团”是指羟基的取代基用来阻断或保护羟基的功能性,合适的保护基团包括,但不限于,乙酰基、苯甲酰基、苄基、对甲氧基苄基和硅烷基等。“羧基保护基团”是指羧基的取代基用来阻断或保护羧基的功能性,一般的羧基保护基包括-CH 2CH 2SO 2Ph,氰基乙基,2-(三甲基硅烷基)乙基,2-(三甲基硅烷基)乙氧基甲基,2-(对甲苯磺酰基)乙基,2-(对硝基苯磺酰基)乙基,2-(二苯基膦基)乙基,硝基乙基,等等。对于保护基团一般的描述可参考文献:T W.Greene,Protective Groups in Organic Synthesis,John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005.
术语“离去基团”或“LG”是指在化学反应中从一较大分子中脱离的原子或官能基,是亲核取代反应与消除反应中应用的术语。在亲核取代反应中,被亲核试剂进攻的反应物称为底物,而从底物分子中带着一对电子断裂出去的原子或原子团称为离去基团。常见的离去基团例如但不限于,卤素原子、酯基、磺酸酯基、硝基、叠氮基或羟基等。
术语“药学上可接受的”是指物质或组合物必须与包含制剂的其它成分和/或用其治疗的哺乳动物化学上和/或毒理学上相容。优选地,本发明所述的“药学上可接受的”是指联邦监管机构或国家政府批准的或美国药典或其他一般认可药典上列举的在动物中、特别是人体中使用的。
术语“载体”包括任何溶剂,分散介质,包衣衣料,表面活性剂,抗氧化剂,防腐剂(例如抗细菌剂、抗真菌剂),等渗剂,盐,药物稳定剂,粘合剂,赋形剂,分散剂,润滑剂,甜味剂,调味剂,着色剂,或其组合物,这些载体都是所属技术领域技术人员的已知的(如Remington's Pharmaceutical Sciences,18th Ed.Mack Printing Company,1990,pp.1289-1329所述)。除了任意常规载体与活性成分不相容的情况外,涵盖其在治疗或药物组合物中的用途。
术语“药物组合物”表示一种或多种本文所述化合物或者其生理学上/药学上可以接受的盐或前体药物与其他化学组分的混合物,其他组分例如生理学上/药学上可以接受的载体、赋形剂、稀释剂、粘合剂、填充剂等辅料,以及抗糖尿病试剂、抗高血糖试剂、抗肥胖症试剂、抗高血压试剂、抗血小板试剂、抗动脉粥样硬化试剂或者降脂试剂等附加治疗剂。药物组合物的目的是促进化合物对生物体的给药。
本发明所使用的术语“前药”,代表一个化合物在体内转化为式(I)所示的化合物。这样的转化受前体药物在血液中水解或在血液或组织中经酶转化为母体结构的影响。本发明前体药物类化合物可以是酯,在现有的发明中酯可以作为前体药物的有苯酯类,脂肪族(C 1-24)酯类,酰氧基甲基酯类,碳酸酯,氨基甲酸酯类和氨基酸酯类。例如本发明里的一个化合物包含羟基,即可以将其酰化得到前体药物形式的化合物。其他的前体药物形式包括磷酸酯,如这些磷酸酯类化合物是经母体上的羟基磷酸化得到的。关于前体药物完整的讨论可以参考以下文献:Higuchi et al.,Pro-drugs as Novel Delivery Systems,Vol.14,A.C.S.Symposium Series;Roche et al.,Bioreversible Carriers in Drug Design,American Pharmaceutical Association and Pergamon Press,1987;Rautio et al.,Prodrugs:Design and Clinical Applications,Nature Reviews Drug Discovery,2008,7,255-270,and Hecker et al.,Prodrugs of Phosphates and Phosphonates,J.Med.Chem.,2008,51,2328-2345。
术语“代谢产物”是指具体的化合物或其盐在体内通过代谢作用所得到的产物。一个化合物的代谢产物可以通过所属领域公知的技术来进行鉴定,其活性可以通过如本发明所描述的那样采用试验的方法进行表征。这样的产物可以是通过给药化合物经过氧化,还原,水解,酰氨化,脱酰氨作用,酯化,脱脂作用,酶裂解等等方法得到。相应地,本发明包括化合物的代谢产物,包括将本发明的化合物与哺乳动物充分接触一段时间所产生的代谢产物。
术语“药学上可接受的盐”是指本发明的化合物的有机盐和无机盐。药学上可接受的盐在所属领域是为我们所熟知的,如文献:Berge et al.,describe pharmaceutically acceptable salts in detail in J.Pharmacol Sci,1997,66,1-19所记载的。药学上可接受的非限定性的盐例子包括与氨基基团反应形成的无机酸盐有盐酸盐、氢溴酸盐、磷酸盐、偏磷酸盐、硫酸盐、硝酸盐、高氯酸盐,和有机酸盐如甲磺酸盐、乙磺酸盐、乙酸盐、三氟乙酸盐、羟基乙酸盐、羟乙基磺酸盐、草酸盐、马来酸盐、酒石酸盐、柠檬酸盐、琥珀酸盐、丙二酸盐、苯磺酸盐、对甲苯磺酸盐、苹果酸盐、富马酸盐、乳酸盐、乳糖酸盐,或通过书籍文献上所记载的其他方法如离子交换法来得到这些盐。其他药学上可接受的盐包括己二酸盐、藻酸盐、抗坏血酸盐、天冬氨酸盐、苯磺酸盐、苯甲酸盐、重硫酸盐、硼酸盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、环戊基丙酸盐、二葡萄糖酸盐、十二烷基硫酸盐、乙磺酸盐、甲酸盐、反丁烯二酸盐、葡庚糖酸盐、甘油磷酸盐、葡萄糖酸盐、 半硫酸盐、庚酸盐、己酸盐、氢碘酸盐、2-羟基-乙磺酸盐、乳糖醛酸盐、月桂酸盐、月桂基硫酸盐、丙二酸盐、2-萘磺酸盐、烟酸盐、硝酸盐、油酸盐、棕榈酸盐、扑酸盐、果胶酸盐、过硫酸盐、3-苯基丙酸盐、苦味酸盐、特戊酸盐、丙酸盐、硬脂酸盐、硫氰酸盐、十一酸盐、戊酸盐,等等。通过适当的碱得到的盐包括碱金属,碱土金属,铵和N +(C 1-4烷基) 4的盐。本发明也拟构思了任何所包含N的基团的化合物所形成的季铵盐。水溶性或油溶性或分散产物可以通过季铵化作用得到。碱金属或碱土金属盐包括钠,锂,钾,钙,镁,等等。药学上可接受的盐进一步包括适当的、无毒的铵,季铵盐和抗平衡离子形成的胺阳离子,如卤化物,氢氧化物,羧化物,硫酸化物,磷酸化物,硝酸化物,C 1-8磺酸化物和芳香磺酸化物。
术语“溶剂化物”是指一个或多个溶剂分子与本发明的化合物所形成的缔合物。形成溶剂化物的溶剂包括,但并不限于,水,异丙醇,乙醇,甲醇,二甲亚砜,乙酸乙酯,乙酸,氨基乙醇。术语“水合物”是指溶剂分子是水所形成的缔合物。
术语“氮氧化物”是指当化合物含几个胺官能团时,可将1个或大于1个的氮原子氧化形成N-氧化物。N-氧化物的特殊实例是叔胺的N-氧化物或含氮杂环氮原子的N-氧化物。可用氧化剂例,如过氧化氢或过酸(例如过氧羧酸)处理相应的胺形成N-氧化物(参见Advanced Organic Chemistry,Wiley Interscience,第4版,Jerry March,pages)。尤其是,N-氧化物可用L.W.Deady的方法制备(Syn.Comm.1977,7,509-514),其中例如在惰性溶剂,例如二氯甲烷中,使胺化合物与3-氯过氧苯甲酸(m-CPBA)反应。
本发明化合物的任何不对称原子(例如,碳等)都可以以外消旋或对映体富集的形式存在,例如(R)-、(S)-、(R,S)-或(S,S)-构型形式存在。在某些实施方案中,各不对称原子在(R)-或(S)-构型方面具有至少50%对映体过量,至少60%对映体过量,至少70%对映体过量,至少80%对映体过量,至少90%对映体过量,至少95%对映体过量,或至少99%对映体过量。如果可能的话,具有不饱和双键的原子上的取代基可以以-(Z)-或-(E)-形式存在。
因此,如本发明所描述的那样,本发明的化合物可以以可能的异构体、旋转异构体、阻转异构体、互变异构体中的一种形式或其混合物的形式存在,例如为基本纯的几何(顺式或反式)异构体、非对映异构体、光学异构体(对映体)、外消旋体或其混合物形式。
可以根据组分的物理化学差异将所得的任何异构体混合物分离成纯或基本纯的几何或光学异构体、非对映异构体、外消旋体,例如通过色谱法和/或分步结晶来进行分离。
可以用已知的方法将任何所得终产物或中间体的外消旋体通过本领域技术人员熟悉的方法拆分成光学对映体,如,通过对获得的其非对映异构的盐进行分离。外消旋的产物也可以通过手性色谱来分离,如,使用手性吸附剂的高压液相色谱(HPLC)。特别地,对映异构体可以通过不对称合成制备(如Jacques,et al.,Enantiomers,Racemates and Resolutions(Wiley Interscience,New York,1981);Principles of Asymmetric Synthesis(2 nd Ed.Robert E.Gawley,Jeffrey Aubé,Elsevier,Oxford,UK,2012);Eliel,E.L.Stereochemistry of Carbon Compounds(McGraw-Hill,NY,1962);and Wilen,S.H.Tables of Resolving Agents and Optical Resolutions p.268(E.L.Eliel,Ed.,Univ.of Notre Dame Press,Notre Dame,IN 1972))。
本发明还包括同位素标记的本发明化合物,其除以下事实外与本发明所述的那些化合物相同:一个或多个原子被原子质量或质量数不同于天然常见原子质量或质量数的原子代替。还可引入本发明化合物中的示例性同位素包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,如 2H, 3H, 13C, 14C, 15N, 16O, 17O, 31P, 32P, 36S, 18F和 37Cl。
包含前述同位素和/或其他原子的其他同位素的本发明化合物以及所述化合物的药学上可接受的盐都包括在本发明范围内。同位素标记的本发明化合物,例如放射性同位素,如 3H和 14C掺入到本发明化合物中可用于药物和/或底物组织分布分析。由于易于制备以及检测,氚代的,即, 3H,以及碳-14,即 14C,同位素特别优选。此外,用重的同位素,如氘,即 2H取代,可提供一些源自更大的代谢稳定性的治疗上的优势,例如增加的体内半衰期或减少的剂量需求。因此,在一些情形下可能是优选的。
本发明使用的立体化学定义和惯例大体上按照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所记载的定义和惯例。本发明化合物可含有不对称中心或手性中心,因此以不同的立体异构形式存在。所预期的是,本发明化合物的所有立体异构形式, 包括但不限于非对映异构体、对映异构体和阻转异构体(atropisomer)及它们的混合物如外消旋混合物,也包含在本发明范围之内。许多有机化合物以光学活性形式存在,即它们具有使平面偏振光的平面发生旋转的能力。当描述具有光学活性的化合物时,使用前缀D和L或R和S来表示就分子中的手性中心(或多个手性中心)而言分子的绝对构型。前缀d和l或(+)和(–)是用于指定化合物所致平面偏振光旋转的符号,其中(–)或l表示化合物是左旋的。前缀为(+)或d的化合物是右旋的。就给定的化学结构而言,除了这些立体异构体互为镜像外,这些立体异构体是相同的。具体的立体异构体也可称为对映异构体,并且所述异构体的混合物通常称作对映异构体的混合物。对映异构体的50:50混合物称为外消旋混合物或外消旋体,当在化学反应或方法中没有立体选择性或立体特异性时,可出现所述外消旋混合物或外消旋体。
依据原料和方法的选择,本发明化合物可以以可能的异构体中的一个或它们的混合物的形式存在,例如作为纯旋光异构体,或作为异构体混合物,如作为外消旋和非对应异构体混合物,这取决于不对称碳原子的数量。旋光性的(R)-或(S)-异构体可使用手性合成子或手性制剂制备,或使用常规技术拆分。如果此化合物含有一个双键,取代基可能为E或Z构型;如果此化合物中含有二取代的环烷基,环烷基的取代基可能为顺式或反式(cis-或trans-)构型。
本发明化合物可以含有不对称中心或手性中心,因此以不同的立体异构体形式存在。所预期的是,本发明化合物的所有立体异构体形式,包括但不限于非对映异构体、对映异构体和阻转异构体(atropisomer)和几何(或构象)异构体及它们的混合物,如外消旋混合物,均在本发明的范围之内。
除非另外指出,本发明描述的结构还表示包括此结构的所有异构体(如,对映体、非对映体阻转异构体(atropisomer)和几何(或构象))形式;例如,各不对称中心的R和S构型,(Z)和(E)双键异构体,以及(Z)和(E)构象异构体。因此,本发明化合物的单个立体化学异构体以及对映体混合物、非对映体混合物和几何异构体(或构象异构体)混合物均在本发明的范围之内。
术语“互变异构体”或“互变异构形式”是指具有不同能量的可通过低能垒(low energy barrier)互相转化的结构异构体。若互变异构是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(protontautomer)(也称为质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键互变异构体(valence tautomer)包括通过一些成键电子的重组来进行的互相转化。酮-烯醇互变异构的具体实例是戊烷-2,4-二酮和4-羟基戊-3-烯-2-酮互变异构体的互变。互变异构的另一个实例是酚-酮互变异构。酚-酮互变异构的一个具体实例是吡啶-4-醇和吡啶-4(1H)-酮互变异构体的互变。除非另外指出,本发明化合物的所有互变异构体形式都在本发明的范围之内。
术语“几何异构体”也称“顺反异构体”,因双键(包括烯烃的双键、C=N双键和N=N双键)或环碳原子的单健不能自由旋转而引起的异构体。
术语“二聚体”是指相同或同一种类的物质,以成双的形态出现,可能具有单一状态时没有的性质或功能。常见的例子包括二聚环戊二烯、二聚氯化亚铜、蔗糖等等。
术语“三聚体”是指三个相同或同一种类的物质,聚合成一个新的分子,该新的分子即为三聚体,是一种低分子量的聚合物。
本发明所使用的术语“受试对象”是指动物。典型地所述动物是哺乳动物。受试对象也是指灵长类动物(例如人)、牛、绵羊、山羊、马、狗、猫、兔、大鼠、小鼠、鱼、鸟等。在某些实施方案中,所述受试对象是灵长类动物。在另外其他实施方案中,所述受试对象是人。
本发明所使用的术语“受治疗者”和“患者”可交换地使用。术语“受治疗者”和“患者”指动物(例如,鸡、鹌鹑或火鸡等鸟类或哺乳动物),特别是包括非灵长类动物在内的“哺乳动物”(例如,牛、猪、马、羊、兔、豚鼠、大鼠、猫、狗和小鼠)和灵长类动物(例如,猴子、黑猩猩和人类),更特别的是人类。在一个实施方案中,受治疗者为非人类动物,例如家畜(例如,马、牛、猪或羊)或宠物(例如,狗、猫、豚鼠或兔)。在另一些实施方案中,“患者”是指人类。
术语“X综合症”,也称作代谢综合症的病症、疾病,其疾患详述于Johannsson et al.,J.Clin.Endocrinol.Metab.,1997,82,727-734中。
术语“肠内改善”是指使双歧杆菌属、乳杆菌属等的有益菌增加、使肠内的有机酸增加、使肠内的腐败 产物减少。
术语“癌症”和“癌的”是指或描述患者中通常以失控的细胞生长为特征的生理学病症。“肿瘤”包含一种或多种癌细胞。癌症的实例包括但不限于癌(carcinoma)、淋巴瘤、胚细胞瘤、肉瘤和白血病,或恶性淋巴增殖性疾病(lymphoid malignancies)。此类癌症的更具体的实例包括鳞状细胞癌(如上皮鳞状细胞癌)、肺癌(包括小细胞肺癌、非小细胞肺癌(NSCLC)、肺腺癌和肺鳞状癌)、腹膜癌、肝细胞癌(hepatocellular cancer)、胃癌(gastric or stomach cancer)(包括胃肠癌)、胰腺癌、恶性胶质瘤、***、卵巢癌、肝癌(liver cancer)、膀胱癌、肝细胞瘤(hepatoma)、乳腺癌、结肠癌、直肠癌、结直肠癌、子宫内膜癌或子宫癌、唾液腺癌、肾癌或肾脏癌(kidney or renal cancer)、***癌、外阴癌、甲状腺癌、肝脏癌(hepatic carcinoma)、***癌、***癌以及头颈癌。
另外,除非其他方面表明,本发明所描述的化合物的结构式包括一个或多个不同的原子的富集同位素。
如本发明所使用的术语“治疗”任何疾病或病症,在其中一些实施方案中指改善疾病或病症(即减缓或阻止或减轻疾病或其至少一种临床症状的发展)。在另一些实施方案中,“治疗”指缓和或改善至少一种身体参数,包括可能不为患者所察觉的身体参数。在另一些实施方案中,“治疗”指从身体上(例如稳定可察觉的症状)或生理学上(例如稳定身体的参数)或上述两方面调节疾病或病症。在另一些实施方案中,“治疗”指预防或延迟疾病或病症的发作、发生或恶化。
本发明化合物的描述
本发明提供了一类具有优异的SGLTs抑制活性的化合物,尤其具有优良的SGLT1抑制活性的化合物,用于制备改善肠内环境的药物;或用于制备治疗糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压的药物。本发明也提供了制备这些化合物的方法、包含这些化合物的药物组合物,以及使用这些化合物和组合物制备治疗哺乳动物,尤其是人类的上述疾病的药物的方法。与已有的同类化合物相比,本发明的化合物不仅具有良好的药理活性,还具有优良的体内代谢动力学性质和体内药效学性质。同时制备方法简单易行,工艺方法稳定,适合工业化生产。因此,本发明提供的化合物相对于目前已有的同类化合物而言,具有更优良的成药性。
具体地说:
一方面,本发明涉及一种化合物,其为式(I)所示化合物或式(I)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药,
Figure PCTCN2019073550-appb-000005
其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、L、n和t具有如本发明所述的定义,条件是,当R 5和R 8同时为H,且R 9
Figure PCTCN2019073550-appb-000006
时,t不为0。
在一些实施方案中,L为-(CR aR b) q-、-CH=CH-(CR aR b) p-、-O-(CR aR b) p-、-NH-(CR aR b) p-、-S-(CR aR b) p-、-S(=O)-(CR aR b) p-或-S(=O) 2-(CR aR b) p-;
q为1、2、3、4、5或6;
p为0、1、2、3、4、5或6;
R a和R b各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基或3-6个原子组成的杂环基;或R a、R b和与它们相连的碳原子一起形成C 3-6碳环或3-6个原子组成的杂环;
R 1为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6 羟基烷基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 1-6烷氨基、C 1-6烷硫基、C 3-6环烷基或C 3-6环烷基-C 1-4亚烷基,其中,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6羟基烷基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 1-6烷氨基、C 1-6烷硫基、C 3-6环烷基和C 3-6环烷基-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 2和R 3各自独立地为H、D、CN、OH、NH 2、-SH、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6烷硫基、C 1-6烷氨基、C 3-6环烷基或3-6个原子组成的杂环基,其中,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6烷硫基、C 1-6烷氨基、C 3-6环烷基和3-6个原子组成的杂环基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R 2、R 3和与它们相连的碳原子一起形成羰基;
或R 2、R 3和与它们相连的碳原子一起形成C 3-6碳环或3-6个原子组成的杂环,其中,所述C 3-6碳环和3-6个原子组成的杂环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 4为H、D、-OR 4a或-SR 4b
R 4a和R 4b各自独立地为H、D、C 1-6烷基、C 3-6环烷基、C 3-6环烷基-C 1-4亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-4亚烷基,其中,所述C 1-6烷基、C 3-6环烷基、C 3-6环烷基-C 1-4亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 10为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-6烷基或C 1-6烷氧基;
R 5为H、D、C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基或5-8个原子组成的杂芳基,其中,所述C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基和5-8个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基或5-8个原子组成的杂芳基,其中,所述C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基和5-8个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
或R 6、R 7和与它们相连的碳原子一起形成C 3-8碳环、3-8个原子组成的杂环、C 6-10芳环或5-8个原子组成的杂芳环,其中,所述C 3-8碳环、3-8个原子组成的杂环、C 6-10芳环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各 自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R 8和R 9各自独立为H、D、R eO-C 1-4亚烷基、R dR cN-C 1-4亚烷基、C 1-8烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基或(5-8个原子组成的杂芳基)-C 1-4亚烷基,其中所述C 1-8烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基和(5-8个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R 8、R 9和与它们相连的氮原子一起形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
R c、R d、R e和R f各自独立地为H、D、C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基或(5-8个原子组成的杂芳基)-C 1-4亚烷基,其中,所述C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基和(5-8个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
或R c、R d和与它们相连的氮原子一起形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、OH、-NH 2、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
n为0、1、2或3;
t为0、1、2、3、4、5或6;条件是,当R 5和R 8同时为H,且R 9
Figure PCTCN2019073550-appb-000007
时,t不为0。
在另一些实施方案中,本发明涉及一种式(I-a)所示结构的化合物或其立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药,
Figure PCTCN2019073550-appb-000008
其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、L、n和t具有如本发明所述的定义。
在另一些实施方案中,本发明涉及一种式(II)所示结构的化合物或其立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药,
Figure PCTCN2019073550-appb-000009
其中,R 1、R 2、R 3、R 4、R 5、R 6、R 7、R 8、R 9、R 10、n和t具有如本发明所述的定义。
在另一些实施方案中,R 1为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4烷氧基、C 1-4羟基烷基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 1-4烷氨基、C 1-4烷硫基、C 3-6环烷基或C 3-6环烷基-C 1-2亚烷基,其中,所述C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4烷氧基、C 1-4羟基烷基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 1-4烷氨基、C 1-4烷硫基、C 3-6环烷基和C 3-6环烷基-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 1为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、甲基、乙基、正丙基、异丙基、丙烯基、丙炔基、甲氧基、乙氧基、羟基甲基、三氟甲基、三氟乙基、一氟甲基、三氟甲氧基、二氟甲氧基、甲氨基、环丙基、环丁基、环戊基、环己基或环丙基-亚甲基,其中,所述甲基、乙基、正丙基、异丙基、丙烯基、丙炔基、甲氧基、乙氧基、羟基甲基、三氟乙基、一氟甲基、二氟甲氧基、甲氨基、环丙基、环丁基、环戊基、环己基和环丙基-亚甲基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 2和R 3各自独立地为H、D、CN、CN、OH、NH 2、-SH、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4烷氧基、C 1-4烷硫基、C 1-4烷氨基、C 3-6环烷基或5-6个原子组成的杂环基,其中,所述C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4烷氧基、C 1-4烷硫基、C 1-4烷氨基、C 3-6环烷基和5-6个原子组成的杂环基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 2和R 3各自独立地为H、D、CN、OH、NH 2、-SH、甲基、乙基、正丙基、异丙基、乙烯基、丙炔基、甲氧基、乙氧基、甲硫基、甲氨基、环丙基、环丁基、环戊基、环己基或5-6个原子组成的杂环基,其中,所述甲基、乙基、正丙基、异丙基、乙烯基、丙炔基、甲氧基、乙氧基、甲硫基、甲氨基、环丙基、环丁基、环戊基、环己基和5-6个原子组成的杂环基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 2、R 3和与它们相连的碳原子一起形成羰基。
在另一些实施方案中,R 2、R 3和与它们相连的碳原子一起形成C 3-6碳环或5-6个原子组成的杂环,其中,所述C 3-6碳环和5-6个原子组成的杂环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 2、R 3和与它们相连的碳原子一起形成环丙烷、环丁烷、环戊烷、环己烷或5-6个原子组成的杂环,其中,所述环丙烷、环丁烷、环戊烷、环己烷和5-6个原子组成的杂环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 4为H、D、-OR 4a或-SR 4b
R 4a和R 4b各自独立地为H、D、C 1-4烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述C 1-4烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-4烷基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 4为H、D、-OR 4a或-SR 4b
R 4a和R 4b各自独立地为H、D、甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 10为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-4烷基或C 1-4烷氧基。
在又一些实施方案中,R 10为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、甲基、乙基、正丙基、异丙基、甲氧基或乙氧基。
在另一些实施方案中,R 5为H、D、C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基,其中,所述C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基和5-6个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基。
在又一些实施方案中,R 5为H、D、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、三氟甲基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基或嘧啶基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基和嘧啶基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基或苯基。
在另一些实施方案中,R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基,其中,所述C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基和5-6个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、 =O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基。
在又一些实施方案中,R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、甲氧基、乙氧基、甲氨基、三氟甲基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基或嘧啶基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、甲氧基、乙氧基、甲氨基、二氟甲氧基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基和嘧啶基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基或苯基。
在另一些实施方案中,R 6、R 7和与它们相连的碳原子一起形成C 3-6碳环、5-6个原子组成的杂环、C 6-10芳环或5-6个原子组成的杂芳环,其中,所述C 3-6碳环、5-6个原子组成的杂环、C 6-10芳环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 6、R 7和与它们相连的碳原子一起形成环丙烷、环丁烷、环戊烷、环己烷、吡咯烷、四氢呋喃、四氢噻吩、四氢吡喃、四氢噻喃、哌啶、吗啉、硫代吗啉、哌嗪、苯环、呋喃、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、噻吩、吡嗪、哒嗪或嘧啶,其中,所述环丙烷、环丁烷、环戊烷、环己烷、吡咯烷、四氢呋喃、四氢噻吩、四氢吡喃、四氢噻喃、哌啶、吗啉、硫代吗啉、哌嗪、苯环、呋喃、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、噻吩、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 8和R 9各自独立为H、D、R eO-C 1-4亚烷基、R dR cN-C 1-4亚烷基、C 1-6烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中所述C 1-6烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、 (5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 8和R 9各自独立为H、D、R dR cN-C 1-4亚烷基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、正戊基、正己基、三氟甲基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、C 6-10芳基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、正戊基、正己基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、C 6-10芳基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OH、-NR cR d、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R 8、R 9和与它们相连的氮原子一起形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R 8、R 9和与它们相连的氮原子一起形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OH、-NR cR d、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R c、R d、R e和R f各自独立地为H、D、C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R c、R d、R e和R f各自独立地为H、D、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、三氟甲基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、 吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
在另一些实施方案中,R c、R d和与它们相连的氮原子一起形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、OH、-NH 2、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
在又一些实施方案中,R c、R d和与它们相连的氮原子一起形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
还在一些实施方案中,本发明涉及以下其中之一的结构,或其立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药,
Figure PCTCN2019073550-appb-000010
Figure PCTCN2019073550-appb-000011
Figure PCTCN2019073550-appb-000012
Figure PCTCN2019073550-appb-000013
另一方面,本发明涉及一种药物组合物,其包含本发明所述的化合物。
在一些实施方案中,本发明所述的药物组合物,其进一步包含药学上可接受的载体、赋形剂、辅剂、媒介物或它们的组合。
在一些实施方案中,本发明所述的药物组合物,进一步包含一种或多种其他附加治疗剂,其中所述其他附加治疗剂选自抗糖尿病药物、抗高血糖药物、抗肥胖症药物、抗高血压药物、食欲抑制药物、降脂药物或其组合。
在另一些实施方案中,本发明所述的药物组合物可以是液体、固体、半固体、凝胶或喷雾剂型。
在另一些实施方案中,本发明所述的抗糖尿病药物和抗高血糖药物分别独立地选自SGLT2抑制剂、双胍类药物、磺酰脲类药物、葡萄糖苷酶抑制剂、PPAR激动剂(过氧化物酶体增殖物激活受体激动剂)、αP2抑制剂(脂肪细胞脂肪酸结合蛋白抑制剂)、PPARα/γ双激活剂(过氧化物酶体增殖物激活受体α/γ双激活剂)、二肽酰肽酶IV抑制剂、格列奈类药物、胰岛素、胰高血糖素样肽-1抑制剂、PTP1B抑制剂(蛋白酪氨酸磷酸酶1B抑制剂)、糖原磷酸化酶抑制剂、葡萄糖-6-磷酸酶抑制剂或其组合。
在另一些实施方案中,本发明所述的抗肥胖症药物选自中枢抗肥胖药、MCH(黒色素浓集激素)受体拮抗剂、神经肽Y受体拮抗剂、***素(cannabinoid)受体拮抗剂、脑肠肽拮抗剂、脂肪酶抑制剂、β3激动剂、11β-HSD1(11β羟基类固醇脱氢酶1)抑制剂、DGAT-1(二酰甘油酰基转移酶1)抑制剂、肽性食欲抑制剂、缩胆囊肽(Cholecystokinin)激动剂、摄食抑制剂或其组合。
在另一些实施方案中,本发明所述的抗高血压药物选自血管紧张素转化酶抑制剂、血管紧张素II受体拮抗剂、钙通道拮抗剂、钾通道开放剂、利尿剂或其组合。
在另一些实施方案中,本发明所述降脂药物选自MTP抑制剂(微粒体甘油三酯转移蛋白抑制剂)、HMGCoA还原酶抑制剂(羟甲基戊二酰辅酶A还原酶抑制剂)、角鲨烯合成酶抑制剂、贝丁酸类降血脂药物(又称贝特类降血脂药物)、ACAT抑制剂(乙酰胆固醇乙酰转移酶抑制剂)、脂加氧酶抑制剂、胆固醇吸收抑制剂、回肠钠离子/胆汁酸协同转运蛋白抑制剂、LDL受体活性的向上调节物、烟酸类降血脂药物、胆汁酸螯合物或其组合。
在又一些实施方案中,本发明所述降脂药物选自普伐他汀、辛伐他汀、阿伐他汀、氟伐他汀、西立伐他汀、埃塔伐他汀、罗素他汀或其组合。
另一方面,本发明涉及本发明所述的化合物或本发明所述的药物组合物在制备药物中的用途,其中,所述药物用于在人或动物体内抑制SGLT1。
另一方面,本发明涉及本发明所述的化合物或本发明所述的药物组合物在制备药物中的用途,其中,所述药物用于改善肠内环境。
另一方面,本发明还涉及本发明所述的化合物或药物组合物在制备药物中的用途,其中,所述药物用于预防或治疗疾病,减轻所述疾病症状或者延缓所述疾病的发展或发作,其中所述疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压。
在一些实施方案中,所述糖尿病并发症为糖尿病性视网膜病、糖尿病性神经病或糖尿病性肾病。
在一些实施方案中,所述高脂血症为高甘油三酯血症。
另一方面,本发明涉及一种使用本发明所述的化合物或药物组合物来抑制SGLT1活性的方法,所述方法包含给予患者所述化合物或所述药物组合物的有效治疗量。
另一方面,本发明涉及一种使用本发明所述的化合物或药物组合物来改善肠内环境的方法,所述方法包含给予患者所述化合物或所述药物组合物的有效治疗量。
另一方面,本发明涉及一种使用本发明所述的化合物或药物组合物用于预防或治疗下列疾病的方法,所述的方法包含给予患者本发明所述的化合物或药物组合物的有效治疗量,其中所述的疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压。并且,本发明提供的上述化合物或其药物组合物可以与其它疗法或治疗剂共同施用。施用方式可以为同时、顺序或以一定时间间隔进行。
实施治疗、预防或延缓等作用所需的化合物或药物组合物的剂量通常取决于施用的具体化合物、患者、具体疾病或病症及其严重程度、给药途径和频率等,并且需要由主治医师根据具体情况判定。例如,在通过经静脉途径施用本发明提供的化合物或药物组合物时,可以每周一次甚至以更长时间间隔进行施用。
另一方面,本发明涉及将本发明所述的化合物或药物组合物用于抑制SGLT1的活性。
另一方面,本发明涉及将本发明所述的化合物或药物组合物用于改善肠内环境。
另一方面,本发明涉及将本发明所述的化合物或药物组合物用于预防或治疗下列疾病,减轻下列疾病症状或者延缓下列疾病的发展或发作,其中所述的疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压。
在一些实施方案中,所述盐是指药学上可接受的盐。术语“药学上可接受的”是指物质或组合物必须与包含制剂的其它成分和/或用其治疗的哺乳动物化学上和/或毒理学上相容。
本发明的化合物还包括这样的化合物的其他盐,该其他盐不一定是药学上可接受的盐,并且可以用作用于制备和/或提纯本发明的化合物和/或用于分离本发明的化合物的对映体的中间体。
可药用的酸加成盐可与无机酸和有机酸形成,例如乙酸盐、天冬氨酸盐、苯甲酸盐、苯磺酸盐、溴化物/氢溴酸盐、碳酸氢盐/碳酸盐、硫酸氢盐/硫酸盐、樟脑磺酸盐、氯化物/盐酸盐、氯茶碱盐、柠檬酸盐、乙二磺酸盐、富马酸盐、葡庚糖酸盐、葡糖酸盐、葡糖醛酸盐、马尿酸盐、氢碘酸盐/碘化物、羟乙基磺酸盐、乳酸盐、乳糖醛酸盐、月桂基硫酸盐、苹果酸盐、马来酸盐、丙二酸盐、扁桃酸盐、甲磺酸盐、甲基硫酸盐、萘甲酸盐、萘磺酸盐、烟酸盐、硝酸盐、十八酸盐、油酸盐、草酸盐、棕榈酸盐、扑酸盐、磷酸盐/磷酸氢盐/磷酸二氢盐、聚半乳糖酸盐、丙酸盐、硬脂酸盐、琥珀酸盐、磺基水杨酸盐、酒石酸盐、甲苯磺酸盐和三氟乙酸盐。
可以由其衍生得到盐的无机酸包括例如盐酸、氢溴酸、硫酸、硝酸、磷酸等。
可以由其衍生得到盐的有机酸包括例如乙酸、丙酸、羟基乙酸、草酸、马来酸、丙二酸、琥珀酸、富马酸、酒石酸、柠檬酸、苯甲酸、扁桃酸、甲磺酸、乙磺酸、对甲苯磺酸、磺基水杨酸等。
可药用碱加成盐可与无机碱和有机碱形成。
可以由其衍生得到盐的无机碱包括,例如铵盐和周期表的I族至XII族的金属。在某些实施方案中,该盐衍生自钠、钾、铵、钙、镁、铁、银、锌和铜;特别适合的盐包括铵、钾、钠、钙和镁盐。
可以由其衍生得到盐的有机碱包括伯胺、仲胺和叔胺、取代的胺包括天然存在的取代的胺、环状胺、碱性离子交换树脂等。某些有机胺包括,例如,异丙胺、苄星青霉素(benzathine)、胆碱盐(cholinate)、二乙醇胺、二乙胺、赖氨酸、葡甲胺(meglumine)、哌嗪和氨丁三醇。
本发明的可药用盐可以用常规化学方法由母体化合物、碱性或酸性部分来合成。一般而言,该类盐可以通过使这些化合物的游离酸形式与化学计量量的适宜碱(如Na、Ca、Mg或K氢氧化物、碳酸盐、碳酸氢盐等)反应,或者通过使这些化合物的游离碱形式与化学计量量的适宜酸反应来进行制备。该类反应通常在水或有机溶剂或二者的混合物中进行。一般地,在适当的情况中,需要使用非水性介质如***、乙酸乙酯、乙醇、异丙醇或乙腈。在例如“Remington′s Pharmaceutical Sciences”,第20版,Mack Publishing Company,Easton,Pa.,(1985);和“药用盐手册:性质、选择和应用(Handbook of Pharmaceutical Salts:Properties,Selection,and Use)”,Stahl and Wermuth(Wiley-VCH,Weinheim,Germany,2002)中可找到另外一些适宜盐的列表。
而且,本发明化合物、包括其盐也可以以其水合物形式获得,或者包括其他用于其结晶的溶剂。本发明化合物可以固有地或通过设计形成具有可药用溶剂(包括水)的溶剂化物;因此,本发明意在包括溶剂化的和未溶剂化的形式。
本发明给出的任何结构式也意欲表示这些化合物未被标记的形式以及同位素标记的形式。同位素标记的化合物具有本发明给出的通式描绘的结构,除了一个或多个原子被具有所选择原子量或质量数的原子替换。可引入本发明化合物中的示例性同位素包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,如 2H, 3H, 11C, 13C, 14C, 15N, 18F, 31P, 32P, 36S, 37Cl或 125I。
另一方面,本发明所述化合物包括用各种同位素标记的本发明所定义的化合物,例如,其中存在放射性同位素,如 3H, 14C和 18F的那些化合物,或者其中存在非放射性同位素,如 2H和 13C。该类同位素标记的化合物可用于代谢研究(使用 14C)、反应动力学研究(使用例如 2H或 3H)、检测或成像技术,如正电子发射断层扫描术(PET)或包括药物或底物组织分布测定的单光子发射计算机断层成像术(SPECT),或可用于患者的放疗中。 18F标记的化合物对PET或SPECT研究而言是特别理想的。同位素标记的式(I)化合物可以通过本领域技术人员熟悉的常规技术或本发明中的实施例和制备过程所描述使用合适的同位素标记试剂替代原来使用过的未标记试剂来制备。
此外,较重同位素特别是氘(即, 2H或D)的取代可提供某些治疗优点,这些优点是由代谢稳定性更高带来的。例如,体内半衰期增加或剂量需求降低或治疗指数得到改善带来的。应当理解,这一上下文中的氘被看做式(I)化合物的取代基。可以用同位素富集因子来定义该类较重同位素特别是氘的浓度。本发明所使用的术语“同位素富集因子”是指所指定同位素的同位素丰度和天然丰度之间的比例。如果本发明化合物的取代基被指定为氘,该化合物对各指定的氘原子而言具有至少3500(各指定氘原子处52.5%的氘掺入)、至少4000(60%的氘掺入)、至少4500(67.5%的氘掺入),至少5000(75%的氘掺入),至少5500(82.5%的氘掺入)、至少6000(90%的氘掺入)、至少6333.3(95%的氘掺入)、至少6466.7(97%的氘掺入)、至少6600(99%的氘掺入)或至少6633.3(99.5%的氘掺入)的同位素富集因子。本发明可药用的溶剂化物包括其中结晶溶剂可以是同位素取代的例如D 2O、丙酮-d 6、或DMSO-d 6的那些溶剂化物。
前面所述内容只概述了本发明的某些方面,但并不限于这些方面,其他方面的内容将在下面作更加具体完整的描述。
本发明的化合物的组合物、制剂和给药
本发明涉及一种药物组合物,其包括本发明所述化合物或实施例中所示结构的化合物,或其立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、二聚体、三聚体及药学上可接受的盐或它们的前药。所述药物组合物进一步包含至少一种药学上可接受的载体、赋形剂、辅剂、媒介物或它们的组合,以及任选地、其它的治疗和/或预防成分。本发明的药物组合物中化合物的量能有效地可探测地抑制钠依赖性葡萄糖转运蛋白(sodium-dependent glucose transporters,SGLTs),尤其是SGLT1的活性。
本发明的化合物存在自由形态,或合适的、作为药学上可接受的衍生物。根据本发明,药学上可接受的衍生物包括,但并不限于,药学上可接受的前药,盐,酯,酯类的盐,或能直接或间接地根据患者的需要给药的其他任何加合物或衍生物,本发明其他方面所描述的化合物,其代谢产物或他的残留物。
药学上可接受的载体可能含有不会过度抑制化合物的生物活性的惰性成分。药学上可接受的载体应生物相容,例如无毒、非炎性、非免疫原性或一旦施用给患者无其它不良反应或副作用。可采用标准制药技术。
像本发明所描述的,本发明所述的药物组合物或药学上可接受的组合物进一步包含药学上可接受的载体、辅剂或赋形剂,像本发明所应用的,包括适合于特有的目标剂型的,任何溶剂、稀释剂、液体赋形剂、分散剂、悬浮剂、表面活性剂、等渗剂、增稠剂、乳化剂、防腐剂、固体粘合剂或润滑剂,等等。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公开了配制药学上可接受的组合物中使用的各种载体及其公知制备方法。除了与本发明的化合物不相容的常规载体媒介,例如会产生不良生物效应或与药学上可接受的组合物中的任何其他组分发生有害的相互作用,其他任何常规的载体媒介及它们的用途也是本发明所考虑的范围。
可用作药学上可接受的载体的物质的一些实例包括但不限于离子交换剂、氧化铝、硬脂酸铝、卵磷脂、血清蛋白(例如人血清白蛋白)、缓冲物质(例如吐温80、磷酸盐、甘氨酸、山梨酸或山梨酸钾)、饱和植物脂肪酸的偏甘油酯混合物、水、盐或电解质(例如硫酸精蛋白、磷酸氢二钠、磷酸氢钾、氯化钠或锌盐)、硅胶、三硅酸镁、聚乙烯吡咯烷酮、聚丙烯酸酯、蜡、聚乙烯-聚氧化丙烯-嵌段共聚物、甲基纤维素、羟丙基甲基纤维素、羊毛脂、糖类(例如乳糖、葡萄糖和蔗糖)、淀粉(例如玉米淀粉和马铃薯淀粉)、纤维素及其衍生物(例如羧甲基纤维素钠、乙基纤维素和醋酸纤维素)、粉状黄蓍胶、麦芽、凝胶、滑石、赋形剂 (例如可可油和栓剂蜡)、油(例如花生油、棉花子油、红花油、芝麻油、橄榄油、玉米油和大豆油)、乙二醇(例如丙二醇或聚乙二醇)、酯(例如油酸乙酯和十二酸乙酯)、琼脂、缓冲剂(例如氢氧化镁和氢氧化铝)、褐藻酸、无热原水、等渗盐水、林格氏溶液(Ringer'ssolution)、乙醇和磷酸盐缓冲液以及其它无毒相容性滑润剂(例如硫酸月桂酯钠和硬脂酸镁)以及根据配制人的判断着色剂、防粘剂、涂层剂、甜味剂和增香剂、防腐剂和抗氧化剂也可存在于组合物中。
本发明的化合物或组合物可以通过任何合适方式给药,可根据受治感染的严重程度经口、直肠、肠胃外、脑池内、***内、腹膜内、局部(如同通过粉剂、药膏或滴剂)、口腔作为口或喷鼻剂等向人或其它动物施用以上所述化合物和药学上可接受的组合物。
供口服的液体剂型包括但不限于药学上可接受的乳剂、微型乳剂、溶液、悬浮剂、糖浆和酏剂。除活性化合物外,液体剂型可能含有本领域常用的惰性稀释剂,例如水或其它溶剂、增溶剂和乳化剂,例如乙醇、异丙醇、碳酸乙酯、乙酸乙酯、苯甲醇、苯甲酸苄酯、丙二醇、1,3-丁二醇、二甲基甲酰胺、油(尤其是棉花子油、花生油、玉米油、胚芽油、橄榄油、蓖麻油和芝麻油)、甘油、四氢糠醇、聚乙二醇和山梨聚糖的脂肪酸酯及其混合物。除惰性稀释剂外,口服组合物也可包括佐剂,例如湿润剂、乳化和悬浮剂、甜味剂、调味剂和增香剂。
可根据已知技术使用适合的分散或湿润剂和悬浮剂配制可注射制剂,例如无菌可注射水或油悬浮剂。无菌可注射制剂也可能是无毒的肠胃外可接受的稀释剂或溶剂中的无菌可注射溶液、悬浮剂或乳剂,例如1,3-丁二醇中的溶液。在可接受的媒介物和溶剂中,可采用的是水、林格氏溶液、U.S.P.和等渗氯化钠溶液。另外,按照惯例采用无菌不挥发性油作为溶剂或悬浮介质。为此,可采用任何无味的不挥发性油,包括合成的单酸甘油脂或甘油二酯。另外,脂肪酸,例如十八烯酸,用于制备注射剂。
例如,可通过细菌保留过滤器过滤或通过加入呈无菌固体组合物形式,使用之前可溶于或分散于无菌水或其它无菌可注射介质中的杀菌剂为可注射制剂灭菌。
为延长本发明所述化合物或组合物的作用,常常希望减缓化合物由皮下或肌肉注射的吸收。这可通过使用水溶性差的晶体或无定形物质的液体悬浮液实现。然后,化合物的吸收速率取决于其溶解速率,而溶解速率又取决于晶体大小和晶形。或者,通过将化合物溶解或悬浮于油媒介物中实现延迟吸收经肠胃外施用的化合物。通过在生物可降解的聚合物例如聚交酯-聚羟基乙酸中形成化合物的微胶囊矩阵制成可注射的储存形式。根据化合物与聚合物之比和采用的特殊聚合物的性质,可控制化合物释放速率。其它生物可降解的聚合物的实例包括聚原酸酯和聚酸酐。也可通过将化合物截留在与身体组织相容的脂质体或微型乳剂中制备可注射的储存制剂。
经直肠或***施用的组合物特别是可通过混合本发明所述化合物和适合的非刺激性赋形剂或载体,例如可可油、聚乙二醇或栓剂蜡制备的栓剂,所述赋形剂或载体在环境温度下为固体但在体温下为液体并因此在直肠或***腔内融化并释放活性化合物。
口服固体剂型包括胶囊、片剂、丸剂、粉剂和颗粒。在这种固体剂型中,活性化合物混有至少一种惰性的药学上可接受的赋形剂或载体例如柠檬酸钠或磷酸二钙和/或a)填料或膨胀剂,例如淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸,b)粘合剂,例如羧基甲基纤维素、藻酸盐、凝胶、聚乙烯吡咯烷酮、蔗糖和***胶,c)保湿剂,例如甘油,d)崩解剂,例如琼脂--琼脂、碳酸钙、马铃薯或木薯淀粉、褐藻酸、某些硅酸盐和碳酸钠,e)溶液阻滞剂,例如石蜡,f)吸收加速剂,例如季铵化合物,g)湿润剂,例如鲸蜡醇和单硬脂酸甘油酯,h)吸收剂,例如高岭土和膨润土,和i)润滑剂,例如滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、硫酸月桂酯钠及其混合物。在为胶囊、片剂和丸剂的情况下,剂型也可包含缓冲剂。
也可使用如乳糖或奶糖以及高分子聚乙二醇等赋形剂将相似类型的固体组合物用作软和硬凝胶胶囊中的填料。可用包衣和壳,例如肠溶衣和制药领域众所周知的其它包衣制备片剂、糖锭、胶囊、丸剂和颗粒的固体剂型。它们可任选含有乳浊剂并且还可具有组合物的性质,以致任选地以延迟方式仅释放活性成分,或优选地,在肠道的某一部分释放。可使用的包埋组合物的实例包括聚合物和蜡。也可使用乳糖或奶糖以及高分子聚乙二醇等赋形剂将相似类型的固体组合物用作软和硬凝胶胶囊中的填料。
活性化合物也可呈现具有一种或多种上述赋形剂的微密封形式。可用包衣和壳,例如肠溶衣、控释包 衣和制药领域中众所周知的其它包衣制备片剂、糖锭、胶囊、丸剂和颗粒的固体剂型。在这种固体剂型中,活性化合物可能混有至少一种惰性稀释剂,例如蔗糖、乳糖或淀粉。一般地,这种剂型也可能包含除惰性稀释剂外的另外的物质,例如压片润滑剂和其它压片辅助剂,例如硬脂酸镁和微晶纤维素。在为胶囊、片剂和丸剂的情况下,剂型也可包含缓冲剂。它们可任选含有乳浊剂并且还可具有组合物的性质,以致任选地以延迟方式仅释放活性成分,或优选地,在肠道的某一部分释放。可使用的包埋组合物的实例包括聚合物和蜡。
本发明所述化合物的局部或经皮施用剂型包括药膏、软膏、乳膏、洗剂、凝胶、粉剂、溶液、喷剂、吸入剂或贴片。活性化合物在无菌条件下与药学上可接受的载体和任何需要的防腐剂或可能需要的缓冲剂一起混合。眼科制剂、耳滴剂和眼药水也被考虑到本发明的范围之内。另外,本发明考虑到具有提供控制化合物向身体递送的附加优点的皮肤贴片的用途。可通过将化合物溶解或分散于恰当介质中制成这种剂型。吸收促进剂也可用于提高化合物通过皮肤的流量。可通过提供速率控制膜或通过将化合物分散于聚合物基质或凝胶中控制速率。
也可经口、肠胃外,通过吸入喷剂经局部、直肠、鼻、口腔、***或通过植入药盒施用本发明所述的组合物。如本发明使用的术语“肠胃外”包括但不限于皮下、静脉内、肌肉、关节内、滑膜腔内、胸骨内、鞘内、肝内、病灶内和颅内注射或输注技术。特别地,经口、腹膜内或静脉内施用组合物。
本发明所述组合物的无菌可注射形式可为水或油悬浮液。这些悬浮液可跟进本领域已知的技术使用适合的分散或湿润剂和悬浮剂制备。无菌可注射制剂也可能是于无毒的可经肠胃外接受的稀释剂或溶剂中的无菌可注射溶液或悬浮液,例如于1,3-丁二醇中的溶液。在可接受的媒介物和溶剂中,可采用的是水、林格氏溶液和等渗氯化钠溶液。另外,按照惯例采用无菌不挥发性油作为溶剂或悬浮介质。为此,可采用任何无味的不挥发性油,包括合成的单酸甘油脂或甘油二酯。另外,正如尤其呈聚氧乙烯化形式的天然药学上可接受的油,例如橄榄油或蓖麻油,脂肪酸例如十八烯酸及其甘油酯衍生物用于制备注射剂。这些油溶液或悬浮液也可能含有长链醇稀释剂或分散剂,例如羧甲基纤维素或在配制药学上可接受的剂型(包括乳剂和悬浮液)中常用的类似分散剂。其它常用表面活性剂,例如Tweens、Spans和在生产药学上可接受的固体、液体或其它剂型中常用的其它乳化剂或生物利用率增强剂也可用于配制的目的。
可以任何口服可接受的剂型,包括但不限于胶囊、片剂、水悬浮液或溶液,口服本发明所述药物组合物。在为供口服片剂的情况下,常用载体包括但不限于乳糖和淀粉。通常还加入润滑剂,例如硬脂酸镁。为了以胶囊形式口服,有用的稀释剂包括乳糖和干玉米淀粉。当口服需要水悬浮液时,活性成分与乳化剂和悬浮剂结合。若需要,还可加入某些甜味剂、增味剂或着色剂。
或者,可以供直肠使用的栓剂形式施用本发明所述的药物组合物。可通过混合试剂和非刺激性赋形剂制备这些药物组合物,所述赋形剂在室温下为固体,但在直肠温度下为液体,因此将在直肠内融化以释放药物。这种物质包括但不限于可可油、蜂蜡和聚乙二醇。
尤其是当治疗目标包括局部滴施易于接近的区域或器官,包括眼部、皮肤或低位肠道疾病时,还可局部施用本发明所述的药物组合物。易于为这些区域或器官的每一个制备适合的局部制剂。
以直肠栓剂制剂(见上文)或适合的灌肠剂制剂可实现对低位肠道的局部滴施。也可使用局部皮肤贴片。
对于局部滴施而言,可将药物组合物配制为含有悬浮或溶于一种或多种载体中的活性组分的适合药膏。适于局部滴施本发明的化合物的载体包括但不限于矿物油、凡士林油、白凡士林、丙二醇、聚氧乙烯、聚氧丙烯化合物、乳化蜡和水。或者,可将药物组合物配制为含有悬浮或溶于一种或多种药学上可接受的载体中的活性组分的适合洗剂或乳膏。适合的载体包括但不限于矿物油、山梨醇酐单硬脂酸酯、聚山梨醇酯60、十六烷基酯蜡、鲸蜡硬脂醇、2-辛基十二醇、苯甲醇和水。
为了眼科使用,可用或不用防腐剂例如苯扎氯铵,将药物组合物配制为在等渗pH调节无菌盐水中的微粉化悬浮液,或特别是等渗pH调节无菌盐水中的溶液。或者,为了眼科使用,可将药物组合物配制为药膏,例如凡士林。
也可通过鼻用气化喷雾剂或吸入施用药物组合物。根据制药领域中众所周知的技术制备这种组合物并 且采用苯甲醇和其它适合的防腐剂、提高生物利用率的吸收促进剂、碳氟化合物和/或其它常规增溶剂或分散剂制备成盐水中的溶液。
可将用于本发明的方法的化合物配制成单位剂型。术语“单位剂型”指适合作为受治疗者的单位剂量的物理分立单位,每单位含有经计算产生预期疗效的预定量的活性物质,任选地与适合的药物载体结合。单位剂型可作单次日剂量或多次日剂量(例如,每日约1-4次或更多次)的其中一次。当使用多次日剂量时,对于每次剂量的单位剂型可相同或不同。
本发明的化合物可以以仅有的药学试剂或结合一个或多个其他附加治疗(药学的)剂来给药,其中联合用药引起可接受的不良反应,这对于糖尿病、糖尿病并发症以及其它相关疾病的治疗具有特殊的意义,所述的这些疾病包括,但并不限于,I型糖尿病、II型糖尿病、糖尿病性视网膜病、糖尿病性神经病、糖尿病性肾病、胰岛素抵抗、高血糖、高胰岛素血症、血液中脂肪酸或甘油水平的升高、高脂血症、肥胖症、高甘油三酯血症、X综合症、糖尿病并发症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压等。本发明所使用的“附加治疗剂”包括抗糖尿病药物、抗高血糖药物、抗肥胖症药物、抗高血压药物、食欲抑制药物、降脂药物或其组合。
其中,本发明所述的抗糖尿病试剂包括,但并不限于SGLT2抑制剂(例如达格列净(dapagliflozin)、卡格列净(canagliflozin)、托格列净(tofogliflozin)、伊格列净(ipragliflozin)、鲁格列净(luseogliflozin)、恩格列净(empagliflozin)),双胍类药物(例如苯乙双胍(phenformin)、二甲双胍(metformin)),磺酰脲类药物(例如醋磺环已脲(acetohexamide)、氯磺丙脲(chlorpropamide)、格列本脲(glibenclamide,优降糖)、格列吡嗪(glipizide,吡磺环已脲)、格列齐特(gliclazide,达美康)、格列美脲(glimepiride)、格列戊脲(glipentide)、格列喹酮(gliquidone)、妥拉磺脲(tolazamide)、甲苯磺丁脲(tolbutamide)及氯茴苯酸(meglitinide)),格列奈类药物(例如瑞格列奈(repaglinide)及那格列奈(nateglinide))、α-葡萄糖苷水解酶抑制剂(例如阿卡波糖(acarbose)),α-葡萄糖苷酶抑制剂(例如酯解素、卡格列波糖(camiglibose)、乙格列酯(emiglitate)、米格列醇(miglitol)、伏格列波糖(voglibose)、普那米星(pradimicin)及沙玻制菌素(salbostatin)),PPAR激动剂(例如巴格列酮(balaglitazone)、环格列酮(ciglitazone)、达格列酮(darglitazone)、恩格列酮(englitazone)、伊沙列酮(isaglitazone)、吡格列酮(pioglitazone)、罗格列酮(rosiglitazone)及曲格列酮(troglitazone)),PPARα/γ双激活剂(例如CLX-0940、GW-1536、GW-1929、GW-2433、KRP-297、L-796449、LR-90、MK-0767及SB-219994),二肽酰肽酶IV(DPP-IV)抑制剂(西格列汀(sitagliptin)、维格列汀(vidagliptin)、阿格列汀(alogliptin)、利格列汀(linagliptin)及沙格列汀(saxagliptin)),胰高血糖素样肽-1(GLP-1)激动剂(乙先素-3(exendin-3)与乙先素-4(exendin-4)),蛋白酪氨酸磷酸酶-1B(PTP1B)抑制剂(曲度奎明、海提索萃取物及由Zhang,S.等人,现代药物发现,12(9/10),373-381(2007)所公开的化合物),胰岛素,胰岛素拟似物,肝糖磷酸化酶抑制剂,VPAC2受体激动剂,葡萄糖激酶活化剂,糖原磷酸化酶抑制剂,葡萄糖-6-磷酸酶抑制剂,αP2抑制剂,乙酰基-CoA羧化酶-2(ACC-2)抑制剂,磷酸二酯酶(PDE)-10抑制剂,二酰基甘油酰基转移酶(DGAT)1或2抑制剂,葡萄糖转运载体4(GLUT4)调节剂及谷氨酰胺-果糖-6-磷酸酰胺转移酶(GFAT)抑制剂。
其中,本发明所述的抗高血糖试剂包括,但并不限于SGLT2抑制剂(例如达格列净(dapagliflozin)、卡格列净(canagliflozin)、托格列净(tofogliflozin)、伊格列净(ipragliflozin)、鲁格列净(luseogliflozin)、恩格列净(empagliflozin)),双胍类药物(例如苯乙双胍(phenformin)、二甲双胍(metformin)),磺酰脲类药物(例如醋磺环已脲(acetohexamide)、氯磺丙脲(chlorpropamide)、格列本脲(glibenclamide,优降糖)、格列吡嗪(glipizide,吡磺环已脲)、格列齐特(gliclazide,达美康)、格列美脲(glimepiride)、格列戊脲(glipentide)、格列喹酮(gliquidone)、妥拉磺脲(tolazamide)、甲苯磺丁脲(tolbutamide)及氯茴苯酸(meglitinide)),格列奈类药物(例如瑞格列奈(repaglinide)及那格列奈(nateglinide)),α-葡萄糖苷水解酶抑制剂(例如阿卡波糖(acarbose)),α-葡萄糖苷酶抑制剂(例如酯解素、卡格列波糖(camiglibose)、乙格列酯(emiglitate)、米格列醇(miglitol)、伏格列波糖(voglibose)、普那米星(pradimicin)及沙玻制菌素(salbostatin)),PPAR激动剂(例如巴格列酮(balaglitazone)、环格列酮(ciglitazone)、达格列酮(darglitazone)、恩格列酮(englitazone)、伊沙列酮(isaglitazone)、吡格列酮(pioglitazone)、罗格列酮(rosiglitazone)及曲格列酮(troglitazone)),PPARα/γ 双激活剂(例如CLX-0940、GW-1536、GW-1929、GW-2433、KRP-297、L-796449、LR-90、MK-0767及SB-219994),二肽基肽酶IV(DPP-IV)抑制剂(例如西格列汀(sitagliptin)、维格列汀(vidagliptin)、阿格列汀(alogliptin)及沙格列汀(saxagliptin)),胰高血糖素样肽-1(GLP-1)激动剂(乙先素-3(exendin-3)与乙先素-4(exendin-4)),蛋白质酪氨酸磷酸酶-1B(PTP1B)抑制剂(曲度奎明、海提索萃取物及由Zhang,S.等人,现代药物发现,12(9/10),373-381(2007)所公开的化合物),胰岛素,胰岛素拟似物,肝糖磷酸化酶抑制剂,VPAC2受体激动剂,葡萄糖激酶活化剂,糖原磷酸化酶抑制剂,葡萄糖-6-磷酸酶抑制剂,αP2抑制剂,乙酰基-CoA羧化酶-2(ACC-2抑制剂),磷酸二酯酶(PDE)-10抑制剂,二酰基甘油酰基转移酶(DGAT)1或2抑制剂,葡萄糖转运载体4(GLUT4)调节剂及谷氨酰胺-果糖-6-磷酸酰胺转移酶(GFAT)抑制剂。
其中,本发明所述的抗肥胖症药物包括,但不限于中枢抗肥胖药(例如右旋芬氟拉明(dexfenfluramine)、芬氟拉明(fenfluramine)、苯叔丁胺(phentermine)、西布茶明(sibutramine)、安非拉酮(amfepramone)、右旋***(d-amphetamine)、氯苯咪吲哚(mazidol)、苯丙醇胺(phenylpropanolamine)、氯苄***(clobenzorex)),MCH受体拮抗剂(例如WO06035967记载的化合物,SB-568849;SNAP-7941、T-226296),神经肽Y受体拮抗剂(例如CP-422935),***素受体拮抗剂(例如利莫那班(rimonabant)、SR-147778),脑肠肽拮抗剂,脂肪酶抑制剂(例如奥利司他(orlistat)、ATL-962),β3激动剂(例如AJ-9677、AZ40140),11β-HSD1抑制剂(例如BVT-3498,INCB13739),DGAT-1抑制剂,肽性食欲抑制剂(例如瘦素(Leptin)、CNTF(睫状神经营养因子)),缩胆囊肽激动剂(例如林替曲特(Lintitript))及摄食抑制剂(例如P-57)。
其中,本发明所述的降脂试剂包括,但并不限于MTP抑制剂,HMG CoA还原酶抑制剂,角鲨烯合成酶抑制剂,贝特类降血脂药物(贝丁酸类降血脂药物),ACAT抑制剂,脂加氧酶抑制剂,胆固醇吸收抑制剂,回肠钠离子/胆汁酸协同转运蛋白抑制剂,LDL受体活性的向上调节物,胆汁酸螯合物或者烟酸类降血脂药物。其中一些实施例是,所述的降脂试剂选自普伐他汀、辛伐他汀、阿伐他汀、氟伐他汀、西立伐他汀、埃塔伐他汀或者罗素他汀。其中,所述的抗肥胖症试剂选自CB-1拮抗剂(例如利莫那班(rimonabant)、泰伦那班(taranabant)、溴乙那班(surinabant)、奥特那班(otenabant)、SLV319与AVE1625),肠-选择性MTP抑制剂(例如地洛他派(dirlotapide)、米曲他匹德(mitratapide)及英普他派(implitapide)),CCKa激动剂,5HT2c激动剂(例如氯卡色林(lorcaserin)),MCR4激动剂,脂肪酶抑制剂(例如赛利司他(Cetilistat)),PYY 3-36,类阿片拮抗剂(例如纳曲酮(naltrexone)),油酰基-雌酮,奥尼匹肽(obinepitide),普兰林肽(pramlintide),特索芬辛(tesofensine),勒帕茄碱,利拉鲁肽(liraglutide),溴麦角环肽,奥利司他(orlistat),艾塞那肽(exenatide),AOD-9604及***(sibutramide)。
其中,本发明所述的适当消炎剂包括生殖道/尿道感染预防与治疗药品,例如酸果蔓(Vaccinium macrocarpon)与酸果蔓衍生物,譬如酸果蔓汁液、酸果蔓萃液或酸果蔓的黄酮醇类。此外,其他的适当消炎剂还包括,但并不限于阿司匹林、非类固醇消炎药、糖皮质类固醇、柳氮磺吡啶和环氧酶II选择性抑制剂等。
本发明化合物及组合物的用途
本发明的化合物或药物组合物中化合物的量可以有效地可探测地抑制钠依赖性葡萄糖转运蛋白(sodium-dependent glucose transporters,SGLTs)的活性,尤其对SGLT1活性抑制明显。因此,本发明的化合物将应用于糖尿病和相关疾病的预防、治疗或者改善这些疾病的症状。
本发明的化合物将应用于,但绝不限于,使用本发明的化合物或药物组合物的有效量对患者给药来预防或治疗患者糖尿病和相关疾病,或者减轻糖尿病和相关疾病症状,或者延缓糖尿病和相关疾病的发展或发作或用于增加高密度脂蛋白的水平。这样的疾病包括,但并不限于糖尿病,尤其是II型糖尿病,以及胰岛素抵抗、高血糖、高胰岛素血症、高脂血症如高甘油三酯血症、糖尿病并发症,如糖尿病性视网膜病、糖尿病性神经病或糖尿病性肾病,肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压。
本发明化合物具有优异的肠内环境改善作用,能使双歧杆菌、乳杆菌等的有益菌增加,使肠内的有机酸增加,使肠内的腐败产物减少。通过盖上肠内环境,可以改善与肠内环境变化相关联的疾病。其中,所述“与肠内环境变化相关联的疾病”包含,但不限于,慢性肾病、伪膜性肠炎/出血性肠炎、感染性肠炎、 溃疡性大肠炎、克罗恩病、肠易激综合征、肥胖、动脉硬化、高血压、吉兰-巴雷综合征、过敏性疾病、糖尿病、多发性硬化症、自身免疫疾病、酒精性肝功能障碍、非酒精性脂肪性肝病、非酒精性脂肪肝炎、非甾体抗炎药起因性的肠炎、压力、郁闷、流行性感冒、牙周病、癌、花粉症、功能性消化不良、瘙痒症等。
此外,本发明化合物或药物组合物还适于预防和治疗糖尿病性后期损伤,例如肾病、视网膜病、神经病、以及心肌梗塞、外周动脉闭合疾病、血栓形成、动脉硬化、炎症、免疫疾病、自身免疫性疾病如AIDS、哮喘、骨质疏松症、癌症、牛皮癣、阿尔茨海默氏症、精神***症和感染性疾病。
本发明的化合物或药学上可接受的药物组合物的“有效量”、“有效治疗量”或“有效剂量”是指处理或减轻一个或多个本发明所提到病症的严重度的有效量。本发明的化合物或药学上可接受的药物组合物在相当宽的剂量范围内是有效的。例如,每天服用的剂量约在0.1mg-1000mg/人范围内,分为一次或数次给药。根据本发明的方法、化合物和药物组合物可以是任何给药量和任何给药途径来有效地用于处理或减轻疾病的严重程度。必需的准确的量将根据患者的情况而改变,这取决于种族,年龄,患者的一般条件,感染的严重程度,特殊的因素,给药方式等。本发明的化合物或药物组合物可以和一个或多个其他治疗剂联合给药,如本发明所讨论的。
一般合成和检测方法
为描述本发明,以下列出了实施例。但需要理解,本发明不限于这些实施例,只是提供实践本发明的方法。
在本说明书中,如果在化学名称和化学结构间存在任何差异,结构是占优的。
一般地,本发明的化合物可以通过本发明所描述的方法制备得到,除非有进一步的说明,其中取代基的定义如式(I)、式(I-a)或式(II)所示。下面的反应方案和实施例用于进一步举例说明本发明的内容。
所属领域的技术人员将认识到:本发明所描述的化学反应可以用来合适地制备许多本发明的其他化合物,且用于制备本发明的化合物的其它方法都被认为是在本发明的范围之内。例如,根据本发明那些非例证的化合物的合成可以成功地被所属领域的技术人员通过修饰方法完成,如适当的保护干扰基团,通过利用其他已知的药物除了本发明所描述的,或将反应条件做一些常规的修改。另外,本发明所公开的反应或已知的反应条件也公认地适用于本发明其他化合物的制备。
化合物的结构是通过核磁共振( 1H-NMR、 13C-NMR或/和 19F-NMR)来确定的。 1H-NMR、 13C-NMR、 19F-NMR化学位移(δ)以百万分之一(ppm)的单位给出。 1H-NMR、 13C-NMR、 19F-NMR的测定是用Bruker Ultrashield-400核磁共振谱仪和Bruker Avance III HD 600核磁共振谱仪,测定溶剂为氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD或MeOH-d 4)或者氘代二甲基亚砜(DMSO-d 6)。用TMS(0ppm)或氯仿(7.25ppm)作为参照标准。当出现多重峰的时候,将使用下面的缩写:s(singlet,单峰),d(doublet,双峰),t(triplet,三重峰),m(multiplet,多重峰),br(broadened,宽峰),dd(doublet of doublets,双二重峰),dt(doublet of triplets,双三重峰),td(triplet of doublets,三双重峰),brs(broadened singlet,宽单峰)。偶合常数J,单位用赫兹(Hz)表示。
制备纯化或制备拆分一般使用Novasep pump 250高效液相色谱仪。
LC-MS的测定用Agilen-6120 Quadrupole LC/MS质谱仪。
柱层析一般使用青岛海洋化工300目~400目硅胶为载体。
本发明的起始原料是已知的,并且可以在市场上购买到得,购买自上海韶远公司(Shanghai Accela Company)、安耐吉公司(Energy Company)、百灵威公司(J&K)、天津阿法埃莎公司(Alfa Company)等公司,或者可以采用或者按照本领域已知的方法来合成。
实施例中无特殊说明,反应均在氮气氛围下进行;
氮气氛围是指反应瓶连接一个约1L容积的氮气气球或钢釜;
氢气氛围是指反应瓶连接一个约1L容积的氢气气球或者是一个约1L容积的不锈钢高压反应釜;
实施例中若无特殊说明,溶液是指水溶液;
实施例中若无特殊说明,反应温度为室温;
实施例中若无特殊说明,室温为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂的体系有:二氯甲烷和甲醇体系,二氯甲烷和乙酸乙酯体系,石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节。
柱层析的洗脱剂的体系包括:A:石油醚和乙酸乙酯体系,B:二氯甲烷和乙酸乙酯体系,C:二氯甲烷和甲醇体系。溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的氨水和醋酸等进行调节。
下面简写词的使用贯穿本发明:
DMSO-d 6:   氘代二甲基亚砜;   CDCl 3:       氘代氯仿;         CD 3OD:     氘代甲醇;
D 2O:       重水;             Allyl:       烯丙基;           Br:       溴;
Mg:        镁;               Cbz:         苄氧羰基;         Ac:       乙酰基;
Bn:        苄基;             Et:          乙基;             Me:        甲基;
Ms:        甲磺酰基;         Boc:         叔丁氧羰基;       PMB:       对甲氧基苄基;
HCl:       氯化氢;           MeOH:       甲醇;             mL:        毫升;
μL:      微升;             M,mol/L:     摩尔/每升;        mmol:      毫摩尔;
g:         克;               mol:         摩尔;             h:         小时;
H 2:        氢气;             min:         分钟;             N 2:        氮气;
MPa:       兆帕;             atm:         标准大气压;       DCM:       二氯甲烷;
HBTU:      O-苯并三氮唑-四甲基脲六氟磷酸酯;
HATU:      O-(7-氮杂苯并***-1-基)-N,N,N,N-四甲基脲六氟磷酸酯;
DBU:       1,8-二氮杂双环[5.4.0]十一碳-7-烯。
一般合成方法
制备本发明公开化合物的典型合成步骤如下面的合成方案1~6所示。除非另外说明,R 2、R 3、R 6、R 7、R c、R d和t具有如本发明所述的定义。
合成方案1:
Figure PCTCN2019073550-appb-000014
(6)所示中间体可以通过合成方案1所公开的方法合成。首先,化合物 (1)与氯甲酸苄酯反应生成化合物 (2)。化合物 (2)与化合物 (3)发生缩合反应生成化合物 (4)。然后,化合物 (4)经催化氢化脱掉氨基上的Cbz保护基生成化合物 (5)。最后,化合物 (5)与氯化氢成盐得到化合物 (6)
合成方案2:
Figure PCTCN2019073550-appb-000015
(10)所示中间体可以通过合成方案2所公开的方法合成。首先,化合物 (7)经还原剂还原生成化合物 (8)。然后,化合物 (8)与烯丙基溴在碱性条件下发生取代反应生成化合物 (9)。最后,化合物 (9)与金属Mg反应得到化合物 (10)
合成方案3:
Figure PCTCN2019073550-appb-000016
(15)所示中间体可以通过合成方案3所公开的方法合成。首先,化合物 (11)与草酰氯反应生成化合物 (12)。然后,化合物 (12)在碱性条件下与二甲羟胺盐酸盐反应生成化合物 (13)。接着,化合物 (13)与化合物 (10)发生格氏反应生成化合物 (14)。最后,化合物 (14)在酸和三乙基硅烷作用下发生还原反应得到化合物 (15)
合成方案4:
Figure PCTCN2019073550-appb-000017
(29)所示中间体或式 (30)所示的中间体可以通过合成方案4所公开的方法合成。首先,化合物 (16)在2,2,6,6-四甲基哌啶氧化物存在下发生催化氧化生成化合物 (17)。化合物 (17)与乙酸酐反应生成化合物 (18)。化合物 (15)与异丙基氯化镁作用后和化合物 (18)经格氏反应生成化合物 (19)。化合物 (19)在酸性条件下与甲醇发生反应生成化合物 (20)。化合物 (20)被氧化剂氧化生成化合物 (21)。化合物 (21)在DBU作用下与甲醛发生反应生成化合物 (22)。然后,化合物 (22)与硼氢化钠发生还原反应生成化合物 (23)。化合物 (23)在酸性条件下关环得到化合物 (24)。化合物 (24)与戴斯马丁氧化剂发生氧化反应得到化合物 (25)。化合物 (25)在酸性条件下与甲醇发生反应生成化合物 (26)。接着,化合物 (26)在低 温条件下与格氏试剂反应生成化合物 (27)。化合物 (27)在催化剂作用下脱去羟基保护基烯丙基生成化合物 (28)。最后,化合物 (28)在催化剂2,2,6,6-四甲基哌啶氧化物存在下与碘苯二乙酸发生氧化反应得到化合物 (29)。化合物 (29)经催化氢化脱去羟基保护基Bn得到化合物 (30)
合成方案5:
Figure PCTCN2019073550-appb-000018
(32)所示化合物可以通过合成方案5所公开的方法合成。首先,化合物 (29)与化合物 (6)发生缩合反应生成化合物 (31)。然后,化合物 (31)经催化氢化脱去羟基保护基Bn得到化合物 (32)
合成方案6:
Figure PCTCN2019073550-appb-000019
(32)所示化合物也可以通过合成方案6所公开的方法合成。首先,化合物 (30)在碱性条件下与乙酸酐反应生成化合物 (33)。接着,化合物 (33)与化合物 (6)发生缩合反应生成化合物 (34)。最后,化合化合物 (34)在碱性条件下脱去羟基保护基Ac得到化合物 (32)
实施例
实施例1 2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰氨基]-N-(2-吡咯烷-1-基乙基)丙酰胺
Figure PCTCN2019073550-appb-000020
步骤1:N-[1,1-二甲基-2-氧代-2-(2-吡咯烷-1-基乙基氨基)乙基]氨基甲酸苄酯
Figure PCTCN2019073550-appb-000021
在室温下,将2-(苄氧基羰基氨基)-2-甲基-丙酸(3.0g,13mmol)溶于二氯甲烷(30mL)中并冷却至0℃,加入HATU(5.6g,14mmol)和N,N-二异丙基乙胺(7.0mL,40mmol),搅拌20分钟。加入2-吡咯烷-1-基乙胺(1.9g,17mmol),升至室温搅拌过夜。用水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物 经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(2.2g,黄色油状物),产率52%。MS(ESI,pos.ion)m/z:334.4[M+H] +
步骤2:2-氨基-2-甲基-N-(2-吡咯烷-1-基乙基)丙酰胺
Figure PCTCN2019073550-appb-000022
在室温下,将N-[1,1-二甲基-2-氧代-2-(2-吡咯烷-1-基乙基氨基)乙基]氨基甲酸苄酯(2.0g,6.0mmol)溶于四氢呋喃(2mL)和无水甲醇(20mL)的混合溶剂中,加入10%钯/碳(0.20g,0.18mmol),氢气氛围下搅拌过夜。抽滤,浓缩,得到标题化合物(1.2g,黄色油状物),产率99%。
步骤3:2-氨基-2-甲基-N-(2-吡咯烷-1-基乙基)丙酰胺二盐酸盐
Figure PCTCN2019073550-appb-000023
在室温下,将2-氨基-2-甲基-N-(2-吡咯烷-1-基乙基)丙酰胺(1.2g,6.0mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(4mL,5M),搅拌1小时。抽滤,滤饼用乙酸乙酯(20mL)淋洗,真空干燥得到标题化合物(1.2g,白色固体),产率73%。
MS(ESI,pos.ion)m/z:200.3[M+H] +
1H NMR(400MHz,D 2O)δ(ppm):3.65(m,2H),3.56(t,2H),3.33(t,2H),3.06(m,2H),2.08(m,2H),1.95(m,2H),1.54(s,6H)。
步骤4:4-(4-溴苯基)丁基-1-醇
Figure PCTCN2019073550-appb-000024
在室温下,将4-(4-溴苯基)丁酸(50.0g,150mmol)溶于四氢呋喃(250mL)中,氮气氛围下冷却至-10℃,滴加硼烷的四氢呋喃溶液(1.0M,300mL,300mmol),升至室温搅拌2小时。倒入冰水(500mL)中,乙酸乙酯(500mL)萃取,有机相用饱和食盐水(200mL)洗涤,无水硫酸钠干燥,抽滤浓缩得到标题化合物(47.0g,无色油状物),产率:98%。
步骤5:1-(4-烯丙氧基丁基)-4-溴-苯
Figure PCTCN2019073550-appb-000025
在室温下,将4-(4-溴苯基)丁基-1-醇(47.0g,205mmol)溶于四氢呋喃(500mL)中,氮气氛围下冷却至-10℃,分批加入氢化钠(11.0g,275mmol),继续搅拌30分钟,滴加烯丙基溴(33.6g,288mmol),升至室温搅拌过夜。倒入冰水(1.0L)中淬灭反应,乙酸乙酯(500mL)萃取,有机相用饱和食盐水(200mL)洗涤,无水硫酸钠干燥,抽滤浓缩。残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/40],得到标题化合物(33.0g,无色油状物),产率:60%。
步骤6:[4-(4-烯丙氧基丁基)苯基]-溴化镁
Figure PCTCN2019073550-appb-000026
在室温下,向反应瓶中依次加入镁条(6.4g,0.26mol)和碘(0.6g,2mmol),将1-(4-烯丙氧基丁基)-4-溴-苯(59.0g,219mmol)溶于四氢呋喃(300mL)中并在氮气氛围下加入其中的10mL溶液,加热至反应引发(碘颜色消失),然后滴加剩余的溶液,在65℃下搅拌20分钟,得到标题化合物(65g,褐色溶液),产率以100%计算,直接进入下一步。
步骤7:5-碘-2-甲基-苯甲酰氯
Figure PCTCN2019073550-appb-000027
在室温下,将5-碘-2-甲基-苯甲酸(50.0g,191mmol)溶于二氯甲烷(500mL)中,氮气氛围下冷却至-10℃,依次滴加草酰氯(25mL,0.29mol)和N,N-二甲基甲酰胺(1.5mL,19mmol),室温下搅拌过夜。浓缩,得到标题化合物(53g,黄色固体),产率100%。
步骤8:5-碘-N-甲氧基-N,2-二甲基-苯甲酰胺
Figure PCTCN2019073550-appb-000028
在室温下,向反应瓶中依次加入5-碘-2-甲基-苯甲酰氯(53.0g,189mmol),二甲羟胺盐酸盐(37.0g,379mmol)和二氯甲烷(500mL),氮气氛围下冷却至0℃,滴加三乙胺(106mL,761mmol),室温下搅拌3.5小时。用饱和食盐水(500mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(54g,黄色油状物),产率93%。
MS(ESI,pos.ion)m/z:306.0[M+H] +
步骤9:[4-(4-烯丙氧基丁基)苯基]-5-碘-2-甲基-苯基)甲基酮
Figure PCTCN2019073550-appb-000029
在室温下,将5-碘-N-甲氧基-N,2-二甲基-苯甲酰胺(50.0g,164mmol)的四氢呋喃(200mL)溶液冷却至-20℃,氮气氛围下,滴加[4-(4-烯丙氧基丁基)苯基]-溴化镁(63.0g,215mmol),在-20℃搅拌1小时,室温下搅拌过夜。冷却至0℃,滴加饱和氯化铵溶液(400mL)淬灭,乙酸乙酯(300mL×2)萃取,合并的有机相用饱和食盐水(300mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/30],得到标题化合物(59.0g,无色油状物),产率83%。
步骤10:2-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-碘-1-甲基-苯
Figure PCTCN2019073550-appb-000030
在室温下,向反应瓶中依次加入[4-(4-烯丙氧基丁基)苯基]-5-碘-2-甲基-苯基)甲基酮(59.0g,136mmol)和三氟乙酸(150mL),氮气氛围下冷却至0℃,依次滴加三乙基硅烷(174mL,1.09mol)和三氟甲磺酸(12.5mL,141mmol),室温搅拌1小时。浓缩,残留物溶于乙酸乙酯(600mL)中,依次用水(500mL)、饱和碳酸氢钠溶液(500mL)和饱和食盐水(500mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[石油醚],得到标题化合物(57g,黄色油状物),产率99%。
1H NMR(400MHz,CDCl 3)δ(ppm):7.52–7.47(m,2H),7.13(d,2H),7.04(d,2H),6.92(d,1H),5.95(m,1H),5.30(dd,1H),5.20(dd,1H),4.02–3.97(m,2H),3.91(s,2H),3.48(t,2H),2.64(t,2H),2.21(s,3H),1.76–1.63(m,4H)。
步骤11:(3R,4S,5R,6R)-3,4,5-三苄氧基-6-(苄氧基甲基)四氢吡喃-2-酮
Figure PCTCN2019073550-appb-000031
在室温下,向反应瓶中依次加入(3R,4S,5R,6R)-3,4,5-三苄氧基-6-(苄氧基甲基)四氢吡喃-2-醇(130g,240mmol)、碳酸氢钠(80.0g,952mmol)、二氯甲烷(1000mL)和水(800mL)。冷却至0℃,加入溴化钾(18.0g,151mmol)和2,2,6,6-四甲基哌啶氧化物(5.6g,36mmol),再一次性加入次氯酸钠溶液(360g,有效氯6.2%,有效氯629mmol),搅拌20分钟。分液,有机相用饱和食盐水(500mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(129g,黄色油状物)。产率100%。
步骤12:[(2R,3R,4S,5R)-3,4,5-三苄氧基-6-氧代-四氢吡喃-2-基]甲基乙酸酯
Figure PCTCN2019073550-appb-000032
在室温下,将冰醋酸(350mL)加入(3R,4S,5R,6R)-3,4,5-三苄氧基-6-(苄氧基甲基)四氢吡喃-2-酮(130g,241mmol)的乙酸酐(200mL,2.12mol)溶液中,氮气氛围下降温至-15℃,滴加浓硫酸(14.0mL,263mmol),控温-15℃~-10℃下搅拌2.5小时。倒入冰水(1.5L)中,乙酸乙酯(1.0L)萃取,有机相依次用水(500mL)、饱和碳酸氢钠溶液(1.0L)和饱和食盐水(500mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/4],得到标题化合物(85g,无色油状物),产率71%。
步骤13:[(2R,3R,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-6-羟基-四氢 吡喃-2-基]甲基乙酸酯
Figure PCTCN2019073550-appb-000033
在室温下,将2-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-碘-1-甲基-苯(30.0g,71.4mmol)溶于四氢呋喃(150mL)中,氮气氛围下冷却至-10℃,滴加异丙基氯化镁的四氢呋喃溶液(39mL,78mmol,2.0M),搅拌1.5小时,然后在氮气氛围下将所得混合物滴加至-10℃的[(2R,3R,4S,5R)-3,4,5-三苄氧基-6-氧代-四氢吡喃-2-基]甲基乙酸酯(25.0g,50.9mmol)的四氢呋喃(150mL)溶液中,搅拌2.5小时。饱和氯化铵溶液(200mL)淬灭反应,乙酸乙酯(300mL×2)萃取,合并的有机相用饱和食盐水(500mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/4],得到标题化合物(32.1g,黄色油状物),产率80%。
步骤14:[(2R,3R,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-6-甲氧基-四 氢吡喃-2-基]甲醇
Figure PCTCN2019073550-appb-000034
在室温下,将[(2R,3R,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-6-羟基-四氢吡喃-2-基]甲基乙酸酯(28.0g,35.6mmol)溶于无水甲醇(300mL)中,加入浓盐酸(9.0mL,0.11mol),搅拌3小时。加入乙酸乙酯(500mL),依次用水(500mL)、饱和碳酸氢钠溶液(200mL)和饱和食盐水(200mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/4],得到标题化合物(23.0g,无色油状物),产率85%。
1H NMR(400MHz,CDCl 3)δ(ppm):7.46–7.27(m,15H),7.20-7.05(m,7H),5.99(m,1H),5.35(dd,1H),5.25 (t,1H),5.02(dt,3H),4.81(d,1H),4.53(d,1H),4.30(t,1H),4.14–3.75(m,10H),3.55–3.44(m,3H),3.24(s,3H),2.67(t,2H),2.34(s,3H),1.79–1.67(m,4H)。
步骤15:[(2S,3S,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-6-甲氧基-四氢 吡喃-2-基]甲醛
Figure PCTCN2019073550-appb-000035
在室温下,向反应瓶中依次加入[(2R,3R,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-6-甲氧基-四氢吡喃-2-基]甲醇(23.0g,30.4mmol)、碳酸氢钠(16.0g,190mmol)、二氯甲烷(250mL)和水(160mL),降温至0℃,依次加入溴化钾(2.4g,20mmol),2,2,6,6-四甲基哌啶氧化物(0.75g,4.8mmol)和次氯酸钠溶液(52g,有效氯5.53%,有效氯81mmol),搅拌15分钟。分液,有机相用饱和食盐水(200mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(22g,黄色油状物),产率96%。
步骤16:(2R,3S,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-2-(羟甲基)-6- 甲氧基-四氢吡喃-2-甲醛
Figure PCTCN2019073550-appb-000036
在室温下,将[(2S,3S,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-6-甲氧基-四氢吡喃-2-基]甲醛(23.0g,30.4mmol)溶于N,N-二甲基甲酰胺(200mL)中并降温至0℃,加入37%甲醛溶液(62.0g,764mmol)和DBU(3.1g,20mmol),升至室温搅拌过夜。加入乙酸乙酯(300mL),依次用水(400mL)和饱和食盐水(200mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(23.0g,黄色油状物),产率96%。
步骤17:[(3S,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-2-(羟甲基)-6-甲 氧基-四氢吡喃-2-基]甲醇
Figure PCTCN2019073550-appb-000037
在室温下,将(2R,3S,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-2-(羟甲基)-6-甲氧基-四氢吡喃-2-甲醛(23.0g,29.3mmol)溶于甲醇(200mL)中并降温至0℃,分批加入硼氢化钠(2.8g,74mmol),继续搅拌10分钟。加入乙酸乙酯(500mL),依次用水(500mL)和饱和食盐水(200mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(23.0g,黄色油状物),产率100%。
步骤18:[(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂双 环[3.2.1]辛烷-1-基]甲醇
Figure PCTCN2019073550-appb-000038
在室温下,将[(3S,4S,5R,6S)-6-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-3,4,5-三苄氧基-2-(羟甲基)-6-甲氧基-四氢吡喃-2-基]甲醇(23.0g,29.2mmol)溶于四氢呋喃(150mL)中,加入一水合对甲苯磺酸(7.0g,37mmol),搅拌过夜。浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/8)],得到标题化合物(12.5g,黄色油状物),产率57%。
步骤19:(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂双 环[3.2.1]辛烷-1-甲酸
Figure PCTCN2019073550-appb-000039
在室温下,将[(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂双环[3.2.1]辛烷-1-基]甲醇(12.5g,16.6mmol)溶于二氯甲烷(130mL)中并降温至0℃,加入戴斯马丁氧化剂(50.0g,117mmol),加热至40℃反应2小时。浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/2],得到标题化合物(10g,白色固体),产率78%。
步骤20:(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂双 环[3.2.1]辛烷-1-甲酸甲酯
Figure PCTCN2019073550-appb-000040
在室温下,将(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂双环[3.2.1]辛烷-1-甲酸(10.0g,13.0mmol)溶于无水甲醇(100mL)中,加入浓硫酸(1.0mL,19mmol),加热至40℃搅拌过夜。加入乙酸乙酯(200mL),依次用水(300mL)和饱和碳酸氢钠溶液(100mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/10],得到标题化合物(7.5g,无色油状物),产率74%。
步骤21:2-[(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂 双环[3.2.1]辛烷-1-基]丙烷-2-醇
Figure PCTCN2019073550-appb-000041
在室温下,将(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂双环[3.2.1]辛烷-1-甲酸甲酯(7.5g,9.6mmol)溶于四氢呋喃(80mL)中,氮气氛围下降温至0℃,滴加甲基溴化镁***溶液(20mL,60mmol,3.0M),升至室温搅拌4小时。降温至0℃,滴加饱和氯化铵溶液(100mL)淬灭,乙酸乙酯(100mL×2)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(7.5g,无色油状物),产率99%。
步骤22:4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三苄氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛 烷-5-基]苯基]甲基]苯基]丁烷-1-醇
Figure PCTCN2019073550-appb-000042
在室温下,将2-[(1S,2S,3S,4R,5S)-5-[3-[[4-(4-烯丙氧基丁基)苯基]甲基]-4-甲基-苯基]-2,3,4-三苄氧基-6,8-二氧杂双环[3.2.1]辛烷-1-基]丙烷-2-醇(7.0g,8.9mmol)溶于无水甲醇(80mL)中,加入二氯化钯(1.0g,5.6mmol),氮气氛围下室温搅拌2小时。抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/2],得到标题化合物(5.1g,无色油状物),产率77%。
步骤23:4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三苄氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛 烷-5-基]苯基]甲基]苯基]丁酸
Figure PCTCN2019073550-appb-000043
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三苄氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁烷-1-醇(2.0g,2.7mmol)溶于二氯甲烷(20mL)中,加入水(4mL)、2,2,6,6-四甲基哌啶氧化物(0.13g,0.79mmol)和碘苯二乙酸(2.2g,6.7mmol),搅拌过夜。分液,有机相用无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/2],得到标题化合物(1.7g,白色固体),产率83%。
步骤24:2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三苄氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环 [3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]-N-(2-吡咯烷-1-基乙基)丙酰胺
Figure PCTCN2019073550-appb-000044
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三苄氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.80g,1.05mmol)溶于N,N-二甲基甲酰胺(6mL)中并降温至0℃,加入HBTU(0.50g,1.3mmol)和N,N-二异丙基乙胺(1.0mL,5.7mmol),搅拌20分钟。加入2-氨基-2-甲基-N-(2-吡咯烷-1-基乙基)丙酰胺二盐酸盐(0.38g,1.4mmol),搅拌过夜。加入水(60mL),乙酸乙酯(100mL×3)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(0.99g,无色油状物),产率99%。
步骤25:2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环 [3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰氨基]-N-(2-吡咯烷-1-基乙基)丙酰胺
Figure PCTCN2019073550-appb-000045
在室温下,将2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三苄氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]-N-(2-吡咯烷-1-基乙基)丙酰胺(0.90g,0.96mmol) 溶于苯甲醚(30mL)中并降温至0℃,缓慢加入无水三氯化铝(1.3g,9.7mmol),搅拌5分钟,升至室温搅拌3小时。倒入冰水(50mL)中,乙酸乙酯(30mL×6)萃取,合并的有机相依次用饱和碳酸氢钠溶液(100mL)和饱和食盐水(100mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=7/1],得到标题化合物(0.28g,白色固体),产率44%。
MS(ESI,pos.ion)m/z:668.5[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):8.65(br.s,1H),8.40(s,1H),7.89(t,1H),7.29(s,1H),7.23(d,1H),7.12–7.04(m,5H),5.50(d,1H),5.32(t,1H),5.02(d,1H),4.89(d,1H),4.21(s,1H),4.03(d,1H),3.91(s,2H),3.81(d,1H),3.72(d,1H),3.50(m,2H),3.19(m,2H),3.02(s,2H),2.18(s,3H),2.13(t,2H),2.04–1.95(m,4H),1.88–1.69(m,4H),1.29(s,6H),1.21(s,3H),1.15(s,3H)。
实施例2 N-[3-(2-甲基氨基乙基氨基)-3-氧代-丙基]-4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰胺
Figure PCTCN2019073550-appb-000046
步骤1:3-(苄氧基羰基氨基)丙酸
Figure PCTCN2019073550-appb-000047
在室温下,将3-氨基丙酸(3.0g,34mmol)溶于二氧六环(20mL)中,加入碳酸钠(11.0g,104mmol)和水(60mL),冷却至0℃,加入氯甲酸苄酯(6.0mL,41mmol),升至室温搅拌过夜。正己烷(100mL)洗涤,水相用稀盐酸(1M)调节pH=1,乙酸乙酯(100mL×2)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(6.9g,无色油状物),产率92%。
步骤2:N-[3-(2-二甲基氨基乙基氨基)-3-氧代-丙基]氨基甲酸苄基酯
Figure PCTCN2019073550-appb-000048
在室温下,将3-(苄氧基羰基氨基)丙酸(3.0g,13mmol)溶于二氯甲烷(30mL)中并降温至0℃,加入HATU(5.9g,15mmol)和N,N-二异丙基乙胺(7.0mL,40mmol),搅拌20分钟。加入N,N-二甲基-1,2-乙二胺(1.5g,17mmol),升至室温搅拌过夜。水(20mL)洗涤,有机相浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(2.0g,黄色油状物),产率51%。
步骤3:3-氨基-N-(2-二甲基氨基乙基)丙酰胺
Figure PCTCN2019073550-appb-000049
在室温下,将N-[3-(2-二甲基氨基乙基氨基)-3-氧代-丙基]氨基甲酸苄基酯(2.0g,6.0mmol)溶于四氢呋喃(2mL)和无水甲醇(20mL)的混合溶剂中,加入10%钯/碳(0.20g,0.19mmol),氢气氛围下搅拌过夜。抽滤,浓缩,得到标题化合物(1.1g,黄色油状物),产率99%。
步骤4:3-氨基-N-(2-二甲基氨基乙基)丙酰胺二盐酸盐
Figure PCTCN2019073550-appb-000050
在室温下,将3-氨基-N-(2-二甲基氨基乙基)丙酰胺(1.1g,6.9mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(4mL,5M),搅拌1小时。抽滤,滤饼用乙酸乙酯(20mL)淋洗,真空干燥得到标题化合物(1.2g,白色固体),产率75%。
MS(ESI,pos.ion)m/z:160.2[M+H] +
1H NMR(400MHz,D 2O)δ(ppm):3.56(t,2H),3.24(dt,4H),2.88(s,6H),2.67(t,2H)。
步骤5:4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5- 基]苯基]甲基]苯基]丁酸
Figure PCTCN2019073550-appb-000051
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三苄氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(1.3g,1.7mmol)溶于四氢呋喃(2mL)和无水甲醇(20mL)的混合溶剂中,加入10%氢氧化钯/碳(1.2g,0.84mmol),氢气氛围下搅拌过夜。抽滤,浓缩,残留物经硅胶柱层析纯化[乙酸乙酯],得到标题化合物(0.52g,白色固体),产率53%。
步骤6:4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛 烷-5-基]苯基]甲基]苯基]丁酸
Figure PCTCN2019073550-appb-000052
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.40g,0.82mmol)溶于四氢呋喃(10mL)中并降温至10℃,加入4-二甲氨基吡啶(5mg,0.04mmol)、乙酸酐(0.70mL,7.5mmol)和三乙胺(1.0mL,7.2mmol),搅拌3小时。加入稀盐酸(1M)调节pH=3,乙酸乙酯(60mL×3)萃取,合并的有机相用饱和食盐水(80mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/1],得到标题化合物(0.35g,白色固体),产率69%。
MS(ESI,pos.ion)m/z:635.5[M+Na] +
步骤7:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[3-(2-甲基氨基乙基氨基)-3-氧代-丙基]氨基]-丁基]苯 基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯
Figure PCTCN2019073550-appb-000053
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.15g,0.25mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.12g,0.29mmol)和N,N-二异丙基乙胺(0.22mL,1.3mmol),搅拌20分钟。加入3-氨基-N-(2-二甲基氨基乙基)丙酰胺二盐酸盐(74mg,0.32mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.11g,白色固体),产率60%。
MS(ESI,pos.ion)m/z:754.6[M+H] +
步骤8:N-[3-(2-甲基氨基乙基氨基)-3-氧代-丙基]-4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基 -1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰胺
Figure PCTCN2019073550-appb-000054
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[3-(2-甲基氨基乙基氨基)-3-氧代-丙基]氨基]-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.11g,0.15mmol)溶于无水甲醇(2mL)中,加入甲醇钠固体(41mg,0.75mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=2/1],得到标题化合物(60mg,白色固体),产率65%。
MS(ESI,pos.ion)m/z:628.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):8.09(s,1H),7.89(t,1H),7.29(s,1H),7.23(d,1H),7.11–7.03(m,5H),5.50(d,1H),5.32(t,1H),5.02(d,1H),4.89(d,1H),4.21(s,1H),4.03(d,1H),3.91(s,2H),3.81(d,1H),3.72(d,1H),3.47(m,2H),3.24(m,2H),2.90(m,2H),2.60(s,6H),2.45(t,2H),2.26(t,2H),2.18(s,3H),2.02(t,2H),1.75(m,2H),1.21(s,3H),1.15(s,3H)。
实施例3 N-(2-二乙基氨基乙基)-2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丙酰胺
Figure PCTCN2019073550-appb-000055
步骤1:N-[2-(2-二乙基氨基乙基氨基)-1,1-二甲基-2-氧代-乙基]氨基甲酸苄基酯
Figure PCTCN2019073550-appb-000056
在室温下,将2-(苄氧基羰基氨基)-2-甲基-丙酸(3.0g,13mmol)溶于二氯甲烷(30mL)中并冷却至0℃,加入HATU(5.6g,14mmol)和N,N-二异丙基乙胺(7.0mL,40mmol),搅拌20分钟。加入N,N-二乙基-1,2-乙二胺(1.9g,16mmol),搅拌过夜。水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(3.8g,黄色油状物),产率90%。
MS(ESI,pos.ion)m/z:336.4[M+H] +
步骤2:2-氨基-N-(2-二乙基氨基乙基)-2-甲基-丙酰胺
Figure PCTCN2019073550-appb-000057
在室温下,将N-[2-(2-二乙基氨基乙基氨基)-1,1-二甲基-2-氧代-乙基]氨基甲酸苄基酯(1.0g,3.0mmol)溶于无水甲醇(20mL)中,加入10%钯/碳催化剂(0.20g,0.18mmol),氢气氛围下搅拌5小时。抽滤,浓缩,得到标题化合物(0.6g,黄色油状物),产率99%。
步骤3:2-氨基-N-(2-二乙基氨基乙基)-2-甲基-丙酰胺二盐酸盐
Figure PCTCN2019073550-appb-000058
在室温下,将2-氨基-N-(2-二乙基氨基乙基)-2-甲基-丙酰胺(0.50g,2.5mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(2mL,5M),搅拌1小时。抽滤,滤饼用乙酸乙酯(20mL)淋洗,真空干燥得 到标题化合物(0.6g,白色固体),产率90%。
MS(ESI,pos.ion)m/z:202.4[M+H] +
步骤4:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-(2-二乙氨基乙氨基)-1,1-二甲基-2-氧代乙基]氨 基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯
Figure PCTCN2019073550-appb-000059
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.15g,0.25mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.12g,0.29mmol)和N,N-二异丙基乙胺(0.22mL,1.3mmol),搅拌20分钟。加入2-氨基-N-(2-二乙基氨基乙基)-2-甲基-丙酰胺二盐酸盐(88mg,0.32mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(90mg,白色固体),产率46%。
MS(ESI,pos.ion)m/z:796.3[M+H] +
步骤5:N-(2-二乙基氨基乙基)-2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基- 乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丙酰胺
Figure PCTCN2019073550-appb-000060
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-(2-二乙氨基乙氨基)-1,1-二甲基-2-氧代乙基]氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(90mg,0.11mmol)溶于无水甲醇(2mL)中,加入甲醇钠固体(30mg,0.55mmol),搅拌2小时。浓缩,残留物直接经薄层色谱制备纯化[DCM/MeOH(v/v)=2/1],得到标题化合物(60mg,白色固体),产率79%。
MS(ESI,pos.ion)m/z:670.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):9.02(br.s,1H),08.23(s,1H),7.93(t,1H),7.29(s,1H),7.23(d,1H),7.17–6.99(m,5H),5.53(d,1H),5.04(d,1H),4.89(d,1H),4.21(s,1H),4.04(d,1H),3.91(s,2H),3.81(d,1H),3.72(m,1H),3.53–3.36(m,5H),3.11(t,2H),3.05(m,4H),2.18(s,3H),2.13(t,2H),1.75(m,2H),1.29(s,6H),1.24(s,3H),1.21(s,3H),1.16(s,6H)。
实施例4 N-(2-二甲氨基乙基)-3-甲基-3-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丁酰胺
Figure PCTCN2019073550-appb-000061
步骤1:3(苄氧基羰基氨基)-3-甲基-丁酸
Figure PCTCN2019073550-appb-000062
在室温下,将3-氨基-3-甲基-丁酸(3.0g,26mmol)溶于二氧六环(20mL)中,加入碳酸钠(8.1g,76mmol)和水(60mL),冷却至0℃,加入氯甲酸苄酯(4.5mL,30mmol),升至室温搅拌过夜。正己烷(100mL)洗涤,水相用稀盐酸(1M)调节pH=1,乙酸乙酯(100mL×2)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(5.4g,无色油状物),产率84%。
步骤2:N-[3-(2-二甲基氨基乙基氨基)-1,1-二甲基-3-氧代-丙基]氨基甲酸苄基酯
Figure PCTCN2019073550-appb-000063
在室温下,将3-(苄氧基羰基氨基)-3-甲基-丁酸(3.0g,12mmol)溶于二氯甲烷(30mL)中并降温至0℃,加入HATU(5.3g,13mmol)和N,N-二异丙基乙胺(7.0mL,40mmol),搅拌20分钟。加入N,N-二甲基-1,2-乙二胺(1.4g,16mmol),升至室温搅拌过夜。水(20mL)洗涤,有机相浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(3.0g,黄色油状物),产率78%。
步骤3:3-氨基-N-(2-二甲基氨基乙基)-3-甲基-丁酰胺
Figure PCTCN2019073550-appb-000064
在室温下,将N-[3-(2-二甲基氨基乙基氨基)-1,1-二甲基-3-氧代-丙基]氨基甲酸苄基酯(3.0g,9.3mmol)溶于无水甲醇(30mL)中,加入10%钯/碳(0.30g,0.28mmol),氢气氛围下搅拌过夜。抽滤,浓缩,得到标题化合物(1.7g,黄色油状物),产率97%。
步骤4:3-氨基-N-(2-二甲基氨基乙基)-3-甲基-丁酰胺二盐酸盐
Figure PCTCN2019073550-appb-000065
在室温下,将3-氨基-N-(2-二甲基氨基乙基)-3-甲基-丁酰胺(1.7g,9.1mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(5mL,5M),搅拌1小时。抽滤,滤饼用乙酸乙酯(20mL)淋洗,真空干燥得到标题化合物(2.0g,白色固体),产率85%。
MS(ESI,pos.ion)m/z:188.2[M+H] +
1H NMR(400MHz,D 2O)δ(ppm):3.53(t,J=6.2Hz,2H),3.24(t,J=6.1Hz,2H),2.84(s,6H),2.55(s,2H),1.32(s,6H)。
步骤5:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[3-(2-甲基氨基乙基氨基)-1,1-二甲基-3-氧代-丙基]氨 基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯
Figure PCTCN2019073550-appb-000066
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.15g,0.25mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.12g,0.29mmol)和N,N-二异丙基乙胺(0.22mL,1.3mmol),搅拌20分钟。加入3-氨基-N-(2-二甲基氨基乙基)-3-甲基-丁酰胺二盐酸盐(83mg,0.32mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.12g,白色固体),产率63%。
MS(ESI,pos.ion)m/z:782.7[M+H] +
步骤6:N-(2-二甲氨基乙基)-3-甲基-3-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙 基)6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丁酰胺
Figure PCTCN2019073550-appb-000067
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[3-(2-甲基氨基乙基氨基)-1,1-二甲基-3-氧代-丙基]氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.12g,0.15mmol)溶于无水甲醇(2mL)中,加入甲醇钠固体(41mg,0.75mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=2/1],得到标题化合物(60mg,白色固体),产率59%。MS(ESI,pos.ion)m/z:656.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):8.07(br.s,1H),7.71(t,1H),7.49(s,1H),7.29(s,1H),7.23(d,1H),7.11-7.03(m,5H),5.51(d,1H),5.02(d,1H),4.88(d,1H),4.21(s,1H),4.03(d,1H),3.90(s,2H),3.81(d,1H),3.72(d,1H),3.47-3.36(m,5H),3.15(m,2H),2.45(s,2H),2.24(s,6H),2.18(s,3H),2.02(t,2H),1.73(m,2H),1.29(s,6H),1.21(s,3H),1.15(s,3H)。
实施例5 N-[3-(二甲氨基)丙基]-2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丙酰胺
Figure PCTCN2019073550-appb-000068
步骤1:N-[2-[3-(二甲基氨基)丙基氨基]-1,1-二甲基-2-氧代-乙基]氨基甲酸苄基酯
Figure PCTCN2019073550-appb-000069
在室温下,将2-(苄氧基羰基氨基)-2-甲基-丙酸(3.0g,13mmol)溶于二氯甲烷(30mL)中并冷却至0℃,加入HATU(5.6g,14mmol)和N,N-二异丙基乙胺(7.0mL,40mmol),搅拌20分钟。加入N,N-二甲基-1,3-丙二胺(1.7g,17mmol),搅拌过夜。水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(4.0g,黄色油状物),产率98%。
步骤2:2-氨基-N-[3-(二甲基氨基)丙基]-2-甲基-丙酰胺
Figure PCTCN2019073550-appb-000070
在室温下,将N-[2-[3-(二甲基氨基)丙基氨基]-1,1-二甲基-2-氧代-乙基]氨基甲酸苄基酯(4.0g,12mmol)溶于无水甲醇(30mL)中,加入10%钯/碳(0.30g,0.28mmol),氢气氛围下搅拌过夜。抽滤,浓缩,得到标题化合物(2.3g,黄色油状物),产率99%。
步骤3:2-氨基-N-[3-(二甲基氨基)丙基]-2-甲基-丙酰胺二盐酸盐
Figure PCTCN2019073550-appb-000071
在室温下,将2-氨基-N-[3-(二甲基氨基)丙基]-2-甲基-丙酰胺(2.3g,12mmol)溶于乙酸乙酯(20mL) 中,加入HCl异丙醇溶液(8mL,5M),搅拌1小时。抽滤,滤饼用乙酸乙酯(20mL)淋洗,真空干燥得到标题化合物(2.5g,白色固体),产率78%。
MS(ESI,pos.ion)m/z:188.2[M+H] +
步骤4:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-[3-(二甲氨基)丙基]-1,1-二甲基-2-氧代-乙基]氨 基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯
Figure PCTCN2019073550-appb-000072
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.10g,0.16mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(80mg,0.21mmol)和N,N-二异丙基乙胺(0.20mL,1.1mmol),搅拌20分钟。加入2-氨基-N-[3-(二甲基氨基)丙基]-2-甲基-丙酰胺二盐酸盐(60mg,0.23mmol),升至室温搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(90mg,白色固体),产率80%。
MS(ESI,pos.ion)m/z:782.6[M+H] +
步骤5:N-[3-(二甲氨基)丙基]-2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙 基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丙酰胺
Figure PCTCN2019073550-appb-000073
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-[3-(二甲氨基)丙基]-1,1-二甲基2-氧代-乙基]氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(90mg,0.11mmol)溶于无水甲醇(2mL)中,加入甲醇钠固体(30mg,0.55mmol),搅拌2小时。浓缩,残留物直接经薄层色谱制备纯化[DCM/MeOH(v/v)=2/1],得到标题化合物(50mg,白色固体),产率66%。
MS(ESI,pos.ion)m/z:656.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):7.95(s,1H),7.70(t,1H),7.29(s,1H),7.23(d,1H),7.12–7.04(m,5H),5.53(d,1H),5.04(d,1H),4.89(d,1H),4.21(s,1H),4.04(d,1H),3.91(s,2H),3.81(d,1H),3.73(t,1H),3.54–3.38(m,4H),3.09(m,2H),2.90(m,2H),2.61(s,6H),2.18(s,3H),2.11(t,2H),1.78–1.67(m,4H),1.29(s,6H),1.21(s,3H),1.16(s,3H)。
实施例6 N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]-2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丙酰胺
Figure PCTCN2019073550-appb-000074
步骤1:N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]氨基甲酸叔丁酯
Figure PCTCN2019073550-appb-000075
在室温下,将(3R)-3-氟吡咯烷盐酸盐(4.1g,33mmol)溶于二甲基亚砜(40mL)中,加入N-(2-溴乙基)氨基甲酸叔丁酯(6.9g,31mmol)和N,N-二异丙基乙胺(11mL,64mmol),加热至40℃搅拌过夜。加入水(150mL),乙酸乙酯(100mL×3)萃取,合并的有机相用饱和食盐水(50mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[乙酸乙酯/石油醚(v/v)=1/1],得到标题化合物(3.0g,黄色油状物),产率42%。
步骤2:2-[(3R)-3-氟吡咯烷-1-基]乙胺
Figure PCTCN2019073550-appb-000076
在室温下,将N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]氨基甲酸叔丁酯(3.5g,15mmol)溶于乙酸乙酯(30mL)中,加入HCl乙酸乙酯溶液(10mL,4M),搅拌2小时。浓缩,残留物溶于甲醇(30mL)中,加入固体碳酸氢钠调节pH=8-9,抽滤,浓缩,得到标题化合物(1.5g,黄色油状物),产率75%。
步骤3:N-[2-[2-[(3R)-3-氟吡咯烷-1-基]乙氨基]-1,1-二甲基-2-氧代-乙基]氨基甲酸苄酯
Figure PCTCN2019073550-appb-000077
在室温下,将2-(苄氧基羰基氨基)-2-甲基-丙酸(1.0g,4.2mmol)溶于二氯甲烷(10mL)中并降温至0℃,加入HATU(1.9g,4.7mmol)和N,N-二异丙基乙胺(2.2mL,13mmol),搅拌20分钟。加入2-[(3R)-3-氟吡咯烷-1-基]乙胺(0.7g,5.3mmol),搅拌过夜。水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(1.1g,无色油状物),产率74%。
MS(ESI,pos.ion)m/z:352.1[M+H] +
步骤4:2-氨基-N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]-2-甲基-丙酰胺
Figure PCTCN2019073550-appb-000078
在室温下,将N-[2-[2-[(3R)-3-氟吡咯烷-1-基]乙氨基]-1,1-二甲基-2-氧代-乙基]氨基甲酸苄酯(1.1g,3.1mmol)溶于无水甲醇(30mL)中,加入10%钯/碳(0.20g,0.21mmol),氢气氛围下搅拌过夜。抽滤,浓缩,得到标题化合物(0.68g,黄色油状物),产率99%。
步骤5:2-氨基-N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]-2-甲基-丙酰胺二盐酸盐
Figure PCTCN2019073550-appb-000079
在室温下,将2-氨基-N-[3-(二甲基氨基)丙基]-2-甲基-丙酰胺(0.68g,3.1mmol)溶于乙酸乙酯(5mL)中,加入HCl乙酸乙酯溶液(3mL,4M),搅拌1小时。抽滤,滤饼用乙酸乙酯(5mL)淋洗,真空干燥得到标题化合物(0.77g,浅黄色固体),产率85%。
MS(ESI,pos.ion)m/z:218.1[M+H] +
步骤6:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-[2-[(3R)-3-氟吡咯烷-1-基]乙基氨基]-1,1-二甲基-2- 氧代-乙基]氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷 -3-基]乙酸酯
Figure PCTCN2019073550-appb-000080
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.15g,0.25mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.18g,0.47mmol)和N,N-二异丙基乙胺(0.22mL,1.3mmol),搅拌20分钟。加2-氨基-N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]-2-甲基-丙酰胺二盐酸盐(93mg,0.32mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=20/1],得到标题化合物(0.15g,白色固体),产率75%。
MS(ESI,pos.ion)m/z:812.7[M+H] +
步骤7:N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]-2-甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基 -1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]丙酰胺
Figure PCTCN2019073550-appb-000081
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-[2-[(3R)-3-氟吡咯烷-1-基]乙基氨基]-1,1-二甲基-2-氧代-乙基]氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.15g,0.18mmol)溶于无水甲醇(2mL)中,加入甲醇钠固体(49mg,0.90mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=3/1],得到标题化合物(100mg,白色固体),产率66%。
MS(ESI,pos.ion)m/z:686.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):8.20(br.s,1H),7.77(br.s,1H),7.29(s,1H),7.23(d,1H),7.12–7.02(m,5H),5.50(d,1H),5.01(d,1H),4.87(d,1H),4.20(s,1H),4.04(d,1H),3.91(s,2H),3.81(d,1H),3.73(t,1H),3.50–3.38(m,4H),2.18(s,3H),2.11(t,2H),1.99(m,1H),1.75(m,2H),1.29(s,6H),1.21(s,3H),1.16(s,3H)。
实施例7 N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]环己基甲酰胺
Figure PCTCN2019073550-appb-000082
步骤1:1-(苄氧基羰基氨基)环己烷甲酸
Figure PCTCN2019073550-appb-000083
在室温下,将1-氨基环己烷羧酸(5.4g,38mmol)溶于二氧六环(30mL)中,加入碳酸钠(11.0g,104mmol)和水(100mL),冷却至0℃,加入氯甲酸苄酯(6.0mL,41mmol),升室温搅拌过夜。正己烷(100mL) 洗涤,水相用稀盐酸(1M)调节pH=2,乙酸乙酯(100mL×2)萃取,合并的有机相用饱和食盐水(100mL)洗涤,无水硫酸钠干燥,抽滤浓缩,得到标题化合物(5.2g,无色油状物),产率50%。
步骤2:N-[1-[2-[(3R)-3-氟吡咯烷-1-基]乙基氨基甲酰基]环己基]氨基甲酸苄酯
Figure PCTCN2019073550-appb-000084
在室温下,将1-(苄氧基羰基氨基)环己烷甲酸(1.3g,4.7mmol)溶于二氯甲烷(15mL)中并降温至0℃,加入HATU(2.0g,5.0mmol)和N,N-二异丙基乙胺(2.3mL,13mmol),搅拌20分钟。加入2-[(3R)-3-氟吡咯烷-1-基]乙胺(0.70g,5.3mmol),升至室温搅拌过夜。水(20mL)洗涤,有机相浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(1.6g,无色油状物),产率87%。
MS(ESI,pos.ion)m/z:392.2[M+H] +
步骤3:1-氨基-N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]环己烷甲酰胺
Figure PCTCN2019073550-appb-000085
在室温下,将N-[1-[2-[(3R)-3-氟吡咯烷-1-基]乙基氨基甲酰基]环己基]氨基甲酸苄酯(1.6g,4.1mmol)溶于无水甲醇(20mL)中,加入10%钯/碳催化剂(0.20g,0.19mmol),氢气氛围下搅拌过夜。抽滤,浓缩,得到标题化合物(1.1g,黄色油状物),产率99%。
步骤4:1-氨基-N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]环己烷甲酰胺二盐酸盐
Figure PCTCN2019073550-appb-000086
在室温下,将1-氨基-N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]环己烷甲酰胺(1.0g,3.9mmol)溶于乙酸乙酯(5mL)中,加入HCl乙酸乙酯溶液(3mL,4M),搅拌1小时。抽滤,滤饼用乙酸乙酯(5mL)淋洗,真空干燥得到标题化合物(1.1g,白色固体),产率86%。
MS(ESI,pos.ion)m/z:258.1[M+H] +
步骤5:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[1-[2-[(3R)-3-氟吡咯烷-1-基]乙基氨基甲酰基]环己基] 氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸
Figure PCTCN2019073550-appb-000087
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.15g,0.25mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.12g,0.32mmol)和N,N-二异丙基乙胺(0.22mL,1.3mmol),搅拌20分钟。加入1-氨基-N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]环己烷甲酰胺二盐酸盐(110mg,0.32mmol),升至室温搅拌过夜。加入水(10mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=20/1],得到标题化合物(0.18g,白色固体),产率86%。MS(ESI,pos.ion)m/z:852.7[M+H] +
步骤6:N-[2-[(3R)-3-氟吡咯烷-1-基]乙基]-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基 -乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基氨基]环己基甲酰胺
Figure PCTCN2019073550-appb-000088
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[1-[2-[(3R)-3-氟吡咯烷-1-基]乙基氨基甲酰基]环己基]氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.18g,0.21mmol)溶于无水甲醇(2mL)中,加入甲醇钠固体(57mg,1.05mmol),搅拌2小时。浓缩,残留物直接经薄层色谱制备纯化[DCM/MeOH(v/v)=3/1],得到标题化合物7(90mg,白色固体),产率58%。
MS(ESI,pos.ion)m/z:726.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):7.92(br.s,1H),7.77(br.s,1H),7.30(s,1H),7.23(d,1H),7.12–7.03(m,5H),5.50(d,1H),5.01(d,1H),4.88(d,1H),4.20(s,1H),4.04(d,1H),3.91(s,2H),3.81(d,1H),3.72(t,1H),3.53–3.38(m,6H),2.23-2.15(m,5H),1.99(m,1H),1.85(d,2H),1.76(m,2H),1.64(m,2H),1.48(m,5H),1.21(s,3H),1.16(s,3H)。
实施例8 N-(2-二甲基氨基乙基)-2-甲基-2-[甲基-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]氨基]丙酰胺
Figure PCTCN2019073550-appb-000089
步骤1:N-[2-(2-二甲基氨基乙基氨基)-1,1,-二甲基-2-氧代-乙基]-N-甲基-甲酸叔丁酯
Figure PCTCN2019073550-appb-000090
在室温下,将2-[-叔丁氧羰基(甲基)氨基]-2-甲基丙酸(2.6g,12mmol)溶于二氯甲烷(40mL)中并冷却至0℃,加入HATU(5.8g,15mmol)和N,N-二异丙基乙胺(6.3mL,36mmol),搅拌20分钟。加入N,N-二甲基-1,2-乙二胺(1.6g,18mmol),升至室温搅拌过夜。用水(30mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=20/1],得到标题化合物(2.0g,黄色油状物),产率58%。
MS(ESI,pos.ion)m/z:288.1[M+H] +
步骤2:N-(2-二甲基氨基乙基)-2-甲基-2-(甲基氨基)丙酰胺二盐酸盐
Figure PCTCN2019073550-appb-000091
在室温下,将N-[2-(2-二甲基氨基乙基氨基)-1,1,-二甲基-2-氧代-乙基]-N-甲基-甲酸叔丁酯(2.0g,7.0mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(4mL,5M),所得混合物搅拌过夜,降温至0℃搅拌30分钟。抽滤,滤饼用乙酸乙酯(5mL)淋洗,真空干燥,得到标题化合物(1.3g,黄色固体),产率71%。
步骤3:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-(2-二甲基氨基乙基氨基)-1,1-二甲基-2-氧代-乙基] 甲基-氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基] 乙酸酯
Figure PCTCN2019073550-appb-000092
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.20g,0.33mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.16g,0.40mmol)和N,N-二异丙基乙胺(0.30mL,1.7mmol),搅拌20分钟。加入N-(2-二甲基氨基乙基)-2-甲基-2-(甲基氨基)丙酰胺二盐酸盐(0.17g,0.66mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.11g,白色固体),产率43%。
MS(ESI,pos.ion)m/z:782.3[M+H] +
步骤4:N-(2-二甲基氨基乙基)-2-甲基-2-[甲基-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1- 甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]氨基]丙酰胺
Figure PCTCN2019073550-appb-000093
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-(2-二甲基氨基乙基氨基)-1,1-二甲基-2-氧-乙基]-甲基-氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.11g,0.14mmol)溶于无水甲醇(3mL)中,加入甲醇钠固体(38mg,0.70mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=4/1],得到标题化合物(55mg,白色固体),产率61%。
MS(ESI,pos.ion)m/z:656.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):8.71(br.s,1H),7.85(t,1H),7.30(s,1H),7.23(d,1H),7.12–7.04(m,5H),5.50(d,1H),5.01(d,1H),4.88(d,1H),4.20(s,1H),4.04(d,1H),3.91(s,2H),3.81(d,1H),3.75–3.69(m,1H),3.45–3.35(m,5H),3.18(s,3H),3.10(t,2H),2.81(s,6H),2.18(s,3H),2.12(t,2H),1.75(m,2H),1.29(s,6H),1.21(s,3H),1.16(s,3H)。
实施例9 N-(2-二甲基氨基乙基)-N,2-二甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]丙酰胺
Figure PCTCN2019073550-appb-000094
步骤1:N-[2-[2-二甲基氨基乙基(甲基)氨基]-1,1,-二甲基-2-氧代-乙基]-甲酸叔丁酯
Figure PCTCN2019073550-appb-000095
在室温下,将2-[-叔丁氧羰基(甲基)氨基]-2-甲基丙酸(2.0g,10mmol)溶于二氯甲烷(30mL)中并冷却至0℃,加入HATU(4.7g,12mmol)和N,N-二异丙基乙胺(5.2mL,30mmol),搅拌20分钟。加入N,N’,N’-三甲基-1,2-乙二胺(1.5g,15mmol),升至室温搅拌过夜。用水(30mL)洗涤,无水硫酸钠干燥,抽滤浓 缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=20/1],得到标题化合物(1.6g,黄色油状物),产率56%。
MS(ESI,pos.ion)m/z:288.0[M+H] +
步骤2:2-氨基-N-(2-二甲基氨基乙基)-N,2-二甲基-丙酰胺二盐酸盐
Figure PCTCN2019073550-appb-000096
在室温下,将N-[2-[2-二甲基氨基乙基(甲基)氨基]-1,1,-二甲基-2-氧-乙基]-甲酸叔丁酯(1.6g,5.6mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(4mL,5M),搅拌过夜,降温至0℃搅拌30分钟。抽滤,滤饼用乙酸乙酯(5mL)淋洗,真空干燥,得到标题化合物(1.0g,黄色固体),产率69%。
步骤3:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-[2-二甲基氨基乙基(甲基)氨基]-1,1-二甲基-2-氧代 -乙基]-氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基] 乙酸酯
Figure PCTCN2019073550-appb-000097
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.20g,0.33mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.16g,0.40mmol)和N,N-二异丙基乙胺(0.30mL,1.7mmol),搅拌20分钟。加入2-氨基-N-(2-二甲基氨基乙基)-N,2-二甲基-丙酰胺二盐酸盐(0.17g,0.66mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.13g,白色固体),产率50%。
MS(ESI,pos.ion)m/z:782.2[M+H] +
步骤4 N-(2-二甲基氨基乙基)-N,2-二甲基-2-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲 基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]丙酰胺
Figure PCTCN2019073550-appb-000098
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[[2-[2-二甲基氨基乙基(甲基)氨基]-1,1-二甲基-2-氧代-乙基]-氨基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.13g,0.17mmol)溶于无水甲醇(3mL)中,加入甲醇钠固体(46mg,0.85mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=4/1],得到标题化合物(42mg,白色固体),产率38%。
MS(ESI,pos.ion)m/z:656.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):8.65(br.s,1H),8.02(s,1H),7.31(s,1H),7.24(d,1H),7.12–7.04(m,5H),5.49(d,1H),5.00(d,1H),4.85(d,1H),4.20(s,1H),4.04(d,1H),3.90(s,2H),3.81(d,1H),3.73–3.69(m,1H),3.44–3.35(m,5H),3.10(s,3H),3.06(t,2H),2.80(s,6H),2.16(s,3H),2.12(t,2H),1.72(m,2H),1.28(s,6H),1.21(s,3H),1.16(s,3H)。
实施例10 (2S)-N-(2-二甲基氨基乙基)-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基- 乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]氮杂环丁烷-2-甲酰胺
Figure PCTCN2019073550-appb-000099
步骤1:(2S)-2-(2-二甲基氨基乙基氨基甲酰基)氮杂环丁烷-1-甲酸叔丁酯
Figure PCTCN2019073550-appb-000100
在室温下,将(2S)-1-叔丁氧羰基氮杂环丁烷-2-甲酸(3.0g,15mmol)溶于二氯甲烷(50mL)中并冷却至0℃,加入HATU(7.1g,18mmol)和N,N-二异丙基乙胺(7.9mL,45mmol),搅拌20分钟。加入N,N-二甲基-1,2-乙二胺(2.0g,23mmol),升至室温搅拌过夜。用水(30mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=20/1],得到标题化合物(2.6g,黄色油状物),产率64%。
MS(ESI,pos.ion)m/z:272.0[M+H] +
步骤2:(2S)-N-(2-二甲基氨基乙基)氮杂环丁烷-2-甲酰胺二盐酸盐
Figure PCTCN2019073550-appb-000101
在室温下,将(2S)-2-(2-二甲基氨基乙基氨基甲酰基)氮杂环丁烷-1-甲酸叔丁酯(2.6g,9.6mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(6mL,5M),搅拌过夜,降温至0℃搅拌30分钟。抽滤,滤饼用乙酸乙酯(10mL)淋洗,真空干燥,得到标题化合物(1.7g,黄色固体),产率72%。
步骤3:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[(2S)-2-(2-二甲基氨基乙基氨基甲酰基)氮杂环丁烷-1- 基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯
Figure PCTCN2019073550-appb-000102
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.20g,0.33mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.16g,0.40mmol)和N,N-二异丙基乙胺(0.30mL,1.7mmol),搅拌20分钟。加入(2S)-N-(2-二甲基氨基乙基)氮杂环丁烷-2-甲酰胺二盐酸盐(0.16g,0.66mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.18g,白色固体),产率71%。
MS(ESI,pos.ion)m/z:766.2[M+H] +
步骤4:(2S)-N-(2-二甲基胺基乙基)-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙 基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]氮杂环丁烷-2-甲酰胺
Figure PCTCN2019073550-appb-000103
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[(2S)-2-(2-二甲基氨基乙基氨基甲酰基)氮杂环丁烷-1-基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.18g,0.23mmol)溶于无水甲醇(5mL)中,加入甲醇钠固体(65mg,1.2mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=4/1],得到标题化合物(0.10g,白色固体),产率68%。
MS(ESI,pos.ion)m/z:640.1[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):9.21(br.s,1H),8.18(s,1H),7.29(s,1H),7.22(d,J=7.8Hz,1H),7.13-7.02(m,5H),5.55(d,J=4.9Hz,1H),5.07(d,J=5.4Hz,1H),4.93(d,J=6.8Hz,1H),4.24(s,1H),4.20–4.15(m,1H),4.03(d,J=7.4Hz,1H),3.92(s,2H),3.82(d,J=7.1Hz,1H),3.75–3.68(m,1H),3.54–3.37(m,4H),3.06(s,2H),2.76(d,J=21.9Hz,6H),2.60–2.52(m,2H),2.39–2.26(m,2H),2.18(s,3H),2.07–1.95(m,2H),1.88(m,1H),1.77-1.72(m,2H),1.21(s,3H),1.15(s,3H).
实施例11 (2S)-N-(2-二甲基氨基乙基)-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000104
步骤1:(2S)-2-(2-二甲基氨基乙基氨基甲酰基)吡咯烷-1-甲酸苄酯
Figure PCTCN2019073550-appb-000105
在室温下,将(2S)-1-苄氧羰基吡咯烷-2-甲酸(4.0g,16mmol)溶于二氯甲烷(50mL)中并冷却至0℃,加入HATU(7.2g,19mmol)和N,N-二异丙基乙胺(8.4mL,48mmol),搅拌20分钟。加入N,N-二甲基-1,2-乙二胺(2.1g,24mmol),升至室温搅拌过夜。用水(50mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=30/1],得到标题化合物(3.8g,黄色油状物),产率74%。
MS(ESI,pos.ion)m/z:320.2[M+H] +
步骤2:(2S)-N-(2-二甲氨基乙基)吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000106
在室温下,向反应瓶中依次加入(2S)-2-(2-二甲基氨基乙基氨基甲酰基)吡咯烷-1-甲酸苄酯(3.8g,12mmol)、10%钯/碳(0.50g,0.47mmol)、四氢呋喃(10mL)和甲醇(40mL),氢气氛围下搅拌反应2小时。抽滤浓缩,得到标题化合物(2.2g,黄色油状物),产率100%。
步骤3:(2S)-N-(2-二甲氨基乙基)吡咯烷-2-甲酰胺二盐酸盐
Figure PCTCN2019073550-appb-000107
在室温下,将(2S)-N-(2-二甲氨基乙基)吡咯烷-2-甲酰胺(2.2g,12mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(6mL,5M),搅拌10分钟,降温至0℃搅拌30分钟。抽滤,滤饼用乙酸乙酯(20mL)淋洗,真空干燥,得到标题化合物(2.8g,黄色固体),产率92%。
步骤4:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[(2S)-2-(2-二甲基氨基乙基氨基甲酰基)吡咯烷-1-基]-4- 氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯
Figure PCTCN2019073550-appb-000108
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.20g,0.33mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.16g,0.40mmol)和N,N-二异丙基乙胺(0.30mL,1.7mmol),搅拌20分钟。加入(2S)-N-(2-二甲氨基乙基)吡咯烷-2-甲酰胺二盐酸盐(0.17g,0.66mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.20g,白色固体),产率79%。
MS(ESI,pos.ion)m/z:780.2[M+H] +
步骤5:(2S)-N-(2-二甲基胺基乙基)-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙 基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000109
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[(2S)-2-(2-二甲基氨基乙基氨基甲酰基)吡咯烷-1-基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.20g,0.26mmol)溶于无水甲醇(5mL)中,加入甲醇钠固体(70mg,1.3mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=4/1],得到标题化合物(0.12g,白色固体),产率72%。
MS(ESI,pos.ion)m/z:654.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):9.80(br.s,1H),8.17(s,1H),7.28(s,1H),7.22(d,J=7.8Hz,1H),7.12-7.02(m,5H),5.55(d,J=4.9Hz,1H),5.06(d,J=5.4Hz,1H),4.92(d,J=6.8Hz,1H),4.22(s,1H),4.20–4.14(m,1H),4.03(d,J=7.4Hz,1H),3.91(s,2H),3.81(d,J=7.1Hz,1H),3.74–3.68(m,1H),3.54–3.37(m,4H),3.05(s,2H),2.71(d,J=21.9Hz,6H),2.57–2.52(m,2H),2.35–2.24(m,2H),2.17(s,3H),2.07–1.95(m,2H),1.90–1.81(m,2H),1.80–1.71(m,3H),1.21(s,3H),1.15(s,3H).
实施例12 (2R)-N-(2-二甲基氨基乙基)-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000110
以(2R)-1-苄氧羰基吡咯烷-2-甲酸代替(2S)-1-苄氧羰基吡咯烷-2-甲酸作为起始物料,参考实施例11描述的方法,得到标题化合物为白色固体。
MS(ESI,pos.ion)m/z:654.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):9.96(br.s,1H),8.15(s,1H),7.29(s,1H),7.22(d,J=7.2Hz,1H),7.17–6.96(m,5H),5.55(d,J=4.9Hz,1H),5.06(d,J=4.8Hz,1H),4.91(d,J=6.3Hz,1H),4.22(s,1H),4.18(d,J=4.7Hz,1H),4.03(d,J=7.9Hz,1H),3.91(s,2H),3.81(d,J=6.6Hz,1H),3.72(m,1H),3.56–3.37(m,4H),3.01(s,2H),2.66(d,J=31.6Hz,6H),2.58-2.52(m,2H),2.32-2.23(m,2H),2.17(s,3H),2.06-1.93(s,2H),1.88-1.80(m,2H),1.80-1.68(m,3H),1.21(s,3H),1.15(s,3H).
实施例13 (2S,4S)-N-(2-二甲基氨基乙基)-4-羟基-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000111
步骤1:(2S,4S)-2-(2-二甲基氨基乙基氨基甲酰基)-4-羟基-吡咯烷-1-甲酸苄酯
Figure PCTCN2019073550-appb-000112
在室温下,将(2S,4R)-1-苄氧羰基-4-羟基-吡咯烷-2-甲酸(5.0g,19mmol)溶于二氯甲烷(80mL)中并冷却至0℃,依次加入HATU(8.7g,23mmol)、N,N-二异丙基乙胺(10mL,57mmol)和N,N-二甲基-1,2-乙二胺(2.4g,27mmol),升至室温搅拌过夜。用水(50mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=15/1],得到标题化合物(2.2g,黄色油状物),产率35%。MS(ESI,pos.ion)m/z:336.0[M+H] +
步骤2:(2S,4S)-N-(2-二甲基氨基乙基)-4-羟基-吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000113
在室温下,向反应瓶中依次加入(2S,4S)-2-(2-二甲基氨基乙基氨基甲酰基)-4-羟基-吡咯烷-1-甲酸苄酯(2.2g,6.6mmol)、10%钯/碳(0.50g,0.47mmol)、四氢呋喃(10mL)和甲醇(40mL),氢气氛围下搅拌反应过夜。抽滤浓缩,得到标题化合物(1.3g,黄色油状物),产率100%。
步骤3:(2S,4S)-N-(2-二甲基氨基乙基)-4-羟基-吡咯烷-2-甲酰胺二盐酸盐
Figure PCTCN2019073550-appb-000114
在室温下,将(2S,4S)-N-(2-二甲基氨基乙基)-4-羟基-吡咯烷-2-甲酰胺(1.3g,6.5mmol)溶于乙酸乙酯(15mL)中,加入HCl异丙醇溶液(2mL,5M),搅拌30分钟。抽滤,滤饼用乙酸乙酯(5mL)淋洗,真 空干燥,得到标题化合物(0.95g,白色固体),产率53%。
MS(ESI,pos.ion)m/z:202.0[M+H] +
步骤4:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[(2S,4S)-2-(2-二甲基氨基乙基氨基甲酰基)-4-羟基-吡咯 烷-1-基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]] 乙酸酯
Figure PCTCN2019073550-appb-000115
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.30g,0.49mmol)溶于二氯甲烷(10mL)中并降温至0℃,依次加入HATU(0.28g,0.74mmol)、N,N-二异丙基乙胺(0.50mL,2.9mmol)和(2S,4S)-N-(2-二甲基氨基乙基)-4-羟基-吡咯烷-2-甲酰胺二盐酸盐(0.20g,0.74mmol),搅拌过夜。加入水(20mL),二氯甲烷(20mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=7/1],得到标题化合物(0.25g,浅黄色固体),产率64%。
MS(ESI,pos.ion)m/z:796.2[M+H] +
步骤5:(2S,4S)-N-(2-二甲基氨基乙基)-4-羟基-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1- 甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000116
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[(2S,4S)-2-(2-二甲基氨基乙基氨基甲酰基)-4-羟基-吡咯烷-1-基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]]乙酸酯(0.25g,0.31mmol)溶于无水甲醇(8mL)中,加入甲醇钠固体(70mg,1.3mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=4/1],得到标题化合物(0.12g,白色固体),产率58%。
MS(ESI,pos.ion)m/z:670.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):9.37(br.s,1H),8.16(s,1H),7.27(s,1H),7.22(d,J=7.8Hz,1H),7.11-7.02(m,5H),5.55(d,J=4.9Hz,1H),5.05(d,J=5.4Hz,1H),4.92(d,J=6.8Hz,1H),4.35(d,J=5.9Hz,1H),4.21(s,1H),4.20–4.15(m,1H),4.02(d,J=7.4Hz,1H),3.90(s,2H),3.80(d,J=7.1Hz,1H),3.76(m,1H),3.73–3.68(m,1H),3.54–3.38(m,4H),3.18(m,1H),3.04(s,2H),2.70(d,J=21.9Hz,6H),2.55–2.51(m,2H),2.38–2.25(m,3H),2.17(s,3H),2.10–1.98(m,2H),1.71(m,1H),1.21(s,3H),1.15(s,3H).
实施例14 (2S,4R)-N-(2-二甲基氨基乙基)-4-氟-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000117
步骤1:(2S,4R)-1-苄氧羰基-4-氟-吡咯烷-2-甲酸
Figure PCTCN2019073550-appb-000118
在室温下,将(2S,4R)-4-氟-吡咯烷-2-甲酸(3.0g,18mmol)溶于1,4-二氧六环(20mL)中,依次加入水(60mL)和碳酸钠(7.5g,71mmol),降温至0℃,滴加CbzCl(3.0mL,21mmol),室温搅拌过夜。停止反应,反应液用正己烷(100mL)洗涤,水相用1M稀盐酸调节pH=2,乙酸乙酯(100mL)萃取,饱和氯化钠溶液(100mL)洗涤,无水硫酸钠干燥,减压浓缩,得到标题化合物(4.7g,无色油状物),产率99%。
步骤2:(2S,4R)-2-(2-二甲基氨基乙基氨基甲酰基)-4-氟-吡咯烷-1-甲酸苄酯
Figure PCTCN2019073550-appb-000119
在室温下,将(2S,4R)-1-苄氧羰基-4-氟-吡咯烷-2-甲酸(4.7g,18mmol)溶于二氯甲烷(50mL)中并冷却至0℃,加入HATU(8.4g,22mmol)和N,N-二异丙基乙胺(9.5mL,54mmol),搅拌20分钟。加入N,N-二甲基-1,2-乙二胺(2.4g,27mmol),升至室温搅拌过夜。用水(50mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=20/1],得到标题化合物(3.0g,黄色油状物),产率51%。
MS(ESI,pos.ion)m/z:338.0[M+H] +
步骤3:(2S,4R)-N-(2-二甲基氨基乙基)-4-氟-吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000120
在室温下,向反应瓶中依次加入(2S,4R)-2-(2-二甲基氨基乙基氨基甲酰基)-4-氟-吡咯烷-1-甲酸苄酯(3.0g,8.9mmol)、10%钯/碳(0.50g,0.47mmol)、四氢呋喃(10mL)和甲醇(40mL),氢气氛围下搅拌反应过夜。抽滤浓缩,得到标题化合物(1.8g,黄色油状物),产率100%。
步骤4:(2S,4R)-N-(2-二甲基氨基乙基)-4-氟-吡咯烷-2-甲酰胺二盐酸盐
Figure PCTCN2019073550-appb-000121
在室温下,将(2S,4R)-N-(2-二甲基氨基乙基)-4-氟-吡咯烷-2-甲酰胺(1.8g,8.9mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(3mL,5M),搅拌30分钟。抽滤,滤饼用乙酸乙酯(10mL)淋洗,真空干燥,得到标题化合物(1.4g,白色固体),产率57%。
MS(ESI,pos.ion)m/z:204.2[M+H] +
步骤5 [(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[(2S,4R)-2-(2-二甲基氨基乙基氨基甲酰基)-4-氟-吡咯烷 -1-基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]]乙 酸酯
Figure PCTCN2019073550-appb-000122
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.33g,0.54mmol)溶于二氯甲烷(10mL)中并降温至0℃,加入HATU(0.26g,0.65mmol)和N,N-二异丙基乙胺(0.50mL,2.9mmol),搅拌20分钟。加入(2S,4R)-N-(2-二甲基氨基乙基)-4-氟-吡咯烷-2-甲酰胺二盐酸盐(0.30g,1.1mmol),搅拌过夜。加入水(20mL),二氯甲烷(20mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.32g,浅黄色固体),产率75%。
MS(ESI,pos.ion)m/z:798.2[M+H] +
步骤6:(2S,4R)-N-(2-二甲基氨基乙基)-4-氟-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1- 甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]吡咯烷-2-甲酰胺
Figure PCTCN2019073550-appb-000123
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[4-[(2S,4R)-2-(2-二甲基氨基乙基氨基甲酰基)-4-氟-吡咯烷-1-基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]]乙酸酯(0.32g,0.40mmol)溶于无水甲醇(8mL)中,加入甲醇钠固体(0.11g,2.0mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=4/1],得到标题化合物(0.17g,白色固体),产率63%。
MS(ESI,pos.ion)m/z:672.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):9.45(br.s,1H),8.37(t,J=5.5Hz,1H),7.29(s,1H),7.23(d,J=7.8Hz,1H),7.12-7.00(m,5H),5.44(s,0.5H),5.30(s,0.5H),5.56(d,J=4.9Hz,1H),5.08(d,J=5.4Hz,1H),4.93(d,J=6.8Hz,1H),4.24(s,1H),4.21–4.16(m,1H),4.04(d,J=7.4Hz,1H),3.92(s,2H),3.83(d,J=7.1Hz,1H),3.76–3.69(m,1H),3.56–3.38(m,4H),3.07(s,2H),2.77(d,J=21.9Hz,6H),2.36–2.18(m,3H),2.35–2.24(m,2H),2.17(s,3H),2.14–1.94(m,2H),1.76(m,2H),1.21(s,3H),1.15(s,3H).
实施例15 (2S)-N-(2-二甲基氨基乙基)-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]哌啶-2-甲酰胺
Figure PCTCN2019073550-appb-000124
步骤1:(2S)-2-(2-二甲基氨基乙基氨基甲酰基)哌啶-1-甲酸叔丁酯
Figure PCTCN2019073550-appb-000125
在室温下,将(2S)-1-叔丁氧羰基哌啶-2-甲酸(3.7g,16mmol)溶于二氯甲烷(50mL)中并冷却至0℃,加入HATU(7.2g,19mmol)和N,N-二异丙基乙胺(8.4mL,48mmol),搅拌20分钟。加入N,N-二甲基-1,2-乙二胺(2.1g,24mmol),升至室温搅拌过夜。用水(30mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=20/1],得到标题化合物(2.7g,黄色油状物),产率56%。
MS(ESI,pos.ion)m/z:300.1[M+H] +
步骤2:(2S)-N-(2-二甲基氨基乙基)哌啶-2-甲酰胺二盐酸盐
Figure PCTCN2019073550-appb-000126
在室温下,将(2S)-2-(2-二甲基氨基乙基氨基甲酰基)哌啶-1-甲酸叔丁酯(2.7g,9.0mmol)溶于乙酸乙酯(20mL)中,加入HCl异丙醇溶液(6mL,5M),搅拌过夜,降温至0℃搅拌30分钟。抽滤,滤饼用乙酸乙酯(10mL)淋洗,真空干燥,得到标题化合物(1.9g,黄色固体),产率78%。
步骤3:[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[(2S)-2-(2-二甲基氨基乙基氨基甲酰基)哌啶-1-基]-4-氧 代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯
Figure PCTCN2019073550-appb-000127
在室温下,将4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三乙酰氧基-1-(1-羟基-1-甲基-乙基)-6,7,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酸(0.20g,0.33mmol)溶于二氯甲烷(5mL)中并降温至0℃,加入HATU(0.16g,0.40mmol)和N,N-二异丙基乙胺(0.30mL,1.7mmol),搅拌20分钟。加入(2S)-N-(2-二甲基氨基乙基)哌啶-2-甲酰胺二盐酸盐(0.18g,0.66mmol),搅拌过夜。加入水(20mL),二氯甲烷(40mL×3)萃取,合并的有机相用饱和食盐水(20mL)洗涤,无水硫酸钠干燥,抽滤浓缩,残留物经硅胶柱层析纯化[二氯甲烷/无水甲醇(v/v)=8/1],得到标题化合物(0.14g,白色固体),产率53%。
MS(ESI,pos.ion)m/z:794.3[M+H] +
步骤4:(2S)-N-(2-二甲基胺基乙基)-1-[4-[4-[[2-甲基-5-[(1S,2S,3S,4R,5S)-2,3,4-三羟基-1-(1-羟基-1-甲基-乙 基)-6,8-二氧杂双环[3.2.1]辛烷-5-基]苯基]甲基]苯基]丁酰基]哌啶-2-甲酰胺
Figure PCTCN2019073550-appb-000128
在室温下,将[(1S,2S,3S,4R,5S)-2,4-二乙酰氧基-5-[3-[[4-[(2S)-2-(2-二甲基氨基乙基氨基甲酰基)哌啶-1-基]-4-氧代-丁基]苯基]甲基]-4-甲基-苯基]-1-(1-羟基-1-甲基-乙基)-6,8-二氧杂双环[3.2.1]辛烷-3-基]乙酸酯(0.14g,0.18mmol)溶于无水甲醇(5mL)中,加入甲醇钠固体(49mg,0.9mmol),搅拌2小时。浓缩,残留物直接经TLC制备纯化[DCM/MeOH(v/v)=4/1],得到标题化合物(76mg,白色固体),产率63%。
MS(ESI,pos.ion)m/z:668.2[M+H] +
1H NMR(400MHz,DMSO-d 6)δ(ppm):9.65(br.s,1H),8.17(s,1H),7.28(s,1H),7.20(d,J=7.8Hz,1H), 7.12-7.01(m,5H),5.53(d,J=4.9Hz,1H),5.06(d,J=5.4Hz,1H),4.91(d,J=6.8Hz,1H),4.23(s,1H),4.18–4.14(m,1H),4.05(d,J=7.3Hz,1H),3.91(s,2H),3.83(d,J=7.0Hz,1H),3.76–3.67(m,1H),3.55–3.39(m,4H),3.07(s,2H),2.77(d,J=21.7Hz,6H),2.63–2.55(m,2H),2.40–2.28(m,2H),2.17(s,3H),2.03–1.92(m,3H),1.87-1.73(m,6H),1.21(s,3H),1.15(s,3H).
测试例
一、SGLT1抑制活性测定
测试目的:
下面的方法是用来测定本发明化合物对SGLT1的抑制活性。
试验材料:
14C-AMG溶液购于PerkinElmer,Cat.No.NEZ080001MC;
α-甲基葡萄糖苷购于Sigma,Cat.No.M9376-100G;
N-甲基-D-葡萄糖胺购于Sigma,Cat.No.M2004-100G;
根皮甙购于Sigma,Cat.No.P3449-1G;
96孔细胞培养板购于Corning,Cat.No.3903。
试验方法:
将3×10 4个Mock-转染的FIP-in CHO细胞和表达人SGLT1基因的CHO细胞分别接种至96孔细胞培养板;培养12小时后,每孔加入150μL无钠缓冲液洗涤细胞1次;每孔加入50μL含有不同浓度化合物的含钠缓冲液和0.5μM[ 14C]-AMG,并在37℃培养箱中孵育1小时,每孔加入150μL的预冷的无钠缓冲液以终止反应;继续用无钠缓冲液洗涤细胞3次并清除孔内残留液体;每孔加入20μL预冷的100mM NaOH,在900rpm下震荡5分钟;每孔加入80μL闪烁液,在600rpm下震荡5分钟后,用液闪仪读板,其结果如表1所示:
表1:本发明实施例提供的化合物SGLT1抑制活性
实施例编号 IC 50(SGLT1)/nM
实施例1 1.73
实施例7 1.16
实施例11 5.75
试验结果显示:本发明化合物对SGLT1具有明显的抑制作用。
二.口服葡萄糖耐量试验和促尿糖***试验
测定目的:
下面的方法是用来测定本发明化合物对改善口服葡萄糖耐量及促进尿糖***的作用。
试验材料:
葡萄糖:成都市科龙化工试剂厂
罗氏生化仪:用于尿糖检测
罗氏卓越型血糖检测仪:用于血糖检测
试验方法1:
将C57BL/6小鼠隔夜禁食15小时后称重、检测空腹血糖浓度,根据体重及空腹血糖分组,然后各给药组分别单次灌胃给予相应受试化合物,给药剂量为1mg/kg,空白对照组给予溶媒,于给药15min后检测血糖值(即0时血糖),检测0时血糖后各组小鼠立即单次灌胃给予葡萄糖(2.5g/kg),于给糖后15min、30min、60min、120min利用尾静脉取血,血糖仪连续检测C57BL/6小鼠血糖浓度,计算糖负荷后120min内血糖曲线下面积(AUC Glu 0-120min)的下降率。
检测完120min血糖后将每组动物分别放入一个代谢笼中,以代谢笼为单位收集给药后2.25-6小时、6-24小时的尿液,并记录每个时间点尿量,尿液收集过程中自由饮食、饮水,尿液收集后离心取上清液使用罗氏全自动生化仪检测各时间点C57BL/6小鼠尿糖浓度。
试验方法2:
将雄性SD大鼠隔夜禁食15小时后称重、检测空腹血糖浓度,根据体重及空腹血糖分组,然后各给药组分别单次灌胃给予相应受试化合物,给药剂量为1mg/kg,空白对照组给予溶媒,于给药30min后检测血糖值(即0时血糖),检测0时血糖后各组小鼠立即单次灌胃给予葡萄糖(4.0g/kg),于给糖后15min、30min、60min利用尾静脉取血,血糖仪连续检测SD大鼠血糖浓度,计算糖负荷后60min内血糖曲线下面积(AUC Glu 0-60min)的下降率。
检测完60min血糖后将每组动物分别放入一个代谢笼中,以代谢笼为单位收集给药后1.5-24小时、24-48小时的尿液,并记录每个时间点尿量,尿液收集过程中自由饮食、饮水,尿液收集后离心取上清液使用罗氏全自动生化仪检测各时间点SD大鼠尿糖浓度。
试验结果显示:本发明化合物在降低血糖水平方面效果显著。
试验结果显示:本发明化合物在促进尿糖***方面的效果显著。
三.静脉注射和口服定量本发明化合物后的药代动力学评价
测定目的:
下面的试验是对本发明化合物在动物体内的药代动力学性质进行评价。
试验方法:
将SD大鼠隔夜禁食15小时后称重,根据体重进行随机分组,受试化合物溶于5%DMSO+5%Koliphor HS 15+90%Saline溶媒进行给药。对于静脉注射给药的试验组,对试验动物给予1mg/kg、2mg/kg或5mg/kg的剂量;对于口服给药的试验组,对试验动物给予5mg/kg的剂量。然后,在给药前0.083小时及给药后的时间点为0.083(仅静脉注射组)、0.25、0.5、1.0、2.0、5.0、7.0和24小时取静脉血(约0.2mL),置于EDTAK 2抗凝管中,在11000rpm离心2分钟,收集血浆,并于-20℃或-70℃下保存直到进行LC/MS/MS分析。测定各时间点血浆中药物浓度,采用WinNonlin 6.3软件非房室模型法计算药动学参数,绘制药时曲线。
试验结果显示,将本发明提供的化合物静脉注射给药或口服给药时,其表现出优良的药代动力学性质。
在本说明书的描述中,参考术语“一个实施例”、“一些实施例”、“示例”、“具体示例”、或“一些示例”等的描述意指结合该实施例或示例描述的具体特征、结构、材料或者特点包含于本发明的至少一个实施例或示例中。在本说明书中,对上述术语的示意性表述不必须针对的是相同的实施例或示例。而且,描述的具体特征、结构、材料或者特点可以在任一个或多个实施例或示例中以合适的方式结合。此外,在不相互矛盾的情况下,本领域的技术人员可以将本说明书中描述的不同实施例或示例以及不同实施例或示例的特征进行结合和组合。
尽管上面已经示出和描述了本发明的实施例,可以理解的是,上述实施例是示例性的,不能理解为对本发明的限制,本领域的普通技术人员在本发明的范围内可以对上述实施例进行变化、修改、替换和变型。

Claims (19)

  1. 一种化合物,其为式(I)所示化合物或式(I)所示化合物的立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药,
    Figure PCTCN2019073550-appb-100001
    其中,
    L为-(CR aR b) q-、-CH=CH-(CR aR b) p-、-O-(CR aR b) p-、-NH-(CR aR b) p-、-S-(CR aR b) p-、-S(=O)-(CR aR b) p-或-S(=O) 2-(CR aR b) p-;
    q为1、2、3、4、5或6;
    p为0、1、2、3、4、5或6;
    R a和R b各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基或3-6个原子组成的杂环基;或R a、R b和与它们相连的碳原子一起形成C 3-6碳环或3-6个原子组成的杂环;
    R 1为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6羟基烷基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 1-6烷氨基、C 1-6烷硫基、C 3-6环烷基或C 3-6环烷基-C 1-4亚烷基,其中,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6羟基烷基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 1-6烷氨基、C 1-6烷硫基、C 3-6环烷基和C 3-6环烷基-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    R 2和R 3各自独立地为H、D、CN、OH、NH 2、-SH、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6烷硫基、C 1-6烷氨基、C 3-6环烷基或3-6个原子组成的杂环基,其中,所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6烷氧基、C 1-6烷硫基、C 1-6烷氨基、C 3-6环烷基和3-6个原子组成的杂环基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    或R 2、R 3和与它们相连的碳原子一起形成羰基;
    或R 2、R 3和与它们相连的碳原子一起形成C 3-6碳环或3-6个原子组成的杂环,其中,所述C 3-6碳环和3-6个原子组成的杂环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    R 4为H、D、-OR 4a或-SR 4b
    R 4a和R 4b各自独立地为H、D、C 1-6烷基、C 3-6环烷基、C 3-6环烷基-C 1-4亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-4亚烷基,其中,所述C 1-6烷基、C 3-6环烷基、C 3-6环烷基-C 1-4亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    R 10为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、C 1-6烷基或C 1-6烷氧基;
    R 5为H、D、C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基或5-8个原子组成的杂芳基,其中,所述C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基和5-8个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
    R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基或5-8个原子组成的杂芳基,其中,所述C 1-6烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷基、C 1-6卤代烷氧基、C 3-8环烷基、3-8个原子组成的杂环基、C 6-10芳基和5-8个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基、C 1-6卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
    或R 6、R 7和与它们相连的碳原子一起形成C 3-8碳环、3-8个原子组成的杂环、C 6-10芳环或5-8个原子组成的杂芳环,其中,所述C 3-8碳环、3-8个原子组成的杂环、C 6-10芳环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    或R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    R 8和R 9各自独立为H、D、ReO-C 1-4亚烷基、R dRcN-C 1-4亚烷基、C 1-8烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基或(5-8个原子组成的杂芳基)-C 1-4亚烷基,其中所述C 1-8烷 基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基和(5-8个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    或R 8、R 9和与它们相连的氮原子一起形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    R c、R d、R e和R f各自独立地为H、D、C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基或(5-8个原子组成的杂芳基)-C 1-4亚烷基,其中,所述C 1-6烷基、C 1-6卤代烷基、C 3-8环烷基、C 3-8环烷基-C 1-4亚烷基、3-8个原子组成的杂环基、(3-8个原子组成的杂环基)-C 1-4亚烷基、C 6-10芳基、C 6-10芳基-C 1-4亚烷基、5-8个原子组成的杂芳基和(5-8个原子组成的杂芳基)-C 1-4亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    或R c、R d和与它们相连的氮原子一起形成3-8个原子组成的杂环或5-8个原子组成的杂芳环,其中,所述3-8个原子组成的杂环和5-8个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、OH、-NH 2、-C(=O)OH、-C(=O)NH 2、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6烷氨基或C 1-6卤代烷氧基;
    n为0、1、2或3;
    t为0、1、2、3、4、5或6;条件是,当R 5和R 8同时为H,且R 9
    Figure PCTCN2019073550-appb-100002
    时,t不为0。
  2. 根据权利要求1所述的化合物,其具有式(II)所示结构:
    Figure PCTCN2019073550-appb-100003
  3. 根据权利要求1或2所述的化合物,其中,R 1为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、甲基、乙基、正丙基、异丙基、丙烯基、丙炔基、甲氧基、乙氧基、羟基甲基、三氟甲基、三氟乙基、一氟甲基、三氟甲氧基、二氟甲氧基、甲氨基、环丙基、环丁基、环戊基、环己基或环丙基-亚甲基,其中, 所述甲基、乙基、正丙基、异丙基、丙烯基、丙炔基、甲氧基、乙氧基、羟基甲基、三氟乙基、一氟甲基、二氟甲氧基、甲氨基、环丙基、环丁基、环戊基、环己基和环丙基-亚甲基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
  4. 根据权利要求1-3任意一项所述的化合物,其中,R 2和R 3各自独立地为H、D、CN、OH、NH 2、-SH、甲基、乙基、正丙基、异丙基、乙烯基、丙炔基、甲氧基、乙氧基、甲硫基、甲氨基、环丙基、环丁基、环戊基、环己基或5-6个原子组成的杂环基,其中,所述甲基、乙基、正丙基、异丙基、乙烯基、丙炔基、甲氧基、乙氧基、甲硫基、甲氨基、环丙基、环丁基、环戊基、环己基和5-6个原子组成的杂环基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
    或R 2、R 3和与它们相连的碳原子一起形成羰基;
    或R 2、R 3和与它们相连的碳原子一起形成环丙烷、环丁烷、环戊烷、环己烷或5-6个原子组成的杂环,其中,所述环丙烷、环丁烷、环戊烷、环己烷和5-6个原子组成的杂环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
  5. 根据权利要求1-4任意一项所述的化合物,其中,R 4为H、D、-OR 4a或-SR 4b
    R 4a和R 4b各自独立地为H、D、甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述甲基、乙基、正丙基、异丙基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、异噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、-C(=O)OH、-SH、=O、甲基、乙基、正丙基、异丙基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
  6. 根据权利要求1-5任意一项所述的化合物,其中,R 10为H、D、F、Cl、Br、I、OH、CN、NO 2、NH 2、甲基、乙基、正丙基、异丙基、甲氧基或乙氧基。
  7. 根据权利要求1-6任意一项所述的化合物,其中,R 5为H、D、C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基,其中,所述C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基和5-6个原子组成的杂芳基各自独立地未被取代或被1、 2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
    R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基,其中,所述C 1-4烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基和5-6个原子组成的杂芳基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基、C 1-4卤代烷氧基、C 3-6环烷基、5-6个原子组成的杂环基、C 6-10芳基或5-6个原子组成的杂芳基;
    或R 6、R 7和与它们相连的碳原子一起形成C 3-6碳环、5-6个原子组成的杂环、C 6-10芳环或5-6个原子组成的杂芳环,其中,所述C 3-6碳环、5-6个原子组成的杂环、C 6-10芳环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基;
    或R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
  8. 根据权利要求1-7任意一项所述的化合物,其中,R 5为H、D、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、三氟甲基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基或嘧啶基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基和嘧啶基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基或苯基;
    R 6和R 7各自独立地为H、D、F、Cl、Br、I、CN、NO 2、OH、NH 2、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、甲氧基、乙氧基、甲氨基、三氟甲基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉 基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基或嘧啶基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、甲氧基、乙氧基、甲氨基、二氟甲氧基、环丙基、环丁基、环戊基、环己基、吡咯烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、苯基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基和嘧啶基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基、二氟甲氧基、环丙基、环丁基、环戊基或苯基;
    或R 6、R 7和与它们相连的碳原子一起形成环丙烷、环丁烷、环戊烷、环己烷、吡咯烷、四氢呋喃、四氢噻吩、四氢吡喃、四氢噻喃、哌啶、吗啉、硫代吗啉、哌嗪、苯环、呋喃、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、噻吩、吡嗪、哒嗪或嘧啶,其中,所述环丙烷、环丁烷、环戊烷、环己烷、吡咯烷、四氢呋喃、四氢噻吩、四氢吡喃、四氢噻喃、哌啶、吗啉、硫代吗啉、哌嗪、苯环、呋喃、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、噻吩、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
    或R 5、R 6和与它们各自相连的原子一起、或R 5、R 7和与它们各自相连的原子一起,形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡咯、吡啶、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
  9. 根据权利要求1-8任意一项所述的化合物,其中,R 8和R 9各自独立为H、D、R eO-C 1-4亚烷基、R dR cN-C 1-4亚烷基、C 1-6烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中所述C 1-6烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基;
    或R 8、R 9和与它们相连的氮原子一起形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中, 所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OR e、-NR cR d、-C(=O)OR e、-C(=O)NHR f、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
  10. 根据权利要求1-9任意一项所述的化合物,其中,R 8和R 9各自独立为H、D、R dR cN-C 1-4亚烷基、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、正戊基、正己基、三氟甲基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、C 6-10芳基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、正戊基、正己基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、C 6-10芳基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OH、-NR cR d、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
    或R 8、R 9和与它们相连的氮原子一起形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、-OH、-NR cR d、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
  11. 根据权利要求1-10任意一项所述的化合物,其中,R c、R d、R e和R f各自独立地为H、D、C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述C 1-4烷基、C 1-4卤代烷基、C 3-6环烷基、C 3-6环烷基-C 1-2亚烷基、5-6个原子组成的杂环基、(5-6个原子组成的杂环基)-C 1-2亚烷基、C 6-10芳基、C 6-10芳基-C 1-2亚烷基、5-6个原子组成的杂芳基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基;
    或R c、R d和与它们相连的氮原子一起形成3-6个原子组成的杂环或5-6个原子组成的杂芳环,其中,所述3-6个原子组成的杂环和5-6个原子组成的杂芳环各自独立地未被取代或被1、2、3或4个取代基所 取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、=O、OH、-NH 2、-C(=O)OH、-C(=O)NH 2、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 1-4卤代烷基、C 1-4烷氧基、C 1-4烷氨基或C 1-4卤代烷氧基。
  12. 根据权利要求1-11任意一项所述的化合物,其中,R c、R d、R e和R f各自独立地为H、D、甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、三氟甲基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基或(5-6个原子组成的杂芳基)-C 1-2亚烷基,其中,所述甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、环丙基、环丁基、环戊基、环己基、C 3-6环烷基-C 1-2亚烷基、吡咯烷基、吡唑烷基、咪唑烷基、噁唑烷基、噻唑烷基、四氢呋喃基、四氢噻吩基、四氢吡喃基、四氢噻喃基、哌啶基、吗啉基、硫代吗啉基、哌嗪基、(5-6个原子组成的杂环基)-C 1-2亚烷基、苯基、苯基-C 1-2亚烷基、呋喃基、吡咯基、吡啶基、吡唑基、咪唑基、***基、四唑基、噁唑基、噁二唑基、1,3,5-三嗪基、噻唑基、噻吩基、吡嗪基、哒嗪基、嘧啶基和(5-6个原子组成的杂芳基)-C 1-2亚烷基各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、-NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基;
    或R c、R d和与它们相连的氮原子一起形成氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪或嘧啶,其中,所述氮丙啶、氮杂环丁烷、吡咯烷、吡唑烷、咪唑烷、噁唑烷、噻唑烷、哌啶、吗啉、硫代吗啉、哌嗪、吡唑、咪唑、***、四唑、噁唑、噁二唑、1,3,5-三嗪、噻唑、吡嗪、哒嗪和嘧啶各自独立地未被取代或被1、2、3或4个取代基所取代,所述取代基独立地选自D、F、Cl、Br、I、CN、NO 2、OH、NH 2、=O、-C(=O)OH、-C(=O)NH 2、甲基、乙基、正丙基、异丙基、乙烯基、乙炔基、三氟甲基、甲氧基、乙氧基、甲氨基、三氟甲氧基或二氟甲氧基。
  13. 根据权利要求1-12任意一项所述的化合物,其具有以下其中之一的结构:
    Figure PCTCN2019073550-appb-100004
    Figure PCTCN2019073550-appb-100005
    Figure PCTCN2019073550-appb-100006
    Figure PCTCN2019073550-appb-100007
    Figure PCTCN2019073550-appb-100008
    Figure PCTCN2019073550-appb-100009
    或其立体异构体、几何异构体、互变异构体、氮氧化物、溶剂化物、代谢产物、药学上可接受的盐、二聚体、三聚体或它们的前药。
  14. 一种药物组合物,其包含权利要求1-13任意一项所述的化合物以及药学上可接受的载体、赋形剂、辅剂、媒介物或它们的组合。
  15. 根据权利要求14所述的药物组合物,进一步包含一种或多种其他附加治疗剂,其中所述其他附加治疗剂选自抗糖尿病药物、抗高血糖药物、抗肥胖症药物、抗高血压药物、食欲抑制药物、降脂药物或其组合。
  16. 根据权利要求15所述的药物组合物,其中,所述的抗糖尿病药物和抗高血糖药物分别独立地选自SGLT2抑制剂、双胍类药物、磺酰脲类药物、葡萄糖苷酶抑制剂、PPAR激动剂、αP2抑制剂、PPARα/γ双激活剂、二肽酰肽酶IV抑制剂、格列奈类药物、胰岛素、胰高血糖素样肽-1抑制剂、PTP1B抑制剂、糖原磷酸化酶抑制剂、葡萄糖-6-磷酸酶抑制剂或其组合;所述的抗肥胖症药物选自中枢抗肥胖药、MCH受体拮抗剂、神经肽Y受体拮抗剂、***素受体拮抗剂、脑肠肽拮抗剂、脂肪酶抑制剂、β3激动剂、11β-HSD1抑制剂、DGAT-1抑制剂、肽性食欲抑制剂、缩胆囊肽激动剂、摄食抑制剂或其组合;所述的降脂药物选自MTP抑制剂、HMGCoA还原酶抑制剂、角鲨烯合成酶抑制剂、贝丁酸类降血脂药物、ACAT抑制剂、脂 加氧酶抑制剂、胆固醇吸收抑制剂、回肠钠离子/胆汁酸协同转运蛋白抑制剂、LDL受体活性的向上调节物、烟酸类降血脂药物、胆汁酸螯合物或其组合;或所述的降脂药物选自普伐他汀、辛伐他汀、阿伐他汀、氟伐他汀、西立伐他汀、埃塔伐他汀、罗素他汀或其组合。
  17. 权利要求1-13任意一项所述的化合物或权利要求14-16任意一项所述的药物组合物在制备药物中的用途,其中,所述药物用于抑制SGLT1;或用于改善肠内环境;或用于预防或治疗疾病、减轻所述疾病症状或者延缓所述疾病的发展或发作,其中所述疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压;
    其中,所述糖尿病并发症为糖尿病性视网膜病、糖尿病性神经病或糖尿病性肾病;所述高脂血症为高甘油三酯血症。
  18. 一种抑制SGLT1;或改善肠内环境;或预防或治疗疾病、减轻所述疾病症状或者延缓所述疾病的发展或发作的方法,包括给患者使用有效量的权利要求1-13任意一项所述的化合物或权利要求14-16任意一项所述的药物组合物,其中所述疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压;
    其中,所述糖尿病并发症为糖尿病性视网膜病、糖尿病性神经病或糖尿病性肾病;所述高脂血症为高甘油三酯血症。
  19. 权利要求1-13任意一项所述的化合物或权利要求14-16任意一项所述的药物组合物用于抑制SGLT1;或用于改善肠内环境;或用于预防或治疗疾病、减轻所述疾病症状或者延缓所述疾病的发展或发作的用途,其中所述疾病为糖尿病、糖尿病并发症、胰岛素抵抗、高血糖、高胰岛素血症、高脂血症、肥胖症、X综合症、动脉粥样硬化、心血管疾病、充血性心力衰竭、低镁血症、低钠血症、肾衰竭、与血浓缩相关的障碍、便秘或者高血压;
    其中,所述糖尿病并发症为糖尿病性视网膜病、糖尿病性神经病或糖尿病性肾病;所述高脂血症为高甘油三酯血症。
PCT/CN2019/073550 2018-01-31 2019-01-29 吡喃葡萄糖基衍生物及其用途 WO2019149178A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US16/965,438 US11186602B2 (en) 2018-01-31 2019-01-29 Glucopyranosyl derivative and use thereof
CN201980008449.3A CN111630058B (zh) 2018-01-31 2019-01-29 吡喃葡萄糖基衍生物及其用途
EP19748264.9A EP3747892A4 (en) 2018-01-31 2019-01-29 GLUCOPYRANOSYL DERIVATIVE AND USE OF IT

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810093154 2018-01-31
CN201810093154.8 2018-01-31

Publications (1)

Publication Number Publication Date
WO2019149178A1 true WO2019149178A1 (zh) 2019-08-08

Family

ID=67478617

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/073550 WO2019149178A1 (zh) 2018-01-31 2019-01-29 吡喃葡萄糖基衍生物及其用途

Country Status (4)

Country Link
US (1) US11186602B2 (zh)
EP (1) EP3747892A4 (zh)
CN (1) CN111630058B (zh)
WO (1) WO2019149178A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021004498A1 (en) * 2019-07-10 2021-01-14 Sunshine Lake Pharma Co., Ltd. Glucopyranosyl derivatives and their uses
WO2021092341A1 (en) * 2019-11-07 2021-05-14 Increvet, Inc. Sodium-glucose linked transporter inhibitors for the management of chronic kidney disease, hypertension, and heart failure in companion animals
US11723929B2 (en) 2017-06-13 2023-08-15 The Regents Of The University Of California Methods of improving cell-based therapy

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006035967A1 (en) 2004-09-30 2006-04-06 Taisho Pharmaceutical Co., Ltd. Pyridine derivatives and their use as medicaments for treating diseases related to mch receptor
US20110218159A1 (en) 2010-03-02 2011-09-08 Philip Manton Brown Methods of using inhibitors of sodium-glucose cotransporters 1 and 2
WO2013038429A2 (en) * 2011-09-13 2013-03-21 Panacea Biotec Ltd. Novel sglt inhibitors
CN104854096A (zh) * 2012-11-20 2015-08-19 莱西肯医药有限公司 钠葡萄糖协同转运蛋白1的抑制剂
CN105461762A (zh) * 2014-09-27 2016-04-06 广东东阳光药业有限公司 吡喃葡萄糖基衍生物及其在医药上的应用
CN104619713B (zh) * 2013-05-24 2017-06-16 四川海思科制药有限公司 氧杂双环衍生物、制备方法及其应用

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2004137245A (ja) 2002-08-23 2004-05-13 Kissei Pharmaceut Co Ltd ピラゾール誘導体、それを含有する医薬組成物、その医薬用途及びその製造中間体
JP2009167104A (ja) 2006-05-02 2009-07-30 Taisho Pharmaceutical Co Ltd フェニル5−チオグリコシド化合物
AU2007252432B2 (en) 2006-05-19 2011-11-17 Taisho Pharmaceutical Co., Ltd. C-phenyl glycitol compound
MX2008016231A (es) 2006-06-29 2009-01-16 Taisho Pharma Co Ltd Compuesto de c-fenil 1-tioglucitol.
US8466113B2 (en) * 2009-02-23 2013-06-18 Taisho Pharmaceutical Co., Ltd. 4-isopropylphenyl glucitol compounds as SGLT1 inhibitors
JP2013224263A (ja) * 2010-08-19 2013-10-31 Taisho Pharmaceutical Co Ltd 4−イソプロピルフェニルグルシトール化合物
EP2607360B1 (en) 2010-08-20 2015-08-19 Taisho Pharmaceutical Co., Ltd. 4-Isopropyl-6-methoxyphenyl glucitol compound

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006035967A1 (en) 2004-09-30 2006-04-06 Taisho Pharmaceutical Co., Ltd. Pyridine derivatives and their use as medicaments for treating diseases related to mch receptor
US20110218159A1 (en) 2010-03-02 2011-09-08 Philip Manton Brown Methods of using inhibitors of sodium-glucose cotransporters 1 and 2
WO2013038429A2 (en) * 2011-09-13 2013-03-21 Panacea Biotec Ltd. Novel sglt inhibitors
CN104854096A (zh) * 2012-11-20 2015-08-19 莱西肯医药有限公司 钠葡萄糖协同转运蛋白1的抑制剂
CN104619713B (zh) * 2013-05-24 2017-06-16 四川海思科制药有限公司 氧杂双环衍生物、制备方法及其应用
CN105461762A (zh) * 2014-09-27 2016-04-06 广东东阳光药业有限公司 吡喃葡萄糖基衍生物及其在医药上的应用

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
"Encyclopedia of Pharmaceutical Technology", 1988, MARCEL DEKKER
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1289 - 1329
BERGE ET AL., J. PHARMACOL SCI, vol. 66, 1977, pages 1 - 19
ELIEL, E.L.: "Stereochemistry of Carbon Compounds", 1962, MCGRAW-HILL
ELIEL, E.WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC.
HECKER ET AL.: "Prodrugs of Phosphates and Phosphonates", J. MED. CHEM., vol. 51, 2008, pages 2328 - 2345, XP008148502, DOI: 10.1021/jm701260b
HIGUCHI ET AL.: "Pro-drugs as Novel Delivery Systems", A.C.S. SYMPOSIUM SERIES, vol. 14
JACQUES ET AL.: "Enantiomers, Racemates and Resolutions", 1981, WILEY INTERSCIENCE
JOHANNSSON ET AL., J. CLIN. ENDOCRINOL. METAB., vol. 82, 1997, pages 727 - 734
L. W. DEADY, SYN. COMM., vol. 7, 1977, pages 509 - 514
MORIYA, R. ET AL., AM J PHYSIOL ENDORINOL METAB, vol. 297, 2009, pages E1358 - E1365
P. J. KOCIENSKI: "Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
RAUTIO ET AL.: "Prodrugs: Design and Clinical Applications", NATURE REVIEWS DRUG DISCOVERY, vol. 7, 2008, pages 255 - 270, XP055227338, DOI: 10.1038/nrd2468
ROBERT E. GAWLEYJEFFREY AUBE: "Principles of Asymmetric Synthesis", 2012, ELSEVIER
ROCHE ET AL.: "Bioreversible Carriers in Drug Design", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
S. P. PARKER: "McGraw-Hill Dictionary of Chemical Terms", 1984, MCGRAW-HILL BOOK COMPANY
SMITH ET AL.: "March's Advanced Organic Chemistry", 2007, JOHN WILEY & SONS
STAHLWERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
T. W. GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
THOMAS SORRELL: "Organic Chemistry", 1999, UNIVERSITY SCIENCE BOOKS
WILEN, S.H.: "Tables of Resolving Agents and Optical Resolutions", 1972, UNIV. OF NOTRE DAME PRESS, pages: 268

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11723929B2 (en) 2017-06-13 2023-08-15 The Regents Of The University Of California Methods of improving cell-based therapy
WO2021004498A1 (en) * 2019-07-10 2021-01-14 Sunshine Lake Pharma Co., Ltd. Glucopyranosyl derivatives and their uses
WO2021092341A1 (en) * 2019-11-07 2021-05-14 Increvet, Inc. Sodium-glucose linked transporter inhibitors for the management of chronic kidney disease, hypertension, and heart failure in companion animals

Also Published As

Publication number Publication date
US11186602B2 (en) 2021-11-30
CN111630058A (zh) 2020-09-04
EP3747892A1 (en) 2020-12-09
CN111630058B (zh) 2022-02-15
EP3747892A4 (en) 2021-11-03
US20210054013A1 (en) 2021-02-25

Similar Documents

Publication Publication Date Title
TWI791087B (zh) 吡喃葡萄糖基衍生物及其用途
CN106892948B (zh) 吡喃葡萄糖基衍生物及其在医药上的应用
CN111630058B (zh) 吡喃葡萄糖基衍生物及其用途
CN111164072A (zh) β-羟基杂环胺及其在治疗高糖血症中的用途
CN113072542B (zh) RORγt抑制剂及其制备方法和用途
US20220411400A1 (en) Chemical compound as thyroid hormone beta receptor agonist and use thereof
WO2020182018A1 (zh) 氮杂环化合物、其制备方法及用途
WO2019024924A1 (zh) 抑制ssao/vap-1的胺类化合物及其在医药上的应用
WO2020083264A1 (zh) 胍类衍生物及其用途
TW201439084A (zh) 作爲丙型肝炎抑制劑的螺環化合物、藥物組合物及它們在藥物中的應用
WO2020233583A1 (zh) 抑制ssao/vap-1的异喹啉酮类化合物及其用途
CN112209908B (zh) 吡喃葡萄糖基衍生物及其用途
WO2015043473A1 (en) Glucopyranosyl derivatives and their uses in medicine
CN113072538B (zh) RORγt抑制剂及其在药物中的应用
CN114728925B (zh) 一种作为ssao/vap-1抑制剂的胺类衍生物及其用途
WO2024008044A1 (zh) 大环拟肽类蛋白酶抑制剂及其用途
WO2023179078A1 (zh) 咪唑并[1,2-a]吡嗪或吡唑并[1,5-a]嘧啶衍生物及其用途
WO2013178064A1 (zh) 吡喃葡萄糖基衍生物、其制备方法及其在医药上的应用
CN113072546A (zh) 五元杂芳衍生物及其制备方法和用途
CN111196806A (zh) 胍类衍生物及其用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19748264

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2019748264

Country of ref document: EP

Effective date: 20200831