NZ626751B2 - Aryl dihydropyridinones and piperidinones as mgat2 inhibitors - Google Patents

Aryl dihydropyridinones and piperidinones as mgat2 inhibitors Download PDF

Info

Publication number
NZ626751B2
NZ626751B2 NZ626751A NZ62675112A NZ626751B2 NZ 626751 B2 NZ626751 B2 NZ 626751B2 NZ 626751 A NZ626751 A NZ 626751A NZ 62675112 A NZ62675112 A NZ 62675112A NZ 626751 B2 NZ626751 B2 NZ 626751B2
Authority
NZ
New Zealand
Prior art keywords
isomer
alkyl
rac
independently selected
group
Prior art date
Application number
NZ626751A
Other versions
NZ626751A (en
Inventor
Saleem Ahmad
Robert Paul Brigance
Dong Cheng
Pratik Devasthale
Jon J Hangeland
R Michael Lawrence
Wei Meng
Huji Turdi
Guohua Zhao
Original Assignee
Bristol Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/688,584 external-priority patent/US8791091B2/en
Application filed by Bristol Myers Squibb Company filed Critical Bristol Myers Squibb Company
Publication of NZ626751A publication Critical patent/NZ626751A/en
Publication of NZ626751B2 publication Critical patent/NZ626751B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/438The ring being spiro-condensed with carbocyclic or heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/80Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D211/84Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen directly attached to ring carbon atoms
    • C07D211/86Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/80Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D211/84Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen directly attached to ring carbon atoms
    • C07D211/90Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/20Spiro-condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/14Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/38Phosphonic acids RP(=O)(OH)2; Thiophosphonic acids, i.e. RP(=X)(XH)2 (X = S, Se)
    • C07F9/40Esters thereof
    • C07F9/4003Esters thereof the acid moiety containing a substituent or a structure which is considered as characteristic
    • C07F9/4006Esters of acyclic acids which can have further substituents on alkyl
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/28Phosphorus compounds with one or more P—C bonds
    • C07F9/535Organo-phosphoranes
    • C07F9/5352Phosphoranes containing the structure P=C-
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/59Hydrogenated pyridine rings

Abstract

Disclosed are dihydropyridinone and piperidinone compounds of formula (I), wherein the substituents are as defined in the specification. The compounds are monoacylglycerol acyltransferase type 2 (MGAT2) inhibitors which may be used as medicaments for the treatment or prophylaxis of diabetes, obesity, and dyslipidemia. Example of a compound of formula (I) are: (S)-3-(1H-tetrazol-5-yl)-4-(p-tolyl)-6-(4-(4,4,4-trifluorobutoxy)phenyl)-6-(trifluoromethyl)-5,6-dihydropyridin-2(1H)-one (S)-N-(4-methoxyphenyl)-2-oxo-4-(p-tolyl)-6-(4-(4,4,4-trifluorobutoxy)phenyl)-6-(trifluoromethyl)-1,2,5,6-tetrahydropyridine-3-carboxamide (S)-2-(methylsulfonyl)-N-(2-oxo-4-(p-tolyl)-6-(4-(4,4,4-trifluorobutoxy)phenyl)-6-(trifluoromethyl)-1,2,5,6-tetrahydropyridin-3-yl)acetamide , and dyslipidemia. Example of a compound of formula (I) are: (S)-3-(1H-tetrazol-5-yl)-4-(p-tolyl)-6-(4-(4,4,4-trifluorobutoxy)phenyl)-6-(trifluoromethyl)-5,6-dihydropyridin-2(1H)-one (S)-N-(4-methoxyphenyl)-2-oxo-4-(p-tolyl)-6-(4-(4,4,4-trifluorobutoxy)phenyl)-6-(trifluoromethyl)-1,2,5,6-tetrahydropyridine-3-carboxamide (S)-2-(methylsulfonyl)-N-(2-oxo-4-(p-tolyl)-6-(4-(4,4,4-trifluorobutoxy)phenyl)-6-(trifluoromethyl)-1,2,5,6-tetrahydropyridin-3-yl)acetamide

Description

-PCT ARYL DIHYDROPYRIDINONES AND PIPERIDINONES AS MGAT2 INHIBITORS CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the priority benefit of U.S. Provisional Application No. 61/566,039 filed December 2, 2011 and the priority benefit of U.S. Non-Provisional Application No. 13/688,584 filed November 29, 2012, both of which are incorporated fully herein by reference.
FIELD OF THE INVENTION
[0002] The present invention provides novel aryl dihydropyridinone and piperidinone compounds, and their analogues thereof, which are MGAT2 inhibitors, compositions containing them, and methods of using them, for example, for the treatment or prophylaxis of diabetes, obesity, dyslipidemia and related conditions.
BACKGROUND OF THE INVENTION The prevalence of obesity and diabetes is increasing at an alarming rate.
According to WHO, in 2008, 70% of the U.S. adult population was overweight, and among them 33% were obese. Parallel to the explosive number of people becoming overweight and obese, in 2008, it was estimated that 12.3% of the U.S. population had elevated blood glucose [http://www.who.int/diabetes/facts/en/]. The obesity/diabetes epidemic is not unique to the U.S. According to WHO (Fact Sheet No. 312, September 2012), 347 million people worldwide have diabetes. Treating obesity and improving glycemic control effectively and safely remain major challenges for modern medicine.
Monoacylglycerol acyltransferase 2 (MGAT2) has emerged as an attractive target for the treatment of obesity and type II diabetes [Yen, C.L. et al., Nat. Med., (4):442-446 (2009)]. MGAT2 is highly and selectively expressed in the small intestine where it exerts a pivotal role in the monoacylglycerol-pathway for the absorption of dietary fat. When dietary fat is ingested, pancreatic lipase digests triglycerides into free fatty acids and 2-monoacylglycerol, which are absorbed by intestinal epithelial enterocytes. Once inside enterocytes, free fatty acids and 2-monoacylglycerol are used as building blocks to resynthesize triglycerides by two sequential acylation steps; first by MGAT and then by DGAT enzyme reactions. Triglycerides are then incorporated into 11841-PCT chylomicrons and secreted into lymph to be utilized as an energy supply for the body.
MGAT2 knockout mice exhibit a healthy metabolic phenotype and show resistance to high-fat diet induced obesity, improvement in insulin sensitivity and decreased fat accumulation in liver and adipose tissue. In addition, genetic deletion of MGAT2 produces mice with increased levels of GLP1 [Yen, C.L. et al., Nat. Med., 15(4):442-446 (2009)]. Taken together, these data show that MGAT2 inhibitors hold promise to treat metabolic disorders such as obesity, type II diabetes and dyslipidemia.
SUMMARY OF THE INVENTION
[0005] The present invention provides aryl dihydropyridinone and piperidinone compounds, and their analogues thereof, which are useful as MGAT2 inhibitors, including stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.
The present invention also provides processes and intermediates for making the compounds of the present invention or stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.
The present invention also provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and at least one of the compounds of the present invention or stereoisomers, tautomers, pharmaceutically acceptable salts, or solvates thereof.
[0008] The compounds of the invention may be used in the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2, such as diabetes, obesity, dyslipidemia and related conditions, such as microvascular and macrovascular complications associated with diabetes, cardiovascular diseases, Metabolic Syndrome and its component conditions, disorders of glucose and lipid metabolism and other maladies.
[0009] The compounds of the invention may be used in therapy.
The compounds of the invention may be used for the manufacture of a medicament for the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2.
The compounds of the invention can be used alone, in combination with other compounds of the present invention, or in combination with one or more other agent(s).
Other features and advantages of the invention will be apparent from the following detailed description and claims. 11841-PCT DETAILED DESCRIPTION OF THE INVENTION I. COMPOUNDS OF THE INVENTION In a first aspect, the present invention provides, inter alia, a compound of Formula (I): 12 14 or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, wherein: designates a single or double bond; x and y can be both a single bond; when x is a double bond, then y is a single 4 16 5 bond and R and R are absent; when y is a double bond, then x is a single bond and R and R are absent; R is independently selected from the group consisting of: -CONH(C alkyl), 4-18 -CONHC haloalkyl, -CONH(CH ) Ph, -CONHCH COC alkyl, 2-8 2 1-8 2 2-8 -(CH ) -(C carbocycle substituted with 0-2 R and 0-2 R ), -(CH ) -(5- to 2 m 3-10 2 m 6-membered heteroaryl comprising: carbon atoms and 1-4 heteroatoms selected from N, e b g NR , O and S; wherein said heteroaryl is substituted with 0-1 R and 0-2 R ), and a C 1-12 hydrocarbon chain substituted with 0-3 R ; wherein said hydrocarbon chain may be straight or branched, saturated or unsaturated; R is independently selected from the group consisting of: C alkyl, C cycloalkyl, and C haloalkyl; 3-4 1-4 R is independently selected from the group consisting of: H, F, Cl, C alkyl and 11841-PCT R and R are independently selected from the group consisting of: H, F, Cl, and C alkyl; when x is a single bond, R and R may be combined with the carbon atom to which they are attached to form a 3- to 6-membered carbocycle; R is independently selected from the group consisting of: H, halo, C alkyl, NO , R , -(CH ) -(X) -(CH ) R , NH , -CONH(C alkyl), -NHCOX SO R , 2 2 n t 2 m 2 1-6 1 2 j j j -NHCOCH PO(OEt) , -NHCOCOR , -NHCOCH(OH)R , -NHCOCH COR , 2 2 2 j f j -NHCONHR , and -OCONR R ; X is independently selected from the group consisting of: O, S, NH, CONH, and NHCO; X is independently C hydrocarbon chain optionally substituted with C alkyl 1 1-4 1-4 or C cycloalkyl; when y is a single bond, R and R may be combined with the carbon atom to which they are attached to form a 3- to 6-membered carbocycle; 11 12 13 14 15 R , R , R , R and R are independently selected from the group consisting of: H, halo, C alkyl substituted with 0-2 R , C alkoxy, C haloalkyl, 1-4 1-4 1-4 f j j j C haloalkoxy, -(CH ) -C cycloalkyl, CN, NR R , OR , SR , NHCO (C alkyl), 1-4 2 m 3-6 2 1-4 NHSO (C alkyl), and a 4- to 6-membered heterocycle comprising: carbon atoms and 2 1-4 1-4 heteroatoms selected from N, NR , O, and S; 11 12 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5 to 6-membered carbocyclic ring or a 5 to 6-membered heterocyclic ring comprising: carbon atoms and 1-3 heteroatoms selected from N, NR , O, and S; 12 13 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5 to 6-membered carbocyclic ring or a 5 to 6-membered heterocyclic ring comprising: carbon atoms and 1-3 heteroatoms selected from N, NR , O, and S; R is independently selected from the group consisting of: H and C alkyl; 11841-PCT R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkoxy, C haloalkyl, C haloalkoxy, N(C alkyl) , 1-6 1-6 1-6 1-4 2 -(CH ) -(X) -(CH ) R , and -(CH ) -(CH O) -(CH ) R ; 2 n t 2 m 2 n 2 m 2 n R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, C alkylthio, 1-10 1-10 1-10 1-10 1-10 C haloalkyltho, N(C alkyl) , -CONH(CH ) H, -O(CH ) O(C alkyl), R , 1-10 1-4 2 2 4-20 2 s 1-6 -(CH ) -(X) -(CH ) R , and -(CH ) -(CH O) -(CH ) R ; 2 n t 2 m 2 n 2 m 2 n R is, at each occurrence, independently selected from the group consisting of: C cycloalkyl substituted with 0-2 R , C cycloalkenyl substituted with 0-2 R , 3-6 3-6 -(CH ) -(phenyl substituted with 0-3 R ), and a 5- to 6-membered heterocycle comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S; wherein said heterocycle is substituted with 0-2 R ; R is, at each occurrence, independently selected from the group consisting of: halo, OH, CN, NO , C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, tetrazolyl, 2 1-4 1-4 1-4 1-4 OBn and phenyl substituted with 0-2 R ; R is, at each occurrence, independently selected from the group consisting of: H, C alkyl, C haloalkyl, benzyl optionally substituted with C alkoxy, CO(C alkyl) 1-8 1-8 1-4 1-4 and COBn; R is, at each occurrence, independently selected from the group consisting of: H and C alkyl; g h i R , R and R are, at each occurrence, independently selected from the group consisting of: halo, C alkyl, C alkoxy, C haloalkyl, and C haloalkoxy; 1-4 1-4 1-4 1-4 R is, at each occurrence, independently selected from the group consisting of: C alkyl, C cycloalkyl and phenyl; 1-4 3-4 n, at each occurrence, is independently 0 or 1; m, at each occurrence, is independently 0, 1, 2, 3, or 4; s, at each occurrence, is independently 1, 2, or 3; and t, at each occurrence, is independently 0 or 1; provided that the following compound is excluded: 11841-PCT Me H In a second aspect, the present invention includes a compound of Formula (I), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the first aspect, wherein: R is independently selected from the group consisting of: -CONHC alkyl, 4-18 -CONH(CH ) Ph, C alkyl substituted with 0-2 R , C alkenyl substituted with 2 1-8 1-12 1-12 a a b 0-2 R , C alkynyl substituted with 0-2 R , -(CH ) -(phenyl substituted with 0-1 R 1-12 2 m and 0-2 R ), -(CH ) -(C cycloalkyl substituted with 0-1 R ), and 2 m 3-6 -(CH ) -(5- to 6-membered heteroaryl substituted with 0-1 R and 0-2 R ), wherein said heteroaryl is selected from: pyridyl, oxazolyl, thiazolyl and .
In a third aspect, the present invention includes a compound of Formula (I), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the first or second aspect, wherein: 11 15 R and R are independently selected from the group consisting of: H, C alkyl and halo; 12 14 R and R are independently selected from the group consisting of: H, halo, C alkyl and C alkoxy; and 1-4 1-4 R is independently selected from the group consisting of: H, halo, C alkyl substituted with 0-1 R , C alkoxy, C haloalkyl, C haloalkoxy, 1-4 1-4 1-4 f j j -(CH ) -C cycloalkyl, CN, NR R , SR , NHCO (C alkyl), NHSO (C alkyl), and 2 m 3-4 2 1-4 2 1-4 a 4- to 6-membered heterocycle comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S.
In a fourth aspect, the present invention provides a compound of Formula (II): 11841-PCT (II) or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof; within the scope of any of the above aspects.
[0017] In a fifth aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the above aspects, wherein: R is independently selected from the group consisting of: C alkyl, C cycloalkyl, -CONHC alkyl, -CONHC haloalkyl, -CONH(CH ) Ph, 3-6 4-18 2-8 2 1-8 -(CH ) -(phenyl substituted with 1 R and 0-2 R ), and a 5- to 6-membered heteroaryl substituted with 0-1 R and 0-2 R , wherein said heteroaryl is selected from: pyridyl, oxazolyl, thiazolyl and ; R is independently selected from the group consisting of: C alkyl and C haloalkyl; R is independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: NH , -CONH(C alkyl), R , -(CH ) -(X) -(CH ) R , -NHCO(CH )SO (C alkyl), 1-6 2 n t 2 m 2 2 1-4 -NHCOCH PO(OEt) , -NHCOCO(C alkyl), -NHCOCH(OH)(C alkyl), 2 2 1-4 1-4 -NHCOCH CO(C alkyl), -NHCONH(C alkyl), and -OCONH(C alkyl); 2 1-4 1-4 1-4 11 15 R and R are independently selected from the group consisting of: H, C alkyl and halo; 11841-PCT 12 14 R and R are independently selected from the group consisting of: H, halo, C alkyl and C alkoxy; 1-4 1-4 R is independently selected from the group consisting of: H, halo, C alkyl substituted with 0-1 C alkoxy, C alkoxy, C haloalkyl, C haloalkoxy, 1-4 1-4 1-4 1-4 -(CH ) -C cycloalkyl, CN, N(C alkyl) , NHCO (C alkyl), NHSO (C alkyl), 2 m 3-4 1-4 2 2 1-4 2 1-4 pyrazolyl, and morpholinyl; 12 13 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5 to 6-membered carbocyclic ring or a 5 to 6-membered heterocyclic ring comprising: carbon atoms and 1-3 heteroatoms selected from N, NR , O, and S; R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, 1-8 1-8 1-8 1-10 -O(CH ) O(C alkyl), N(C alkyl) , -CONH(CH ) H, 2 s 1-6 1-4 2 2 6-20 -(CH ) (C cycloalkyl), -(CH ) (C cycloalkenyl), -O(CH ) (C cycloalkyl), 2 m 3-6 2 m 4-6 2 m 3-6 4-C alkoxy-Ph, -O(CH ) Ph, morpholinyl, pyridyl, 2-C alkoxy-pyridinyl, 1-4 2 m 1-4 pyrimidinyl, pyrazinyl, and -O-pyrimidinyl; R is, at each occurrence, independently selected from the group consisting of: halo, C alkyl, C alkoxy, C haloalkyl, and C haloalkoxy; 1-4 1-4 1-4 1-4 m, at each occurrence, is independently 0, 1, 2 or 3; and s, at each occurrence, is independently 1, 2, or 3.
In a sixth aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the above aspects, wherein: R is independently selected from the group consisting of: C alkyl, -CONHC alkyl, -CONH(CH ) Ph, and ; 4-18 2 1-8 R is independently selected from the group consisting of: NH , 11841-PCT -CONH(C alkyl), -NHCOCH PO(OEt) , -NHCO(CH )SO (C alkyl), R , OR , 1-6 2 2 2 2 1-4 -CONHR , and -NHCOR ; R is independently selected from the group consisting of: H, halo, C alkyl and C alkoxy; R is independently selected from the group consisting of: H, halo, C alkyl substituted with 0-1 C alkoxy, C alkoxy, C haloalkyl, C haloalkoxy, 1-4 1-4 1-4 1-4 -(CH ) -C cycloalkyl, CN, N(C alkyl) , NHCO (C alkyl), NHSO (C alkyl), 2 m 3-4 1-4 2 2 1-4 2 1-4 pyrazolyl, and morpholinyl; 12 13 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5 to 6-membered carbocyclic ring or a 5 to 6-membered saturated heterocyclic ring comprising: carbon atoms and 1-2 oxygen atoms; R is independently selected from the group consisting of: H and C alkoxy; R is, at each occurrence, independently selected from the group consisting of: halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) O(C alkyl), 1-6 1-6 1-6 1-10 2 s 1-6 -CONH(CH ) H, -(CH ) (C cycloalkyl), -(CH ) (C cycloalkenyl), 2 6-20 2 m 3-6 2 m 4-6 -O(CH ) (C cycloalkyl), phenoxy, benzoxy, morpholinyl, 2-C alkoxy-pyridinyl, 2 m 3-6 1-4 pyrimidinyl, pyrazinyl and -O-pyrimidinyl; and R is, at each occurrence, independently selected from the group consisting of: C cycloalkyl substituted with 0-2 R , -(CH ) -(phenyl substituted with 0-3 R ), and a 3-6 2 m heteroaryl selected from: oxazolyl, isoxazolyl, thiazolyl, pyrazolyl, imidazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, and pyrazinyl; wherein said heteroaryl is substituted with 0-2 R .
In a seventh aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the above aspects, wherein: R is ; 11841-PCT R is independently selected from the group consisting of: NH , -CONH(C alkyl), OPh, -CONH(C cycloalkyl), -CONHPh, -CONH-(2-halo-Ph), 1-4 3-6 -CONH-(3-halo-Ph), -CONH-(4-halo-Ph), -CONH-(4-C alkyl-Ph), -CONH(4-OH-Ph), -CONH-(3-C alkoxy-Ph), -CONH-(4-C alkoxy-Ph), -CONH-(4-C haloalkyl-Ph), 1-4 1-4 1-4 -CONH-(4-C haloalkoxy-Ph), -CONH-(4-CN-Ph), -CONH-(4-tetrazolyl-Ph), -CONH-(3-haloC alkyl-Ph), -CONH-(3-haloC alkoxy-Ph), -CONH(CH ) Ph, 1-4 1-4 2 2 -CONH(4-(4-C alkoxy-Ph)-thiazolyl), -CONH(1-C alkyl-pyrazolyl), -CONH(5-C alkoxy-pyridyl), 1-4 1-4 -CONH(6-C alkoxy-pyridyl), -CONH(5-C alkoxy-pyrazinyl), 1-4 1-4 -CONH(6-C alkoxy-pyridazinyl), -NHCO(CH )SO (C alkyl), -NHCOPh, 1-4 2 2 1-4 -NHCO(2-C alkyl-Ph), -NHCO(3-C alkyl-Ph), -NHCO(4-C alkyl-Ph), 1-4 1-4 1-4 -NHCO(2-halo-Ph), -NHCO(3-halo-Ph), -NHCO(2-C haloalkyl-Ph), -NHCO(2-C haloalkoxy-Ph), -NHCO(2-halohalo-Ph), -NHCO(2-halohalo-Ph), -NHCO(oxazolyl), -NHCO(isoxazolyl), -NHCO(3-C alkyl-isoxazolyl), -NHCO(4-C alkyl-isoxazolyl), -NHCO(3-C alkoxy-isoxazolyl), 1-4 1-4 -NHCO(4-C alkoxy-isoxazolyl), -NHCO(3-halo-isoxazolyl), -NHCO(3-OBn-isoxazolyl), -NHCO(3-(2-halo-Ph)-isoxazolyl), -NHCO(3-(3-halo-Ph)-isoxazolyl), -NHCO(5-C alkyl-1H-pyrazolyl), imidazolyl, -NHCO(5-C alkyl-1,3,4-oxadiazolyl), -NHCO(1-C alkyl-1,2,3-triazolyl), 1-4 1-4 -NHCO(6-C alkoxy-pyridyl), -NHCO(pyrazinyl), -NHCO(6-halo-pyridazinyl), -C haloalkyl-1,3,4-oxadiazolyl, 3-NO -1H-1,2,4-triazolyl, tetrazolyl and 1-4 2 -C alkyl-tetrazolyl; R is independently selected from the group consisting of: halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -CONH(CH ) H, C cycloalkyl, 1-6 1-6 1-8 2 6-20 3-6 C cycloalkenyl, -O(CH ) (C cycloalkyl), phenoxy, benzoxy, pyrimidinyl, pyrazinyl 4-6 2 m 3-6 and -O-pyrimidinyl; and R is independently selected from the group consisting of: halo and C alkyl. 11841-PCT In an eighth aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the above aspects, wherein: R is independently selected from the group consisting of: CF and Me; R is independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: NH , -CONHMe, OPh, -CONH(cyclopropyl), -CONH(cyclobutyl), -CONH(cyclopentyl), -CONH(cyclohexyl), -CONHPh, -CONH(4-F-Ph), -CONH(2-Cl-Ph), -CONH(4-Cl-Ph), -CONH(4-Me-Ph), -CONH(4-OH-Ph), -CONH(3-OMe-Ph), -CONH(4-OMe-Ph), -CONH(4-CF -Ph), -CONH(4-OCF -Ph), -CONH(1-Me-pyrazolyl), -CONH(4-(1H-tetrazolyl)-Ph), -CONH(4-(2H-tetrazolyl)-Ph), -CONH(3-FMe-Ph), -CONH(3-FOMe-Ph), -CONH(CH ) Ph, -CONH(5-OMe-pyridyl), -CONH(6-OMe-pyridyl), -CONH(5-OMe-pyrazinyl), -CONH(6-OMe-pyridazinyl), -NHCO(CH )SO Me, -NHCOPh, -NHCO(2-Me-Ph), -NHCO(3-Me-Ph), -NHCO(4-Me-Ph), -NHCO(2-Cl-Ph), -NHCO(3-Cl-Ph), -NHCO(2-ClF-Ph), -NHCO(2-ClF-Ph), -NHCO(isoxazolyl), -NHCO(3-Me-isoxazolyl), -NHCO(4-Me-isoxazolyl), -NHCO(3-OMe-isoxazolyl), -NHCO(3-Br-isoxazolyl), -NHCO(3-(2-Cl-Ph)-isoxazolyl), -NHCO(3-(3-F-Ph)-isoxazolyl), -NHCO(3-OBn-isoxazolyl), 1H-imidazolyl, -NHCO(5-Me-1,3,4-oxadiazolyl), -NHCO(1-Me-1,2,3-triazolyl), -NHCO(6-OMe-pyridyl), -NHCO(6-Cl-pyridazinyl), 5-CF -1,3,4-oxadiazolyl, 1H-tetrazolyl, 1H-tetrazolyl, and 2H-tetrazolyl; 11 15 R and R are independently selected from the group consisting of: H, Me, F, and Cl; R is independently selected from the group consisting of: H, F, Cl, Me and OMe; R is independently selected from the group consisting of: H, F, Cl, Br, Me, OMe, OEt, CH OMe, CF , CH CF , OCHF , OCF , CN, N(Me) , cyclopropyl and 2 3 2 3 2 3 2 cyclopropylmethyl; 11841-PCT 12 13 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5 to 6-membered carbocyclic ring or a 5 to 6-membered saturated heterocyclic ring comprising: carbon atoms and 1-2 oxygen atoms; R is H; R is, at each occurrence, independently selected from the group consisting of: n-pentyl, methoxy, n-butoxy, i-butoxy, i-pentoxy, -O(CH ) CF , -O(CH ) CF CF , 2 1-6 3 2 1-4 2 3 -CONH(CH ) H, cyclopropyl, cyclopentenyl, cyclohexenyl, 2 6-20 -O(CH ) (cyclopentyl), phenoxy, benzoxy, pyrimidinyl, pyrazinyl and -O-pyrimidinyl; and R is F.
In a ninth aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the first, second, third, fourth, fifth and sixth apsects, wherein: R is ; R is independently selected from CF and CH ; 6 c c c R is independently selected from: R , -CONHR , -NHCOR , and -NHCOCH SO (C alkyl); 2 2 1-4 R is independently selected from: -O(CH ) CF , -O(CH ) CF CF , 2 1-6 3 2 1-4 2 3 -CONH(CH ) H, cyclopentenyl, cyclohexenyl, -O(CH ) (cyclopentyl), 2 6-20 2 2 phenoxy, benzoxy, pyrimidinyl, pyrazinyl and -O-pyrimidinyl; R is, at each occurrence, independently selected from the group consisting of: -(CH ) -(phenyl substituted with 0-3 R ), and a heteroaryl selected from: oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, and pyrazinyl; wherein said heteroaryl is substituted with 0-2 R ; and 11841-PCT R is, at each occurrence, independently selected from the group consisting of: halo, OH, CN, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, tetrazolyl and 1-4 1-4 1-4 1-4 OBn.
[0022] In another aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of any of the first, second, third, fourth, fifth, sixth and ninth apsects, wherein: R is ; R is independently selected from CF and CH ; 6 c c c R is independently selected from: R , -CONHR , -NHCOR , and -NHCOCH SO (C alkyl); 2 2 1-4 11 12 14 15 R , R , R and R are H; R is independently selected from the group consisting of: H, C alkyl, C haloalkyl, C alkoxy, and C haloalkoxy; and 1-4 1-4 1-4 R is independently selected from: -O(CH ) CF and -O(CH ) CF CF . 2 1-6 3 2 1-4 2 3 In another aspect, the present invention includes a compound of Formula (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the fourth or fifth aspect, wherein: R is ; R is independently selected from CF and CH ; R and R are H; R is independently 5-membered nitrogen heteroaryl; 11 12 14 15 R , R , R and R are H; R is independently selected from the group consisting of: H, C alkyl, C haloalkyl, C alkoxy, and C haloalkoxy; and 1-4 1-4 1-4 11841-PCT R is independently selected from: -O(CH ) CF and -O(CH ) CF CF . 2 1-6 3 2 1-4 2 3 In another aspect, the present invention includes a compound of Formula (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the fourth or ffith aspect, wherein: R is ; R is independently selected from CF and CH ; R and R are H; R is independently selected from: 1H-imidazolyl, 1H-tetrazolyl, 1H-tetrazolyl, and 2H-tetrazolyl; 11 12 14 15 R , R , R and R are H; R is independently selected from the group consisting of: H, Me, OMe, and OCHF ; and R is independently selected from: -O(CH ) CF and -O(CH ) CF CF . 2 1-6 3 2 1-4 2 3 Also described herein is a compound of Formula (I): 12 14 or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, wherein: designates a single or double bond; x and y can be both a single bond; when x is a double bond, then y is a single 4 16 5 bond and R and R are absent; when y is a double bond, then x is a single bond and R and R are absent; R is independently selected from the group consisting of: 11841-PCT -(CH ) -(C carbocycle substituted with 0-3 R ) or a C hydrocarbon chain 2 m 3-10 1-12 substituted with 0-3 R ; wherein said hydrocarbon chain may be straight or branched, saturated or unsaturated; R is independently selected from the group consisting of: C alkyl and C haloalkyl; R is independently selected from the group consisting of: H, halo, C alkyl and R and R are independently selected from the group consisting of: H, halo and C alkyl; when x is a single bond, R and R may be combined with the carbon atom to which they are attached to form a 3- to 6-membered carbocycle; R is independently selected from the group consisting of: H, halo, C alkyl, CN, NO , and -(CH ) -(X) -(CH ) R ; 2 2 n t 2 m X is independently selected from the group consisting of: O, S, NH, CONH and NHCO; when y is a single bond, R and R may be combined with the carbon atom to which they are attached to form a 3- to 6-membered carbocycle; 11 12 13 14 15 R , R , R , R and R are independently selected from the group consisting of: H, halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, CN and a 5- to 1-4 1-4 1-4 1-4 6-membered heterocycle comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S; R is independently selected from the group consisting of: H and C alkyl; R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkoxy, C haloalkyl, C haloalkoxy, N(C alkyl) , 1-6 1-6 1-6 1-4 2 -(CH ) -(X) -(CH ) R , and -(CH ) -(CH O) -(CH ) R ; 2 n t 2 m 2 n 2 m 2 n R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, 1-6 1-6 1-6 1-6 -(CH ) -(X) -(CH ) R , and -(CH ) -(CH O) -(CH ) R ; 2 n t 2 m 2 n 2 m 2 n R is independently selected from the group consisting of: C cycloalkyl substituted with 0-2 R , phenyl substituted with 0-3 R , and a 5- to 6-membered 11841-PCT heterocycle comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S; wherein said heterocycle is substituted with 0-2 R ; R is, at each occurrence, independently selected from the group consisting of: halo, OH, CN, C alkyl, C alkoxy, C haloalkyl, and C haloalkoxy; 1-4 1-4 1-4 1-4 R is independently selected from the group consisting of: H, C alkyl, benzyl, CO(C alkyl) and COBn; R is independently selected from the group consisting of: H and C alkyl; n, at each occurrence, is independently 0 or 1; m, at each occurrence, is independently 0, 1, 2, 3 or 4; and t, at each occurrence, is independently 0 or 1; provided that the following compounds are excluded: CN CN Ph Ph N O N O Me Me Me H H H , , , MeO Br CN CN N O N O Me Me
[0026] Also described herein is a compound of Formula (I), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, as described in paragraph [0025], wherein: R is independently selected from the group consisting of: C alkyl substituted with 0-2 R , -(CH ) -(phenyl substituted with 0-3 R ), and -(CH ) -(C cycloalkyl 2 m 2 m 3-6 substituted with 0-2 R ). 11841-PCT Also described herein is a compound of Formula (I), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, as described in paragraph [0025] or [0026], wherein: 11 15 R and R are independently selected from the group consisting of: H and halo; 12 14 R and R are independently selected from the group consisting of: H, C alkyl and C alkoxy; and 1-4 1-4 R is independently selected from the group consisting of: H, halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, CN and morpholinyl. 1-4 1-4 1-4
[0028] Also described herein is a compound of Formula (II): (II) or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof; as described in any of paragraphs [0025] to [0027].
[0029] Also described herein is a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, as described in any of paragraphs [0025 to 0028], wherein: R is independently selected from the group consisting of: C alkyl, C cycloalkyl, -(CH ) Ph and ; 3-6 2 m R is independently selected from the group consisting of: C alkyl and C haloalkyl; R is H; R is H; R is independently selected from the group consisting of: CN, NO , 11841-PCT -CONH(C alkyl), -CONHPh, -CONH-(3-halo-Ph), -CONH-(4-halo-Ph), -CONH-(4-C alkyl -Ph), -CONH-(3-C alkoxy-Ph), 1-4 1-4 -CONH-(4-C alkoxy-Ph), -CONH-(4-C haloalkyl-Ph), 1-4 1-4 -CONH-(4-C haloalkoxy-Ph), -CONH-(3-haloC alkyl-Ph), 1-4 1-4 -CONH-(3-haloC alkoxy-Ph), -CONH(CH ) Ph, and 2H-tetrazolyl; 1-4 2 2 11 15 R and R are independently selected from the group consisting of: H and halo; 12 14 R and R are independently selected from the group consisting of: H, C alkyl and C alkoxy; 1-4 1-4 R is independently selected from the group consisting of: H, halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, CN and morpholinyl; 1-4 1-4 1-4 R is independently selected from the group consisting of: halo, OH, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) O(C alkyl), 1-6 1-6 1-6 2 m 1-4 N(C alkyl) , -O(CH ) (C cycloalkyl), 4-C alkoxy-Ph, -O(CH ) Ph, 1-4 2 2 m 3-6 1-4 2 m pyridinyl, 2-C alkoxy-pyridinyl, pyrimidinyl, pyrazinyl and -O-pyrimidinyl; and m, at each occurrence, is independently 0, 1, 2 or 3.
Also described herein is a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, as described in any of paragraphs [0025] to [0029], wherein: R is independently selected from the group consisting of: C alkyl and R is independently selected from the group consisting of: CN, -CONH(C alkyl), -CONHPh, -CONH-(3-halo-Ph), -CONH-(4-halo-Ph), -CONH-(4-C alkyl -Ph), -CONH-(3-C alkoxy-Ph), 1-4 1-4 -CONH-(4-C alkoxy-Ph), -CONH-(4-C haloalkyl-Ph), 1-4 1-4 -CONH-(4-C haloalkoxy-Ph), -CONH-(3-haloC alkoxy-Ph), -CONH(CH ) Ph, 1-4 1-4 2 2 and 2H-tetrazolyl; 11841-PCT R is independently selected from the group consisting of: H, C alkyl and C alkoxy; R is independently selected from the group consisting of: H and C alkoxy; R is independently selected from the group consisting of: H, halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy and CN; 1-4 1-4 1-4 R is independently selected from the group consisting of: C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) O(C alkyl), 1-6 1-6 1-6 2 m 1-4 -O(CH ) (C cycloalkyl), phenoxy, benzoxy, 2-C alkoxy-pyridinyl, 2 m 3-6 1-4 pyrimidinyl, pyrazinyl and -O-pyrimidinyl; and m, at each occurrence, is independently 0, 1, 2 or 3; provided that the following compounds are excluded: Me OMe Me MeO CN CN N O N O Me H Me H In a tenth aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the ninth aspect, wherein: R is ; R is independently selected from the group consisting of: -CONHPh, -CONH-(4-halo-Ph), -CONH-(4-C alkyl -Ph), -CONH-(3-C alkoxy-Ph), 1-4 1-4 -CONH-(4-C alkoxy-Ph), -CONH-(4-C haloalkyl-Ph), 1-4 1-4 -CONH-(4-C haloalkoxy-Ph), -CONH-(3-haloC alkoxy-Ph), -CONH(CH ) Ph, 1-4 1-4 2 2 and 2H-tetrazolyl; and 11841-PCT R is independently selected from the group consisting of: C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) (C cycloalkyl), phenoxy, 1-6 1-6 1-6 2 m 3-6 benzoxy, pyrimidinyl, pyrazinyl and -O-pyrimidinyl.
[0032] In an eleventh aspect, the present invention includes a compound of Formula (I) or (II), or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, within the scope of the ninth aspect, wherein: R is independently selected from the group consisting of: CF and Me; R is independently selected from the group consisting of: -CONHPh, -CONH-(4-F-Ph), -CONH-(4-Cl-Ph), -CONH-(4-Me-Ph), -CONH-(3-OMe-Ph), -CONH-(4-OMe-Ph), -CONH-(4-CF -Ph), -CONH-(4-OCF -Ph), -CONH-(3-FOMe-Ph), -CONH(CH ) Ph, and 2H-tetrazolyl; 11 15 R and R are independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: H, Me and OMe; R is independently selected from the group consisting of: H, F, Cl, Me, OMe, OEt, CF , OCHF , OCF and CN; 3 2 3 R is H; and R is independently selected from the group consisting of: n-pentyl, methoxy, n-butoxy, i-butoxy, -O(CH ) CF , -O(CH ) (cyclopentyl), phenoxy, benzoxy, 2 1-3 3 2 2 pyrimidinyl, pyrazinyl and -O-pyrimidinyl.
In a further aspect the present invention provides a compound selected from the exemplified examples or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
[0034] In another aspect, the present invention provides a compound selected from any subset list of compounds or a single compound from the exemplified examples within the scope of any of the above aspects.
In another aspect, the present disclosure provides a compound selected from: (S)(1H-tetrazolyl)(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one, 11841-PCT (S)-N-(4-methoxyphenyl)oxo(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl)- 6-(trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide, (S)(2H-tetrazolyl)(p-tolyl)(4-((6,6,6-trifluorohexyl)oxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one, (S)oxo(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl)-N-(4- (trifluoromethoxy)phenyl)(trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide, (S)-N-(6-methoxypyridinyl)oxo(p-tolyl)(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide, (S)-N-cyclopropyloxo(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide, (S)-N-(4-hydroxyphenyl)oxo(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide, (S)(4-(difluoromethoxy)phenyl)(1H-tetrazolyl)(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-5,6-dihydropyridin-2(1H)-one, (S)methyl-N-(2-oxo(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-1,2,5,6-tetrahydropyridinyl)isoxazolecarboxamide, (S)methyl-N-(2-oxo(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-1,2,5,6-tetrahydropyridinyl)-1,3,4-oxadiazolecarboxamide, N -heptyl-N -(4-methoxyphenyl)methyloxo(p-tolyl)-1,2,3,6- tetrahydropyridine-2,5-dicarboxamide, (S)(1H-tetrazolyl)(p-tolyl)(4-((6,6,6-trifluorohexyl)oxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one, (S)(5,6,7,8-tetrahydronaphthalenyl)(1H-tetrazolyl)(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-5,6-dihydropyridin-2(1H)-one, (S)(methylsulfonyl)-N-(2-oxo(p-tolyl)(4-(4,4,4-trifluorobutoxy)phenyl)- 6-(trifluoromethyl)-1,2,5,6-tetrahydropyridinyl)acetamide, (S)(1H-tetrazolyl)(4-(4,4,4-trifluorobutoxy)phenyl)(4-(2,2,2- trifluoroethyl)phenyl)(trifluoromethyl)-5,6-dihydropyridin-2(1H)-one, and (S)-N-(5-methoxypyrazinyl)oxo(p-tolyl)(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide; or a pharmaceutically acceptable salt thereof. 11841-PCT In another embodiment, R is independently -CONH(C alkyl), 4-18 -CONHC haloalkyl, or -CONH(CH ) Ph. 2-8 2 1-8 In another embodiment, R is -(CH ) -(C carbocycle substituted with 0-2 2 m 3-10 R and 0-2 R ), or -(CH ) -(5- to 6-membered heteroaryl comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O and S; wherein said heteroaryl is substituted with 0-1 R and 0-2 R ).
In another embodiment, R is -(CH ) -(C carbocycle substituted with 0-2 2 m 3-10 R and 0-2 R ).
In another embodiment, R is -(CH ) -(phenyl substituted with 0-2 R and 0-2 R ).
In another embodiment, R is -(CH ) -(5- to 6-membered heteroaryl comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O and S; wherein said heteroaryl is substituted with 0-1 R and 0-2 R ).
In another embodiment, R is a C hydrocarbon chain substituted with 0-3 1-12 R ; wherein said hydrocarbon chain may be straight or branched, saturated or unsaturated.
In another embodiment, R is independently: C alkyl, C cycloalkyl, 1-6 3-6 -CONHC alkyl, -CONHC haloalkyl, -CONH(CH ) Ph, -(CH ) -(phenyl 4-18 2-8 2 1-8 2 m substituted with 1 R and 0-2 R ), or a 5- to 6-membered heteroaryl substituted with 0-1 R and 0-2 R , wherein said heteroaryl is selected from: pyridyl, oxazolyl, thiazolyl and In another embodiment, R is independently: C alkyl, -CONHC alkyl, 1-6 4-18 -CONH(CH ) Ph, or . 2 1-8 In another embodiment, R is . 11841-PCT In another embodiment, R is independently .
In another embodiment, R is independently C alkyl or C haloalkyl. 1-4 1-4 In another embodiment, R is C alkyl.
[0048] In another embodiment, R is C haloalkyl.
In another embodiment, R is independently CF or Me.
In another embodiment, R is CF .
In another embodiment, R is Me.
[0052] In another embodiment, R is independently H or F.
In another embodiment, R is H.
In another embodiment, R is F.
In another embodiment, R is independently H or F.
[0056] In another embodiment, R is H.
In another embodiment, R is F.
In another embodiment, R is independently H or F.
In another embodiment, R is H.
[0060] In another embodiment, R is F.
In another embodiment, R is independently C alkyl, R , or -(CH ) -(X) -(CH ) R . 2 n t 2 m In another embodiment, R is independently -CONH(C alkyl), j j j j j -NHCOX SO R , -NHCOCOR , -NHCOCH(OH)R , -NHCOCH COR , -NHCONHR , or 1 2 2 -OCONR R .
In another embodiment, R is independently NH , -CONH(C alkyl), 2 1-6 11841-PCT R , -(CH ) -(X) -(CH ) R , -NHCO(CH )SO (C alkyl), -NHCOCO(C alkyl), 2 n t 2 m 2 2 1-4 1-4 -NHCOCH(OH)(C alkyl), -NHCOCH CO(C alkyl), -NHCONH(C alkyl), or 1-4 2 1-4 1-4 -OCONH(C alkyl).
In another embodiment, R is independently NH , -CONH(C alkyl), 2 1-6 c c c c -NHCO(CH )SO (C alkyl), R , OR , -CONHR , or -NHCOR . 2 2 1-4 In another embodiment, R is independently NH , -CONH(C alkyl), 2 1-6 c c c c -NHCO(CH )SO (C alkyl), R , OR , -CONHR , or -NHCOR . 2 2 1-4 6 c c c In another embodiment, R is independently R , -CONHR , -NHCOR , or - NHCOCH SO (C alkyl). 2 2 1-4
[0067] In another embodiment, R is independently 5-membered nitrogen heteroaryl.
In another embodiment, R is independently: 1H-imidazolyl, 1H-tetrazolyl, 1H-tetrazolyl, or 2H-tetrazolyl.
In another embodiment, R is independently H, C alkyl or halo.
[0070] In another embodiment, R is independently H, Me, F, or Cl.
In another embodiment, R is H.
In another embodiment, R is C alkyl.
In another embodiment, R is Me.
In another embodiment, R is halo.
[0075] In another embodiment, R is independently F or Cl.
In another embodiment, R is independently H, halo, C alkyl or C alkoxy.
In another embodiment, R is independently H, F, Cl, Me and OMe.
[0078] In another embodiment, R is H.
In another embodiment, R is C alkyl.
In another embodiment, R is Me.
In another embodiment, R is C alkoxy.
In another embodiment, R is OMe. 11841-PCT In another embodiment, R is halo.
In another embodiment, R is independently F or Cl.
In another embodiment, R is independently: H, halo, C alkyl substituted with 0-1 R , C alkoxy, C haloalkyl, C haloalkoxy, C cycloalkyl, CN, NR R , 1-4 1-4 1-4 3-4 SR , NHCO (C alkyl), NHSO (C alkyl), or a 4- to 6-membered heterocycle 2 1-4 2 1-4 comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S.
In another embodiment, R is independently: H, halo, C alkyl substituted with 0-1 C alkoxy, C alkoxy, C haloalkyl, C haloalkoxy, CN, C cycloalkyl, 1-4 1-4 1-4 1-4 3-4 N(C alkyl) , NHCO (C alkyl), NHSO (C alkyl), pyrazolyl, or morpholinyl. 1-4 2 2 1-4 2 1-4 In another embodiment, R is independently: H, halo, C alkyl substituted with 0-1 C alkoxy, C alkoxy, C haloalkyl, C haloalkoxy, CN or 1-4 1-4 1-4 1-4 C cycloalkyl.
In another embodiment, R is independently: NR R , NHCO (C alkyl), 2 1-4 NHSO (C alkyl), or a 4- to 6-membered heterocycle comprising: carbon atoms and 1-4 2 1-4 heteroatoms selected from N, NR , O, and S.
In another embodiment, R is independently H, halo, C alkyl or C alkoxy.
[0090] In another embodiment, R is independently H, F, Cl, Me and OMe.
In another embodiment, R is H.
In another embodiment, R is C alkyl.
In another embodiment, R is Me.
In another embodiment, R is C alkoxy.
[0095] In another embodiment, R is OMe.
In another embodiment, R is halo.
In another embodiment, R is independently F or Cl.
In another embodiment, R is independently H, C alkyl or halo. 11841-PCT In another embodiment, R is independently H, Me, F, or Cl.
In another embodiment, R is H.
In another embodiment, R is C alkyl.
In another embodiment, R is Me.
[00103] In another embodiment, R is halo.
In another embodiment, R is independently F or Cl.
In another embodiment, R is H.
In another embodiment, R is C alkyl.
In another embodiment, X is independently O, S, or NH.
In another embodiment, X is independently O or S.
In another embodiment, X is O.
In another embodiment, X is independently CONH or NHCO.
[00111] In another embodiment, X is CONH.
In another embodiment, X is NHCO.
In another embodiment, R is, at each occurrence, independently: C alkyl, 1-10 C alkoxy, C haloalkyl, C haloalkoxy, C alkylthio, C haloalkyltho, 1-10 1-10 1-10 1-10 1-10 -CONH(CH ) H, -O(CH ) O(C alkyl), R , -(CH ) -(X) -(CH ) R , or 2 4-20 2 s 1-6 2 n t 2 m -(CH ) -(CH O) -(CH ) R . 2 n 2 m 2 n In another embodiment, R is, at each occurrence, independently: halo, OH, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) O(C alkyl), 1-8 1-8 1-8 1-10 2 s 1-6 N(C alkyl) , -CONH(CH ) H, -(CH ) (C cycloalkyl), 1-4 2 2 6-20 2 m 3-6 -(CH ) (C cycloalkenyl), -O(CH ) (C cycloalkyl), 4-C alkoxy-Ph, 2 m 4-6 2 m 3-6 1-4 -O(CH ) Ph, morpholinyl, pyridyl, 2-C alkoxy-pyridinyl, pyrimidinyl, pyrazinyl, 2 m 1-4 or -O-pyrimidinyl.
In another embodiment, R is, at each occurrence, independently: halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) O(C alkyl), 1-6 1-6 1-6 1-10 2 s 1-6 -CONH(CH ) H, -(CH ) (C cycloalkyl), -(CH ) (C cycloalkenyl), 2 6-20 2 m 3-6 2 m 4-6 11841-PCT -O(CH ) (C cycloalkyl), phenoxy, benzoxy, morpholinyl, 2-C alkoxy-pyridinyl, 2 m 3-6 1-4 pyrimidinyl, pyrazinyl or -O-pyrimidinyl.
In another embodiment, R is, at each occurrence, independently: halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -CONH(CH ) H, 1-6 1-6 1-6 1-8 2 6-20 C cycloalkyl, C cycloalkenyl, -O(CH ) (C cycloalkyl), phenoxy, benzoxy, 3-6 4-6 2 m 3-6 pyrimidinyl, pyrazinyl and -O-pyrimidinyl.
In another embodiment, R is, at each occurrence, independently: -O(CH ) CF and -O(CH ) CF CF . 2 1-6 3 2 1-4 2 3
[00118] In another embodiment, R is, at each occurrence, independently: C cycloalkyl substituted with 0-2 R , C cycloalkenyl substituted with 0-2 R , or 3-6 3-6 -(CH ) -(phenyl substituted with 0-3 R ).
In another embodiment, R is, at each occurrence, independently: C cycloalkyl substituted with 0-2 R or C cycloalkenyl substituted with 0-2 R . 3-6 3-6
[00120] In another embodiment, R is, at each occurrence, independently -(CH ) -(phenyl substituted with 0-3 R ),.
In another embodiment, R is, at each occurrence, independently C cycloalkyl substituted with 0-2 R .
In another embodiment, R is, at each occurrence, independently C cycloalkenyl substituted with 0-2 R .
In another embodiment, R is, at each occurrence, independently a 5- to 6-membered heterocycle comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S; wherein said heterocycle is substituted with 0-2 R .
In another embodiment, R is, at each occurrence, independently -(CH ) -(phenyl substituted with 0-3 R ), or a heteroaryl selected from: oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, and pyrazinyl; wherein said heteroaryl is substituted with 0-2 R .
In another embodiment, R is, at each occurrence, independently a heteroaryl selected from: oxazolyl, isoxazolyl, pyrazolyl, imidazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, and pyrazinyl; wherein said heteroaryl is substituted with 0-2 R . 11841-PCT In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 10 µM, using the MGAT2 SPA assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 5 µM, using the MGAT2 SPA assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 1 µM, using the MGAT2 SPA assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 0.5 µM, using the MGAT2 SPA assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 10 µM, using the MGAT2 LCMS assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 5 µM, using the MGAT2 LCMS assay.
[00132] In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 2.5 µM, using the MGAT2 LCMS assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 1 µM, using the MGAT2 LCMS assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 0.5 µM, using the MGAT2 LCMS assay.
In another embodiment, the compounds of the present invention have hMGAT2 IC values ≤ 0.1 µM, using the MGAT2 LCMS assay.
II. OTHER EMBODIMENTS OF THE INVENTION In another embodiment, the present invention provides a composition comprising at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
In another embodiment, the present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and at least one of the 11841-PCT compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
In another embodiment, the present invention provides a pharmaceutical composition, comprising: a pharmaceutically acceptable carrier and a therapeutically effective amount of at least one of the compounds of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
In another embodiment, the present invention provides a process for making a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
[00140] In another embodiment, the present invention provides an intermediate for making a compound of the present invention or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
In another embodiment, the present invention provides a pharmaceutical composition further comprising additional therapeutic agent(s). In a preferred embodiment, the present invention provides pharmaceutical composition, wherein the additional therapeutic agent is, for example, a dipeptidyl peptidase-IV (DPP4) inhibitor (for example a member selected from saxagliptin, sitagliptin, vildagliptin and alogliptin).
Also described herein is a method for the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.
Examples of diseases or disorders associated with the activity of the MGAT2 that can be prevented, modulated, or treated according to the present invention include, but are not limited to, diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, nonalcoholic fatty liver disease (NAFLD) including nonalcoholic steatohepatitis (NASH), retinopathy, neuropathy, nephropathy, delayed wound healing, atherosclerosis and its sequelae, abnormal heart function, myocardial ischemia, stroke, Metabolic Syndrome, hypertension, obesity, dyslipidemia, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low high- 11841-PCT density lipoprotein (HDL), high low-density lipoprotein (LDL), non-cardiac ischemia, lipid disorders, and glaucoma.
Also described herein is a method for the treatment and/or prophylaxis of diabetes, hyperglycemia, gestational diabetes, obesity, dyslipidemia, and hypertension, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.
Also described herein is a method for the treatment and/or prophylaxis of diabetes, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.
Also described herein is a method for the treatment and/or prophylaxis of hyperglycemia, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.
Also described herein is a method for the treatment and/or prophylaxis of obesity, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.
Also described herein is a method for the treatment and/or prophylaxis of dyslipidemia, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.
Also described herein is a method for the treatment and/or prophylaxis of hypertension, comprising administering to a patient in need of such treatment and/or prophylaxis a therapeutically effective amount of at least one of the compounds of the 11841-PCT present invention, alone, or, optionally, in combination with another compound of the present invention and/or at least one other type of therapeutic agent.
In another embodiment, the present invention provides a compound of the present invention for use in therapy.
[00151] In another embodiment, the present invention provides a compound of the present invention for use in therapy for the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2.
In another embodiment, the present invention also provides the use of a compound of the present invention for the manufacture of a medicament for the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2, wherein the diseases and disorders are selected from diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, nonalcoholic fatty liver disease (NAFLD), retinopathy, neuropathy, nephropathy, delayed wound healing, atherosclerosis and its sequelae, abnormal heart function, myocardial ischemia, stroke, Metabolic Syndrome, hypertension, obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low high-density lipoprotein (HDL), high low-density lipoprotein (LDL), non-cardiac ischemia, lipid disorders, or glaucoma.
Also described herein is a method for the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2, comprising: administering to a patient in need thereof a therapeutically effective amount of a first and second therapeutic agent, wherein the first therapeutic agent is a compound of the present invention.
Preferably, the second therapeutic agent, for example, dipeptidyl peptidase-IV (DPP4) inhibitor (for example a member selected from saxagliptin, sitagliptin, vildagliptin, linagliptin and alogliptin).
[00154] In another embodiment, the present invention provides a combined preparation of a compound of the present invention and additional therapeutic agent(s) for simultaneous, separate or sequential use in therapy.
In another embodiment, the present invention provides a combined preparation of a compound of the present invention and additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2. 11841-PCT Where desired, the compound of the present invention may be used in combination with one or more other types of antidiabetic agents and/or one or more other types of therapeutic agents which may be administered orally in the same dosage form, in a separate oral dosage form or by injection. The other type of antidiabetic agent that may be optionally employed in combination with the MGAT2 inhibitor of the present invention may be one, two, three or more antidiabetic agents or antihyperglycemic agents which may be administered orally in the same dosage form, in a separate oral dosage form, or by injection to produce an additional pharmacological benefit.
The antidiabetic agents used in the combination with the MGAT2 inhibitor of the present invention include, but are not limited to, insulin secretagogues or insulin sensitizers, other MGAT2 inhibitors, or other antidiabetic agents. These agents include, but are not limited to, dipeptidyl peptidase IV (DP4) inhibitors (for example, sitagliptin, saxagliptin, alogliptin, linagliptin and vildagliptin), biguanides (for example, metformin and phenformin), sulfonyl ureas (for example, glyburide, glimepiride and glipizide), glucosidase inhibitors (for example, acarbose, miglitol), PPARγ agonists such as thiazolidinediones (for example, rosiglitazone and pioglitazone), PPAR α/γ dual agonists (for example, muraglitazar, tesaglitazar and aleglitazar), glucokinase activators, GPR40 receptor modulators (e.g. TAK-875), GPR119 receptor modulators (for example, MBX-2952, PSN821, and APD597), sodium-glucose transporter-2 (SGLT2) inhibitors (for example, dapagliflozin, canagliflozin and remagliflozin), 11b-HSD-1 inhibitors (for example MK-0736, BI35585, BMS-823778, and LY2523199), amylin analogs such as pramlintide, and/or insulin.
The MGAT2 inhibitor of the present invention may also be optionally employed in combination with one or more hypophagic and/or weight-loss agents such as diethylpropion, phendimetrazine, phentermine, orlistat, sibutramine, lorcaserin, pramlintide, topiramate, MCHR1 receptor antagonists, oxyntomodulin, naltrexone, Amylin peptide, NPY Y5 receptor modulators, NPY Y2 receptor modulators, NPY Y4 receptor modulators, cetilistat, 5HT2c receptor modulators, and the like. The compounds of the present invention may also be employed in combination with an agonist of the glucagon-like peptide-1 receptor (GLP-1 R), such as exenatide, liraglutide, GPR-1(1-36) amide, GLP-1(7-36) amide, GLP-1(7-37), which may be administered via injection, intranasal, or by transdermal or buccal devices. 11841-PCT The MGAT2 inhibitor of the present invention may also be optionally employed in combination with one or more other types of therapeutic agents, such as DGAT inhibitors, LDL lowering drugs such as statins (inhibitors of HMG CoA reductase) or inhibitors of cholesterol absorption, modulators of PCSK9, drugs that increase HDL such as CETP inhibitors.
The present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof. This invention encompasses all combinations of preferred aspects of the invention noted herein. It is understood that any and all embodiments of the present invention may be taken in conjunction with any other embodiment or embodiments to describe additional embodiments. It is also understood that each individual element of the embodiments is its own independent embodiment.
Furthermore, any element of an embodiment is meant to be combined with any and all other elements from any embodiment to describe an additional embodiment.
III. CHEMISTRY Throughout the specification and the appended claims, a given chemical formula or name shall encompass all stereo and optical isomers and racemates thereof where such isomers exist. Unless otherwise indicated, all chiral (enantiomeric and diastereomeric) and racemic forms are within the scope of the invention. Many geometric isomers of C=C double bonds, C=N double bonds, ring systems, and the like can also be present in the compounds, and all such stable isomers are contemplated in the present invention. Cis- and trans- (or E- and Z-) geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms. The present compounds can be isolated in optically active or racemic forms. Optically active forms may be prepared by resolution of racemic forms or by synthesis from optically active starting materials. All processes used to prepare compounds of the present invention and intermediates made therein are considered to be part of the present invention. When enantiomeric or diastereomeric products are prepared, they may be separated by conventional methods, for example, by chromatography or fractional crystallization. Depending on the process conditions the end products of the present invention are obtained either in free (neutral) or salt form.
Both the free form and the salts of these end products are within the scope of the 11841-PCT invention. If so desired, one form of a compound may be converted into another form. A free base or acid may be converted into a salt; a salt may be converted into the free compound or another salt; a mixture of isomeric compounds of the present invention may be separated into the individual isomers. Compounds of the present invention, free form and salts thereof, may exist in multiple tautomeric forms, in which hydrogen atoms are transposed to other parts of the molecules and the chemical bonds between the atoms of the molecules are consequently rearranged. It should be understood that all tautomeric forms, insofar as they may exist, are included within the invention.
As used herein, the term “alkyl” or “alkylene” is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms. For examples, “C to C alkyl” or “C alkyl” (or alkylene), 1 12 1-12 is intended to include C , C , C , C , C , C , C , C , C , C , C and C alkyl groups; 1 2 3 4 5 6 7 8 9 10 11 12 “C to C alkyl” or “C alkyl” (or alkylene), is intended to include C , C , C , C , C , 4 18 4-18 4 5 6 7 8 C , C , C , C , C , C , C , C , C , and C alkyl groups. Additionally, for 9 10 11 12 13 14 15 16 17 18 example, “C to C alkyl” or “C alkyl” denotes alkyl having 1 to 6 carbon atoms. 1 6 1-6 Alkyl group can be unsubstituted or substituted with at least one hydrogen being replaced by another chemical group. Example alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, t- butyl), and pentyl (e.g., n-pentyl, isopentyl, neopentyl). When “C alkyl” or “C alkylene” is used, it is intended to denote a direct bond.
Alkenyl” or “alkenylene” is intended to include hydrocarbon chains of either straight or branched configuration having the specified number of carbon atoms and one or more, preferably one to two, carbon-carbon double bonds that may occur in any stable point along the chain. For example, “C to C alkenyl” or “C alkenyl” (or alkenylene), 2 6 2-6 is intended to include C , C , C , C , and C alkenyl groups. Examples of alkenyl 2 3 4 5 6 include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 2-butenyl, 3-butenyl, 2- pentenyl, 3, pentenyl, 4-pentenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 2-methyl- 2-propenyl, and 4-methylpentenyl.
“Alkynyl” or “alkynylene” is intended to include hydrocarbon chains of either straight or branched configuration having one or more, preferably one to three, carbon-carbon triple bonds that may occur in any stable point along the chain. For example, “C to C alkynyl” or “C alkynyl” (or alkynylene), is intended to include C , 2 6 2-6 2 11841-PCT C , C , C , and C alkynyl groups; such as ethynyl, propynyl, butynyl, pentynyl, and 3 4 5 6 hexynyl.
When the term “hydrocarbon chain” is used, it is intended to include “alkyl”, “alkenyl” and “alkynyl”, unless otherwise specified.
[00166] The term "alkoxy" or "alkyloxy" refers to an -O-alkyl group. For example, "C to C alkoxy" or "C alkoxy" (or alkyloxy), is intended to include C , C , C , C , 1 6 1-6 1 2 3 4 C , and C alkoxy groups. Example alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), and t-butoxy. Similarly, "alkylthio" or "thioalkoxy" represents an alkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example methyl-S- and ethyl-S-.
"Halo" or "halogen" includes fluoro, chloro, bromo, and iodo. "Haloalkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more halogens. Examples of haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, trichloromethyl, pentafluoroethyl, pentachloroethyl, 2,2,2-trifluoroethyl, heptafluoropropyl, and heptachloropropyl. Examples of haloalkyl also include "fluoroalkyl" that is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms, substituted with 1 or more fluorine atoms.
"Haloalkoxy" or "haloalkyloxy" represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through an oxygen bridge. For example, "C haloalkoxy", is intended to include C , C , C , C , C , and C haloalkoxy 1-6 1 2 3 4 5 6 groups. Examples of haloalkoxy include, but are not limited to, trifluoromethoxy, 2,2,2-trifluoroethoxy, and pentafluorothoxy. Similarly, "haloalkylthio" or "thiohaloalkoxy" represents a haloalkyl group as defined above with the indicated number of carbon atoms attached through a sulphur bridge; for example trifluoromethyl-S-, and pentafluoroethyl-S-.
The term "cycloalkyl" refers to cyclized alkyl groups, including mono-, bi- or poly-cyclic ring systems. For example, "C to C cycloalkyl" or "C cycloalkyl" is 3 6 3-6 intended to include C , C , C , and C cycloalkyl groups. Example cycloalkyl groups 3 4 5 6 include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and 11841-PCT norbornyl. Branched cycloalkyl groups such as 1-methylcyclopropyl and 2-methylcyclopropyl are included in the definition of "cycloalkyl". The term "cycloalkenyl" refers to cyclized alkenyl groups. C cycloalkenyl is intended to include C , C , and C cycloalkenyl groups. Example cycloalkenyl groups include, but are not 4 5 6 limited to, cyclobutenyl, cyclopentenyl, and cyclohexenyl.
As used herein, “carbocycle,” “carbocyclyl,” or “carbocyclic residue” is intended to mean any stable 3-, 4-, 5-, 6-, 7-, or 8-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, 12-, or 13-membered bicyclic or tricyclic ring, any of which may be saturated, partially unsaturated, unsaturated or aromatic. Examples of such carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, cyclooctadienyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane (decalin), [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, anthracenyl, and tetrahydronaphthyl (tetralin). As shown above, bridged rings are also included in the definition of carbocycle (e.g., [2.2.2]bicyclooctane). Preferred carbocycles, unless otherwise specified, are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl, indanyl, and tetrahydronaphthyl. When the term “carbocycle” is used, it is intended to include “aryl.” A bridged ring occurs when one or more, preferably one to three, carbon atoms link two non-adjacent carbon atoms. Preferred bridges are one or two carbon atoms. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.
As used herein, the term “bicyclic carbocycle” or “bicyclic carbocyclic group” is intended to mean a stable 9- or 10-membered carbocyclic ring system that contains two fused rings and consists of carbon atoms. Of the two fused rings, one ring is a benzo ring fused to a second ring; and the second ring is a 5- or 6-membered carbon ring which is saturated, partially unsaturated, or unsaturated. The bicyclic carbocyclic group may be attached to its pendant group at any carbon atom which results in a stable structure. The bicyclic carbocyclic group described herein may be substituted on any carbon if the resulting compound is stable. Examples of a bicyclic carbocyclic group are, but not limited to, naphthyl, 1,2-dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, and indanyl. 11841-PCT “Aryl” groups refer to monocyclic or bicyclic aromatic hydrocarbons, including, for example, phenyl, and naphthyl. Aryl moieties are well known and described, for example, in Hawley’s Condensed Chemical Dictionary (15 ed.), R.J.
Lewis, ed., J. Wiley & Sons, Inc., New York, 2007. “C aryl” refers to phenyl and 6-10 naphthyl.
The term "benzyl," as used herein, refers to a methyl group on which one of the hydrogen atoms is replaced by a phenyl group.
As used herein, the term "heterocycle," "heterocyclyl," or "heterocyclic group" is intended to mean a stable 3-, 4-, 5-, 6-, or 7-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, 12-, 13-, or 14-membered polycyclic heterocyclic ring that is saturated, partially unsaturated, or fully unsaturated, and that contains carbon atoms and 1, 2, 3 or 4 heteroatoms independently selected from the group consisting of N, O and S; and including any polycyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., N→O and S(O) , wherein p is 0, 1 or 2). The nitrogen atom may be substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined). The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure. The heterocyclic rings described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. A nitrogen in the heterocycle may optionally be quaternized. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1. When the term "heterocycle" is used, it is intended to include heteroaryl.
[00175] Examples of heterocycles include, but are not limited to, acridinyl, azetidinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, imidazolopyridinyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, 11841-PCT isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isothiazolopyridinyl, isoxazolyl, isoxazolopyridinyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolopyridinyl, oxazolidinylperimidinyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolopyridinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2-pyrrolidonyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrazolyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-1,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thiazolopyridinyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, and xanthenyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles.
Examples of 5- to 10-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, morpholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, triazolyl, benzimidazolyl, 1H-indazolyl, benzofuranyl, benzothiofuranyl, benztetrazolyl, benzotriazolyl, benzisoxazolyl, benzoxazolyl, oxindolyl, benzoxazolinyl, benzthiazolyl, benzisothiazolyl, isatinoyl, isoquinolinyl, octahydroisoquinolinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, isoxazolopyridinyl, quinazolinyl, quinolinyl, isothiazolopyridinyl, thiazolopyridinyl, oxazolopyridinyl, imidazolopyridinyl, and pyrazolopyridinyl.
Examples of 5- to 6-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, morpholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles. 11841-PCT As used herein, the term "bicyclic heterocycle" or "bicyclic heterocyclic group" is intended to mean a stable 9- or 10-membered heterocyclic ring system which contains two fused rings and consists of carbon atoms and 1, 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O and S. Of the two fused rings, one ring is a 5- or 6-membered monocyclic aromatic ring comprising a 5-membered heteroaryl ring, a 6-membered heteroaryl ring or a benzo ring, each fused to a second ring. The second ring is a 5- or 6-membered monocyclic ring which is saturated, partially unsaturated, or unsaturated, and comprises a 5-membered heterocycle, a 6-membered heterocycle or a carbocycle (provided the first ring is not benzo when the second ring is a carbocycle).
The bicyclic heterocyclic group may be attached to its pendant group at any heteroatom or carbon atom which results in a stable structure. The bicyclic heterocyclic group described herein may be substituted on carbon or on a nitrogen atom if the resulting compound is stable. It is preferred that when the total number of S and O atoms in the heterocycle exceeds 1, then these heteroatoms are not adjacent to one another. It is preferred that the total number of S and O atoms in the heterocycle is not more than 1.
Examples of a bicyclic heterocyclic group are, but not limited to, quinolinyl, isoquinolinyl, phthalazinyl, quinazolinyl, indolyl, isoindolyl, indolinyl, 1H-indazolyl, benzimidazolyl, 1,2,3,4-tetrahydroquinolinyl, 1,2,3,4-tetrahydroisoquinolinyl, 5,6,7,8-tetrahydro-quinolinyl, 2,3-dihydro-benzofuranyl, chromanyl, 1,2,3,4-tetrahydro-quinoxalinyl, and 1,2,3,4-tetrahydro-quinazolinyl.
As used herein, the term "aromatic heterocyclic group" or "heteroaryl" is intended to mean stable monocyclic and polycyclic aromatic hydrocarbons that include at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrroyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyl, tetrazolyl, indazolyl, 1,2,4-thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, indolinyl, benzodioxolanyl, and benzodioxane. Heteroaryl groups are substituted or unsubstituted.
The nitrogen atom is substituted or unsubstituted (i.e., N or NR wherein R is H or another substituent, if defined). The nitrogen and sulfur heteroatoms may optionally be oxidized (i.e., N→O and S(O) , wherein p is 0, 1 or 2). 11841-PCT Examples of 5- to 6-membered heteroaryls include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, imidazolyl, imidazolidinyl, tetrazolyl, isoxazolyl, oxazolyl, oxadiazolyl, oxazolidinyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyl.
[00183] Bridged rings are also included in the definition of heterocycle. A bridged ring occurs when one or more, preferably one to three, atoms (i.e., C, O, N, or S) link two non-adjacent carbon or nitrogen atoms. Examples of bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and a carbon-nitrogen group. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.
The term "counter ion" is used to represent a negatively charged species such as chloride, bromide, hydroxide, acetate, and sulfate or a positively charged species such as sodium (Na+), potassium (K+), ammonium (R NH + where n=0-4 and m=0-4) and the like.
When a dotted ring is used within a ring structure, this indicates that the ring structure may be saturated, partially saturated or unsaturated.
As used herein, the term "amine protecting group" means any group known in the art of organic synthesis for the protection of amine groups which is stable to an ester reducing agent, a disubstituted hydrazine, R4-M and R7-M, a nucleophile, a hydrazine reducing agent, an activator, a strong base, a hindered amine base and a cyclizing agent.
Such amine protecting groups fitting these criteria include those listed in Wuts, P. G. M. and Greene, T.W. Protecting Groups in Organic Synthesis, 4th Edition, Wiley (2007) and The Peptides: Analysis, Synthesis, Biology, Vol. 3, Academic Press, New York (1981), the disclosure of which is hereby incorporated by reference. Examples of amine protecting groups include, but are not limited to, the following: (1) acyl types such as formyl, trifluoroacetyl, phthalyl, and p-toluenesulfonyl; (2) aromatic carbamate types such as benzyloxycarbonyl (Cbz) and substituted benzyloxycarbonyls, 1-(p-biphenyl)methylethoxycarbonyl, and 9-fluorenylmethyloxycarbonyl (Fmoc); (3) aliphatic carbamate types such as tert-butyloxycarbonyl (Boc), ethoxycarbonyl, diisopropylmethoxycarbonyl, and allyloxycarbonyl; (4) cyclic alkyl carbamate types such as cyclopentyloxycarbonyl and adamantyloxycarbonyl; (5) alkyl types such as 11841-PCT triphenylmethyl and benzyl; (6) trialkylsilane such as trimethylsilane; (7) thiol containing types such as phenylthiocarbonyl and dithiasuccinoyl; and (8) alkyl types such as triphenylmethyl, methyl, and benzyl; and substituted alkyl types such as 2,2,2-trichloroethyl, 2-phenylethyl, and t-butyl; and trialkylsilane types such as trimethylsilane.
As referred to herein, the term "substituted" means that at least one hydrogen atom is replaced with a non-hydrogen group, provided that normal valencies are maintained and that the substitution results in a stable compound. Ring double bonds, as used herein, are double bonds that are formed between two adjacent ring atoms (e.g., C=C, C=N, or N=N).
In cases wherein there are nitrogen atoms (e.g., amines) on compounds of the present invention, these may be converted to N-oxides by treatment with an oxidizing agent (e.g., mCPBA and/or hydrogen peroxides) to afford other compounds of this invention. Thus, shown and claimed nitrogen atoms are considered to cover both the shown nitrogen and its N-oxide (N→O) derivative.
When any variable occurs more than one time in any constituent or formula for a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0-3 R, then said group may optionally be substituted with up to three R groups, and at each occurrence R is selected independently from the definition of R.
When a bond to a substituent is shown to cross a bond connecting two atoms in a ring, then such substituent may be bonded to any atom on the ring. When a substituent is listed without indicating the atom in which such substituent is bonded to the rest of the compound of a given formula, then such substituent may be bonded via any atom in such substituent.
Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, and/or other problem or complication, commensurate with a reasonable benefit/risk ratio. 11841-PCT As used herein, "pharmaceutically acceptable salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic groups such as amines; and alkali or organic salts of acidic groups such as carboxylic acids. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, and isethionic, and the like.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington: The Science and Practice of Pharmacy, 22 Edition, Allen, L. V. Jr., Ed.; Pharmaceutical Press, London, UK (2012), the disclosure of which is hereby incorporated by reference.
In addition, compounds of formula I may have prodrug forms. Any compound that will be converted in vivo to provide the bioactive agent (i.e., a compound of formula I) is a prodrug as described herein. Various forms of prodrugs are well known in the art. For examples of such prodrug derivatives, see: a) Bundgaard, H., ed., Design of Prodrugs, Elsevier (1985), and Widder, K. et al., eds., Methods in Enzymology, 112:309-396, Academic Press (1985); b) Bundgaard, H., Chapter 5, "Design and Application of Prodrugs," A Textbook of Drug Design and Development, pp. 113-191, Krosgaard-Larsen, P. et al., eds., Harwood Academic Publishers (1991); c) Bundgaard, H., Adv. Drug Deliv. Rev., 8:1-38 (1992); 11841-PCT d) Bundgaard, H. et al., J. Pharm. Sci., 77:285 (1988); e) Kakeya, N. et al., Chem. Pharm. Bull., 32:692 (1984); and f) Rautio, J (Editor). Prodrugs and Targeted Delivery (Methods and Principles in Medicinal Chemistry), Vol 47, Wiley-VCH, 2011.
Compounds containing a carboxy group can form physiologically hydrolyzable esters that serve as prodrugs by being hydrolyzed in the body to yield formula I compounds per se. Such prodrugs are preferably administered orally since hydrolysis in many instances occurs principally under the influence of the digestive enzymes. Parenteral administration may be used where the ester per se is active, or in those instances where hydrolysis occurs in the blood. Examples of physiologically hydrolyzable esters of compounds of formula I include C alkyl, C alkylbenzyl, 1-6 1-6 4-methoxybenzyl, indanyl, phthalyl, methoxymethyl, C alkanoyloxy-C alkyl (e.g., 1-6 1-6 acetoxymethyl, pivaloyloxymethyl or propionyloxymethyl), C alkoxycarbonyloxy-C alkyl (e.g., methoxycarbonyl-oxymethyl or 1-6 1-6 ethoxycarbonyloxymethyl, glycyloxymethyl, phenylglycyloxymethyl, (5-methyloxo-1,3-dioxolenyl)-methyl), and other well known physiologically hydrolyzable esters used, for example, in the penicillin and cephalosporin arts. Such esters may be prepared by conventional techniques known in the art.
[00197] Preparation of prodrugs is well known in the art and described in, for example, King, F.D., ed., Medicinal Chemistry: Principles and Practice, The Royal Society of Chemistry, Cambridge, UK (2 edition, reproduced, 2006); Testa, B. et al., Hydrolysis in Drug and Prodrug Metabolism. Chemistry, Biochemistry and Enzymology, VCHA and Wiley-VCH, Zurich, Switzerland (2003); Wermuth, C.G., ed., The Practice of Medicinal Chemistry, 3 edition, Academic Press, San Diego, CA (2008).
The present invention is intended to include all isotopes of atoms occurring in the present compounds. Isotopes include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of 13 14 hydrogen include deuterium and tritium. Isotopes of carbon include C and C.
Isotopically-labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to 11841-PCT those described herein, using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
The term "solvate" means a physical association of a compound of this invention with one or more solvent molecules, whether organic or inorganic. This physical association includes hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. The solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement. The solvate may comprise either a stoichiometric or nonstoichiometric amount of the solvent molecules.
"Solvate" encompasses both solution-phase and isolable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates.
Methods of solvation are generally known in the art.
Abbreviations as used herein, are defined as follows: "1 x" for once, "2 x" for twice, "3 x" for thrice, "°C" for degrees Celsius, "eq" for equivalent or equivalents, "g" for gram or grams, "mg" for milligram or milligrams, "L" for liter or liters, "mL" for milliliter or milliliters, "μL" for microliter or microliters, "N" for normal, "M" for molar, "mmol" for millimole or millimoles, "min" for minute or min, "h" for hour or h, "rt" for room temperature, "RT" for retention time, "atm" for atmosphere, "psi" for pounds per square inch, "conc." for concentrate, "aq" for "aqueous", "sat" or "sat'd " for saturated, "MW" for molecular weight, "mp" for melting point, "MS" or "Mass Spec" for mass spectrometry, "ESI" for electrospray ionization mass spectroscopy, "HR" for high resolution, "HRMS" for high resolution mass spectrometry, "LCMS" for liquid chromatography mass spectrometry, "HPLC" for high pressure liquid chromatography, "RP HPLC" for reverse phase HPLC, "TLC" or "tlc" for thin layer chromatography, "NMR" for nuclear magnetic resonance spectroscopy, "nOe" for nuclear Overhauser effect spectroscopy, " H" for proton, "δ" for delta, "s" for singlet, "d" for doublet, "t" for triplet, "q" for quartet, "m" for multiplet, "br" for broad, "Hz" for hertz, and "α", "β", "R", "S", "E", "Z" and "ee" are stereochemical designations familiar to one skilled in the art.
Me methyl Et ethyl Pr propyl 11841-PCT i-Pr isopropyl Bu butyl i-Bu isobutyl t-Bu tert-butyl Ph phenyl Bn benzyl Hex hexanes MeOH methanol EtOH ethanol i-PrOH or IPA isopropanol AcOH or HOAc acetic acid Ag CO silver carbonate silver acetate AgOAc CDCl deutero-chloroform CHCl chloroform cDNA complimentary DNA DCC N,N′-dicyclohexylcarbodiimide DIAD diisopropyl azodicarboxylate DMA dimethylamine DME dimethylether DMF dimethyl formamide DMSO dimethyl sulfoxide DMAP 4-dimethylaminopyridine EDTA ethylenediaminetetraacetic acid EtOAc ethyl acetate Et O diethyl ether AlCl aluminum chloride Boc tert-butyloxycarbonyl CH Cl dichloromethane CH CN or ACN acetonitrile Cs CO cesium carbonate 11841-PCT HCl hydrochloric acid H SO sulfuric acid K CO potassium carbonate potassium cyanide mCPBA or m-CPBA meta-chloroperbenzoic acid Pd/C palladium on carbon PhSO Cl benzenesulfonyl chloride diisopropylethylamine i-Pr NEt PS polystyrene SFC Supercritical Fluid Chromatography SiO silica oxide SnCl tin(II) chloride TBAT tetrabutylammonium triphenydifluorosilicate TEA triethylamine TFA trifluoroacetic acid THF tetrahydrofuran KOAc potassium acetate MgSO magnesium sulfate NaCl sodium chloride NaH sodium hydride NaHCO sodium bicarbonate NaOH sodium hydroxide Na SO sodium sulfite Na SO sodium sulfate NH ammonia NH Cl ammonium chloride NH OH ammonium hydroxide LG leaving group Pd dba tris(dibenzylideneacetone)dipalladium(0) selectFluor N-fluoro-N′-methyl-triethylenediamine bis(tetrafluoroborate) 11841-PCT The compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis. The compounds of the present invention can be synthesized using the methods described below, together with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art. Preferred methods include, but are not limited to, those described below. The reactions are performed in a solvent or solvent mixture appropriate to the reagents and materials employed and suitable for the transformations being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformations proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention.
The novel compounds of this invention may be prepared using the reactions and techniques described in this section. Also, in the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and workup procedures, are chosen to be the conditions standard for that reaction, which should be readily recognized by one skilled in the art. Restrictions to the substituents that are compatible with the reaction conditions will be readily apparent to one skilled in the art and alternate methods must then be used.
SYNTHESIS The compounds of Formula (I) may be prepared by the exemplary processes described in the following schemes and working examples, as well as relevant published literature procedures that are used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter and in the working examples.
Protection and de-protection in the processes below may be carried out by procedures generally known in the art (see, for example, Wuts, P. G. M. and Greene, T.W. Protecting Groups in Organic Synthesis, 4th Edition, Wiley (2007)). General methods of organic synthesis and functional group transformations are found in: Trost, B.M. and Fleming, I., eds., Comprehensive Organic Synthesis: Selectivity, Strategy & Efficiency in Modern Organic Chemistry, Pergamon Press, New York, NY (1991); Smith, M. B. and March, J., 11841-PCT March’s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure. 6th Edition, Wiley & Sons, New York, NY (2007); Katritzky, A.R. and Taylor, R. J. K., eds., Comprehensive Organic Functional Groups Transformations II, 2nd Edition, Elsevier Science Inc., Tarrytown, NY (2004); Larock, R.C., Comprehensive Organic Transformations, VCH Publishers, Inc., New York, NY (1999), and references therein. 3 4 6 For example, compounds of Formula (II), where R = R = H and R = CN, can be made according to Scheme 1. Ketones 1 and 2 and 2-cyanoethyl acetate (3) are heated to between 80 °C and 110 °C with ammonium acetate in a solvent such as DMF or DMSO. Ketones 1 and 2 may be different or the same.
Scheme 1 Alternatively, compounds of Formula (II), where R = R = H, may be made according to Scheme 2. α-Bromoketone 4 is combined with triphenylphosphine in a solvent such as THF, CH Cl or 1,4-dioxane at temperatures between room temperature and reflux. The intermediate triphenylphosphonium bromide is treated with base, such as NaOH, in a solvent such as methanol and water to form the phosphorous ylide 5. The phosphorous ylide 5 is heated to 80 °C with ketone 2 in a suitable solvent such as THF or DMSO to give α,β-unsat'd ketone 6, which may exist as a mixture of E/Z isomers.
Microwave irradiation may be employed to shorten the reaction time. α,β-Unsat'd ketone 6 is treated with concentrated aq NH OH in a solvent such as DMSO in a sealed vessel to provide amine 7. Alternatively, alkene 6 may be treated with NH in a solvent such as DMSO or DMSO and methanol in a sealed vessel to provide amine 7. Amine 7 is couple with carboxylic acid 8 using a variety of amide bond forming reactions. For example, carboxylic acid 8 may be converted to the corresponding acid chloride using oxalyl 11841-PCT chloride in a solvent such as CH Cl and catalytic DMF. Alternatively, when R is an amide or a heterocycle, the carboxylic acid 8 may be activated using triphenylphosphine and trichloroacetonitrile in a suitable solvent such as CH Cl . The acid chlorides thus formed are combined with amine 7 in a suitable solvent such as CH Cl or CH Cl and 2 2 2 2 DMF in the presence of a base, preferably pyridine. When R is CF , cyclization of amide 9 to a compound of Formula (I) typically occurs during the work-up procedure for amide 9; for example, when an EtOAc solution of amide 9 is washed with sat'd aq NaHCO . When cyclization does not occur under these conditions, cyclization may be affected by stirring amide 9 in the presence of a weak base such as piperidine in a suitable solvent such as EtOH at a temperature between room temperature and reflux. 11841-PCT Scheme 2 An alternate synthesis to α,β-unsaturated ketone 6, where R is -(CH ) - (phenyl substituted with 0-3 R ) and m = 0, is shown in Scheme 3. Aryl bromide 10 and α,β-unsat'd ester 11 are coupled using palladium (II) acetate, tetrabutylammonium chloride and dicyclohexylamine in DMA at 110 °C. α,β-Unsat'd ester 12 is combined with O,N-dimethylhydroxyl-amine 13 in the presence of a strong base such as iso-propylmagnesium bromide in an aprotic solvent such as THF. α,β-Unsat'd amide 14 is combined with aryl magnesium halide 15 to provide α,β-unsat'd ketone 6. The identity of the halide in the aryl magnesium halide is dependent upon availability of the aryl halide used to make the Grignard reagent; typically the halide is chloride or bromide. 11841-PCT Scheme 3 Pd(OAc) Et NCl O dicylcohexylmethylamine DMA O H CH 11 12 13 2 15 R R O R R O 12 14 iPrMgBr R R R R N CH 11 15 11 13 3 R R R CH 14 15 6
[00207] Non-commercial α,α,α-trifluoroketones 2, where R = CF , may be made from the corresponding aldehyde 16 as shown in Scheme 4. Aldehyde 16 is reacted with trimethyl-(trifluoromethyl)silane in the presence of a fluoride source, for example cesium fluoride, using a suitable solvent such dimethoxyethane at room temperature. Other fluoride sources, such as potassium hydrogen fluoride or tetrabutylammonium difluorotriphenylsilicate, and other solvents, such as THF or acetonitrile and methanol, may also be employed. Trifluoromethyl alcohol 17 is oxidized using, for example, Dess-Martin periodinane in a suitable solvent such as CH Cl .
Scheme 4 OH O (CH ) SiCF oxidize 3 3 3 1 2 1 2 R R R R 16 17: R = CF 2: R = CF Ketones of formula 2 may be made according to Scheme 5. For example, aryl halide 18, where X = bromine and the aryl group is a suitable chemical moiety to form a Grignard reagent, is combined with magnesium metal in the presence of an initiator such as iodine in a suitable solvent such as THF. Other alkyl halides having a chemical moiety suitable for formation of Grignard reagents, other halides such as chlorine or iodine, other 11841-PCT solvents such as diethyl ether or 1,4-dioxane, and other initiators such as 1,2-dibromoethene, may be employed as determined by one skilled in the art. Grignard reagent 19 is combined with amide 20 in a suitable solvent such as THF to provide ketone 2. Other solvents such as 1,4-dioxane or diethyl ether may be employed as determined by one skilled in the art.
Scheme 5 solvent R MgX + 18 19 2 3 4 1
[00209] Compounds of Formula 23 having R = R = H, R = -(CH ) -(phenyl b b c substituted with 0-3 R ) where m = 0, and at least one R = -(CH ) -(X) -(CH ) -R 2 n t 2 m where n = m = t = 0, or n = m = 0 and t = 1 when X = O or NH, and R is a suitable chemical moiety to participate in palladium cross coupling reactions, may be made according to Scheme 6. Compound 21 is heated with boronic acid 22, where R = H, in the presence of a palladium catalyst and base using a suitable solvent such as 1,4-dioxane, toluene, DMF with or without water. Boronic acid 22 may be substituted with alternative boronic acid analogs such as boronate esters, trifluoroborates, and others known to those skilled in the art. Palladium catalyst commonly employed include, but are not restricted to, Pd(PPh ) and PdCl (dppf). Other palladium catalyst known to those skilled in the art 3 4 2 may be employed. Bases commonly employed include, but are not restricted to, K PO and K CO . Other bases known to those skilled in the art may also be employed. When n = m = 0 and t = 1 when X = O or NH, biarylethers or biarylamines such as 23 can be obtained from 21 when G = OH. Alternatively, biaryl ethers and amines can also be obtained from 21, when G = boronic acid or equivalent, via metal-catalyzed coupling with suitable phenols or amines. 11841-PCT Scheme 6 13 13 12 14 12 14 R R R R 11 15 11 15 R R R R Pd catalyzed 6 coupling 6 R -B(OR) N O 22 N O G = Cl, Br, I, OMs, OTs, OTf, OH, NH 3 4 1 Compounds of Formula 27 having R = R = H, R = -(CH ) -(phenyl b b c substituted with 0-3 R ) where m = 0, and at least one R = -(CH ) -(X) -(CH ) -R 2 n t 2 m where n = 0, t = 1, m = 1-4 and X = O, may be made according to Scheme 7. Bromide 24 is treated with tris(dibenzylideneacetone)palladium (0) in the presence of bis(1,1-dimethylethyl)[2′,4′,6′-tris(1-methylethyl)[1,1′-biphenyl]yl]- phosphine (t-butyl-Xphos) using 1,4-dioxane and water as solvent and KOH as base. Phenol 25 and alcohol 26 were stirred in the presence of triphenylphosphine and DIAD in a suitable solvent such as CH Cl . 11841-PCT Scheme 7 Carboxylic acid 8, where R = CONHR , may be made according to Scheme 8. The mono-ester of malonic acid 28, where PG = benzyl group, and amine 29 are coupled together using standard amide bond forming conditions. For example, treatment of carboxylic acid 28 with oxalyl chloride in CH Cl and DMF provides the acid chloride. The acid chloride is then combined with amine 29 in the presence of pyridine in a suitable solvent such as CH Cl . Other amide bond forming reaction known to those skilled in the art may be employed. The benzyl group is removed using a combination of hydrogen gas and 10% palladium on carbon in a suitable solvent such as methanol or methanol and EtOAc. Other PG moieties and methods for their removal known to those skilled in the art may be employed. 11841-PCT Scheme 8 1. amide coupling 2. deprotection NH -R R O OH 28 29 8: R = CONHR Compounds of Formula (I), where R and R are combined with the carbon atom to which they are attached to form a 3-6 membered carbocycle, or R = R = F, may be made according to Scheme 9. For example, to synthesize compounds for Formula (I) where R and R are combined with the carbon atom to which they are attached to form a 3-membered ring (i.e., cyclopropyl), β-ketoester 30 is stirred at room temperature with 1,2-dibromoethane in the presence of a base, for example K CO , in a suitable solvent such as DMF to provide the cyclopropyl β-ketoester 31. Cyclopropyl β-ketoester 31 is stirred with a suitable amine, such as benzyl amine, in the presence of a suitable Lewis acid, such as TiCl , in a solvent such as CH Cl starting at 0 °C then warming to room 4 2 2 temperature. Other amines, Lewis acids, solvents and temperatures may be used as determined by those skilled in the art. The use of benzylamine provides imine 32, where PG = benzyl. Imine 32 is alkylated with, for example, trimethyl(trifluoromethyl)silane in the presence of a fluoride source such as potassium hydrogen fluoride and TFA, using acetonitrile and DMF. Other fluoride sources, such as tetrabutylammonium difluorotriphenylsilicate or cesium fluoride, other acids such as HOAc or HCl, and other solvents may be employed as determined by those skilled in the art. Use of trimethyl(trifluoromethyl)silane provides amino ester 33, where R = CF . Ester hydrolysis of amino ester 33 was done in the presence of lithium iodide in refluxing pyridine to provide amino acid 34. Use of other hydrolysis conditions known to those skilled in the art may be employed. Cyclization of amino acid 34 to β-lactam 35 was affected by activating the carboxylic acid of amino acid 34 with oxalyl chloride in a suitable solvent such as CH Cl containing catalytic DMF. The cyclization occurred spontaneously at room temperature to provide β-lactam 35. Other methods for activating the carboxylic acid may be employed as determined by those skilled in the art. β-Lactam is arylated using an organometallic reagent. Organometallic reagents may include, for example, Grignard reagents or organolithium reagents, formed from a suitably 11841-PCT substituted phenyl ring containing a halide atom able to react with either elemental magnesium to form a Grignard reagent or with an alkyl lithium reagent to form an phenyl lithium reagent via transmetallation. Exact conditions required to form these phenyl organometallic species must determined by those skilled in the art. A suitable aprotic solvent is used, for example, THF. Other suitable solvents may be employed as determined by those skilled in the art. The reaction is carried out between room temperature and reflux depending upon the identity of the organometallic reagent employed and the substitution pattern on β-lactam 35. The β-amino ketone 37 thus formed is deprotected using hydrogen gas and 10% palladium on carbon in a suitable solvent such as methanol containing 4.4% formic acid to provide β-amino ketone 38.
Other conditions to remove the benzyl group may be employed as determined by those skilled in the art. β-Amino ketone 38 is acylated with carboxylic acid 8 using conditions described in Scheme 2 to give the β-keto amide 39. Stirring β-keto amide 39 with a base such as sodium ethoxide in a suitable solvent such as ethanol at room temperature provides compounds having Formula (I). 11841-PCT Scheme 9 Compounds of Formula (I), where y is a single bond and R = F may be made according to Scheme 10. This synthesis is exemplified for a compound of Formula (I) where R = CN. Thus, a compound of Formula (I), where x is a single bond and R = CN, was heated to 80 °C in the presence of a fluorinating reagent, 1-fluorohydroxy-1,4-diazoniabicyclo[2,2,2]octane bis(tetrafluoroborate), in a suitable solvent such as DMF in the presence of a base such as Na CO to provide compounds of Formula (I), where y is a single bond, R = F and R = CN. Other fluorinating reagents, solvents and bases may be employed as determined by one skilled in the art. 11841-PCT Scheme 10 12 14 12 14 11 15 fluorination 11 15 1 N O (I): x = single bond (I): y = single bond R = F Compounds of Formula (I), where x and y are both single bonds and R = H, can be made according to Scheme 11. Reduction of compounds of Formula (I), where x equals a single bond and y equals a double bond, is carried out using a suitable catalyst such as palladium on carbon under an atmosphere of hydrogen gas at suitable pressure, such as 50 psi, to effect reduction of the double bond y to a single bond. Suitable solvents include, but are not restricted to, methanol.
Scheme 11 Compounds of Formula (II), single enantiomer, where R = R =H, can be made according to Scheme 12. Ketone 2 was stirred with 2-methylpropanesulfinamide in the presence of a suitable Lewis acid, such as Ti(OEt) , in a solvent such as THF at refluxed tempereature provides imine 40. Other Lewis acids, solvents and temperatures may be used as determined by those skilled in the art. Imine 40 is alkylated with ketone 1 11841-PCT in the presence of a base, such as LiHMDS, KHMDS, NaHMDS, or LDA in an aprotic solvent such as THF or ether at a temperature ranging from -78 °C to ambient to provide β-amino ketone 41 as a mixture of two diastereomers, which can be separated by silica gel chromatography to give the desired isomer 42. Other metal enolates (such as titanium enolate), solvents, and temperatures may be used as determined by those skilled in the art (T. P. Tang, J. A Ellman, J. Org. Chem. 1999, 64, 12-13, J. Org. Chem. 2002, 67, 7819- 7832). Preferably, chiral S- or Rmethylpropanesulfinamide can be optionally used to generate each of the optically pure enantiomers of imine 40 that can allow for chiral induction to prepare diastereomerically enriched ketone 42. In these cases, the product mixture can be further purified by silica gel chromatography to obtain desired products with diaseteromeric excess of >97%. β-amino ketone 42 thus formed is deprotected using HCl in a suitable solvent such as MeOH to provide β-amino ketone 43. Other conditions to remove the t-butylsulfinyl group may be employed as determined by those skilled in the art. β-Amino ketone 43 is acylated with carboxylic acid 8 using conditions described in Scheme 2 to give the β-keto amide 44. Stirring β-keto amide 44 with a base such as sodium ethoxide in a suitable solvent such as ethanol at room temperature provides compounds having Formula (II). 11841-PCT Scheme 12 13 14 NH O R imine S 2 12 N 1 R R S formation NH 1 1 2 2 1 2 alkylation 13 NH O R NH O R diastereomer separated by 2 12 R silica gel chromatography R R deprotection 6 13 13 enriched diastereomer 12 14 amide 11 15 NH O R coupling R R cyclization 13 R R N O (I): R = R = H Alternatively, compounds of Formula (II), where R = R =H, may be made according to Scheme 13. Ketone 2 can be reacted with 2-methylpropanesulfinamide in the presence of a suitable Lewis acid, such as Ti(OEt) , in a solvent such as THF at a temperature ranging from ambient to reflux to provide imine 45. Other Lewis acids, solvents and temperatures may be used as determined by those skilled in the art. Imine 45 is alkylated with the enolate of an ester in a suitable aprotic solvent such as THF or ether starting at -78 °C then warming to 0 °C or room temperature to provide protected β- amino ketone 46 as a mixture of two diastereomers, which can be separated by silica gel chromatography to give each individual chiral compound. The generation of the ester enolate is achieved by treating the ester, such as methyl acetate, with a suitable base such as LHMDS, KHMDS, NaHMDS, or LDA in an aprotic solvent such as THF or ether at a temperature ranging from -78 °C to ambient. Other metal enolates (such as titanium enolate), solvents, and temperatures may be used as determined by those skilled in the art (T. P. Tang, J. A Ellman, J. Org. Chem. 1999, 64, 12-13, J. Org. Chem. 2002, 67, 7819- 11841-PCT 7832). Preferably, chiral S- or Rmethylpropanesulfinamide can be optionally used to generate each of the optically pure enantiomers of imine 45 that can allow for chiral induction to prepare diastereomerically enriched ester 46. In these cases, the product mixture can be further purified by silica gel chromatography to obtain desired products with diaseteromeric excess of >97%. The tert-butyl sulfinyl group of 46 is removed using acids such as HCl and TFA in a suitable solvent such as MeOH or dioxane to generate amino ester 47. Other conditions to remove the t-butylsulfinyl group may be employed as determined by those skilled in the art. β-Amino ketone 47 is acylated with carboxylic acid 8 using conditions described in Scheme 2 to give the β-keto amide 48.
Stirring β-keto amide 48 with a base such as sodium ethoxide in a suitable solvent such as ethanol at room temperature to 80 C provides cyclic enol 49. Other conditions can also by used to effect the cyclization as determined by those skilled in the art. Compound 49, when treated with stoichiometric amount of a chlorinating agent, such as POCl , at elevated temperature in an inert solvent such as toluene, is converted to mono-chloride 50. Chloride 50 can then react with various boronic reagents through a Suzuki-type of cross coupling reaction to generate compounds of Formula (II). The choices of boronic reagents, catalysts, ligands, bases, solvents and temperatures are well documented in the literature and can be selected appropriately by those skilled in the art. 11841-PCT Scheme 13 3 4 1 Compounds of Formula (II), where R = R =H and R = CONHC alkyl or 4-18 CONH(CH ) Ph can be made according to Scheme 14. The phosphorous ylide 5 is 2 1-8 heated, using microwave irradiation, to 150 °C with α-ketoester 52 in a suitable solvent such as THF or DMSO to give α,β-unsat'd ketone 53. α,β-Unsat'd ketone 53 is treated with concentrated aq NH OH in a solvent such as DMSO in a sealed vessel to provide amine 54. Alternatively, alkene 53 may be treated with NH in a solvent such as DMSO or DMSO and methanol in a sealed vessel to provide amine 54. Amine 54 is couple with carboxylic acid 8 using a variety of amide bond forming reactions. For example, carboxylic acid 8 may be converted to the corresponding acid chloride using oxalyl chloride in a solvent such as CH Cl and catalytic DMF. Alternatively, the carboxylic acid 8 may be activated using 1-chloro-N,N,2-trimethylprop-enamine in a suitable solvent such as CH Cl . The acid chlorides thus formed are combined with amine 54 in a suitable solvent such as CH Cl or CH Cl and DMF in the presence of a base, 2 2 2 2 preferably pyridine. Other amide bond forming reaction known to those skilled in the art may be employed. Cyclization of amide 55 and susequent hydrolysis to a the carboxylic acid 56 typically occurs by stirring amide 55 in the presence of a weak base such as 11841-PCT piperidine in a suitable solvent such as EtOH at a temperature between room temperature and reflux or a base such as litium hydroxide in a suitable solvent such as THF and water at room temperature. Alternatively, when R is a nitrile, cyclization of amide 55 typically occurs by stirring amide 55 in the presence of a base such as lithium hydroxide in a suitable solvent such as THF and water at room temperature. Hydrolysis is then carried out under acidic conditions using a strong acid such as HCl in a suitable solvent such as acetic acid at temperatures between room temperature and 50 C. Carboxylic acid 56 and an amine are coupled together using standard amide bond forming conditions. For example, treatment of carboxylic acid 56 and an amine with HOBt, EDC and DIEA in the presence of pyridine in a suitable solvent such as DCM at room temperature provides compounds having Formula (II). Other amide bond forming reaction known to those skilled in the art may be employed.
Scheme 14 11841-PCT Compounds of formula 60 having R = alkyl and R = cyano can be synthesized by coupling acid 56 with N, O-dimethylhydroxylamine using typical amide bond forming reactions. For example, carboxylic acid 56 is coupled to N, O- dimethylhydroxylamine using EDC in the presence of base, preferably N- methylmopholine, in a suitable solvent, such as CH Cl , to provide the Weinreb amide 57. Other amide forming reactions known to those skilled in the art may also be employed. The intermediate Weinreb amide 57 is reacted with ethynylmagnesium bromide in an aprotic solvent such as THF at 0-35 C to provide the acylacetylide intermediate 58. Compounds of formula 60 are obtained when acylacetylide intermediate 58 is reacted with hydrazine 59 in the presence of a base such as TEA in a suitable solvent such as EtOH.
Scheme 15 Compounds of formula 63 having R = alkyl and R = cyano can be synthesized by first coupling acid 56 with β−ketoamine 61 using typical amide bond forming reactions. For example, carboxylic acid 56 is coupled to α−ketoamine 61 using EDC and HOBt in the presence of base, preferably DIEA, in a suitable solvent, such as CH Cl , to provide β-ketoamide 62. Other amide forming reactions known to those skilled in the art may also be employed. 5-Alkyl substituted oxazole 63 is then obtained 11841-PCT via the dehydrative cyclization of β-ketoamide 62 using a dehydrating agent, preferably POCl3, in the presence of a suitable base, such as DIEA, in a suitable solvent such as dichloroethane at a temperature of 80-120 C.
Scheme 16 IV. BIOLOGY In mammals, there are two triglyceride synthesis pathways: glycerol phosphate pathway and monoacylglycerol pathway. The former is mainly responsible for energy storage in the peripheral tissues such as fat, liver, skeletal muscle; the latter is essential for the dietary fat absorption which takes place in the small intestine. When dietary fat is ingested, pancreatic lipase digests triglycerides into free fatty acids and 2- monoacylglycerol, which are absorbed by intestinal epithelial enterocytes. Once inside enterocytes, free fatty acids and 2-monoacylglycerol are used as building blocks to resynthesize triglycerides by two sequential acylation steps; first by MGAT and then by DGAT enzyme reactions. Triglycerides are then incorporated into chylomicrons and secreted into lymph to be utilized as an energy supply for the body.
Monoacylglycerol acyltransferase 2 (MGAT2) is a membrane bound acyltransferase that belongs to diacylglycerol acyltransferase 2 (DGAT2) gene family. It 11841-PCT is highly and selectively expressed in the small intestine. Genetic deletion of MGAT2 in mice decreased the rate of absorption for the orally ingested triglycerides, indicating that MGAT2 plays an important role for the intestinal MGAT/DGAT pathway [Yen, C.L. et al, Nat. Med., 15(4):442-446 (2009); Okawa, M. et al., Biochem. Biophys. Res. Commun., 390(3):377-381 (2009)]. When chronically challenged with a high fat diet, in contrast to wild type mice that became obese, MGAT2 knockout mice resisted the impact of high-fat feeding and demonstrated with a lower body weight, less adiposity, and less hepatic fat accumulation. In contrast to hyperinsulinemic wild type mice after high-fat challenge, MGAT2 deletion normalizes the insulin level and decreased fasting glucose. In the glucose tolerance test, they also had an improved glucose excursion. Consistent with their improved glycemic profile, MGAT2 knockout mice also had an increased level of GLP1, an incretin gut hormone that profoundly impacts glucose metabolism[Yen, C.L. et al., Nat. Med., 15(4):442-446 (2009)]. Taken together, it is expected that inhibition of MGAT2 through pharmacological intervention would provide the same benefit as demonstrated in the knock-out mice, e.g., resistance to weight gain, or conversely, reduction in fat body mass. In addition, MGAT2 inhibition would lead to an improved insulin sensitivity and glucose metabolism which either leads to a decrease in the incidence of Type II diabetes, or a treatment of diabetic condition.
It is also desirable and preferable to find compounds with advantageous and improved characteristics compared with known anti-diabetic agents, in one or more of the following categories that are given as examples, and are not intended to be limiting: (a) pharmacokinetic properties, including oral bioavailability, half life, and clearance; (b) pharmaceutical properties; (c) dosage requirements; (d) factors that decrease blood drug concentration peak-to-trough characteristics; (e) factors that increase the concentration of active drug at the receptor; (f) factors that decrease the liability for clinical drug-drug interactions; (g) factors that decrease the potential for adverse side-effects, including selectivity versus other biological targets; and (h) improved therapeutic index with less propensity for hypoglycemia.
As used herein, the term "patient" encompasses all mammalian species.
[00224] As used herein, the term "subject" refers to any human or non-human organism that could potentially benefit from treatment with a MGAT2 inhibitor.
Exemplary subjects include human beings of any age with risk factors for metabolic 11841-PCT disease. Common risk factors include, but are not limited to, age, sex, weight, family history, or signs of insulin resistance such as acanthosis nigricans, hypertension, dyslipidemia, or polycystic ovary syndrome (PCOS).
As used herein, "treating" or "treatment" cover the treatment of a disease-state in a mammal, particularly in a human, and include: (a) inhibiting the disease-state, i.e., arresting it development; and/or (b) relieving the disease-state, i.e., causing regression of the disease state.
As used herein, "prophylaxis" or "prevention" cover the preventive treatment of a subclinical disease-state in a mammal, particularly in a human, aimed at reducing the probability of the occurrence of a clinical disease-state. Patients are selected for preventative therapy based on factors that are known to increase risk of suffering a clinical disease state compared to the general population. "Prophylaxis" therapies can be divided into (a) primary prevention and (b) secondary prevention. Primary prevention is defined as treatment in a subject that has not yet presented with a clinical disease state, whereas secondary prevention is defined as preventing a second occurrence of the same or similar clinical disease state.
As used herein, "risk reduction" covers therapies that lower the incidence of development of a clinical disease state. As such, primary and secondary prevention therapies are examples of risk reduction.
[00228] "Therapeutically effective amount" is intended to include an amount of a compound of the present invention that is effective when administered alone or in combination to inhibit MGAT2 and/or to prevent or treat the disorders listed herein.
When applied to a combination, the term refers to combined amounts of the active ingredients that result in the preventive or therapeutic effect, whether administered in combination, serially, or simultaneously.
A. ASSAY METHODS MGAT SPA Assay MGAT2 enzyme was assayed using membranes isolated from Sf9 cells expressing the recombinant human MGAT2 cDNA with 2-monooleoylglycerol and [ H]- oleoyl-CoA as substrates as described by Seethala et al. [Anal. Biochem., 383(2):144-150 (Dec. 15, 2008)]. Briefly, the assays were conducted in 384-well plates in a total volume 11841-PCT of 30 µL at 25 °C. In each assay, 200 ng of recombinant human MGAT2 membrane was incubated with 10 μM of 2-monooleoylglycerol and 15 μM of [ H]-oleoyl-CoA in 100 mM potassium phosphate (pH 7.4) for 20 min with various concentrations of compounds delivered in DMSO. The assay was terminated by the addition of 20 μl of Stopping Solution (7.5 mg/ml Yittrium Oxide Polylysine beads, 3.3 mg/ml Fraction V BSA and 200 μM Mercuric chloride in 50 mM HEPES, pH 7.4). The signal was measured 1 h after quenching the reaction using LEADSEEKER for 5 minutes. To calculate the degree of inhibition, the zero level of enzyme activity (blank) was defined by the above assay procedure using membrane form Sf9 cell uninfected with baculovirus (Naive) and the 100% level of MGAT2 enzyme activity was defined by human MGAT2 assay with the vehicle DMSO. The IC s of inhibitors were determined by logistic 4 parameter equation in XL-fit.
MGAT LCMS Assay
[00230] The MGAT enzyme reactions were performed in CORNING Falcon 96-well Polypropylene plates, in a total volume of 60 µL of 50 mM Potassium Phosphate buffer pH 7.4, containing a final concentration of 100 µM 2-oleoylglycerol, 15 µM oleoyl- Coenzyme A and 0.0013 µg/µL Human or Mouse MGAT-2 or 0.0026 µg/µL Rat recombinant MGAT-2 membranes expressed in Sf9 cells. Assay plates were run through a fully automated robotics system and shaken for 5 seconds every minute for a total 10 minutes. The reactions were then quenched with 120 µL of ice cold methanol containing 1 µg/mL 1,2-distearoyl-rac-glycerol as the internal standard. Plates were shaken for 2 minutes and spun down to remove protein precipitation. After the spin, samples were transferred to LC/MS compatible PCR plates. For LC/MS analysis, a ThermoFisher Surveyor pump, utilizing a Waters Symmetry C8, 50 x 2.1 mm column, was used for the chromatography of enzyme products. The buffer system consists of 0.1% formic acid in water with a mobile phase consisting 0.1% formic acid in methanol. The shallow gradient is 90-100% mobile phase in 0.2 min with a total run time of 2.3 min. The first 0.5 minutes of each injection was diverted to waste to eliminate the presence of Phosphate buffer in the enzymatic reaction. The column was run at 0.6 mL/min and a temperature of 65 °C. Mass spectrometry analysis of the samples was performed on a ThermoFisher Quantum Triple quad utilizing APCI (+) as the mode of ionization. Data 11841-PCT was acquired in Single Ion Monitoring (SIM) mode analyzing Diolein =m/z 603.6 (PRODUCT) and 1,2- distearoyl-rac-glycerol (IS)= m/z 607.6. The ratio of Diolein to internal standard (Peak Area Ratio) is utilized to calculate IC values.
[00231] The exemplified Examples disclosed below were tested in the MGAT2 in vitro assays described above and were found having MGAT2 inhibitory activity. Table 1 below lists human MGAT2 IC values measured for the following examples. “NT” denotes “Not tested”.
Table 1 Example h-MGAT2 IC (nM) SPA Assay LCMS Assay 1 651 2049 2 85 5 2-1 35 8 2-2 1400 261 3 1804 NT 4 5871 NT 309 21 6 15 7 6-1 1435 NT 6-2 14 2 7 818 NT 8 NT 7 9 33330 121 2716 138 11 48 13 12 NT 374 13 3333 35 14 NT 2050 -1 3306 12 11841-PCT -2 33330 431 -3 33330 756 -4 33330 2049 16-1 25 7 16-2 8430 416 17 1931 NT 18 5762 NT 19 2905 NT 3456 NT 21 913 NT 22 801 NT 23 1292 NT 24 1731 NT 4569 NT 26 6428 NT 27 2519 NT 28 564 NT 29 7602 NT 774 NT 31 1510 NT 32 9956 NT 33 167 12 34 656 126 5199 13 36 396 57 37 263 15 38 2143 NT 39 401 20 40 2546 164 41 170 15 42 403 77 43 5387 60 11841-PCT 44 1458 NT 45 569 NT 46 871 NT 47 756 NT 48 226 7 49 127 64 50 242 95 51 63 13 52 477 99 53 111 3 54 258 20 55 846 20 56 6550 NA 57 412 16 58 275 29 59 53 4 60 441 12 61 709 NT 62 156 24 63 116 28 64 761 280 65 96 3 66 731 265 67 1935 253 68 7523 1282 69 9230 983 70 3404 NA 71 2931 1917 72 3519 214 73 684 15 74 2476 697 75 873 209 11841-PCT 76 5073 370 77 89 27 78 151 4 79 1856 45 80 310 5 81 55 4 82 133 2 83 89 1 84 115 2 85 1465 7 86 201 1 87 NT 1215 88 28 5 89 731 20 90 815 18 91 916 2 92 NT 136 93 37 30 94 32 4 95 95 43 96 NT 1431 97 NT 1199 98 154 69 99 NT 795 100 NT 1575 101 379 108 102 NT 1088 103 NT 645 104 NT 103 105 672 24 106 NT 552 107 NT 93 11841-PCT 108 NT 189 109 NT 39 110 11 5 111 9 7 112 1297 204 113 158 8 114 NT 704 115 NT 40 116 48 4 117 22 18 118 32 4 119 94 21 120 58 6 121 231 20 122 14 1 123 743 5 124 4264 55 125 NT 282 126 1874 33 127 655 10 128 NT 328 129 NT 222 130 20 1 131 NT 1050 132 NT 163 133 1518 127 134 NT 168 135 NT 778 136 NT 328 137 NT 252 138 NT 5799 140 NT 356 11841-PCT 141 NT 459 142 93 10 143 156 4 144 175 16 145 130 25 146 397 25 147 91 14 148 102 9 149 53 6 150 11 3 151 NT 5115 152 NT 252 153 NT 247 154 34 8 155 NT 484 156 NT 254 157 168 84 158 302 75 159 NT 16710 160 NT 5401 161 NT 1261 162 NT 214 163 1176 47 164 NT 162 165 60 8 166 237 102 167 8 2 168 NT 113 169 NT 421 170 NT 210 171 724 57 172 16 3 11841-PCT 173 43 63 174 237 60 175 NT 264 176 NT 3 177 NT 399 178 47 78 179 24 2 180 36 30 181 NT 2049 182 NT 2049 183 NT 50 184 NT 76 185 NT 121 186 NT 229 187 NT 4 188 NT 1524 189 NT 1159 190 NT 155 191 NT 797 192 NT 433 193 NT 46 194 NT 6 195 NT 33 196 NT 826 197 NT 178 198 NT 16 199 NT 16 200 NT 37 201 NT 24 202 NT 89 203 NT 59 204 NT 41 11841-PCT 205 NT 13 206 NT 78 207 NT 11 208 NT 30 209 NT 37 210 NT 2 211 NT 97 212 NT 416 213 NT 1096 214 NT 12 215 NT 11 216 NT 440 217 NT 118 218 NT 166 219 NT 172 220 NT 13 221 NT 152 222 NT 733 223 NT 112 224 NT 414 225 NT 889 226 NT 206 227 NT 63 228 NT 7 229 NT 25 230 NT 1532 231 NT 34 232 NT 6 233 NT 639 234 NT 40 235 NT 22 236 NT 489 11841-PCT 237 NT 130 238 NT 7 239 NT 15 240 NT 7 242 NT 44 243 NT 14 244 NT 61 245 NT 46 246 NT 36 247 NT 51 248 NT 8 249 NT 185 250 NT 243 251 NT 3 252 NT 7 253 NT 411 254 NT 170 255 NT 702 256 NT 22 257 NT 421 258 NT 1147 259 NT 15 260 NT 56 261 NT 1222 262 NT 96 263 NT 103 264 NT 46 265 NT 40 266 NT 10 267 NT 796 268 NT 8 269 NT 171 11841-PCT 270 NT 407 271 NT 12 272 NT 112 273 21 10 274 NT 1 275 NT 2 276 NT 2 277 NT 2 278 NT 2 279 NT 2 280 NT 2 281 NT 3 282 NT 3 283 NT 3 284 NT 4 285 NT 4 286 NT 4 287 NT 13 288 NT 19 289 NT 19 290 NT 56 291 NT 178 292 NT 304 293 NT 7 294 NT 84 295 NT 183 296 NT 2 297 NT 639 298 NT 2 299 NT 11 300 NT 11 301 NT 19 11841-PCT 302 NT 105 303 NT 28 304 NT 6 305 NT 94 306 NT 3 307 NT 3 308 NT 61 309 NT 1 310 NT 4 311 NT 3 312 NT 21 313 NT 4 314 NT 109 The compounds of the present invention possess activity as inhibitors of MGAT2, and, therefore, may be used in the treatment of diseases associated with MGAT2 activity. Via modulation of MGAT2, the compounds of the present invention may preferably be employed to modulate, either enhance or decrease the production/secretion of insulin and/or gut hormones, such as GLP1, GIP, CCK, PYY, PP, Amylin.
Accordingly, the compounds of the present invention can be administered to mammals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to, treating, preventing, or slowing the progression of diabetes and related conditions, microvascular complications associated with diabetes, macrovascular complications associated with diabetes, cardiovascular diseases, Metabolic Syndrome and its component conditions, inflammatory diseases and other maladies.
Consequently, it is believed that the compounds of the present invention may be used in preventing, inhibiting, or treating diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, retinopathy, neuropathy, nephropathy, wound healing, atherosclerosis and its sequelae (acute coronary syndrome, myocardial infarction, angina pectoris, peripheral vascular disease, intermittent claudication, myocardial ischemia, stroke, heart failure), Metabolic Syndrome, 11841-PCT hypertension, obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low HDL, high LDL, lipid disorders, PCOS, and glaucoma.
Metabolic Syndrome or "Syndrome X" is described in Ford et al., J. Am. Med.
Assoc., 287:356-359 (2002) and Arbeeny et al., Curr. Med. Chem. - Imm., Endoc. & Metab. Agents, 1:1-24 (2001).
V. PHARMACEUTICAL COMPOSITIONS, FORMULATIONS AND COMBINATIONS The compounds of this invention can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice.
The term "pharmaceutical composition" means a composition comprising a compound of the invention in combination with at least one additional pharmaceutically acceptable carrier. A "pharmaceutically acceptable carrier" refers to media generally accepted in the art for the delivery of biologically active agents to animals, in particular, mammals, including, i.e., adjuvant, excipient or vehicle, such as diluents, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
[00237] Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include, without limitation: the type and nature of the active agent being formulated; the subject to 11841-PCT which the agent-containing composition is to be administered; the intended route of administration of the composition; and the therapeutic indication being targeted.
Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Allen, L. V. Jr. et al. Remington: The Science and Practice of Pharmacy (2 Volumes), 22nd Edition (2012), Pharmaceutical Press.
The dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
By way of general guidance, the daily oral dosage of each active ingredient, when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day. Intravenously, the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.
Compounds of this invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
[00240] The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g., oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices.
[00241] Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 2000 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be 11841-PCT present in an amount of about 0.1-95% by weight based on the total weight of the composition.
A typical capsule for oral administration contains at least one of the compounds of the present invention (250 mg), lactose (75 mg), and magnesium stearate (15 mg). The mixture is passed through a 60 mesh sieve and packed into a No. l gelatin capsule.
A typical injectable preparation is produced by aseptically placing at least one of the compounds of the present invention (250 mg) into a vial, aseptically freeze-drying and sealing. For use, the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation.
The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of the present invention, alone or in combination with a pharmaceutical carrier. Optionally, compounds of the present invention can be used alone, in combination with other compounds of the invention, or in combination with one or more other therapeutic agent(s), e.g., an antidiabetic agent or other pharmaceutically active material.
The compounds of the present invention may be employed in combination with other MGAT2 inhibitors or one or more other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-diabetic agents, anti- hyperglycemic agents, anti-hyperinsulinemic agents, anti-retinopathic agents, anti- neuropathic agents, anti-nephropathic agents, anti-atherosclerotic agents, anti-ischemic agents, anti-hypertensive agents, anti-obesity agents, anti-dyslipidemic agents, anti- dyslipidemic agents, anti-hyperlipidemic agents, anti-hypertriglyceridemic agents, anti- hypercholesterolemic agents, anti-restenotic agents, anti-pancreatic agents, lipid lowering agents, anorectic agents, memory enhancing agents, anti-dementia agents, or cognition promoting agents, appetite suppressants, treatments for heart failure, treatments for peripheral arterial disease and anti-inflammatory agents.
Where desired, the compound of the present invention may be used in combination with one or more other types of antidiabetic agents and/or one or more other types of therapeutic agents which may be administered orally in the same dosage form, in a separate oral dosage form or by injection. The other type of antidiabetic agent that may 11841-PCT be optionally employed in combination with the MGAT2 inhibitor of the present invention may be one, two, three or more antidiabetic agents or antihyperglycemic agents which may be administered orally in the same dosage form, in a separate oral dosage form, or by injection to produce an additional pharmacological benefit.
[00247] The antidiabetic agents used in the combination with the compound of the present invention include, but are not limited to, insulin secretagogues or insulin sensitizers, other MGAT2 inhibitors, or other antidiabetic agents. These agents include, but are not limited to, dipeptidyl peptidase IV (DP4) inhibitors (for example, sitagliptin, saxagliptin, alogliptin, vildagliptin and the like), biguanides (for example, metformin, phenformin and the like), sulfonyl ureas (for example, glyburide, glimepiride, glipizide and the like), glucosidase inhibitors (for example, acarbose, miglitol, and the like), PPARγ agonists such as thiazolidinediones (for example, rosiglitazone, pioglitazone, and the like), PPAR α/γ dual agonists (for example, muraglitazar, tesaglitazar, aleglitazar, and the like), glucokinase activators (as described in Fyfe, M.C.T. et al., Drugs of the Future, 34(8):641-653 (2009) and incorporated herein by reference), GPR40 receptor modulators, GPR119 receptor modulators (MBX-2952, PSN821, APD597 and the like), SGLT2 inhibitors (dapagliflozin, canagliflozin, remagliflozin and the like), amylin analogs such as pramlintide, and/or insulin. Reviews of current and emerging therapies for the treatment of diabetes can be found in: Mohler, M.L. et al., Medicinal Research Reviews, 29(1):125-195 (2009), and Mizuno, C.S. et al., Current Medicinal Chemistry, 15:61-74 (2008).
The compounds of the present invention may also be optionally employed in combination with agents for treating complication of diabetes. These agents include PKC inhibitors and/or AGE inhibitors.
[00249] The compounds of the present invention may also be optionally employed in combination with one or more hypophagic agents such as diethylpropion, phendimetrazine, phentermine, orlistat, sibutramine, lorcaserin, pramlintide, topiramate, MCHR1 receptor antagonists, oxyntomodulin, naltrexone, Amylin peptide, NPY Y5 receptor modulators, NPY Y2 receptor modulators, NPY Y4 receptor modulators, cetilistat, 5HT2c receptor modulators, and the like. The compound of structure I may also be employed in combination with an agonist of the glucagon-like peptide-1 receptor (GLP-1 R), such as exenatide, liraglutide, GPR-1(1-36) amide, GLP-1(7-36) amide, 11841-PCT GLP-1(7-37) (as disclosed in U.S. Patent No. 5,614,492 to Habener, the disclosure of which is incorporated herein by reference), which may be administered via injection, intranasal, or by transdermal or buccal devices. Reviews of current and emerging therapies for the treatment of obesity can be found in: Melnikova, I. et al., Nature Reviews Drug Discovery, 5:369-370 (2006); Jones, D., Nature Reviews: Drug Discovery, 8:833-834 (2009); Obici, S., Endocrinology, 150(6):2512-2517 (2009); and Elangbam, C.S., Vet. Pathol., 46(1):10-24 (2009).
The compounds of the present invention may also be optionally employed in combination with one or more other types of therapeutic agents, such as DGAT inhibitors, LDL lowering drugs such as statins (inhibitors of HMG CoA reductase) or inhibitors of cholesterol absorption, modulators of PCSK9, drugs increase HDL such as CETP inhibitors.
The above other therapeutic agents, when employed in combination with the compounds of the present invention may be used, for example, in those amounts indicated in the Physicians' Desk Reference, as in the patents set out above, or as otherwise determined by one of ordinary skill in the art.
Particularly when provided as a single dosage unit, the potential exists for a chemical interaction between the combined active ingredients. For this reason, when the compound of the present invention and a second therapeutic agent are combined in a single dosage unit they are formulated such that although the active ingredients are combined in a single dosage unit, the physical contact between the active ingredients is minimized (that is, reduced). For example, one active ingredient may be enteric coated.
By enteric coating one of the active ingredients, it is possible not only to minimize the contact between the combined active ingredients, but also, it is possible to control the release of one of these components in the gastrointestinal tract such that one of these components is not released in the stomach but rather is released in the intestines. One of the active ingredients may also be coated with a material that affects a sustained-release throughout the gastrointestinal tract and also serves to minimize physical contact between the combined active ingredients. Furthermore, the sustained-released component can be additionally enteric coated such that the release of this component occurs only in the intestine. Still another approach would involve the formulation of a combination product in which the one component is coated with a sustained and/or enteric release polymer, 11841-PCT and the other component is also coated with a polymer such as a low viscosity grade of hydroxypropyl methylcellulose (HPMC) or other appropriate materials as known in the art, in order to further separate the active components. The polymer coating serves to form an additional barrier to interaction with the other component.
[00253] These as well as other ways of minimizing contact between the components of combination products of the present invention, whether administered in a single dosage form or administered in separate forms but at the same time by the same manner, will be readily apparent to those skilled in the art, once armed with the present disclosure.
The compounds of the present invention can be administered alone or in combination with one or more additional therapeutic agents. By "administered in combination" or "combination therapy" it is meant that the compound of the present invention and one or more additional therapeutic agents are administered concurrently to the mammal being treated. When administered in combination, each component may be administered at the same time or sequentially in any order at different points in time.
Thus, each component may be administered separately but sufficiently closely in time so as to provide the desired therapeutic effect.
The compounds of the present invention are also useful as standard or reference compounds, for example as a quality standard or control, in tests or assays involving the MGAT2 enzyme. Such compounds may be provided in a commercial kit, for example, for use in pharmaceutical research involving MGAT2 or anti-diabetic activity. For example, a compound of the present invention could be used as a reference in an assay to compare its known activity to a compound with an unknown activity. This would ensure the experimentor that the assay was being performed properly and provide a basis for comparison, especially if the test compound was a derivative of the reference compound. When developing new assays or protocols, compounds according to the present invention could be used to test their effectiveness.
The compounds of the present invention may also be used in diagnostic assays involving MGAT2.
The present invention also encompasses an article of manufacture. As used herein, article of manufacture is intended to include, but not be limited to, kits and packages. The article of manufacture of the present invention, comprises: (a) a first container; (b) a pharmaceutical composition located within the first container, wherein 11841-PCT the composition, comprises: a first therapeutic agent, comprising: a compound of the present invention or a pharmaceutically acceptable salt form thereof; and, (c) a package insert stating that the pharmaceutical composition can be used for the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2 (as defined previously). In another embodiment, the package insert states that the pharmaceutical composition can be used in combination (as defined previously) with a second therapeutic agent for the treatment and/or prophylaxis of multiple diseases or disorders associated with MGAT2. The article of manufacture can further comprise: (d) a second container, wherein components (a) and (b) are located within the second container and component (c) is located within or outside of the second container. Located within the first and second containers means that the respective container holds the item within its boundaries.
The first container is a receptacle used to hold a pharmaceutical composition.
This container can be for manufacturing, storing, shipping, and/or individual/bulk selling.
First container is intended to cover a bottle, jar, vial, flask, syringe, tube (e.g., for a cream preparation), or any other container used to manufacture, hold, store, or distribute a pharmaceutical product.
The second container is one used to hold the first container and, optionally, the package insert. Examples of the second container include, but are not limited to, boxes (e.g., cardboard or plastic), crates, cartons, bags (e.g., paper or plastic bags), pouches, and sacks. The package insert can be physically attached to the outside of the first container via tape, glue, staple, or another method of attachment, or it can rest inside the second container without any physical means of attachment to the first container.
Alternatively, the package insert is located on the outside of the second container. When located on the outside of the second container, it is preferable that the package insert is physically attached via tape, glue, staple, or another method of attachment. Alternatively, it can be adjacent to or touching the outside of the second container without being physically attached.
The package insert is a label, tag, marker, etc. that recites information relating to the pharmaceutical composition located within the first container. The information recited will usually be determined by the regulatory agency governing the area in which the article of manufacture is to be sold (e.g., the United States Food and Drug 11841-PCT Administration). Preferably, the package insert specifically recites the indications for which the pharmaceutical composition has been approved. The package insert may be made of any material on which a person can read information contained therein or thereon. Preferably, the package insert is a printable material (e.g., paper, plastic, cardboard, foil, adhesive-backed paper or plastic, etc.) on which the desired information has been formed (e.g., printed or applied).
Other features of the invention will become apparent in the course of the following descriptions of exemplary embodiments that are given for illustration of the invention and are not intended to be limiting thereof.
VI. EXAMPLES The following Examples are offered as illustrative, as a partial scope and particular embodiments of the invention and are not meant to be limiting of the scope of the invention. In the following Examples, the compounds of Examples 1, 3-5, 7, 14-15, 17-20, 22-37, 39, 40, 42-47, 53, 63-69, 71-81, 89, 91, 107-109, 138, 144-147, 160, 177, 184, 215-227, 230, 256-264 and 269 are reference examples. The compounds of the other Examples illustrate the presently claimed invention. Abbreviations and chemical symbols have their usual and customary meanings unless otherwise indicated. Unless otherwise indicated, the compounds described herein have been prepared, isolated and characterized using the schemes and other methods disclosed herein or may be prepared using the same.
HPLC/MS, PREPARATORY/ANALYTICAL HPLC, AND CHIRAL SEPARATION METHODS EMPLOYED IN CHARACTERIZATION OR PURIFICATION OF EXAMPLES Analytical HPLC/MS (unless otherwise noted) was performed on Shimadzu SCL-10A liquid chromatographs and Waters MICROMASS ZQ Mass Spectrometers (Desolvation Gas: Nitrogen; Desolvation Temp. 250 °C; Ion Source Temp: 120 °C; Positive Electrospray conditions) using the following methods: Linear Gradient of 0% to100% solvent B over 2 min, with 1 minute hold at 100% B, Linear Gradient of 0% to100% solvent B over 4 min, with 1 minute hold at 100% B; 11841-PCT UV visualization at 220 nm; Column: PHENOMENEX Luna C18 (2) 30mm × 4.6 mm; 5μ particle (heated to Temp. 40 °C); Flow rate: 1.0 mL/min (2 min gradient) or 0.8 ml/min (4 min gradient); Solvent A: 10% ACN, 90% water, 0.1% TFA; or, 10% MeOH, 90% water, 0.1% TFA Solvent B: 90% ACN, 10% water, 0.1% TFA; or, 90% MeOH, 10% water, 0.1% TFA.
[00264] Preparatory HPLC (unless otherwise noted) was performed on a Shimadzu SCL-10A liquid chromatograph with a linear gradient of 20-100% Solvent B over 10 to min, with either a 2 to 5 min hold at 100% Solvent B as determined by on skilled in the art; UV visualization at 220 nm; Column: PHENOMENEX Luna Axia 5μ C18 30 × 100 mm; Flow rate: 20 mL/min; Solvent A: 10% ACN, 90% water, 0.1% TFA; or 10% MeOH, 90% water, 0.1% TFA; Solvent B: 90% ACN, 10% water, 0.1% TFA; or 90% MeOH, 10% water, 0.1% TFA.
Preparatory chiral SFC chromatography (unless otherwise noted) was performed on a Berger Multigram II SFC chromatograph using one of the following methods: Preparative chiral SFC method A: Column: CHIRALCEL OD-H, 30 × 250mm ID, 5 μ Flow rate: 90 mL/min, 100 bar BP, 40 °C Mobile Phase: 15% Methanol /85% CO Detector Wavelength: 254 nm Injection Vol and Sample Solution: 0.5 mL of 4.65 g in 35 mL Methanol (133 mg/mL) Preparative chiral SFC method B: Instrument: Berger SFC MGII (HPW-2501) 11841-PCT Column: CHIRALPAK IA 25 × 3 cm ID, 5 µm Flow rate: 85.0 mL/min Mobile Phase: 85/15/0.1,CO /IPA/DEA, 150 bar Detector Wavelength: 225 nm (Lamda max) Sample Prep and Inj. Volume: 300 µL of ~13 mg / 0.5 mL IPA (~26 mg/mL) Preparative chiral SFC method C Column: CHIRALPAK IA 25 × 3 cm ID, 5 µm Flow rate: 90 mL/min Mobile Phase: 85/15/0.1,CO /MeOH/DEA, 150 bar Detector Wavelength: 270 nm (Lambda max) Sample Prep and Inj. Volume: 300 µL of ~90 mg / 2 mL MeOH (~45 mg/mL) Preparative chiral SFC method D Flow rate: 40 mL/min, 100 Bar, 35 °C Mobile Phase: 20% Methanol/80% CO Detector Wavelength: 224 nm (Lambda max) Injection Volume: 300 µL Sample Preparation: 10 mg dissolved in 0.5 mL MeCN (20 mg/mL); 17 mg dissolved in 0.5 mL MeCN (34 mg/mL)
[00266] Analytical chiral SFC chromatography (unless otherwise noted) was performed on an Aurora Analytical SFC or Berger Analytical SFC using one of the following methods: Analytical chiral SFC method A: Column: CHIRALCEL OD-H, 4.6 × 250mm ID, 5 μm Flow rate: 3.0 mL/min, 100 bar BP, 35 °C.
Mobile Phase: 15% Methanol/85% CO Detector Wavelength: 220 nm Sample Solution: 1 mg/mL in methanol (concentrated/reconstituted) Injection Volume: 10 μL Analytical chiral SFC method B: Column: CHIRALPAK IA 250 × 4.6 mm ID, 5µm Flow rate: 2.0 mL/min 11841-PCT Mobile Phase: 85/15/0.1, CO /IPA/DEA, 150 bar Detector Wavelength: 225 nm (Lamda max) Injection Volume: 10 µL Analytical chiral SFC method C: Column: CHIRALPAK IA 250 × 4.6 mm ID, 5 µm Flow rate: 3.0 mL/min Mobile Phase: 65/35/0.1, CO /MeOH/DEA, 150 bar Detector Wavelength: 270 nm (Lambda max) Injection Volume: 10 µL Analytical chiral SFC method D: Column: CHIRALCEL OD, 250 × 4.6 mm ID, 10 µm Flow rate: 2.0 mL/min, 100 bar, 35 °C Mobile Phase: 20% Methanol/80% CO Detector Wavelength: 223 nm Injection Volume: 10 µL NMR EMPLOYED IN CHARACTERIZATION OF EXAMPLES H NMR spectra (unless otherwise noted) were obtained with JEOL or Bruker FOURIER transform spectrometers operating at 400 MHz or 500 MHz. H-nOe experiments were performed in some cases for regiochemistry elucidation with a 400 MHz Bruker FOURIER Transform spectrometer.
Spectral data are reported as chemical shift (multiplicity, number of hydrogens, coupling constants in Hz) and are reported in ppm (δ units) relative to either an internal standard (tetramethyl silane = 0 ppm) for H NMR spectra, or are referenced to the residual solvent peak (2.49 ppm for CD SOCD H, 3.30 ppm for CD HOD, 1.94 for CHD CN, 7.26 ppm for CHCl , 5.32 ppm for CDHCl ).
Microwave instrumentation employed in heating reactions.
BIOTAGE Initiator 2.5, maximum power 400 W, reaction volume range 0.2 - 10 mL. Reactions are run in sealed pressure vessels specially manufactured for this instrument.
Example 1. 6-Methyloxophenylp-tolyl-1,2,5,6-tetrahydropyridinecarbonitrile 11841-PCT A solution of heptanone (51 mg, 0.45 mmol), 1-p-tolylethanone (60 mg, 0.45 mmol), ethylcyanoacetate (51 mg, 0.45 mmol) and ammonium acetate (42 mg, 0.55 mmol) in anhydrous DMF (0.6 mL) was heated with stirring at 100 C for 16 h. The reaction was diluted with EtOAc and washed with water. The solvent was removed in vacuo and the product was purified by preparative HPLC (MeOH/H O/TFA) to provide the desired product (8 mg, 6%) as a white solid. LCMS Anal. Calc'd for C H N O 19 24 2 297.41, found [M+H] 297.2. H NMR (500 MHz, CDCl ) δ 0.89 (t, J = 6.87 Hz, 3 H), 1.24 - 1.40 (m, 9 H), 1.51 - 1.71 (m, 2 H) 2.42 (s, 3 H), 2.73 - 2.97 (m, 2 H), 6.09 (s, 1 H), 7.30 (d, J = 7.70 Hz, 2 H), 7.52 (d, J = 7.70 Hz, 2 H).
Example 2. 3-(1H-Tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one Intermediate 2A. 4-(4,4,4-Trifluorobutoxy)benzaldehyde F C O To a solution of 4-hydroxybenzaldehyde (20 g, 164 mmol) and 4,4,4- trifluorobutanol (25 g, 195 mmol) in anhydrous CH Cl (500 mL) at 0 °C under Ar was added a solution of PPh (51.5 g, 196 mmol) in CH Cl (200 mL) over 15 min, and 3 2 2 11841-PCT then DIAD (36.4 g, 180 mmol) in anhydrous CH Cl (150 mL) was added dropwise. The mixture was stirred at 0 °C for 0.5 h. The reaction was warmed to rt and stirred for another 3 h. The solvent was removed in vacuo and the residue was triturated with CH Cl three times to remove insoluble solids. The combined CH Cl washings were 2 2 2 2 concentrated and the residue was purified by silica gel chromatography (330 g silica gel, eluted with EtOAc in hexanes) to provide Intermediate 2A (27 g, 71%) as a light brown oil. LCMS Anal. Calc'd for C H F O 232.20, found [M+H] 233.0. 11 11 3 2 Intermediate 2B. 2,2,2-Trifluoro(4-(4,4,4-trifluorobutoxy)phenyl)ethanol F C O To the solution of Intermediate 2A (26.7 g, 114 mmol) and trimethyl(trifluoromethyl)silane (16.9 g, 119 mmol) in anhydrous DME (112 mL) was added CsF (500 mg, 3.29 mmol). The reaction was stirred at rt for 16 h. To the mixture was added 4 N aq HCl (114 mL) and the reaction was stirred at rt for 2.5 h. The reaction was diluted with EtOAc (300 mL) and washed with water, sat'd aq NaHCO , sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated to provide Intermediate 2B (42.5 g, 122%) as an oil. The crude product was used without further purification.
LCMS Anal. Calc'd for C H F O 302.21, found [M-H] 301.2. 12 12 6 2 Intermediate 2C. 2,2,2-Trifluoro(4-(4,4,4-trifluorobutoxy)phenyl)ethanone F C O To a solution of Intermediate 2B (115 mmol) in anhydrous CH Cl (320 mL) was added Dess-Martin periodinane (50.2 g, 118 mmol) portionwise at 0 °C. The reaction was stirred at 0 °C for 0.5 h then at rt for 3 h. To the reaction was added 100 mL of sat'd aq Na CO and 250 mL of EtOAc. The reaction was stirred for another 2 h. The insoluble material was removed by filtration. The layers were separated. The organic layer was washed with sat'd aq Na2CO3. Additional solids that formed upon standing overnight were removed. The organic solution was washed with sat'd aq NaCl, dried 11841-PCT over anhydrous MgSO , filtered and concentrated to provide a dark brown liquid, which was purified by silica gel chromatography (220 g silica gel, elute with EtOAc in hexanes to provide Intermediate 2C (26 g, 76%) as a colorless oil.
Intermediate 2D. Triphenylphosphonium p-tolylcarbonylylide To a refluxing solution of PPh (6.15 g, 23.47 mmol) in anhydrous THF (220 mL) under argon was added dropwise a solution of 2-bromop-tolylethanone (5 g, 23.47 mmol) in THF (60 mL). The reaction was refluxed for 2.5 h and then cooled to rt.
The precipitate was collected by filtration and rinsed with diethyl ether. The solids were suspended in 1:1 MeOH and H O (500 mL), and then 2 N aq NaOH (55 mL) was added.
The reaction was stirred at rt for 16 h. MeOH was removed in vacuo and the aq solution was extracted with CHCl . The combined organic extracts were washed with sat'd aq NaCl, dried over anhydrous Na SO , filtered and concentrated to provide Intermediate 2D (9 g, 97%) as a white solid. LCMS Anal. Calc'd for C H OP 394.44, found [M+H] 27 23 395.2.
Intermediate 2E. (Z)-4,4,4-Trifluorop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)but enone O CF Intermediate 2D (5.13 g, 13 mmol) and Intermediate 2C (3.90 g, 13 mmol) were suspended in DMSO (15 mL). The reaction was heated to 160 °C for 1000 s under microwave conditions. The reaction was cooled to rt and diluted with EtOAc (60 mL).
The mixture was washed with water and sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated. The crude product was purified by silica gel chromatography 11841-PCT (120 g silica gel, elute with EtOAc in hexanes to provide Intermediate 2E (5.9 g, 98%) as a light brown oil.
Intermediate 2F, isomer 1. (R)Amino-4,4,4-trifluorop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)-butanone O CF Intermediate 2F, isomer 2. (S)Amino-4,4,4-trifluorop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)-butanone O CF To a solution of Intermediate 2E (2.1 g, 5.04 mmol) in DMSO (50 mL) was added 15 N aq NH OH (25 mL). The mixture was stirred in sealed pressure vessel for 2 days. The reaction was diluted with EtOAc (60 mL), washed with water and sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated. The residue was purified by silica gel chromatography column (eluted with EtOAc in hexanes to provide racemic Intermediate 2F (2.2 g, 101%) as a white solid. LCMS Anal. Calc'd for C H F NO 21 21 6 2 433.39, found [M+H] 434.2. Separation of the individual enantiomers of Intermediate 2F was carried out using preparative chiral SFC method A: Racemic Intermediate 2F (2200 mg) provided Intermediate 2F, isomer 1 (817 mg) and Intermediate 2F, isomer 2 (790 mg). Enantiomeric purity determination of Intermediate 2F, isomer 1 and 2 was carried out using analytical SFC method A. Intermediate 2F, isomer 1: RT = 2.2 min, 99% ee.
Intermediate 2F, isomer 2: RT = 2.8 min, 99% ee. X-ray crystal data collected for the camphorsulfonic acid salt of Intermediate 2F, isomer 1 showed the chiral center to have 11841-PCT the R-configuration; therefore, the chiral center for Intermediate 2F, isomer 2 has the S- configuration.
Intermediate 2G. 2-(1H-Tetrazolyl)-N-(1,1,1-trifluorooxop-tolyl(4-(4,4,4- trifluorobutoxy)-phenyl)butanyl)acetamide O CF To a solution of Intermediate 2F (789 mg, 1.82 mmol) in anhydrous THF (9 ml) at 0 °C was added DCC (1.13 g, 5.46 mmol). 2-Tetrazole acetic acid (700 mg, 5.46 mmol) was added dropwise as a suspension in anhydrous THF (8 mL). The reaction was stirred at 0 °C for 1 h and then at rt overnight. The reaction was filtered and solids were rinsed with THF. The filtrate was diluted with EtOAc (40 mL), washed with sat’d Na CO and sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated to 2 3 4 provide Intermediate 2G (1.5 g, 152%) as a reddish brown solid. Intermediate 2G was used in the next step without further purification. LCMS Anal. Calc'd for C H F N O 24 23 6 5 3 543.46, found [M+H] 543.9.
Example 2 To a solution of Intermediate 2G (1.5 g) in EtOH (11 mL) was added piperidine (0.33 mL). The reaction was heated to 78 °C for 16 h in a sealed vial. The reaction was cooled to rt and the solvent was removed in vacuo. The residue was purified by preparative HPLC (MeOH/H O/TFA). Fractions containing the product were dried in vacuo and the product was re-dissolved in MeOH and concentrated again. The oily brown product was re-dissolved in CH Cl (5 mL) and concentrated in vacuo to provide Example 2 (552 mg, 57% over 2 steps) as a reddish foam. LCMS Anal. Calc'd for C H F N O 525.45, found [M+H] 526.2. H NMR (500 MHz, CD OD) δ 7.61 (d, J = 24 21 6 5 2 9.1 Hz, 2H), 7.11 (d, J = 8.0 Hz, 2H), 7.07 - 7.01 (m, 2H), 6.90 (d, J = 8.3 Hz, 2H), 4.10 11841-PCT (t, J = 6.1 Hz, 2H), 3.84 - 3.64 (m, 2H), 2.48 - 2.35 (m, 2H), 2.30 (s, 3H), 2.14 - 2.00 (m, 2H).
Example 2-1. (S)(1H-Tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one Separation of the individual enantiomers of Example 2 was carried out using preparative chiral SFC method C: Racemic Example 2 (89 mg) provided Example 2-1 (21 mg). Enantiomeric purity determination of Example 2-1 was carried out using analytical SFC method C. RT = 6.0 min, 99% ee.
Example 2-1 can be alternatively obtained from Intermediate 2F, isomer 2 using a sequence similar to one used for the conversion of Intermediate 2F to Example 2.
Example 2-2. (R)(1H-Tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one Separation of the individual enantiomers of Example 2 was carried out using preparative chiral SFC method C: Racemic Example 2 (89 mg) provided Example 2-2 (22 mg). Enantiomeric purity determination of Example 2-2 was carried out using analytical SFC method C. RT = 15.1 min, 99% ee.
Example 2-2 can be alternatively obtained from Intermediate 2F, isomer 1 using a sequence similar to one used for the conversion of Intermediate 2F to Example 2. 11841-PCT Example 3. 6-(4-Bromophenyl)oxop-tolyl(trifluoromethyl)-1,2,5,6- tetrahydropyridinecarbonitrile Intermediate 3A. (Z)(4-Bromophenyl)-4,4,4-trifluorop-tolylbutenone Intermediate 2D (1.84 g, 4.74 mmol) and 1-(4-bromophenyl)-2,2,2- trifluoroethanone (1.2 g, 4.74 mmol) were dissolved in THF (8 mL) and heated to 150 °C for 1000 s under microwave conditions. The reaction was cooled to rt and the solvent was removed in vacuo. The residue was purified by silica gel chromatography (24 g silica gel, elute with EtOAc in hexanes) to yield Intermediate 3A (1.51 g, 86%) as a light brown solid. LCMS Anal. Calc'd for C H BrF O 369.18, found [M+H] 371.0. 17 12 3 Intermediate 3B. 3-Amino(4-bromophenyl)-4,4,4-trifluorop-tolylbutanone To a solution of Intermediate 3A (1.53 g, 4.14 mmol) in DMSO (23 mL) was added 15 N aq NH OH (10 mL). The reaction vessel was sealed and the reaction was stirred at rt for 16 h. Additional DMSO (3 mL) and 15 N aq NH OH (1 mL) were added 11841-PCT and the reaction was stirred at rt for 16 h. Addition of DMSO (4 mL) was repeated and the reaction was stirred for another 16 h. The solution was concentrated in vacuo followed by lyophilization to provide Intermediate 3B (1.8 g, 100%) as a brown oil.
LCMS Anal. Calc'd for C H BrF NO 386.21, found [M+H] 388.1. 17 15 3 Intermediate 3C. 2-Cyanoacetyl chloride To a mixture of 2-cyanoacetic acid (893 mg, 10.5 mmol) and DMF (16 µL) in anhydrous CH Cl (20 mL) at 0 C was added dropwise a 2 M solution of oxalyl chloride (6 mL, 12 mmol) in CH Cl . The reaction was stirred at 0 °C for 20 min, and then warmed to rt and stirred for 2 h. The solvent was removed in vacuo to provide Intermediate 3C, which was used in the next reaction without purification.
Example 3
[00287] To a solution of Intermediate 3B (1.6 g, 4.1 mmol) in anhydrous CH Cl (15 mL) at 0 °C was added a solution of Intermediate 3C (1.1 g, 10.5 mmol), pyridine (0.85 mL), and DMAP (20 mg) in anhydrous CH Cl (5 mL),. The reaction was stirred at 0 °C for 20 min, then at rt for 2.5 h. The solvent was removed in vacuo. The crude product was dissolved in EtOAc (30 mL), washed with sat'd aq Na CO and sat'd aq NaCl, dried over anhydrous Na SO , filtered and concentrated to yield a brown solid. The solid was triturated with CH Cl /diethyl ether to provide Example 3 (1.27 g) as an off white solid.
The supernatant was evaporated and the residue was purified by silica gel chromatography (eluted with EtOAc in hexanes) to provide a second batch of Example 3 (0.22 g). The combined yield was 1.49 g (80%). LCMS Anal. Calc'd for C H BrF N O 14 3 2 435.24, found [M+H] 437.0. H NMR (500 MHz, CDCl ) δ 2.42 (s, 3 H), 3.45 - 3.67 (m, 2 H), 7.30 (d, J = 7.70 Hz, 2 H), 7.39 (d, J = 8.25 Hz, 2 H), 7.45 (d, J = 7.70 Hz, 2 H), 7.60 (d, J = 8.80 Hz, 2 H).
Example 4. 6-(4-Hydroxyphenyl)oxop-tolyl(trifluoromethyl)-1,2,5,6-tetrahydro- pyridinecarbonitrile 11841-PCT Example 3 (1.49 g, 3.42 mmol), Pd dba (78 mg, 0.086 mmol), KOH (403 mg, 7.2 mmol) and bis(1,1-dimethylethyl)[2′,4′,6′-tris(1-methylethyl)[1,1′-biphenyl] yl]- phosphine (73 mg, 0.17 mmol) were placed in a microwave vial. The vial was evacuated and back filled with Ar. 1,4-Dioxane (23 mL) and water (13 mL) were added to the vial and the reaction mixture was heated to 140 °C for 15 min under microwave conditions. The reaction was cooled to rt, neutralized with 1 N HCl, diluted with water, and extracted with EtOAc. The combined organic extracts were washed with water and sat'd aq NaCl, dried over anhydrous Na SO , filtered and concentrated. The residue was purified by silica gel chromatography to provide Example 4 (1.14 g, 85%) as a light brown solid. LCMS Anal. Calc'd for C H F N O 372.34, found [M+H] 373.2. H 15 3 2 2 NMR (500 MHz, DMSO-d ) δ 2.30 (s, 3 H), 3.65 (q, J = 18.15 Hz, 2 H), 6.73 (d, J = 8.25 Hz, 2 H), 7.28 (d, J = 8.25 Hz, 2 H), 7.38 (d, J = 8.80 Hz, 2 H), 7.47 (d, J = 8.25 Hz, 2 H), 9.63 (s, 1 H), 9.70 (s, 1 H).
Example 5. 6-(4-Butoxyphenyl)oxop-tolyl(trifluoromethyl)-1,2,5,6- tetrahydropyridinecarbonitrile To a solution of Example 4 (20 mg, 0.054 mmol) and n-butanol (6 mg, 0.081 mmol) in anhydrous CH Cl (0.3 mL) under Ar was added sequentially triphenylphosphine (21 mg, 0.081 mmol) in CH Cl (0.3 mL) and DEAD (14 mg, 0.081 mmol) in CH Cl (0.3 mL). The reaction mixture was stirred at 0 °C under argon for 20 min, then at rt for 2 h. The solvent was evaporated in vacuo and the product was purified 11841-PCT by preparative HPLC (CH CN/H O/TFA) to provide Example 5 (5.6 mg, 24%). LCMS Anal. Calc'd for C H F N O 428.45, found [M+H] 429.3. H NMR (500 MHz, 24 23 3 2 2 CDCl ) δ 0.98 (t, J = 7.42 Hz, 3 H), 1.41 - 1.56 (m, 2 H), 1.73 - 1.84 (m, 2 H), 2.41 (s, 3 H), 3.56 (q, J = 18.15 Hz, 2 H), 3.92-4.03 (m, 2 H), 6.94 (d, J = 8.25 Hz, 3 H), 7.29 (d, J = 8.25 Hz, 2 H), 7.37 (d, J = 8.80 Hz, 2 H), 7.45 (d, J = 7.70 Hz, 2 H).
Example 6. N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoro-methyl)-1,2,5,6-tetrahydropyridinecarboxamide Intermediate 6A. Benzyl 3-(4-(methylamino)phenylamino)oxopropanoate O O CH To a solution of monobenzyl malonate (12.2 g, 63.1 mmol) and DMF (90 µL) in anhydrous CH Cl (100 mL) at 0 °C was added 2 M oxalyl chloride (35 mL, 70 mmol) in CH Cl . The reaction was stirred at 0 °C for 30 min, then at rt for 2.5 h. The solvent was removed in vacuo to provide freshly prepared acid chloride. This was dissolved in anhydrous CH Cl (50 mL) and added dropwise to a solution of 4-methoxyaniline (7.76 g, 63 mmol) in anhydrous CH Cl (50 mL) at 0 °C followed by the addition of pyridine (5.35 mL, 66.2 mmol). The reaction was stirred at 0 °C for 0.5 h, then at rt overnight.
The reaction was washed with water and sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated. The residue was triturated with EtOAc/ CH Cl to yield the first batch of Intermediate 6A as a light brown solid (6.95 g). The supernatant was evaporated and the residue was purified by silica gel chromatography (eluted with EtOAc in hexanes) to provide a second batch of Intermediate 6A as a light brown solid (7.4 g). 11841-PCT The combined yield was 14.4 g (76%). LCMS Anal. Calc'd for C H N O 298.34, 17 18 2 3 found [M+H] 300.2.
Intermediate 6B. 3-(4-Methoxyphenylamino)oxopropanoic acid H C O O N OH To a solution of Intermediate 6A (14.4 g, 4.8 mmol) in 10:1 EtOAc/MeOH (220 mL) was added 10% Pd/C (250 mg). The reaction mixture was stirred vigorously under an atmosphere of hydrogen (40 psi) for 2 h. More 10% Pd/C (250 mg) was added and the reaction was stirred under 50 psi hydrogen for another 1 h. Additional 10% Pd/C (500 mg) was added and the reaction was stirred under 50 psi hydrogen for an additional 1 h. The reaction was filtered through a pad of Celite and the filtrate was concentrated in vacuo to yield Intermediate 6B (11.1 g, 96%) as an off-white solid. LCMS Anal.
Calc'd for C10H11NO4 209.20, found [M+H] 210.1.
Intermediate 6C. N -(4-Methoxyphenyl)-N -(1,1,1-trifluorooxop-tolyl(4-(4,4,4- trifluoro-butoxy)phenyl)-butanyl)malonamide O CF O CH HN O To triphenylphosphine (8.42 g, 32.1 mmol) in anhydrous CH Cl (70 mL) was added Intermediate 6B (2.24 g, 10.7 mmol) in anhydrous CH Cl (30 mL) followed by trichloroacetonitrile (1.86 g, 12.8 mmol). The mixture was stirred at rt for 3 h. The freshly prepared acid chloride was added to a solution of Intermediate 2F (1.13 g, 2.61 mmol) in anhydrous CH Cl (20 mL), followed by the addition of pyridine (1.04 mL, 1.02 g, 12.85 mmol). The reaction was stirred at rt under argon overnight. The reaction was cooled to 0 °C and MeOH (40 mL) was added. The reaction was stirred at 0 °C for 11841-PCT min, then at rt for 30 min. The solvent was removed in vacuo and the crude product was purified by silica gel chromatography (120 g silica gel, eluted with EtOAc in hexanes). Product and triphenylphosphine oxide co-eluted. Fractions containing both were combined and evaporated to dryness. The solids were triturated with hexanes/EtOAc to remove most of the triphenylphosphine oxide. The product was again purified by silica gel chromatography (40 g silica gel, eluted with EtOAc in hexanes) to provide Intermediate 6C (1.03 g, 63%) as a brown oil. LCMS Anal. Calc'd for C H F N O 624.57, found [M+H] 625.3. 31 30 6 2 5 Example 6 To a solution of Intermediate 6C (1.03 g, 1.65 mmol) in MeOH (10 mL) was added piperidine (100 µL, 1.01 mmol). The reaction was stirred at 75 °C for 1 h. The solvent was removed in vacuo and the crude product was purified by silica gel chromatography (120 g silica gel, eluted with EtOAc in hexanes). Mixed fractions from the first column were purified again by silica gel chromatography (40 g silica gel, eluted with EtOAc in hexanes) to provide Example 6 (546 mg, 55%) as a off-white solid.
LCMS Anal. Calc'd for C H F N O 606.56, found [M+H] 607.3. H NMR (500 MHz, 31 28 6 2 4 CD OD) δ 1.97-2.08 (m, 2 H), 2.30 (s, 3 H), 2.32-2.44 (m, 2 H), 3.45-3.67 (m, 2 H), 3.72 (s, 3 H), 4.06 (t, J = 6.05 Hz, 2 H), 6.77 (d, J = 9.35 Hz, 2 H), 6.97 (d, J = 8.80 Hz, 2 H), 7.13-7.24 (m, 4 H), 7.28 (d, J = 8.25 Hz, 2 H), 7.52 (d, J = 8.80 Hz, 2 H).
Example 6-1. (R)-N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoro-methyl)-1,2,5,6-tetrahydropyridinecarboxamide
[00294] Example 6-1 was prepared using a procedure analogous to Example 6 by replacing Intermediate 2F with Intermediate 2F, isomer 1. 11841-PCT Example 6-2. (S)-N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoro-methyl)-1,2,5,6-tetrahydropyridinecarboxamide Example 6-2 was prepared using a procedure analogous to Example 6 by replacing Intermediate 2F with Intermediate 2F, isomer 2.
Example 7. 6-(4-(6-Ethoxypyridinyl)phenyl)oxop-tolyl(trifluoromethyl)- 1,2,5,6-tetrahydropyridinecarbonitrile
[00296] To a solution of Example 3 (20 mg, 0.046 mmol), 6-ethoxypyridinyl boronic acid (11 mg, 0.07 mmol) and tetrakis(triphenylphosphine) palladium(0) (5 mg, 10 mol%) in DMF (0.6 mL) sparged with Ar was added 2 N aq K CO (46 µL, 0.096 mmol).
The vessel was sealed and the reaction heated to 80 °C for 22 h. The reaction was cooled to rt and the product was purified twice by preparative HPLC (CH CN/H O/TFA and CH OH/H O/TFA sequentially) to provide Example 7 (10 mg, 45%) as a light brown solid. LCMS Anal. Calc'd for C H F N O 477.48, found [M+H] 478.3. H NMR (500 27 22 3 3 2 MHz, CD OD) δ 1.40 (t, J = 6.87 Hz, 3 H), 2.40 (s, 3 H), 3.71 - 3.88 (m, 2 H), 4.37 (q, J = 7.15 Hz, 2 H), 6.94 (d, J = 8.25 Hz, 1 H), 7.33 (d, J = 7.70 Hz, 2 H), 7.54 (d, J = 8.25 Hz, 2 H), 7.66 - 7.78 (m, 4 H), 8.05 (dd, J = 8.80, 2.20 Hz, 1 H), 8.42 (d, J = 2.20 Hz, 1 H).
Example 8. (S)(2H-Tetrazolyl)p-tolyl(4-(6,6,6-trifluorohexyloxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one 11841-PCT Intermediate 8A. 4-(6,6,6-Trifluorohexyloxy)benzaldehyde
[00297] To a suspension of 4-hydroxybenzaldehyde (488 mg, 4 mmol) and 6-bromo- 1,1,1-trifluorohexane (657 mg, 3 mmol) in MeCN (10 mL) was added K CO (829 mg, 6.00 mmol). The resulting mixture was reflux overnight. Insoluble material was filtered off and rinsed with MeCN. The combined filtrate was concentrated to afford a white solid. This white solid was partitioned between EtOAc and 1 N NaOH solution. The organic layer was separated, washed with sat’d NH Cl, dried over MgSO , filtered and concentrated to afford Intermediate 8A as a clear liquid. LCMS Anal. Calc’d for C H F O 260.10, found [M+H] 261.0. 13 15 3 2 Intermediate 8B. 2,2,2-Trifluoro(4-(6,6,6-trifluorohexyloxy)phenyl)ethanone Intermediate 8B was prepared using a procedure analogous to Intermediate 2C except that Intermediate 2A was replaced with Intermediate 8A. H NMR (500 MHz, CDCl ) δ 8.06 - 8.02 (m, 2 H), 6.99 - 6.97 (m, 1 H), 4.08 (t, J = 6.2 Hz, 2 H), 2.19 - 2.06 (m, 2 H), 1.92 - 1.82 (m, 2 H), 1.71 - 1.55 (m, 4 H).
Intermediate 8C. (S,E)Methyl-N-(2,2,2-trifluoro(4-(6,6,6- trifluorohexyloxy)phenyl)ethylidene)propanesulfinamide 11841-PCT To a solution of Intermediate 8B (717 mg, 2.184 mmol) and (S) methylpropanesulfinamide (529 mg, 4.37 mmol) in THF (10 mL) was added tetraethoxytitanium (1993 mg, 8.74 mmol) in THF (20 mL). The resulting mixture was reflux for 5 h. TLC (20% EtOAc in hexane) indicated the starting ketone was completely consumed. The solvent was evaporated to afford a yellow oil. This yellow oil was dissolved in EtOAc and then washed with saturated NaHCO (25 mL) and a large amount of white precipitation formed which was removed by filtering through a bed of Celite®.
The white precipitation was rinsed with EtOAc. The combined EtOAc solution was washed again with saturated NaHCO , dried (MgSO ) and concentrated. The crude product was purified by silica gel chromatography (40 g silica gel, eluted with EtOAc in hexanes) to afford Intermediate 8C (620 mg, 66%).
Intermediate 8D. (S)Methyl-N-((S)-1,1,1-trifluorooxop-tolyl(4-(6,6,6- trifluorohexyloxy)phenyl)butanyl)propanesulfinamide To a solution of 1-(p-tolyl)ethanone (609 mg, 4.31 mmol) in THF (10 mL) was cooled to -78 °C and to this solution was added lithium bis(trimethylsilyl)amide (4.31 mL, 4.31 mmol). The resulting mixture was stirred at -78 °C for 20 min and then Intermediate 8C (620 mg, 1.437 mmol) in THF (3 mL) was added dropwise. The resulting mixture was stirred at -78 °C for 1.5 h and then at 0 °C for 1.5 h. The reaction was quenched with NH Cl and concentrated. The crude product was purified by silica gel chromatography (80 g silica gel, eluted with EtOAc in hexanes) to afford Intermediate 8D 11841-PCT (482 mg, 59%) as the slower eluting diastereomer on silica gel column. LCMS Anal.
Calc’d for C H F NO S 565.21, found [M+H] 566.0. 27 33 6 3 Intermediate 8E. (S)Amino-4,4,4-trifluorop-tolyl(4-(6,6,6- trifluorohexyloxy)phenyl)butanone To a solution of Intermediate 8D (482 mg, 0.852 mmol) in MeOH (4 mL) was added 4 M HCl (1 mL, 4.00 mmol) in dioxane. The resulting mixture was stirred at rt for 2 h and then concentrated. The residue was taken up in EtOAc, washed with saturated NaHCO and brine, dried (MgSO ), filtered and concentrated to afford Intermediate (386 mg, 58%) as a colorless oil, which was used for the subsequent reaction without further purification. LCMS Anal. Calc’d for C H F NO 461.18, found [M+H] 461.9. 23 25 6 2 Example 8
[00302] To a solution of Intermediate 8E (140 mg, 0.303 mmol) and 2-(2H-tetrazol yl)acetic acid (117 mg, 0.910 mmol) in THF (3 mL) at 0 °C was added DCC (188 mg, 0.910 mmol). The resulting mixture was stirred overnight. The solvent was evaporated and the crude mixture was taken up in EtOAc. The organic solution was washed with saturated NaHCO , 1 N HCl and brine, dried (MgSO ), filtered and concentrated to afford a brown oil. This oil was dissolved in EtOH (3 mL) and added piperidine (300 µL, 3.03 mmol). The resulting mixture was stirred at 80 °C overnight. The reaction mixture was diluted with MeOH and purified by preparative HPLC (MeCN/H O/TFA) to yield Example 8 (86 mg, 51%) as a solid. LCMS Anal. Calc’d for C26H25F6N5O2 553.19, found [M+H] 554.0. H NMR (500 MHz, CDCl ) δ 7.46 (d, J = 8.8 Hz, 2 H), 7.19 (d, J = 8.0 Hz, 3 H), 7.02 - 6.93 (m, 4 H), 3.99 (t, J = 6.3 Hz, 2 H), 3.68 - 3.56 (m, 2 H), 2.39 (s, 3 H), 2.19 - 2.07 (m, 2 H), 1.89 - 1.79 (m, 2 H), 1.71 - 1.63 (m, 2 H), 1.62 - 1.53 (m, 2 11841-PCT Example 9. 3-(2-Ethyl-2H-tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one To Example 2 (30 mg, 0.057 mmol) in anhydrous CH Cl (0.6 mL) was added iodoethane (9 mg, 0.057 mmol) and triethylamine (24 µL, 0.17 mmol). The reaction was heated to 120 °C for 10 min under microwave conditions. The solvent was evaporated in vacuo and the residue was purified by preparative HPLC (CH CN/H O/TFA) to provide Example 9 (2.3 mg, 7%) as a light brown solid. LCMS Anal. Calc'd for C H F N O 26 25 6 5 2 553.50, found [M+H] 554.3. H NMR (500 MHz, CDCl3) δ 1.51 (t, J = 7.42 Hz, 2 H), 1.98 - 2.13 (m, 2 H), 2.21 - 2.39 (m, 5 H), 3.51 (d, J = 17.60 Hz, 1 H), 3.72 (d, J = 17.60 Hz, 1 H), 4.04 (t, J = 6.05 Hz, 2 H), 4.56 (q, J = 7.52 Hz, 2 H) 6.87 (d, J = 8.25 Hz, 2 H), 6.95 (d, J = 8.80 Hz, 2 H), 7.02 (d, J = 8.25 Hz, 2 H), 7.46 (d, J = 8.80 Hz, 2 H), 7.93 (s, 1 H).
Example 10. 4-p-Tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)(5- (trifluoromethyl)-1,3,4-oxadiazolyl)-5,6-dihydropyridin-2(1H)-one To Example 2 (29 mg, 0.055 mmol) in anhydrous CH Cl (0.3 mL) was added a solution of trifluoroacetic anhydride (23.2 mg, 0.11 mmol) in anhydrous CH Cl (0.2 mL) dropwise. The mixture was stirred at rt for 4 h and then concentrated. The residue was purified by preparative HPLC to provide Example 10 (19 mg, 58%) as a white solid.
LCMS Anal. Calc'd for C H F N O 593.44, found [M+H] 594.2. H NMR (500 MHz, 26 20 9 3 3 11841-PCT CD OD) δ 1.97 - 2.11 (m, 2 H), 2.31 (s, 3 H), 2.34 - 2.44 (m, 2 H), 3.64 - 3.88 (m, 2 H), 4.08 (t, J = 6.05 Hz, 2 H), 6.98 (d, J = 8.25 Hz, 2 H), 7.03 (d, J = 8.80 Hz, 2 H), 7.16 (d, J = 8.25 Hz, 2 H), 7.57 (d, J = 8.80 Hz, 2 H).
Example 11. 6-Methyloxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)-N-(4- (trifluoromethoxy)-phenyl)-1,2,5,6-tetrahydropyridinecarboxamide O CF Intermediate 11A. 1-Bromo(4,4,4-trifluorobutoxy)benzene F C O To a solution of 4-bromophenol (2.2 g, 12.7 mmol) and 4-bromo-1,1,1- trifluorobutane (2.4 g, 12.7 mmol) in anhydrous DMF (15 mL) was added K CO (3.5 g, .4 mmol). The mixture was stirred at rt overnight. The mixture was diluted with EtOAc (120 mL) and the solids were removed by filtration. The filtrate was washed with water and sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated. The residue was purified by silica gel chromatography (40 g silica gel, eluted with EtOAc in hexanes) to provide Intermediate 11A (3.05 g, 85%) as a colorless oil.
Intermediate 11B. (E)-Ethyl 3-(4-(4,4,4-trifluorobutoxy)phenyl)butenoate F C O Intermediate 11A (2.81 g, 9.93 mmol), (E)-ethyl butenoate (1.25 g, 10.9 mmol), palladium(II) acetate (0.11 g, 0.5 mmol), tetraethylammonium chloride (1.65 g, 11841-PCT 9.9 mmol), N-cyclohexyl-N-methylcyclohexanamine (2.91 g, 14.9 mmol) and dimethylacetamide (30 ml) were placed in a oven-dried vial and sparged with argon for 5 min. The vial was sealed and the reaction heated to 110 °C for 6 h. The reaction was cooled to rt, diluted with EtOAc (75 mL), washed with water and sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated. The residue was purified by silica gel chromatography eluting with EtOAc in hexanes to provide Intermediate 11B (1.81 g, 49%) as a colorless oil. LCMS Anal. Calc'd for C H F O 316.32, found [M+H] 317.2. 16 19 3 3 Intermediate 11C. (E)-N-Methoxy-N-methyl(4-(4,4,4-trifluorobutoxy)phenyl)but enamide N CH F C O A solution of Intermediate 11B (2.67 g, 8.44 mmol) and N,O- dimethylhydroxylamine hydrochloride (1.65 g, 16.9 mmol) in anhydrous THF (35 mL) was cooled to -61°C (CHCl /dry ice) under an atmosphere of Ar. To this solution was added 0.5 M isopropylmagnesium chloride (16.9 mL, 33.8 mmol) in THF slowly via a syringe. The reaction was stirred at -61 °C for 1.5 h, warmed to -20 °C (sat'd aq NaCl/ice) and stirred for 40 min, then warmed to 0 °C and stirred for 20 min. The reaction was poured into 10 mL of sat'd aq NH Cl and 12 mL of water, and then extracted with EtOAc. The organic layer was washed with sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated. The residue was purified by silica gel chromatography eluting with EtOAc in hexanes to provide Intermediate 11C (1.62 g, 58%) as a light brown oil. LCMS Anal. Calc'd for C H F NO 331.33, found [M+H] 332.2. 16 20 3 3 Intermediate 11D. (E)p-Tolyl(4-(4,4,4-trifluorobutoxy)phenyl)butenone O CF 11841-PCT To a solution of Intermediate11C (1.62 g, 4.89 mmol) in anhydrous THF (25 mL) at -78 °C was added dropwise 0.5 M p-tolylmagnesium bromide in ether (25 mL, 12.5 mmol). The reaction was stirred at -78 °C for 40 min, then gradually warmed to rt.
The reaction was poured into 1:1 sat'd aq NH Cl and water (60 mL). The mixture was extracted with EtOAc. The organic layer was washed with sat'd aq NaCl, dried over MgSO , filtered and concentrated. The residue was purified by silica gel chromatography (eluted with EtOAc in hexanes) to provide Intermediate 11D (1.34 g, 76%) as a light brown solid. LCMS Anal. Calc'd for C H F O 362.39, found [M+H] 363.2. H NMR 21 21 3 2 (500 MHz, CDCl ) δ 2.01 - 2.11 (m, 2 H), 2.29 - 2.37 (m, 2 H), 2.42 (s, 3 H), 2.58 (s, 3 H), 4.07 (t, J = 6.05 Hz, 2 H), 6.92 (d, J = 8.80 Hz, 2 H), 7.14 (s, 1 H), 7.23 - 7.34 (m, J = 8.25 Hz, 2 H), 7.55 (d, J = 8.80 Hz, 2 H), 7.90 (d, J = 8.25 Hz, 2 H).
Intermediate 11E. 3-Aminop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)butanone H C NH H C O CF
[00309] Ammonia was bubble into a solution of Intermediate 11D (1.34 mg, 0.64 mmol) in EtOH (20 mL) and DMSO (12 mL) for 10 min at 0 °C. The reaction was stirred at rt overnight in a sealed pressure vessel. Analytical HPLC showed reaction to have progressed only ca. 10%. The mixture was cooled to -15 °C, then ammonia was bubbled through for 7 min. The vessel was sealed and the reaction was stirred at rt overnight.
Analytical HPLC showed the reaction to be ca. 25-30% complete. The mixture was diluted with EtOAc (75 mL), washed with water and sat'd aq NaCl, dried over anhydrous MgSO , filtered and concentrated in vacuo. The residue was purified by silica gel chromatography to provide Intermediate11E (281 mg, 20%) as a light brown oil. LCMS Anal. Calc'd for C H F NO 379.42, found [M+H] 380.3. 21 24 3 2 Intermediate 11F. 3-Oxo(4-(trifluoromethoxy)phenylamino)propanoic acid F C O O N OH 11841-PCT By sequential application of the procedures for Intermediates 6A and 6B, 4- trifluoromethoxyaniline (2.3 g, 13 mmol) was converted to Intermediate 11F (2.8 g, 11 mmol), which was isolated as a white solid. LCMS Anal. Calc'd for C H F NO 263.17, 8 3 4 found [M+H] 264.1.
Intermediate 11G. N -(4-Oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)butanyl)- N -(4-(trifluoromethoxy)-phenyl)malonamide O CF O CF HN O To a solution of Intermediate 11F (41.6 mg, 0.158 mmol) in anhydrous CH Cl (0.6 mL) was added PPh (124 mg, 0.474 mmol) followed by dropwise addition 2 2 3 of trichloroacetonitrile (27.4 mg, 0.19 mmol). The reaction was stirred at rt for 3 h. To the freshly prepared acid chloride was added a solution of Intermediate 11E (20 mg, 0.053 mmol) in anhydrous CH Cl (0.3 mL) followed by pyridine (19 μL, 0.237 mmol).
The mixture was stirred at rt overnight. The solvent was removed in vacuo. The product was purified by preparative HPLC (MeOH/H O/TFA) to yield Intermediate 11G (11 mg, 33%) as a brown solid. LCMS Anal. Calc'd for C H F N O 624.57, found [M+H] 31 30 6 2 5 625.4.
Example 11
[00312] To a solution of Intermediate 11G (11 mg, 0.018 mmol) in MeOH (0.8 ml) was added piperidine (15 μL). The reaction was stirred at 75 °C for 1.5 h. The product was isolated by preparative HPLC (CH CN/H O/TFA) to provide Example 11 (5.2 mg, 44%) as a brown solid. LCMS Anal. Calc'd for C H F N O 606.56, found [M+H] 31 28 6 2 4 607.4. H NMR (500 MHz, CD OD) δ 1.68 (s, 3 H), 1.95 - 2.07 (m, 2 H), 2.27 (s, 3 H), 2.30 - 2.41 (m, 2 H), 3.11 - 3.26 (m, 2 H), 4.03 (t, J = 6.05 Hz, 2 H), 6.91 (d, J = 8.80 Hz, 11841-PCT 2 H), 7.08 - 7.18 (m, 4 H), 7.22 (d, J = 8.25 Hz, 2 H), 7.38 (d, J = 8.80 Hz, 2 H), 7.45 (d, J = 8.80 Hz, 2 H).
Example 12. 3-(2H-Tetrazolyl)p-tolyl(trifluoromethyl)(1-(5,5,5- trifluoropentyl)-1H-pyrazolyl)-5,6-dihydropyridin-2(1H)-one Intermediate 12A. 4-Iodo(5,5,5-trifluoropentyl)-1H-pyrazole
[00313] To a stirred solution of 4-iodo-1H-pyrazole (337 mg, 1.737 mmol) in DMF (10 mL) was added sodium hydride (104 mg, 2.61 mmol). After 30 min, 5-bromo-1,1,1- trifluoropentane (427 mg, 2.085 mmol) was added. The reaction was stirred at rt for 2 h. 3:1 hexane:ether and water were added. The organic layer was washed with H2O, dried over MgSO , filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (24 g silica gel, eluted with 0-60% EtOAc in hexanes) to give the desired product (460 mg, 83%) as clear oil. LCMS Anal. Calc’d for C H F IN 8 10 3 2 318.0, found [M+H] 319.0. H NMR (400 MHz, CDCl ) δ 7.49 (s, 1 H), 7.40 (s, 1 H), 4.12 (t, J = 6.9 Hz, 2 H), 2.00 - 2.15 (m, 2 H), 1.85 - 1.96 (m, 2 H), 1.47 - 1.61 (m, 2 H).
Intermediate 12B. 2,2,2-Trifluoro(1-(5,5,5-trifluoropentyl)-1H-pyrazolyl)ethanone To a stirred solution of Intermediate 12A (460 mg, 1.446 mmol) in tetrahydrofuran (5 mL) at 0 °C was added isopropylmagnesium chloride (0.795 mL, 1.591 mmol) quickly. After 30 min, additional 0.25 eq of iPrMgCl was added and after 11841-PCT 30min, the mixure was cooled to -78 °C. 2,2,2-Trifluoro(piperidinyl)ethanone (288 mg, 1.591 mmol) was added quickly and the reaction was warmed to rt and stirred for 3 h. The reaction was quenched with sat’d aq NH Cl and diluted with EtOAc. The organic layer was washed with sat’d aq NH Cl, dried over MgSO , filtered and concentrated in vacuo. The crude product was purified by silica gel chromatography (40 g silica gel, eluted with 0-100% EtOAc in hexanes) to give the desired product (265 mg, 64%) as clear oil. H NMR (500 MHz, CDCl ) δ 8.08 (s, 2 H), 4.22 (t, J = 7.0 Hz, 2 H), 2.07 - 2.20 (m, 2 H), 1.98 - 2.05 (m, 2 H), 1.55 - 1.65 (m, 2 H).
Example 12 Example 12 was prepared using a procedure analogous to Example 2 by replacing Intermediate 2C with Intermediate 12B. LCMS Anal. Calc’d for C22H21F6N7O 513.2, found [M+H] 514.3. H NMR (500 MHz, CDCl ) δ 7.75 (s, 1 H), 7.63 (s, 1 H), 7.54 - 7.59 (m, 1 H), 7.06 (d, J = 8.0 Hz, 2 H), 6.88 (d, J = 8.0 Hz, 2 H), 4.16 (t, J = 7.0 Hz, 2 H), 3.58 (d, J = 18.2 Hz, 1 H), 3.39 (d, J = 17.9 Hz, 1 H), 2.28 (s, 3 H), 2.02 - 2.17 (m, 2 H), 1.93 (quin, J = 7.4 Hz, 2 H), 1.46 - 1.55 (m, 2 H).
Example 13. 3-Nitrop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)- ,6-dihydropyridin-2(1H)-one Intermediate 13A. 2-Nitro-N-(1,1,1-trifluorooxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)butanyl)-acetamide 11841-PCT O CF To a solution of Intermediate 2F (130 mg, 0.3 mmol) in anhydrous THF (1 mL) at 0 °C was added DCC (204 mg, 0.99 mmol) followed by a solution of 2-nitroacetic acid (104 mg, 0.99 mmol) in anhydrous THF (0.5 mL) dropwise. The reaction was stirred at 0 °C for 1 h then at rt overnight. The reaction was heated to 70 °C for 4 h. The reaction was cooled to rt and diluted with EtOAc (3 mL). The solids were filtered off and the filtrate was concentrated in vacuo. The residue was purified by silica gel chromatography (eluted with EtOAc in hexanes) to provide Intermediate 13A (129 mg, 83%) as a brown oil. LCMS Anal. Calc'd for C H F N O 520.42, found [M+H] 521.1. 23 22 6 2 5 Example 13 To a solution of Intermediate 13A (126 mg, 0.24 mmol) in MeOH (2 mL) was added piperidine (35 µL). The reaction mixture was heated at 75 °C for 1.5 h. The product was isolated by preparative HPLC (MeOH/H O/TFA) to provide Example 13 (21 mg, 17%) as a off-white solid. LCMS Anal. Calc'd for C H F N O 502.41, found 23 20 3 2 4 [M+H] 503.1. H NMR (500 MHz, CD OD) δ 1.98 - 2.08 (m, 2 H), 2.28 - 2.45 (m, 5 H), 3.58 (d, J = 17.60 Hz, 1 H), 3.76 (d, J = 17.60 Hz, 1 H), 4.08 (t, J = 6.05 Hz, 2 H), 7.02 (d, J = 8.80 Hz, 2 H), 7.12 - 7.19 (m, 2 H), 7.22 - 7.30 (m, 2 H), 7.53 (d, J = 8.80 Hz, 2 Example 14. 8-(4-Methoxyphenyl)oxo(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoro-methyl)azaspiro[2.5]octenecarbonitrile 11841-PCT Intermediate 14A. Methyl 3-oxo(4-(4,4,4-trifluorobutoxy)phenyl)propanoate
[00318] To a solution of methyl 3-(4-hydroxyphenyl)oxopropanoate (7.8 g, 40.2 mmol) and 4,4,4-trifluorobutanol (5.15 g, 40.2 mmol) in CH Cl (201 mL) at 0 °C was added triphenylphosphine (12.6 g, 48.2 mmol). The mixture was stirred for a few min, and then DIAD (9.37 mL, 48.2 mmol) was added. The reaction was stirred at 0 °C for 30 min, then warmed to rt and stirred for 3 days. The reaction was loaded directly onto a 300 g silica gel column and eluted with EtOAc in hexanes. Fractions containing the product were combined and concentrated to provide Intermediate 14A (9.9 g, 80%) as a colorless solid. H NMR (500 MHz, CDCl ) δ 2.03 - 2.13 (m, 2 H), 2.26 - 2.39 (m, 2 H), 3.75 (s, 3 H), 3.96 (s, 2 H), 4.09 (t, J = 6.05 Hz, 2 H), 6.92 - 6.96 (m, 2 H), 7.86 - 7.97 (m, 2 H).
Intermediate 14B. Methyl 1-(4-(4,4,4-trifluorobutoxy)benzoyl)cyclopropanecarboxylate To a solution of Intermediate 14A (6.0 g, 19.7 mmol) in DMF (197 mL) was added K CO (8.18 g, 59.2 mmol) followed by 1,2-dibromoethane (2.6 mL, 30 mmol).
The reaction mixture was stirred at rt for 2 days. The reaction was reduced in volume in vacuo, then diluted with EtOAc, washed with a 1:1 solution of sat'd aq NaCl and water, 11841-PCT dried over anhydrous MgSO , filtered and concentrated. The product was isolated by silica gel chromatography (220 g silica gel, eluted with EtOAc in hexanes) to provide Intermediate 14B (4.2 g, 61%) as a colorless oil. H NMR (500 MHz, CDCl ) δ 1.45 - 1.49 (m, 2 H), 1.56 - 1.61 (m, 2 H), 2.03 - 2.14 (m, 2 H), 2.26 - 2.39 (m, 2 H), 3.60 (s, 3 H), 4.09 (t, J = 6.05 Hz, 2 H), 6.90 - 6.94 (m, 2 H), 7.86 - 7.93 (m, 2 H).
Intermediate 14C. Methyl 1-((benzylimino)(4-(4,4,4- trifluorobutoxy)phenyl)methyl)cyclopropane carboxylate
[00320] To a solution of Intermediate 14B (4.2 g, 13 mmol) and benzylamine (5.5 mL, 50 mmol) in diethyl ether (63 mL) at 0 °C was added 1.0 M TiCl in CH Cl (7.6 mL, 7.5 4 2 2 mmol). The reaction was stirred at 0 °C for a few minutes, then warmed to rt and stirred for 16 h. Celite was added to the reaction and the solids were removed by filtering through a bed of Celite . The filtrate was concentrated and purified by silica gel chromatography (120 g silica gel, eluted with EtOAc in hexanes) to provide Intermediate 14C (4.6 g, 83%) as a colorless oil. H NMR (500 MHz, CDCl ) δ 7.82 (d, J = 8.8 Hz, 2H), 7.42 - 7.38 (m, 2H), 7.34 (t, J = 7.7 Hz, 2H), 7.27 - 7.24 (m, 1H), 6.87 (d, J = 8.8 Hz, 2H), 4.90 (s, 2H), 4.04 (t, J = 6.0 Hz, 2H), 3.69 (s, 3H), 2.39 - 2.26 (m, 2H), 2.11 - 2.02 (m, 2H), 1.78 - 1.72 (m, 2H), 1.11 (d, J = 3.3 Hz, 2H).
Intermediate 14D. Methyl 1-(1-(benzylamino)-2,2,2-trifluoro(4-(4,4,4- trifluorobutoxy)phenyl)ethyl)-cyclopropane carboxylate 11841-PCT HN CF To a solution of Intermediate 14C (4.6 g, 11 mmol) in acetonitrile (22 mL) and DMF (2.6 mL, 33 mmol) at 0 °C was added neat TFA (1.1 mL, 13.7 mmol) followed by potassium hydrogen fluoride (0.64 g, 8.2 mmol). The reaction was stirred for ca. 5 min, then trimethyl(trifluoromethyl)silane (2.4 mL, 16.4 mmol) was added and the reaction was warmed to rt and stirred for 16 h. Sat'd aqueous NaHCO was added to the reaction and the mixture was extracted with EtOAc. The combined organic extracts were washed with sat'd aq NaCl, dried over Na SO , filtered and concentrated. The residue was purified by silica gel chromatography (220 g silica gel, eluted with EtOAc in hexanes) to provide Intermediate 14D (1.8 g, 31%) as an oil. LCMS Anal. Calc'd for C H F NO 489.45, found [M+H] 490.1. 24 25 6 3 Intermediate 14E. 1-(1-(Benzylamino)-2,2,2-trifluoro(4-(4,4,4- trifluorobutoxy)phenyl)ethyl)-cyclopropane carboxylic acid HN CF To a solution Intermediate 14D (558 mg, 1.140 mmol) in pyridine (5.7 mL) was added lithium iodide (1.53 g, 11.4 mmol). The reaction was heated to reflux overnight. The reaction was cooled to rt and diluted with water and EtOAc. The aqueous layer was made acidic with 1 N HCl and the layers were separated. The organic layer was washed with sat'd aq NaCl, dried over MgSO , filtered and concentrated to provide Intermediate 14E (506 mg, 84%) which was used in the next step without further purification. LCMS Anal. Calc'd for C23H23F6NO3 475.42, found [M+H] 476.2. 11841-PCT Intermediate 14F. 5-Benzyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl) azaspiro[2.3]-hexanone
[00323] To a solution of Intermediate 14E (506 mg, 1.1 mmol) in CH Cl (11 mL) was added oxalyl chloride (0.12 mL, 2.2 mmol) followed by several drops of DMF. The reaction was stirred at rt for 16 h. The solvent was removed in vacuo and the residue was purified by silica gel chromatography (24 g silica gel, eluted with EtOAc in hexanes) to provide Intermediate 14F (389 mg, 72%) as a yellow oil. LCMS Anal. Calc'd for C H F NO 457.41, found [M+H] 458.2. 23 21 6 2 Intermediate 14G. (1-(1-(Benzylamino)-2,2,2-trifluoro(4-(4,4,4- trifluorobutoxy)phenyl)ethyl)-cyclopropyl)-(4-methoxyphenyl)methanone HN CF F CH
[00324] Magnesium turnings (130 mg, 5.4 mmol) were suspended in anhydrous THF (13 mL). 4-Bromoanisole (1 g, 670 μL, 5.4 mmol) was added followed by a few drops of 1,2-dibromoethane. The reaction mixture was stirred at rt for 1.5 h after which most of the magnesium had dissolved. The reaction was warmed to 50 °C for 30 min, then cooled to rt. Final concentration of the Grignard reagent was 0.4 M assuming complete conversion. To a cold solution (-40 °C) of Intermediate 14F (200 mg, 0.44 mmol) in THF (4.4 mL) was added the freshly prepared Grignard reagent. The reaction was warmed from -40 °C to rt, then heated to reflux overnight. The reaction was cooled to room temperature and quenched with sat'd aq NH Cl. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic extracts were washed with sat'd aq NaCl, dried over MgSO , filtered and concentrated. The product was 11841-PCT isolated using preparative HPLC (MeOH/H O/TFA). Fractions containing the product were combined and reduced in volume in vacuo. The resulting aqueous solution was made basic with sat'd aq NaHCO and extracted with EtOAc. The combined organic extracts were washed with sat'd aq NaCl, dried over Na SO , filtered and concentrated to provide Intermediate 14G (6.1 mg, 2.5%). LCMS Anal. Calc'd for C H F NO 565.55, 29 6 3 found [M+H] 566.3.
Intermediate 14H. (1-(1-Amino-2,2,2-trifluoro(4-(4,4,4- trifluorobutoxy)phenyl)ethyl)cyclopropyl)(4-methoxy-phenyl)methanone H N CF F CH To a solution of Intermediate 14G (6 mg, 10.6 µmol) in MeOH (1 mL) containing 4.4% formic acid was added 10% palladium on carbon (2 mg, 2 µmol). The reaction was stirred at rt overnight. The reaction was filtered through Celite . The filtrate was made basic with sat'd aq NaHCO , and then evaporated to remove MeOH.
The aqueous residue was diluted with sat'd aq NaCl and extracted with EtOAc. The combined organic extracts were washed with sat'd aq NaCl, dried over Na SO , filtered and concentrated to provide Intermediate 14H (4 mg, 71%) as a tan glass. LCMS Anal.
Calc'd for C H F NO 475.42, found [M+H] 476.2. 23 23 6 3 Intermediate 14I. 2-Cyano-N-(2,2,2-trifluoro(1-(4-methoxybenzoyl)cyclopropyl) (4-(4,4,4-trifluorobutoxy)-phenyl)ethyl)acetamide HN CF F CH To a solution of 2-cyanoacetic acid (320 mg, 3.8 mmol) in CH2Cl2 (9.1 mL) was added oxalyl chloride (523 mg, 360 μL, 4.1 mmol) followed by a few drops of DMF.
The reaction was stirred at rt for 1 h. Final concentration of 2-cyanoacetyl chloride was 11841-PCT 0.4 M assuming complete conversion. To a solution of Intermediate 14H (4 mg, 8.4 µmol) and pyridine (2.0 µL, 0.025 mmol) in CH Cl (100 µL) at 0 °C was added the freshly prepared 0.4 M 2-cyanoacetyl chloride in CH Cl (0.032 mL, 0.013 mmol). The reaction was warmed to rt and stirred for 16 h. The reaction was quenched with one drop of MeOH, diluted with EtOAc and washed with 1:1 sat'd aq NaCl and water, dried over MgSO , filtered and concentrated to provide Intermediate 14I (7 mg, 153%), which was used in the next step without further purification. LCMS Anal. Calc'd for C H F N O 26 24 6 2 4 542.47, found [M+H] 543.3.
Example 14 To a solution of Intermediate 14I (7 mg, 0.013 mmol) in EtOH (2 mL) was added 25% w/w sodium methoxide (0.015 mL, 0.065 mmol). The reaction was stirred at rt for 1.5 h. The reaction was made acidic with a few drops of 1 N aqueous HCl and then concentrated. The product was isolated by preparative HPLC (MeOH/H OTFA). The fraction containing the product was reduced in volume in vacuo, made basic with sat'd aq NaHCO and extracted with EtOAc. The combined organic extracts were washed with sat'd aq NaCl, dried over MgSO , filtered and concentrated to provide Example 14 (2.9 mg, 42%). LCMS Anal. Calc'd for C H F N O 524.46, found [M+H] 525.2. H NMR 26 22 6 2 3 (500 MHz, CD OD) δ 0.65 (t, J = 8.80 Hz, 2 H), 1.18 - 1.25 (m, 2 H), 1.99 - 2.08 (m, 2 H), 2.31 - 2.43 (m, 2 H), 3.85 (s, 3 H), 4.09 (t, J = 6.05 Hz, 2 H), 7.03 (d, J = 8.80 Hz, 2 H), 7.06 (br. s., 2 H), 7.23 (br. s., 2 H), 7.58 (d, J = 9.35 Hz, 2 H).
Examples 15-1 and Example 15-2. 3-Fluoro(4-methoxyphenyl)oxo(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-1,2,3,6-tetrahydropyridinecarbonitrile To a solution of the Example 77 (70 mg, 0.140 mmol) in DMF (1.4 mL) was added Na CO (30 mg, 0.28 mmol) followed by 1-fluorohydroxy-1,4- 11841-PCT diazoniabicyclo[2,2,2]octane-bis-(tetrafluoroborate) (196 mg, 0.28 mmol). The reaction was heated to 80 °C overnight. The reaction was cooled to rt and diluted with EtOAc and water. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with sat'd aq NaCl, dried over MgSO , filtered and concentrated in vacuo. The residue was purified by preparative HPLC (MeOH/H OTFA) to yield the product (44 mg, 61%) as a mixture of two diastereoisomers. The two diastereoisomers were separated by preparative chiral SFC method B to provide Example -1 (14.6 mg, 20%) and Example 15-2 (17.2 mg, 23%). Data for Example 15-1: LCMS Anal. Calc'd for C H F N O 516.41, found [M+H] 517.2. H NMR (500 MHz, 24 19 7 2 3 CD OD) δ 2.00 - 2.08 (m, 2 H), 2.32 - 2.44 (m, 2 H), 3.85 (s, 3 H), 4.10 (t, J = 6.05 Hz, 2 H), 6.92 (d, J = 1.10 Hz, 1 H), 7.02 (d, J = 8.80 Hz, 2 H), 7.07 (d, J = 9.35 Hz, 2 H), 7.50 (d, J = 8.25 Hz, 2 H), 7.57 (d, J = 8.80 Hz, 2 H). Analytical chiral HPLC method B: RT = 3.23 min, 99% ee. Data for Example 15-2: LCMS Anal. Calc'd for C H F N O 24 19 7 2 3 516.41, found [M+H] 517.2. H NMR (500 MHz, CD OD) δ 2.00 - 2.08 (m, 2 H), 2.32 - 2.44 (m, 2 H), 3.85 (s, 3 H), 4.10 (t, J = 6.05 Hz, 2 H), 6.92 (d, J = 1.10 Hz, 1 H), 7.02 (d, J = 8.80 Hz, 2 H), 7.07 (d, J = 9.35 Hz, 2 H), 7.50 (d, J = 8.25 Hz, 2 H), 7.57 (d, J = 8.80 Hz, 2 H). Analytical chiral HPLC method B: RT = 4.84 min, 99% ee.
Examples 15-3 and Example 15-4. 3-Fluoro(4-methoxyphenyl)oxo(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-1,2,3,6-tetrahydropyridinecarbonitrile Application of the method described for Example 15-1 and Example 15-2 provided Example 15-3 (4.6 mg, 43%) and Example 15-4 (4.6 mg, 43%) from the R- isomer of Example 65 (10 mg, 0.02 mmol). Data for Example 15-3: LCMS Anal. Calc'd for C H F N O 516.41, found [M+H] 517.1. H NMR (500 MHz, CD OD) δ 2.00 - 24 19 7 2 3 3 2.08 (m, 2 H) 2.31 - 2.44 (m, 2 H) 3.85 (s, 3 H) 4.10 (t, J = 6.05 Hz, 2 H) 6.92 (d, J = 1.65 Hz, 1 H) 7.02 (d, J = 8.80 Hz, 2 H) 7.07 (d, J = 8.80 Hz, 2 H) 7.50 (d, J = 8.25 Hz, 11841-PCT 2 H) 7.57 (d, J = 8.80 Hz, 2 H). Analytical chiral HPLC method B: RT = 5.13 min, 99% ee. Data for Example 15-4: LCMS Anal. Calc'd for C H F N O 516.41, found [M+H] 24 19 7 2 3 517.1. H NMR (500 MHz, CD OD) δ 1.99 - 2.07 (m, 2 H) 2.30 - 2.43 (m, 2 H) 3.84 (s, 3 H) 4.09 (t, J = 6.05 Hz, 2 H) 6.78 (s, 1 H) 7.02 (d, J = 8.80 Hz, 2 H) 7.05 (d, J = 9.35 Hz, 2 H) 7.54 (dd, J = 8.25, 4.95 Hz, 4 H). Analytical chiral HPLC method B: RT = 5.13 min, 99% ee.
Example 16-1 and Example 16-2. N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)piperidinecarboxamide O CH F C H To a solution of Example 6-2 (90 mg, 0.15 mol) in MeOH (2 mL) was added % palladium on carbon (5 mg). The mixture was stirred under hydrogen (50 psi) overnight. Additional palladium on carbon (5 mg) was added and the reaction was stirred under hydrogen (50 psi) for an additional 1 h. The catalyst was removed by filtration through a pad of Celite and the solution was concentrated in vacuo. The products were partly separated by preparative HPLC (CH CN/H O/TFA). Fractions containing the two products were combined and separated by chiral HPLC method D to provide Example 16-1 and Example 16-2. Data for Example 16-1: LCMS Anal. Calc'd for C H F N O 31 30 6 2 4 608.57, found [M+H] 609.2. H NMR (400 MHz, CD OD) δ 1.97 - 2.14 (m, 2 H), 2.25 (s, 3 H), 2.32 - 2.46 (m, 2 H), 2.54 - 2.69 (m, 2 H), 3.17 (td, J = 11.82, 3.85 Hz, 1 H), 3.60 (d, J = 11.54 Hz, 1 H), 3.71 (s, 3 H), 4.10 (t, J = 6.05 Hz, 2 H), 6.76 (d, J = 8.79 Hz, 2 H), 6.99 - 7.12 (m, 6 H), 7.13 - 7.23 (m, 2 H), 7.55 (d, J = 8.79 Hz, 2 H). Analytical chiral HPLC method D RT = 6.75 min, 99% ee. Data for Example 16-2: LCMS Anal.
Calc'd for C31H30F6N2O4 608.57, found [M+H] 609.2. H NMR (400 MHz, CD3OD) δ 7.52 (d, J = 8.8 Hz, 2H), 7.25 - 7.20 (m, 2H), 7.19 - 7.15 (m, 2H), 7.14 - 7.10 (m, 2H), 7.02 - 6.97 (m, 2H), 6.81 - 6.75 (m, 2H), 4.07 (t, J = 6.0 Hz, 2H), 3.94 - 3.84 (m, 1H), 3.73 (s, 3H), 3.60 (d, J = 12.1 Hz, 1H), 2.90 (dd, J = 14.8, 3.3 Hz, 1H), 2.45 - 2.32 (m, 11841-PCT 2H), 2.31 (dd, 1H, overlaps with peak at δ 2.27), 2.27 (s, 3H), 2.08 - 1.98 (m, 2H).
Preparative chiral HPLC method D: RT = 10.9 min, 99% ee.
Examples 17-101 expressed by Formula (IIa), unless noted in the table, may be made by one skilled in the art by appropriate application of the procedures described 11 15 for Examples 1-16. R to R are hydrogen, unless noted in the table. 11841-PCT INFORMAL Table 2 12 14 11 15 (IIa) 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 17 CH CN 459.2* R = morpholine 18 CH CN 345.1* R = CH R = CH 19 CH CN 377.2* R = OCH CH 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R CH CN 401.2* R = t-Bu N 13 21 CH 360.2 N R = CH 22 CF CN 371.2 R = CH 23 CH CN 331.3 R = CH 24 CF CN 391.1 R = CH CF CN 375.2 R = CH 26 CF CN 393.2 R = CH 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 27 CF CN 375.2 R = CH 28 CF CN 413.3 R = CH 29 CF CN 387.2 R = CH CF CN 387.2 R = CH 31 CF CN 400.2 R = CH 32 CF CN 435.2 R = CH 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 33 CF CN 483.3 2.03 - 2.12 (m, 2 H) 2.25 - R = CH Rac 2.36 (m, 2 H) 2.42 (s, 3 H) F C O 3.51 - 3.68 (m, 2 H) 4.04 (t, J = 5.50 Hz, 2 H) 6.94 (d, J = 8.80 Hz, 2 H) 7.31 (d, J = 7.70 Hz, 2 H) 7.40 (d, J = 8.80 Hz, 2 H) 7.47 (d, J = 7.70 Hz, 2 H) 7.64 (br. s., 1 34 CF CN 471.4 R = CH Rac [M-H]- CF CN 491.3 2.06 - 2.18 (m, 2 H) 2.41 (s, 3 R = CH Rac H) 2.81 (t, J = 7.70 Hz, 2 H) 3.45 - 3.68 (m, 2 H) 3.97 (t, J = 6.32 Hz, 2 H) 6.55 (s, 1 H) 6.93 (d, J = 8.80 Hz, 2 H) 7.21 (d, J = 7.15 Hz, 3 H) 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 7.25 - 7.31 (m, 4 H) 7.37 (d, J = 8.25 Hz, 2 H) 7.45 (d, J = 8.25 Hz, 2 H). 36 CF CN 469.3 1.16 (dd, J = 12.10, 8.25 Hz, R = CH Rac 2 H) 1.54 (dd, J = 7.42, 4.67 Hz, 2 H) 1.60 - 1.70 (m, 2 H) 1.81 (q, J = 6.60 Hz, 4 H) 1.96 (m, J = 7.70 Hz, 1 H) 2.41 (s, 3 H) 3.45 - 3.64 (m, 2 H) 3.98 (t, J = 6.60 Hz, 2 H) 6.50 (s, 1 H) 6.94 (d, J = 9.35 Hz, 2 H) 7.23 - 7.32 (m, 2 H) 7.37 (d, J = 8.80 Hz, 2 H) 7.44 (d, J = 8.25 Hz, 2 H). 37 CF CN 443.3 R = CH 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 38 CF 430.1 R = CH 39 CF CN 387.2 R = CH S-isomer 40 CF CN 463.2 R = CH F 13 41 CF 499.2 R = CH 42 CF CN 427.4 0.85 - 0.96 (m, 3 H) 1.33 (d, J R = CH Rac = 2.75 Hz, 4 H) 1.47 - 1.69 (m, 4 H) 2.41 (s, 3 H) 2.56 - 2.67 (m, 2 H) 3.45 - 3.66 (m, 2 H) 6.46 (br. s., 1 H) 7.20 - 7.31 (m, 4 H) 7.37 (d, J = 7.15 Hz, 2 H) 7.45 (d, J = 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 6.60 Hz, 2 H). 43 CF CN 434.3 R = CH 44 CF CN 450.3 R = CH 45 CF CN 449.3 R = CH 46 CF CN 451.3 R = CH 47 CF CN 463.3 R = CH F 13 48 CF 561.4 2.36 (s, 3 H) 3.40 - 3.59 (m, 2 R = CH Rac H) 6.82 (s, 1 H) 6.93 (t, J = 8.79 Hz, 2 H) 7.01 - 7.13 (m, 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R H) 7.15 - 7.20 (m, 3 H) 7.34 - 7.45 (m, 4 H) 7.48 (d, J = 8.79 Hz, 2 H) 9.86 (s, 1 H).
F 13 49 CF 547.4 2.30 (s, 3 H) 3.57 - 3.81 (m, 2 R = CH Rac H) 6.96 (t, J = 8.79 Hz, 2 H) 7.17 (d, J = 7.70 Hz, 2 H) 7.27 - 7.38 (m, 4 H) 7.83 (m, 4 H) 9.11 (s, 2 H) 9.16 (s, 1 F 13 50 CF 547.4 R = CH 51 CF 539.5 0.90 (t, J = 6.87 Hz, 3 H) R = CH Rac 1.28 - 1.40 (m, 4 H) 1.58 - 1.71 (m, 2 H) 2.35 (s, 3 H) 2.55 - 2.71 (m, 2 H) 3.30 - 3.60 (m, 2 H) 6.63 (s, 1 H) 6.92 (t, J = 8.52 Hz, 2 H) 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 7.05 - 7.12 (m, 2 H) 7.14 - 7.20 (m, 2 H) 7.22 - 7.31 (m, 2 H) 7.37 - 7.49 (m, 4 H) 9.94 (s, 1 H).
F 13 52 CF 563.4 R = CH 53 CF CN 483.2 1.93 - 2.09 (m, 2 H) 2.27 - R = CH S-isomer 2.38 (m, 2 H) 2.40 (s, 3 H) F C O 3.62 - 3.78 (m, 2 H) 4.06 (t, J = 6.05 Hz, 2 H) 7.00 (d, J = 8.80 Hz, 2 H) 7.33 (d, J = 7.70 Hz, 2 H) 7.44 - 7.58 (m, 4 H).
F 13 54 CF 575.3 R = CH 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 55 CF 571.4 O R = CH F C O 56 CF O 591.3 R = CH Rac N F C O 57 CF 605.4 2.02 (dd, J = 9.90, 6.05 Hz, 2 R = CH Rac H) 2.28 - 2.40 (m, 5 H) 2.45 - F C O 2.58 (m, 2 H) 3.12 - 3.35 (m, 2 H) 3.37 - 3.60 (m, 2 H) 4.05 (t, J = 6.05 Hz, 2 H) 6.91 - 7.01 (m, 4 H) 7.09 - 7.20 (m, 5 H) 7.21 - 7.26 (m, 2 H) 7.50 (d, J = 8.80 Hz, 2 58 CF 607.3 1.96 - 2.07 (m, 2 H) 2.26 - R = CH Rac CH 2.32 (m, 3 H) 2.32 - 2.42 (m, F C O 2 H) 3.45 - 3.67 (m, 2 H) 4.06 (t, J = 6.05 Hz, 2 H) 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 6.61 (dd, J = 8.25, 2.20 Hz, 1 H) 6.83 (d, J = 8.25 Hz, 1 H) 6.98 (d, J = 8.80 Hz, 2 H) 7.03 (s, 1 H) 7.09 (t, J = 8.25 Hz, 1 H) 7.16 (d, J = 8.25 Hz, 2 H) 7.28 (d, J = 8.25 Hz, 2 H) 7.52 (d, J = 8.80 Hz, 2 H). 59 CF 577.3 1.95 - 2.08 (m, 2 H) 2.28 (s, 3 R = CH Rac H) 2.32 - 2.42 (m, 2 H) 3.42 - F C O 3.70 (m, 2 H) 4.05 (t, J = 6.05 Hz, 2 H) 6.97 (d, J = 8.80 Hz, 2 H) 7.00 - 7.06 (m, 1 H) 7.15 (d, J = 8.25 Hz, 2 H) 7.20 (t, J = 7.97 Hz, 2 H) 7.30 (dd, J = 16.77, 7.97 Hz, 4 H) 7.52 (d, J = 8.80 Hz, 2 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 60 CF 611.3 R = CH F C O 61 CF 611.3 R = CH N Cl F C O 62 CF Cl 611.3 R = CH F C O 63 CF CN 469.1 2.40 (s, 3 H) 2.63 - 2.75 (m, 2 R = CH Rac H) 3.65 - 3.77 (m, 2 H) 4.24 (t, J = 6.05 Hz, 2 H) 7.01 (d, J = 9.35 Hz, 2 H) 7.33 (d, J = 7.70 Hz, 2 H) 7.50 (d, J = 8.25 Hz, 2 H) 7.54 (d, J = 8.80 Hz, 2 H). 64 CF CN 499.2 R = OCH F C O 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 65 CF CN 499.2 1.95 - 2.07 (m, 2 H) 2.29 - R = OCH Rac 2.43 (m, 2 H) 3.88 (s, 3 H) F C O 3.92 (d, J = 16.51 Hz, 1 H) 4.06 (t, J = 6.05 Hz, 2 H) 4.55 (d, J = 16.23 Hz, 1 H) 6.96 (d, J = 9.08 Hz, 2 H) 7.01 (d, J = 8.80 Hz, 2 H) 7.52 (d, J = 9.08 Hz, 2 H) 7.97 (d, J = 9.08 Hz, 2 H) 66 CF CN 494.2 R = CN F C O 67 CF CN 487.2 R = F F C O 68 CF CN 503.2 R = Cl F C O 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 69 CF CN 494.2 R = CN F C O 70 CF H 458.2 R = CH F C O 71 CF CN all H 469.2 F C O 72 CF CN 503.1 R = Cl F C O 12 13 73 CF CN 519.2 8.15 (d, J = 1.7 Hz, 1H), 8.01 R and R Rac - 7.95 (m, 2H), 7.92 (d, J = F C O 8.0 Hz, 1H), 7.65 - 7.55 (m, 5H), 7.05 - 7.00 (m, 2H), 4.07 (t, J = 6.1 Hz, 2H), 3.85 (ABq, J = 18.1 Hz, 2H), 2.42 - 2.29 (m, 2H), 2.06 - 1.98 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R (m, 2H) 74 CF CN 487.2 R = F F C O 75 CF CN 537.2 R = CF F C O 76 CF CN 483.2 R = CH F C O 77 CF CN 499.2 1.97 - 2.06 (m, 2 H) 2.27 - R = OCH S-isomer 2.42 (m, 2 H) 3.66 (d, J = F C O 17.90 Hz, 1 H) 3.75 (d, J = 17.90 Hz, 1 H) 3.87 (s, 3 H) 4.06 (t, J = 6.05 Hz, 2 H) 6.96 - 7.02 (m, 2 H) 7.03 - 7.08 (m, 2 H) 7.52 (d, J = 8.80 Hz, 2 H) 7.61 - 7.68 (m, 2 H) 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 78 CF CN 535.1 7.69 - 7.63 (m, 2H), 7.54 (d, J R = OCHF Rac = 9.1 Hz, 2H), 7.31 - 7.25 F C O (m, 2H), 7.01 (m, 2H), 6.96 (t, J = 73.5 Hz, 1 H), 4.07 (t, J = 6.1 Hz, 2H), 3.78 - 3.67 (m, 2H), 2.44 - 2.30 (m, 2H), 2.07 - 1.97 (m, 2H) 79 CF CN 553.1 R = OCF F C O 80 CF CN 513.2 R = OCH CH F C O 81 CF CN R = F 517.2 7.51 (d, J = 9.1 Hz, 2H), 7.36 3 11 Rac 13 (t, J = 8.8 Hz, 1H), 7.04 - R = OCH F C O 6.97 (m, 2H), 6.91 - 6.83 (m, 2H), 4.08 (t, J = 6.1 Hz, 2H), 3.87 (s, 3H), 3.65 (s, 2H), 2.46 - 2.29 (m, 2H), 2.08 - 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 1.98 (m, 2H) 82 CF 591.2 7.52 (d, J = 8.8 Hz, 2H), 7.28 R = CH Rac (d, J = 8.2 Hz, 2H), 7.15 (d, J F C O = 7.7 Hz, 2H), 7.19 (d, J = 8.8 Hz, 2H), 7.02 (d, J = 8.2 Hz, 2H), 6.98 (d, J = 7.7 Hz, 2H), 4.06 (t, J = 6.0 Hz, 2H), 3.64 (d, J = 17.0 Hz, 1H), 3.48 (d, J = 17.0 Hz, 1H), 2.39 - 2.32 (m, 2H), 2.29 (s, 3H), 2.24 (s, 3H), 2.06 - 1.98 (m, 2H). 83 CF O 661.2 7.53 (d, J = 8.8 Hz, 2H), 7.46 R = CH O CF Rac - 7.41 (m, 2H), 7.31 - 7.24 F C O (m, J = 8.2 Hz, 2H), 7.13 (d, J = 8.8 Hz, 2H), 7.17 (d, J = 7.7 Hz, 2H), 7.01 - 6.96 (m, 2H), 4.07 (t, J = 6.0 Hz, 2H), 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 3.65 (d, J = 17.0 Hz, 1H), 3.50 (d, J = 17.0 Hz, 1H), 2.43 - 2.33 (m, 2H), 2.30 (s, 3H), 2.07 - 1.99 (m, 2H). 84 CF O 625.2 7.52 (d, J = 8.8 Hz, 2H), 7.30 R = CH O CH Rac - 7.24 (m, 3H), 7.17 (d, J = F C O 7.7 Hz, 2H), 7.01 - 6.90 (m, 4H), 4.06 (t, J = 6.0 Hz, 2H), 3.82 - 3.77 (s, 3H), 3.64 (d, J = 17.0 Hz, 1H), 3.49 (d, J = 17.0 Hz, 1H), 2.42 - 2.33 (m, 2H), 2.30 (s, 3H), 2.08 - 1.96 (m, 2H). 85 CF 609.2 7.56 - 7.50 (ab quartet, J = R = CH Rac 9.3 Hz, 2H), 7.30 - 7.20 (m, F C O 3H), 7.16 (d, J = 8.2 Hz, 2H), 7.05 (t, J = 8.2 Hz, 1H), 7.01 - 6.95 (m, J = 8.8 Hz, 2H), 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 6.91 (dd, J = 8.2, 1.6 Hz, 1H), 4.07 (t, J = 6.0 Hz, 2H), 3.64 (d, J = 17.6 Hz, 1H), 3.49 (d, J = 17.0 Hz, 1H), 2.43 - 2.32 (m, 2H), 2.30 (s, 3H), 2.16 (s, 3H), 2.08 - 1.97 (m, 2H). 86 CF CF 645.2 7.58 - 7.48 (m, 6H), 7.27 (d, J R = CH Rac = 8.2 Hz, 2H), 7.15 (d, J = F C O 8.2 Hz, 2H), 7.00 - 6.95 (m, 2H), 4.06 (t, J = 6.0 Hz, 2H), 3.65 (d, J = 17.6 Hz, 1H), 3.50 (d, J = 17.0 Hz, 1H), 2.43 - 2.31 (m, 2H), 2.28 (s, 3H), 2.09 - 1.95 (m, 2H). 87 CF 538.2 R = CH F C O S-isomer 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 88 CF O 579.2 2.31 (s, 3 H) 3.45 - 3.69 (m, 2 R = CH O CH Rac H) 3.70 - 3.76 (s, 3 H) 4.56 F C O (q, J = 8.25 Hz, 2 H) 6.78 (d, J = 9.35 Hz, 2 H) 7.07 (d, J = 9.35 Hz, 2 H) 7.13 - 7.22 (m, 4 H) 7.29 (d, J = 8.25 Hz, 2 H) 7.59 (d, J = 8.80 Hz, 2 H). 89 CF CN 455.1 R = CH F C O 90 CF H 498.1 2.28 (s, 3 H) 3.63 - 3.82 (m, 2 R = CH Rac H) 4.58 (q, J = 8.61 Hz, 2 H) F C O 6.88 (d, J = 8.25 Hz, 2 H) 7.10 (dd, J = 12.65, 8.80 Hz, 4 H) 7.65 (d, J = 8.80 Hz, 2 91 CF CN R = F 517.2 7.51 (d, J = 9.1 Hz, 2H), 7.36 3 11 S-isomer 13 (t, J = 8.8 Hz, 1H), 7.03 - R = OCH F C O 6.99 (m, 2H), 6.90 - 6.84 (m, 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 2H), 5.49 (s, 1H), 4.08 (t, J = 6.1 Hz, 2H), 3.87 (s, 3H), 3.65 (s, 2H), 2.46 - 2.28 (m, 2H), 2.10 - 1.97 (m, 2H) 92 CF H 556.3 1.34 (t, J = 6.87 Hz, 3 H) R = OCH CH Rac 1.96 - 2.09 (m, 2 H) 2.26 - F C O 2.43 (m, 2 H) 3.70 (s, 2 H) 3.98 (q, J = 6.78 Hz, 2 H) 4.06 (t, J = 6.05 Hz, 2 H) 6.78 (d, J = 8.80 Hz, 2 H) 6.92 (d, J = 8.80 Hz, 2 H) 7.01 (d, J = 8.80 Hz, 2 H) 7.58 (d, J = 8.80 Hz, 2 H). 93 CF H 578.3 1.96 - 2.12 (m, 2 H) 2.28 - R = OCHF Rac 2.45 (m, 2 H) 3.63 - 3.85 (m, F C O 2 H) 4.07 (t, J = 6.05 Hz, 2 H) 6.95 - 7.08 (m, 7 H) 7.59 (d, J = 8.80 Hz, 2 H). 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 94 CF O 661.3 1.97 - 2.08 (m, 2 H) 2.29 (s, 3 R = CH O CF S-isomer H) 2.32 - 2.40 (m, 2 H) 3.50 F C O (d, J = 17.05 Hz, 1 H) 3.65 (d, J = 17.60 Hz, 1 H) 4.06 (t, J = 6.05 Hz, 2 H) 6.98 (d, J = 8.80 Hz, 2 H) 7.14 (dd, J = 16.77, 8.52 Hz, 4 H) 7.28 (d, J = 8.25 Hz, 2 H) 7.43 (d, J = 9.35 Hz, 2 H) 7.52 (d, J = 8.80 Hz, 2 H). 95 CF H 560.3 1.95 - 2.08 (m, 2 H) 2.28 - R = F S-isomer 13 2.42 (m, 2 H) 3.58 - 3.71 (m, R = OCH F C O 2 H) 3.76 (s, 3 H) 4.06 (t, J = 6.05 Hz, 2 H) 6.59 (dd, J = 12.92, 2.47 Hz, 1 H) 6.69 (dd, J = 8.80, 2.75 Hz, 1 H) 6.91 - 6.96 (m, 1 H) 7.00 (d, J = 8.80 Hz, 2 H) 7.57 (d, J = 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 8.80 Hz, 2 H). 96 CF H 526.3 R = CH F C O 97 CF H All H 512.2 F C O 98 CF H 580.3 7.60 (t, J = 8.2 Hz, 4H), 7.18 R = CF Rac (d, J = 8.2 Hz, 2H), 7.03 (d, J F C O = 8.8 Hz, 2H), 4.08 (t, J = 6.0 Hz, 2H), 3.80 (d, J = 18.1 Hz, 1H), 3.69 (d, J = 18.1 Hz, 1H), 2.45 - 2.31 (m, 2H), 2.11 - 1.93 (m, 2H). 99 CF H 546.2 R = Cl F C O 11841-PCT INFORMAL 1 2 6 11 15 Example [M+H] HNMR (400 MHz, MeOD) R R R R -R 100 CF H 542.3 R = OCH F C O 101 CF H 546.2 R = Cl F C O *Data reported as molecular weight of compound based on electrospray mass spec results 11841-PCT Example 102. N -(4-Methoxyphenyl)methyloxop-tolyl-N -(4,4,4-trifluorobutyl)- 1,2,3,6-tetrahydropyridine-2,5-dicarboxamide Intermediate 102A. (E)-Ethyl 2-methyloxop-tolylbutenoate A solution of Intermediate 2D (3.02 g, 7.65 mmol) and ethyl 2-oxopropanoate (0.74 g, 6.37 mmol) in THF (12 mL) in a 5 mL microwave vial equipped with a magnetic stirrer was heated at 150 °C under microwave conditions for 20 min. The solvent was removed under vacuum and the residue was purified by silica gel chromatography (80 g silica gel, eluted with EtOAc in hexanes) to provide the desired product (1.067 g, 72%) as a yellow oil. LCMS Anal. Calc’d for C H O 232.11, found [M+H] 233.1. H NMR 14 16 3 (500 MHz, CDCl ) δ ppm 7.87 (d, J = 8.25 Hz, 2 H), 7.69 (q, J = 1.56 Hz, 1 H), 7.28 (d, J = 7.98 Hz, 2 H), 4.30 (q, J = 7.15 Hz, 2 H), 2.42 (s, 3 H), 2.16 (d, J = 1.38 Hz, 3 H), 1.36 (t, J = 7.02 Hz, 3 H).
Intermediate 102B. Ethyl 2-aminomethyloxop-tolylbutanoate To a solution of Intermediate 102A (1.067 g, 4.59 mmol) in DMSO (20 mL) under argon was added NH OH (18.04 mL, 271 mmol) and the reaction mixture was stirred at rt overnight. The reaction mixture was diluted with EtOAc (75 mL) and washed sequentially with water (40 mL) and brine (20 mL). The organic phase was dried over MgSO and concentrated in vacuo to give a yellow oil. The oil was purified by silica gel 11841-PCT chromatography (120 g silica gel) to provide the desired product (0.632 g, 55%) as a clear oil. LCMS Anal. Calc’d for C H NO 249.14, found [M+H] 250.1. H NMR (500 14 19 3 MHz, CDCl ) δ 7.83 (d, J = 8.25 Hz, 2 H), 7.24 (d, J = 7.98 Hz, 2 H), 4.14 (dd, J = 7.15, 2.75 Hz, 2 H), 3.65 (d, J = 17.61 Hz, 1 H), 3.20 (d, J = 17.61 Hz, 1 H), 2.40 (s, 3 H), 2.18 - 2.27 (m, 2 H), 1.39 (s, 3 H), 1.19 (t, J = 7.01 Hz, 3 H).
Intermediate 102C. Ethyl 2-(3-(4-methoxyphenylamino)oxopropanamido)methyl- 4-oxop-tolylbutanoate
[00334] To a solution of Intermediate 6B (0.388 g, 1.855 mmol) in DCM (10 mL) under argon was added 1-chloro-N,N,2-trimethylpropenamine (0.293 g, 2.192 mmol) and the reaction mixtrue was stirred at rt for 20 min. A solution of Intermediate 102B (0.4204 g, 1.686 mmol) in DCM (1.000 mL) followed by pyridine (0.409 mL, 5.06 mmol) were added and the reaction mixture was stirred at rt for 2.5 h. The reaction mixture was concentrated to give a dark oil which was dissolved in EtOAc (15 mL) and washed with water (5 mL). The organic phase was dried over MgSO4, filtered, and concentrated. The residue was purified by silica gel chromatography (80 g silica gel) to provide the desired product (0.650 g, 88%) as an orange oil. LCMS Anal. Calc’d for C24H28N2O6 440.19, found [M+H] 441.2. H NMR (500 MHz, CDCl3) δ 7.83 (d, J = 8.25 Hz, 2 H), 7.24 (d, J = 7.98 Hz, 2 H), 4.14 (dd, J = 7.15, 2.75 Hz, 2 H), 3.65 (d, J = 17.61 Hz, 1 H), 3.20 (d, J = 17.61 Hz, 1 H), 2.40 (s, 3 H), 2.18 - 2.27 (m, 2 H), 1.39 (s, 3 H), 1.19 (t, J = 7.01 Hz, 3 H).
Intermediate 102D. 5-(4-Methoxyphenylcarbamoyl)methyloxop-tolyl-1,2,3,6- tetrahydropyridinecarboxylic acid 11841-PCT To a solution of Intermediate 102C (0.033 g, 0.075 mmol) in THF (8 mL) and water (1.600 mL) was added lithium hydroxide monohydrate (3.77 mg, 0.090 mmol) and the reaction mixture was stirred at rt for 1 h. The reaction mixture was acidified with AcOH (5 drops) and diluted with EtOAc (10 mL) and water (3 mL). The phases were separated and the aqueous phase was extracted with EtOAc (10 mL). The organic phases were combined and dried over MgSO , filtered, and concentrated in vacuo to give the desired product (0.0213 g, 72%) as a white solid. LCMS Anal. Calc’d for C H N O 22 22 2 5 394.15, found [M+H] 395.0.
Example 102 To a solution of Intermediate 102D (0.0213 g, 0.054 mmol) in DCM (2 mL) under argon was added EDC (0.014 g, 0.076 mmol), HOBT (9.92 mg, 0.065 mmol), 4,4,4-trifluorobutanamine (8.24 mg, 0.065 mmol), and DIEA (0.019 mL, 0.108 mmol).
The reaction mixture was stirred at rt for 3 days. The reaction mixture was diluted with EtOAc (5 mL) and the solution was washed with water (2 mL) and brine (2 mL). The organic phase was dried over MgSO , filtered, and concentrated in vacuo to give an orange solid which was purified by preparative HPLC (ACN/H O/TFA) to afford the desired product (4.6 mg, 15%) as a pale yellow solid. LCMS Anal. Calc’d for C H F N O 503.2, found [M+H] 504.1. H NMR (500 MHz, CDCl ) δ 8.46 (br. s., 1 26 28 3 3 4 3 H), 7.61 (br. s., 1 H), 7.24 - 7.32 (m, 4 H), 7.16 (d, J = 7.83 Hz, 2 H), 6.79 (d, J = 8.84 Hz, 2 H), 3.75 (s, 3 H), 3.56 (d, J = 17.18 Hz, 1 H), 3.22 - 3.37 (m, 2 H), 3.14 (br. s., 1 H), 2.73 (d, J = 17.18 Hz, 1 H), 2.34 (s, 3 H), 2.00 - 2.15 (m, 2 H), 1.70 - 1.80 (m, 2 H), 1.54 (s, 3 H).
Example 103. N-(4-Cyanophenyl)-5,5-difluorooxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide 11841-PCT Intermediate 103A. Methyl 2,2-difluorooxo(4-(4,4,4- trifluorobutoxy)phenyl)propanoate F C O To a solution of Intermediate 14A (3 g, 9.86 mmol) and selectFluor (10.48 g, 29.6 mmol) in acetonitrile (10 mL) was added 1 M methanolic tetrabutylammonium hydroxide (19.72 mL, 19.72 mmol). The reaction mixture was heated to 82 °C for 10 min under microwave conditions. The reaction was diluted with 1:1 ACN and MeOH and filtered rinsing with 1:1 ACN in MeOH (50 mL). The filtrate was evaporated to dryness and the crude product was purified by silica gel chromatography (80 g silica gel, elute with EtOAc in hexanes) to yield the desired product (2.34 g, 69%) as a clear, colorless oil. H NMR (500 MHz, CDCl ) δ 8.08 (d, J = 9.08 Hz, 2 H), 6.98 (d, J = 9.08 Hz, 2 H), 4.13 (t, J = 6.05 Hz, 2 H), 3.93 (s, 3 H), 2.27 - 2.39 (m, 2 H), 2.07 - 2.15 (m, 2 H).
Intermediate 103B. 2,2-Difluoro(piperidinyl)(4-(4,4,4- trifluorobutoxy)phenyl)propane-1,3-dione F C O
[00338] Piperidine (175 µL, 1.763 mmol) was slowly added to Intermediate 103A (500 mg, 1.470 mmol) at rt. The reaction was stirred at rt for 3 h. The reaction was diluted with CH Cl , loaded onto a 12 g SiO column and eluted with EtOAc in hexanes. 2 2 2 Fractions containing the product were combined and evaporated to dryness to provide the 11841-PCT product (520 mg, 81%) as a clear oil. LCMS Anal. Calc’d for C H F NO 393.14, 18 20 5 3 found [M+H] 394.2. H NMR (500 MHz, CDCl ) δ 8.10 (d, J = 9.1 Hz, 2 H), 6.98 - 6.91 (m, 2 H), 4.10 (t, J = 5.9 Hz, 2 H), 3.61 - 3.56 (m, 2 H), 3.54 - 3.50 (m, 2 H), 2.39 - 2.26 (m, 2 H), 2.14 - 2.05 (m, 2 H), 1.69 - 1.61 (m, 2 H), 1.60 - 1.51 (m, 4 H).
Intermediate 103C. N-(2,2-Difluorooxo(piperidinyl)(4-(4,4,4- trifluorobutoxy)phenyl)propylidene)methylpropanesulfinamide F C O To a solution of Intermediate 103B (1.97 g, 5.01 mmol) and 2-methylpropane- 2-sulfinamide (1.821 g, 15.02 mmol) in anhydrous THF (25.04 mL) was added Ti(OEt) (5.19 mL, 25.04 mmol). The reaction was heated at refluxed temperature overnight. The reaction was cooled to rt, poured into brine, diluted with EtOAc, and stirred for 30 min.
The titanium oxide was removed by filtering through a plug of Celite®. The filtrate layers were separated and the organic layer was washed with brine, dried over Na SO , filtered, and concentrated. The residue was purified by silica gel chromatography (80 g silica gel, eluted with EtOAc in hexanes to yield the desired product (1.62 g, 59%) as a yellow oil. H NMR (500 MHz, CDCl3) δ 7.56 (d, J = 8.80 Hz, 2 H), 6.95 (d, J = 9.08 Hz, 2 H), 4.07 (t, J = 6.05 Hz, 2 H), 3.62 - 3.69 (m, 1 H), 3.52 - 3.58 (m, 1 H), 3.39 (t, J = 5.09 Hz, 2 H), 2.26 - 2.37 (m, 2 H), 2.05 - 2.11 (m, 2 H), 1.53 - 1.70 (m, 7 H), 1.25 (s, 9 H).
Intermediate 103D. 2-Methyl-N-(1,1,1,3,3-pentafluorooxo(piperidinyl)(4- (4,4,4-trifluorobutoxy)phenyl)butanyl)propanesulfinamide F C NH F C O 11841-PCT To a solution of TBAT (3.43 g, 6.36 mmol) in DMF (5.89 mL) was added a solution of Intermediate 103C (1.17 g, 2.356 mmol) in THF (5.89 mL). The solution was cooled to 0 °C, and then 2 M TMSCF (3.53 mL, 7.07 mmol) in THF was added dropwise. The reaction was stirred at 0 °C for 1 h, and then quenched with brine (20 mL) at 0 °C. The mixture was warmed to rt and diluted with water and EtOAc. The layers were separated. The aqueous layer was washed with EtOAc. The combined organic layers were washed with brine, dried over Na SO , filtered and concentrated. The residue was purified by silica gel chromatography (80 g silica gel, eluted with EtOAc in hexanes) to afford the desired product (773 mg, 58%) as a yellow gum. LCMS Anal. Calc’d for C H F N O S 566.18, found [M+H] 567.1. H NMR (500 MHz, CD OD) δ 7.69 (d, J 23 30 8 2 3 3 = 8.53 Hz, 2 H), 6.91 (d, J = 9.08 Hz, 2 H), 4.03 (t, J = 5.91 Hz, 2 H), 3.50 - 3.62 (m, 2 H), 3.34 - 3.45 (m, 2 H), 2.23 - 2.35 (m, 2 H), 1.98 - 2.07 (m, 2 H), 1.48 - 1.69 (m, 6 H), 1.26 (s, 9 H).
Intermediate 103E. N-Benzylmethyl-N-(1,1,1,3,3-pentafluorooxo(piperidin yl)(4-(4,4,4-trifluorobutoxy)phenyl)butanyl)propanesulfinamide F C N F C O A solution of Intermediate 103D (461 mg, 0.814 mmol) in anhydrous DMF (0.5 mL) was added to a suspension of NaH (65 mg, 1.627 mmol) (60% in mineral oil) at 0 °C. After stirring for 5 min, BnBr (0.484 mL, 4.07 mmol) was added. The reaction mixture was warmed to rt and stirred for 1 h. The reaction was diluted with EtOAc, washed with brine, dried over Na2SO4, filtered and concentrated. The residue was purified by silica gel chromatography (4 g silica gel, eluted with EtOAc in hexanes) to yield the desired product (260 mg, 44%) as a yellow gum. H NMR (500 MHz, CDCl ) δ 7.75 (d, J = 8.5 Hz, 2 H), 7.33 - 7.27 (m, 3 H), 7.14 (dd, J = 7.3, 2.1 Hz, 2 H), 6.80 (d, J = 9.1 Hz, 2 H), 4.46 - 4.38 (m, 2 H), 4.00 - 3.95 (m, 2 H), 3.72 (br. s., 1 H), 3.51 (br. s., 1 H), 3.35 - 3.10 (m, 2 H), 2.37 - 2.25 (m, 2 H), 2.09 - 1.99 (m, 2 H), 1.70 - 1.39 (m, 6 H), 1.34 - 1.29 (m, 9 H). 11841-PCT Intermediate 103F. 2-Methyl-N-(1,1,1,3,3-pentafluorooxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)butanyl)propanesulfinamide F C NH F C O
[00342] Mg turnings were suspended in 0.1 N aq HCl for a few minutes, rinsed with water, MeOH and dried under vacuum. A flame dried flask equipped with a stir bar was charged with Mg turnings (0.243 g, 10 mmol), anhydrous THF (4.4 mL) and 4- bromotoluene (1.71 g, 10 mmol) in anhydrous THF (4.4 mL) followed by several drops of 1,2-dibromoethane. The reaction initiated in a few minutes and the mixture became warm. The approximate concentration of the Grignard reagent is 1 M. The mixture was diluted with 10 mL anhydrous THF to produce a clear solution of p-tolylmagnesium bromide (~0.5 M). To a solution of Intermediate 103E (260 mg, 0.396 mmol) in THF (3959 µL) at 0 °C was added the freshly prepared p-tolylmagnesium bromide (3959 µL, 1.980 mmol). The reaction was warmed to rt, concentrated to half of the original volume, and stirred for 3 h. The reaction was cooled to 0 °C, quenched with sat'd aq NH Cl, and then diluted with EtOAc. The layers were separated and the organic layer was washed with brine, dried over MgSO , filtered and concentrated. The residue was purified by silica gel chromatography (24 g silica gel, eluted with EtOAc in hexanes) to afford the desired product (60 mg, 24%) as a yellow gum. H NMR (500 MHz, CDCl ) δ 7.73 (d, J = 8.0 Hz, 2H), 7.55 (d, J = 8.5 Hz, 2H), 7.19 (d, J = 8.0 Hz, 2H), 6.83 (d, J = 8.8 Hz, 2H), 4.19 (s, 1H), 4.01 (br. s., 2H), 2.40 (s, 3H), 2.31 (dd, J = 16.2, 10.2 Hz, 2H), 2.10 - 1.99 (m, 2H), 1.25 (s, 9H).
Intermediate 103G. 3-Amino-2,2,4,4,4-pentafluorop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)butanone, HCl F C NH F C O 11841-PCT To a solution of Intermediate 103F (30 mg, 0.052 mmol) in MeOH (0.5 mL) was added 4.0 M HCl (0.026 mL, 0.105 mmol) in dioxane. The reaction was stirred at rt for 1 h. The reaction was concentrated and the crude product was used in the next step without further purification. LCMS Anal. Calc’d for C H F NO 469.13, found [M+H] 21 19 8 2 470.1.
Intermediate 103H. Ethyl 5,5-difluorooxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)(trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxylate
[00344] Intermediate 103G (268 mg, 0.530 mmol) was dissolved in MeOH and then passed through NaHCO resin (500 mg; 0.9 mmol). The solution was concentrated to yield the free base (230 mg). The free base was dissolved in 9:1 CH Cl /pyridine (3 mL) added ethyl 3-chlorooxopropanoate (160 mg, 1.060 mmol). The resulting mixture was stirred at rt for 24 h and then concentrated. The residue was taken up with EtOH (1 mL) and treated with piperidine (20 uL). The resulting mixture was stirred at 65 °C for 24 h and then concentrated. The residue was purified by preparative HPLC (MeOH/H O/TFA) to yield the desired product (100 mg, 33%) as a foam. LCMS Anal.
Calc’d for C H F NO 565.15, found [M+H] 566.1. 26 23 8 4 Intermediate 103I. 5,5-Difluorooxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxylic acid F OH 11841-PCT To a solution of Intermediate 103H (100 mg, 0.177 mmol) in MeOH (2 mL) was added 2 M LiOH (0.4 mL, 0.800 mmol). The resulting mixture was stirred at 100 °C for 15 min. LC-MS indicated the product to be the major component along with a trace amount of SM and 14% of the corresponding methyl ester. 2 M LiOH (100 uL) was then added and the reaction was stirred at 100 °C for an additional 15 min. The reaction was concentrated, acidified with 1 N HCl, and extracted with EtOAc. The organic layer was washed with brine, dried over MgSO , filtered and concentrated to yield the desired product (75 mg, 78%) as a solid. LCMS Anal. Calc’d for C H F NO 537.12, found 24 19 8 4 [M+H] 537.9.
Example 103 To a solution of Intermediate 103I (20 mg, 0.037 mmol) in MeCN (1 mL) was added 3H-[1,2,3]triazolo[4,5-b]pyridinol (7.60 mg, 0.056 mmol), 4-aminobenzonitrile (6.59 mg, 0.056 mmol), and EDC (10.70 mg, 0.056 mmol). The resulting mixture was stirred at rt overnight. The reaction mixture was diluted with MeCN and purified by preparative HPLC (MeCN/H O/TFA) to yield the desired product (11 mg, 46%) as a solid. LCMS Anal. Calc’d for C H F N O 637.16, found [M+H] 637.9. H NMR (500 31 23 8 3 3 MHz, CD OD) δ 7.67 (d, J = 9.08 Hz, 2 H), 7.56 - 7.63 (m, 4 H), 7.22 - 7.26 (m, 2 H), 7.15 - 7.20 (m, 2 H), 7.04 - 7.09 (m, 2 H), 4.12 (t, J = 6.05 Hz, 2 H), 2.33 - 2.44 (m, 2 H), 2.30 (s, 3 H), 2.02 - 2.09 (m, 2 H).
Example 104. (S)Aminop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one Intermediate 104A. (S)(1,3-Dioxoisoindolinyl)-N-(1,1,1-trifluorooxop-tolyl- 2-(4-(4,4,4-trifluorobutoxy)phenyl)butanyl)acetamide 11841-PCT O CF To a solution of 2-(1,3-dioxoisoindolinyl)acetic acid (592 mg, 2.88 mmol) in dry DCM (10 mL) was added PPh (2269 mg, 8.65 mmol) fairy rapidly, and then CCl CN (500 mg, 3.46 mmol) was added dropwise. The mixture was stirred at rt for 3 h and a solution of Intermediate 2F, isomer 2 (500 mg, 1.15 mmol) in dry DCM (3 mL), followed by pyridine (0.3 mL) were added. The mixture was stirred at rt overnight. The mixture was diluted with DCM (10 mL) and washed with sat’d NaHCO (2 × 8 mL). The organic layer was dried over anhydrous MgSO , filtered and concentrated. The residue was purified by silica gel chromatography to yield the desired product (620 mg, 87%).
LCMS Anal. Calc’d for C H F N O 620.17, found [M+H] 621.3. 31 26 6 2 5 Intermediate 104B. (S)(2-Oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-1,2,5,6-tetrahydropyridinyl)isoindoline-1,3-dione Intermediate 104A (145 mg, 0.234 mmol) was dissolved in MeOH (2.2 mL) and then 1 N NaOH (0.2 mL) was added. The mixture was stirred at 69 °C for 45 min and added another 0.2 mL of 1 N NaOH. The reaction was heated at 130 °C under microwave conditions for 10 min. The mixture was neutralized with 1 N HCl and then concentrated. The residue was purified by silica gel chromatography (12 g silica gel, 11841-PCT eluted with EtOAc in hexanes) to yield the desired product (80 mg, 57%). LCMS Anal.
Calc’d for C H F N O 602.16, found [M+H] 603.3. 31 24 6 2 4 Example 104
[00349] To a solution of Intermediate 104B (80 mg, 0.133 mmol) in 1 mL of EtOH was added 1 mL of 2 N MeNH in MeOH. The mixture was stirred at 67 °C for 24 h.
The solvent was removed in vacuo and the crude product was purified by silica gel chromatography (4 g silics gel, eluted with EtOAc in hexanes) to yield the desired product (32.3 mg, 52%) as a light brown solid. LCMS Anal. Calc’d for C H F N O 23 22 6 2 2 472.16, found [M+H] 473.2. H NMR (500 MHz, CD OD) δ 7.49 (d, J = 8.8 Hz, 2 H), 7.31 - 7.17 (m, 4 H), 7.04 - 6.92 (m, 2 H), 4.08 (s, 2 H), 3.46 (d, J = 16.2 Hz, 1 H), 3.21 (d, J = 16.2 Hz, 1 H), 2.44 - 2.30 (m, 5 H), 2.09 - 1.99 (m, 2 H).
Example 105. (S)Methyl-N-(2-oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-1,2,5,6-tetrahydropyridinyl)benzamide To a solution of 104 (15 mg, 0.032 mmol) in dry DCM (0.5 mL) was added 2- methylbenzoyl chloride (5.4 mg, 0.035 mmol) and pyridine (2.8 μL, 0.035 mmol). The mixture was stirred at rt for 2 h and then concentrated. The residue was purified by preparative HPLC (MeCN/H O/TFA) to yield the desired product. LCMS Anal. Calc’d for C31H28F6N2O3 590.20, found [M+H] 591.3. H NMR (500 MHz, CD3OD) δ 7.56 (d, J = 8.80 Hz, 2 H), 7.11 - 7.29 (m, 8 H), 6.98 (d, J = 9.08 Hz, 2 H), 4.06 (t, J = 6.05 Hz, 2 H), 3.73 (d, J = 17.06 Hz, 1 H), 3.46 (d, J = 16.78 Hz, 1 H), 2.33 (s, 3 H), 2.30 - 2.42 (m, 2 H), 2.22 (s, 3 H), 1.98 - 2.06 (m, 2 H).
Example 106. (S)Phenoxyp-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one 11841-PCT Intermediate 106A. (S)Phenoxy-N-(1,1,1-trifluorooxop-tolyl(4-(4,4,4- trifluorobutoxy)phenyl)butanyl)acetamide O CF To a mixture of triphenylphosphine (138 mg, 0.318 mmol) in dry DCM (0.8 mL) was added 2-phenoxyacetic acid (26 mg, 0.173 mmol), followed by trichloroacetonitrile (30 mg, 0.208 mmol). The mixture was stirred at rt for 2.5 h. To the mixture was added a solution of Intermediate 2F, isomer 2 (30 mg, 0.069 mmol) in dry DCM (0.5 mL) followed by pyridine (17 μL, 0.208 mmol). The reaction was stirred at rt overnight. The mixture was concentrated and purified by preparative HPLC (MeOH/H O/TFA) to yield the desired product (27 mg, 69%) as a brown oil. LCMS Anal. Calc’d for C H F NO 567.18, found [M+H] 568.3. 29 27 6 4 Example 106 To a solution of Intermediate 106A (26 mg, 0.046 mmol) in MeOH (0.5 mL) was added 1 N NaOH (60 μL). The mixture was heated at 130 °C under microwave conditions for 10 min. The mixture was neutralized with 1 N HCl and concentrated. The residue was purified by preparative HPLC (MeCN/H O/TFA) to yield the desired product (1.5 mg, 6%) as a light brown oil. LCMS Anal. Calc’d for C H F NO 549.17, found 29 25 6 3 11841-PCT [M+H] 550.3. H NMR (500 MHz, CD OD) δ 7.59 (d, J = 8.80 Hz, 2 H), 7.33 (d, J = 8.25 Hz, 2 H), 7.13 (d, J = 8.25 Hz, 2 H), 7.01 - 7.08 (m, 4 H), 6.85 - 6.90 (m, 1 H), 6.38 (dd, J = 8.67, 0.96 Hz, 2 H), 4.10 - 4.15 (m, 2 H), 3.71 - 3.76 (m, 1 H), 3.61 - 3.67 (m, 1 H), 2.34 - 2.45 (m, 2 H), 2.30 (s, 3 H), 2.02 - 2.11 (m, 2 H).
Example 107. 6-(4-Butoxyphenyl)methyloxop-tolyl-1,2,5,6-tetrahydropyridine- 3-carbonitrile Intermediate 107A. (Z)-N-(1-(4-Butoxyphenyl)ethylidene)methylpropane sulfinamide To a stirred solution of 1-(4-butoxyphenyl)ethanone (3 g, 15.60 mmol) and 2- methylpropanesulfinamide (2.84 g, 23.41 mmol) in THF (50 mL) was added tetraethoxytitanium (8.90 g, 39.0 mmol). The reaction was heated to 70 °C for 3 d. Cold water was added and the reaction was stirred vigorously for 20 min. The mixture was filtered through Celite® and the filtrated was diluted with EtOAc. The organic solution was washed with H O, brine, dried over MgSO , filtered and concentrated. The crude product was by silica gel chromatography (220 g silica gel, eluted with EtOAc in hexanes) to yield the desired product (4 g, 87%) as a yellow oil. H NMR (500 MHz, CDCl ) δ 7.88 (d, J = 8.80 Hz, 2 H), 6.91 (d, J = 8.80 Hz, 2 H), 4.02 (t, J = 6.46 Hz, 2 H), 2.73 (s, 3 H), 1.75 - 1.83 (m, 2 H), 1.46 - 1.56 (m, 2 H), 1.32 (s, 9 H), 0.99 (t, J = 7.43 Hz, 3 H). 11841-PCT Intermediate 107B. Methyl 3-(4-butoxyphenyl)(1,1- dimethylethylsulfinamido)butanoate O HN To a stirred solution of diisopropylamine (3.86 mL, 27.1 mmol) in THF (40 mL) at -78 °C was added n-butyllithium (22.57 mL, 27.1 mmol) dropwise. The reaction was slowly warmed up to -20 °C and stirred for 45 min. The reaction was cooled to -78 °C and methyl acetate (2.006 g, 27.1 mmol) was added dropwise. After 30 min, chlorotitanium triisopropoxide (8.09 mL, 33.8 mmol) in THF (40 mL) was added dropwise. The reaction was stirred at -78 °C for 1 h. Intermediate 107A (4 g, 13.54 mmol) in THF (15 mL) was added dropwise and the reaction was stirred at -78 °C for 2 h.
The reaction was quenched by addition of sat’d NH Cl solution and stirred vigorously while warmed to rt. The mixture was filtered through Celite® and washed with EtOAc.
The filtrate was washed with H O, brine, dried over MgSO , filtered and concentrated.
The crude product was purified by silica gel chromatography (220 g silica gel, eluted with EtOAc in hexanes) to yield the desired product (4.33 g, 87%) as a light yellow oil.
LCMS Anal. Calc’d for C H NO S 369.20, found [M+H] 370.2. H NMR (500 MHz, 19 31 4 CDCl ) δ 7.31 (d, J = 8.80 Hz, 2 H), 6.85 (d, J = 8.80 Hz, 2 H), 3.95 (t, J = 6.46 Hz, 2 H), 3.62 (s, 3 H), 2.05 (s, 2 H), 1.74 - 1.80 (m, 2 H), 1.74 (s, 3 H), 1.44 - 1.54 (m, 2 H), 1.29 (s, 9 H), 0.98 (t, J = 7.43 Hz, 3 H).
Intermediate 107C. Methyl 3-amino(4-butoxyphenyl)butanoate O NH To a stirred solution of Intermediate 107B (1 g, 2.71 mmol) in MeOH (6 mL) was added 4 N HCl (3.38 mL, 13.53 mmol). The reaction was stirred at rt for 4 h. The reaction mixture was concentrated in vacuo and diluted with EtOAc. The organic layer was washed with saturated NaHCO , dried over MgSO , filtered and concentrated to H NMR (500 MHz, CDCl ) δ yield the desired product (700 mg, 97%) as a yellow oil. 3 11841-PCT 7.38 (d, J = 8.80 Hz, 2 H), 6.86 (d, J = 8.80 Hz, 2 H), 3.95 (t, J = 6.46 Hz, 2 H), 3.59 (s, 3 H), 1.73 - 1.80 (m, 2 H), 1.53 (s, 2 H), 1.45 - 1.52 (m, 2 H), 1.19 (s, 3 H), 0.98 (t, J = 7.29 Hz, 3 H).
Intermediate 107D. Methyl 3-(4-butoxyphenyl)(2-cyanoacetamido)butanoate O HN O To a stirred solution of 2-cyanoacetic acid (128 mg, 1.507 mmol) in CH Cl (5 mL) was added oxalyl chloride (0.754 mL, 1.507 mmol) and 1 drop of DMF. The reaction was stirred at rt for 1 h and then concentrated. The resulting acid chloride was dissolved in CH Cl (1 mL) and was then added to Intermediate 107C (200 mg, 0.754 mmol) and pyridine (0.183 mL, 2.261 mmol)in CH Cl (5 mL). The reaction was stirred at rt for 5 h. The reaction mixture was concentrated and the residue was diluted with EtOAc. The organic layer was washed with saturated NH Cl, dried over MgSO , filtered and concentrated. The crude product was purified by silica gel chromatography (24 g silica gel, eluted with EtOAc in hexanes) to yield the desired product (218 mg, 87%) as a clear oil. H NMR (500 MHz, CDCl ) δ 7.22 (d, J = 8.80 Hz, 2 H), 6.85 (d, J = 8.80 Hz, 2 H), 3.94 (t, J = 6.46 Hz, 2 H), 3.65 (s, 3 H), 3.30 (s, 2 H), 3.03 (d, J = 15.13 Hz, 1 H), 2.85 (d, J = 15.13 Hz, 1 H), 1.80 (s, 3 H), 1.71 - 1.78 (m, 2 H), 1.44 - 1.53 (m, 2 H), 0.97 (t, J = 7.29 Hz, 3 H).
Intermediate 107E. 6-(4-Butoxyphenyl)hydroxymethyloxo-1,2,5,6- tetrahydropyridinecarbonitrile To a stirred solution of Intermediate 107D (218 mg, 0.656 mmol) in MeOH (5 mL) was added 4.37 M NaOMe (0.750 mL, 3.28 mmol) in methanol solution. The reaction was heated to 55 °C for 5 h. The reaction mixture was concentrated and the 11841-PCT residue was diluted with EtOAc. The organic layer was washed with 1 M HCl, dried over MgSO , filtered and concentrated to yield the desired product (200 mg, 102%) as a light yellow solid. LCMS Anal. Calc’d for C H N O 300.15, found [M+H] 301.1. 17 20 2 3 Intermediate 107F. 6-(4-Butoxyphenyl)chloromethyloxo-1,2,5,6- tetrahydropyridinecarbonitrile To a stirred solution of Intermediate 107E (130 mg, 0.433 mmol) in ClCH CH Cl (3 mL) was added POCl (0.056 mL, 0.606 mmol) and Hunig's base (0.113 2 2 3 mL, 0.649 mmol). The reaction was stirred at rt for 30 min and then heated to 85 °C for 3 h. The reaction mixture was concentrated and the residue was diluted with EtOAc. The organic layer was washed with saturated NH Cl, dried over MgSO , filtered and concentrated. The crude product was purified by silica gel chromatography (24 g silica gel, eluted with EtOAc in hexanes) to yiled the desired product (102 mg, 74%) as a white solid. LCMS Anal. Calc’d for C H ClN O 318.11, found [M+H] 319.2. H NMR 17 19 2 2 (500 MHz, CDCl ) δ 7.23 (d, J = 8.80 Hz, 2 H), 6.90 (d, J = 8.80 Hz, 2 H), 3.97 (t, J = 6.60 Hz, 2 H), 2.05 (s, 2 H), 1.74 - 1.82 (m, 2 H), 1.69 (s, 3 H), 1.46 - 1.55 (m, 2 H), 0.99 (t, J = 7.43 Hz, 3 H).
Example 107 To a stirred solution of Intermediate 107F (11 mg, 0.035 mmol), p- tolylboronic acid (5.63 mg, 0.041 mmol), and 1,1'-bis(di-tert-butylphosphino)ferrocene palladium dichloride (2.270 mg, 3.45 µmol) in dioxane (1 mL) was added CsF (10.48 mg, 0.069 mmol). The reaction was degassed for 10 min and heated to 80 °C for 2 h. The reaction mixture was concentrated. The residue was diluted with EtOAc, washed with H O, brine, dried over MgSO , filtered and concentrated. The crude material was purified by preparative HPLC (MeOH/H O/TFA) to yield the desired product (5.7 mg, 44%) as a brown solid. LCMS Anal. Calc’d for C H N O 374.20, found [M+H] 375.2. 24 26 2 2 H NMR (400 MHz, CD OD) δ 7.41 (d, J = 8.14 Hz, 2 H), 7.32 (d, J = 8.80 Hz, 2 H), 11841-PCT 7.28 (d, J = 8.14 Hz, 2 H), 6.89 (d, J = 8.80 Hz, 2 H), 3.96 (t, J = 6.38 Hz, 2 H), 3.46 (d, J = 18.05 Hz, 1 H), 3.24 (d, J = 17.83 Hz, 1 H), 2.38 (s, 3 H), 1.69 - 1.78 (m, 2 H), 1.65 (s, 3 H), 1.43 - 1.55 (m, 2 H), 0.97 (t, J = 7.48 Hz, 3 H).
Example 108. 6-(5-Hexyloxazolyl)methyloxop-tolyl-1,2,5,6- tetrahydropyridinecarbonitrile Intermediate 108A. Ethyl 2-(2-cyanoacetamido)methyloxop-tolylbutanoate To a solution of Intermediate 3C (2.192 g, 21.18 mmol) in DCM (30 mL) under argon was added Intermediate 102C (1.2 g, 4.81 mmol), pyridine (0.779 mL, 9.63 mmol) and DMAP (0.059 g, 0.481 mmol). The reaction mixture was stirred at rt for 1 h.
The reaction mixture was loaded onto silica gel (25 g) and washed with EtOAc (4 × 50 mL). The filtrate was concentrated in vacuo to a dark red oil. The oil was dissolved in EtOAc (100 mL) and washed with water (2 × 50 mL). The organic phase was dried over MgSO and concentrated in vacuo. The residue was purified by silica gel chromatography (80 g silica gel, eluted with EtOAc in hexanes) to afford the desired product (1.28 g, 84%) as yellow oil. LCMS Anal. Calc’d for C H N O 316.1, found 17 20 2 4 [M+H] 317.1. H NMR (500 MHz, CDCl ) δ 7.82 (d, J = 8.0 Hz, 2 H), 7.39 (s, 1 H), 7.24 - 7.29 (m, 2 H), 4.45 (d, J = 18.2 Hz, 1 H), 4.22 - 4.31 (m, 2 H), 3.47 (d, J = 18.2 Hz, 1 H), 3.28 - 3.38 (m, 2 H), 2.42 (s, 3 H), 1.74 (s, 3 H), 1.24 (t, J = 7.0 Hz, 3 H). 11841-PCT Intermediate 108B. Ethyl 5-cyanomethyloxop-tolyl-1,2,3,6-tetrahydropyridine- 2-carboxylate Intermediate 108A (1.2837 g, 4.06 mmol) was dissolved in a mixture of THF (15 mL) and water (3.00 mL) in a 25 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer. Lithium hydroxide monohydrate (0.204 g, 4.87 mmol) was added and the reaxction mixture was stirred at rt for 1 h. The reaction mixture was acidified to pH 4 with AcOH, diluted with EtOAc (50 mL) and the solution was washed with water (20 mL). The organic phase was dried over MgSO and concentrated in vacuo to give the desired product (1.40 g, 115%) as yellow oil. LCMS Anal. Calc’d for C H N O 298.1, 17 18 2 3 found [M+H] 299.1.
Intermediate 108C. 5-Cyanomethyloxop-tolyl-1,2,3,6-tetrahydropyridine carboxylic acid Intermediate 108B (1.211 g, 4.06 mmol) was dissolved in acetic acid (95 mL) in a 250 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer. HCl (11.67 mL, 142 mmol) was added and the reaction mixture was stirred at 60 °C overnight.
The reaction mixture was concentrated in vacuo to give the desired product as a pale yellow solid. LCMS Anal. Calc’d for C H N O 270.1, found [M+H] 271.0. H NMR 14 2 3 (500 MHz, CDCl ) δ 7.48 (d, J = 8.3 Hz, 2 H), 7.23 (d, J = 7.7 Hz, 2 H), 3.39 (d, J = 18.2 Hz, 1 H), 2.83 (d, J = 18.2 Hz, 1 H), 2.35 (s, 3 H), 1.54 (s, 3 H). 11841-PCT Intermediate 108D. Ethyl 5-hexyloxazolecarboxylate Ethyl 2-isocyanoacetate (1.0 g, 8.84 mmol) was dissolved in DMF (5 mL) in a 25 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer and an Ar inlet. DBU (1.999 mL, 13.26 mmol) and heptanoyl chloride (1.770 mL, 11.49 mmol) were added and the reaction mixture was stirred at 80 °C for 6 h. The reaction mixture was poured into water(50 mL) and extracted with EtOAc (3 × 30 mL). The phases were seperated and the organic phase was dried over MgSO and concentrated in vacuo to a a dark oil. The desired product was isolated by distillattion at 90-95°C at 0.3 mmHg as a clear oil (0.89 g, 45%). LCMS Anal. Calc’d for C H NO 225.1, found [M+H] 226.1. H NMR (500 12 19 3 MHz, CDCl ) δ 7.76 (s, 1H), 4.37 (q, J = 7.2 Hz, 2H), 3.03 (t, J = 7.6 Hz, 2H), 1.71 - 1.64 (m, 2H), 1.38 (t, J = 7.2 Hz, 3H), 1.35 - 1.26 (m, 6H), 0.86 (t, J = 6.3 Hz, 3H).
Intermediate 108E. 1-Aminooctanone, HCl salt Intermediate 108D (0.8889 g, 3.95 mmol) was stirred in HCl (16.44 mL, 99 mmol) in a 100 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer.
The reaction mixture was heated to 100 °C and stirred for 6 h. The reaction mixture was concentrated in vacuo to a tan solid that was triturated in ether (15 mL). The slurry was filtered and was washed with ether (3 mL). The filter cake was dried under vacuum to give the desired product (0.57 g, 80%) as an off-white solid. H NMR (500 MHz, MeOD) δ 3.97 (s, 2 H), 2.56 (t, J = 7.4 Hz, 2 H), 1.62 (quin, J = 7.4 Hz, 2 H), 1.26 - 1.40 (m, 6 H), 0.88 - 0.93 (m, 3 H).
Intermediate 108F. 5-Cyanomethyloxo-N-(2-oxooctyl)p-tolyl-1,2,3,6- tetrahydropyridinecarboxamide 11841-PCT Intermediate 108C (0.1 g, 0.370 mmol) was dissolved in DCM (4 mL) in a 10 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer and an Ar inlet.
EDC (0.099 g, 0.518 mmol), HOBT (0.068 g, 0.444 mmol), Intermediate 108E (0.080 g, 0.444 mmol) and DIEA (0.194 mL, 1.110 mmol) were added and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated in vacuo to an orange oil that was purified by silica gel chromatography (40 g silica gel, eluted with 0-100% EtOAc in hexanes) to give the desired product (0.09 g, 61%) as yellow oil.
Example 108 Intermediate 108F (0.045 g, 0.114 mmol) was dissolved in 1,2-dichloroethane (2 mL) in a 10 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer and an Ar inlet. POCl (0.042 mL, 0.455 mmol) and DIEA (0.089 mL, 0.512 mmol) were added and the reaction mixture was stirred at 85 °C overnight. The reaction mixture was cooled in an ice/water bath and was quenched with water (3 mL). The mixture was extracted with EtOAc (2 × 5 mL) and the organic phases were combined, dried over MgSO and concentrated in vacuo to a brown oil. The crude product was purified by preparative HPLC (MeOH/H O/TFA) to yield the desired product (3.5 mg, 8%) as a yellow solid. LCMS Anal. Calc’d for C H N O 377.2, found [M+H] 378.2. H NMR 23 27 3 2 (500 MHz, CDCl ) δ 7.60 (d, J = 8.0 Hz, 2 H), 7.31 (d, J = 8.0 Hz, 2 H), 6.81 (br. s., 1 H), 3.65 (d, J = 17.9 Hz, 1 H), 3.09 (d, J = 17.9 Hz, 1 H), 2.63 (t, J = 7.4 Hz, 2 H), 2.43 (s, 3 H), 1.76 (s, 3 H), 1.57 - 1.67 (m, 2 H), 1.23 - 1.42 (m, 6 H), 0.89 (t, J = 6.7 Hz, 3 H).
Example 109. 6-(1-Heptyl-1H-pyrazolyl)methyloxop-tolyl-1,2,5,6- tetrahydropyridinecarbonitrile 11841-PCT Intermediate 109A. 5-Cyano-N-methoxy-N,2-dimethyloxop-tolyl-1,2,3,6- tetrahydropyridinecarboxamide Intermediate 108C (0.25 g, 0.925 mmol) was dissolved in DCM (15 mL) in a mL 1 neck rb flask that was equipped with a magnetic stirrer and an Ar inlet. N,O- Dimethylhydroxylamine (0.062 g, 1.017 mmol), EDC (0.195 g, 1.017 mmol) and N- methylmorpholine (0.112 mL, 1.017 mmol) were added and the reaction mixture was stirred at rt overnight. The reaction solvent was removed under vacuum. The residue was dissolved in EtOAc (10 mL) and washed with water (5 mL). The organic phaase was dried over MgSO and concentrated in vacuo to a dark solid. The crude product was purified by silica gel chromatography (40 g silica gel, eluted with 0-100% EtOAc in hexanes) to give the desired product (0089 g, 31%) as a white solid. LCMS Anal. Calc’d for C H N O 313.3, found [M+H] 314.1. H NMR (500 MHz, CDCl ) δ 7.67 (d, J = 17 19 3 3 3 8.3 Hz, 2 H), 7.30 (d, J = 8.0 Hz, 2 H), 3.81 (s, 3 H), 3.69 - 3.75 (m, 1 H), 3.22 (s, 3 H), 2.72 (d, J = 17.6 Hz, 1 H), 2.42 (s, 3 H), 1.63 (s, 3 H).
Intermediate 109B. 6-Methyloxopropioloylp-tolyl-1,2,5,6-tetrahydropyridine carbonitrile 11841-PCT Intermediate 109A (0.0887 g, 0.283 mmol) was dissolved in tetrahydrofuran (1 mL) in a 25 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer and an Ar inlet. Ethynylmagnesium bromide (5.66 mL, 2.83 mmol) was added dropwise and the reaction mixture was stirred at 35 °C for 4 h. The reaction mixture was cooled to rt, quenched with sat’d. aq NH Cl (5 mL) and extracted with EtOAc (2 × 10 mL). The organic phases were combined and concentrated in vacuo. The residue was purified by silica gel chromatography (24 g silica gel, eluted with 0-100% EtOAc in hexanes) to give the desired product (0.06 g, 78%) as an orange solid. LCMS Anal. Calc’d for C H N O 278.1, found [M+H] 279.0. 17 14 2 2 Example 109 Intermediate 109B (0.0613 g, 0.220 mmol) was dissolved in ethanol (3 mL) in a 25 mL 1 neck pear shaped flask that was equipped with a magnetic stirrer.
Heptylhydrazine, HCl (0.073 g, 0.441 mmol) and TEA (0.061 mL, 0.441 mmol) were added and the reaction mixture was stirred at rt overnight. The reaction mixture was concentrated and purified by preparative HPLC (MeOH/H O/TFA) to yield the desired product (3.2 mg, 4%) as a white solid. LCMS Anal. Calc’d for C H N O 390.2, found 24 30 4 [M+H] 391.2. H NMR (500 MHz, CDCl ) δ 7.55 (d, J = 8.3 Hz, 2 H), 7.25 - 7.30 (m, 2 H), 6.35 (s, 1 H), 6.15 (d, J = 1.7 Hz, 1 H), 3.98 - 4.08 (m, 2 H), 3.57 (d, J = 17.9 Hz, 1 H), 3.01 (d, J = 17.9 Hz, 1 H), 2.41 (s, 3 H), 1.81 (quin, J = 7.3 Hz, 2 H), 1.70 - 1.77 (m, 2 H), 1.68 (s, 3 H), 1.16 - 1.34 (m, 6 H), 0.87 (t, J = 7.0 Hz, 3 H).
Examples 110-273 expressed by Formula (II), unless noted in the table, may be made by one skilled in the art by appropriate application of the procedures described 11 15 for Examples 1-16 and Examples 102-. R to R are hydrogen, unless noted in the table. 11841-PCT INFORMAL 12 14 11 15 (II) 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 N OMe 110 CF H 13 608.1 8.13 (d, J = 2.5 Hz, 1H), 3 R = CH S-isomer 7.93 (dd, J = 9.1, 2.8 Hz, F C O 1H), 7.46 (d, J = 9.1 Hz, 2H), 7.22 (s, 1H), 7.20 - 7.15 (m, 2H), 7.13 - 7.08 (m, 2H), 6.94 (d, J = 9.1 Hz, 2H), 6.67 (d, J = 9.1 Hz, 1H), 4.02 (t, J = 5.9 Hz, 2H), 3.90 (br. s., 3H), 3.56 - 3.38 (m, 2H), 2.39 - 2.25 (m, 5H), 2.10 - 2.01 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 111 CF H 13 645.2 7.88 (d, J = 8.5 Hz, 2H), 7.60 3 R = CH S-isomer (d, J = 8.8 Hz, 2H), 7.54 (d, J F C O = 8.5 Hz, 2H), 7.29 (d, J = 8.0 Hz, 2H), 7.16 (d, J = 7.7 Hz, 2H), 7.00 (d, J = 8.8 Hz, 2H), 4.07 (t, J = 5.9 Hz, 2H), 3.75 - 3.46 (m, 2H), 2.43 - 2.32 (m, 2H), 2.29 (s, 3H), 2.11 - 1.97 (m, 2H). 112 CF H 13 480.0 3 R = CH 113 CF H 13 526.1 9.07 (d, J = 1.1 Hz, 1H), 3 R = CH S-isomer 7.63 (d, J = 8.5 Hz, 2H), F C O 7.16 - 7.09 (m, J = 8.0 Hz, 2H), 7.09 - 7.03 (m, 2H), 6.93 - 6.86 (d, J = 7.7 Hz, 2H), 4.12 (t, J = 6.1 Hz, 2H), 4.00 (s, 1H), 3.92 (d, J = 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 17.6 Hz, 1H), 3.77 (d, J = 17.6 Hz, 1H), 2.46 - 2.34 (m, 2H), 2.30 (s, 3H), 2.12 - 2.02 (m, 2H). 114 CF H 13 524.1 3 R = CH S-isomer F C O 115 CF H 577.2 R = CH S-isomer F C O NH 13 116 CF H 554.2 7.42 (d, J = 8.5 Hz, 2H), N R = CH Rac 7.16 (d, J = 8.0 Hz, 2H), F C O 7.10 (br. s., 1H), 6.95 (dd, J = 15.0, 8.4 Hz, 4H), 3.97 (t, J = 6.3 Hz, 2H), 3.59 (d, J = .5 Hz, 2H), 2.36 (s, 3H), 2.16 - 2.04 (m, 2H), 1.86 - 1.77 (m, 2H), 1.69 - 1.59 (m, 2H), 1.59 - 1.50 (m, 2H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 0.93 - 0.79 (m, 3H) NH 13 117 CF H 540.1 7.43 (d, J = 8.5 Hz, 2H), N R = CH Rac 7.17 (d, J = 7.7 Hz, 2H), 7.03 - 6.91 (m, 5H), 3.99 (t, J = 5.9 Hz, 2H), 3.65 - 3.53 (m, 2H), 2.37 (s, 3H), 2.23 - 2.11 (m, 2H), 1.92 - 1.82 (m, 2H), 1.82 - 1.72 (m, 2H) NH 13 118 CF H 576.1 7.44 (d, J = 8.5 Hz, 2H), N R = CH Rac 7.17 (d, J = 8.0 Hz, 2H), 6.96 (dd, J = 11.4, 8.7 Hz, 4H), 6.80 (br. s., 1H), 4.07 - 4.01 (m, 2H), 3.59 (d, J = 7.4 Hz, 2H), 2.37 (s, 3H), 2.33 - 2.19 (m, 2H), 2.10 (dd, J = 9.5, 5.6 Hz, 2H) 119 CF H 541.2 7.44 (d, J = 8.3 Hz, 2H), R = CH S-isomer 7.21 - 7.16 (m, 2H), 7.14 - F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 7.09 (m, 2H), 6.96 (d, J = 8.8 Hz, 2H), 4.06 (t, J = 5.9 Hz, 3H), 3.58 - 3.27 (m, 2H), 2.48 - 2.27 (m, 5H), 2.09 (dd, J = 9.5, 5.9 Hz, 2H), 0.97 - 0.80 (m, 2H), 0.75 - 0.61 (m, 2H), 0.38 (d, J = 3.6 Hz, 2H) O 13 120 CF H 555.2 7.43 (d, J = 8.5 Hz, 2H), R = CH S-isomer 7.20 - 7.03 (m, 5H), 6.97 - F C O 6.88 (m, 2H), 4.36 - 4.22 (m, 1H), 4.08 - 3.98 (m, 2H), 3.53 - 3.27 (m, 2H), 2.44 - 2.16 (m, 8H), 2.12 - 2.02 (m, 2H), 1.78 - 1.51 (m, 4H) 121 CF H 583.3 7.49 - 7.37 (m, 2H), 7.22 - R = CH S-isomer 7.07 (m, 4H), 6.97 - 6.84 (m, F C O 3H), 6.74 (d, J = 8.0 Hz, 1H), 4.10 - 3.95 (m, 3H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 3.77 - 3.62 (m, 1H), 3.52 - 3.26 (m, 2H), 2.42 - 2.25 (m, 6H), 2.15 - 1.96 (m, 3H), 1.86 - 1.64 (m, 2H), 1.64 - 1.44 (m, 4H), 1.35 - 1.16 (m, 3H), 1.14 - 0.93 (m, 4H) OH 13 122 CF H 593.3 7.55 (d, J = 8.5 Hz, 2H), R = CH S-isomer 7.32 (d, J = 8.3 Hz, 2H), F C O 7.20 (d, J = 8.0 Hz, 2H), 7.14 - 7.09 (d, J = 8.8 Hz, 2H), 7.02 - 6.97 (d, J = 8.8 Hz, 2H), 6.68 - 6.62 (m, 2H), 4.08 (t, J = 6.1 Hz, 2H), 3.67 (d, J = 17.1 Hz, 1H), 3.51 (d, J = 17.1 Hz, 1H), 2.45 - 2.35 (m, 2H), 2.34 (s, 3H), 2.09 - 2.00 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 123 CF H 515.2 7.57 - 7.50 (m, J = 8.8 Hz, R = CH S-isomer 2H), 7.27 - 7.23 (m, J = 8.3 F C O Hz, 2H), 7.23 - 7.19 (m, J = 8.3 Hz, 2H), 7.02 - 6.97 (m, J = 9.1 Hz, 2H), 4.08 (t, J = 6.1 Hz, 2H), 3.60 (d, J = 17.1 Hz, 1H), 3.46 (d, J = 17.1 Hz, 1H), 2.57 (s, 3H), 2.45 - 2.37 (m, 2H), 2.36 (s, 3H), 2.11 - 1.99 (m, 2H).
OMe 13 124 CF F 643.3 7.90 (s, 1H), 7.62 (d, J = 8.8 R = CH Rac Hz, 2H), 7.25 - 7.17 (m, 6H), F C O 6.98 (d, J = 8.8 Hz, 2H), 6.95 (br. s., 1H), 6.78 (d, J = 9.1 Hz, 2H), 4.05 (t, J = 5.9 Hz, 2H), 3.81 - 3.71 (m, 3H), 2.44 - 2.27 (m, 5H), 2.13 - 2.03 (m, 2H) 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 NH 13 125 CF H 512.1 7.45 (d, J = 8.5 Hz, 3H), N R = CH Rac 7.13 (d, J = 7.7 Hz, 2H), 6.93 (d, J = 7.7 Hz, 4H), 4.18 (t, J = 6.5 Hz, 2H), 3.68 - 3.52 (m, 2H), 2.67 - 2.56 (m, 2H) NH 13 126 CF H 466.2 N R = CH NH 13 127 CF H 480.1 7.50 - 7.40 (m, 4H), 7.16 (d, N R = CH Rac J = 7.7 Hz, 2H), 7.09 (br. s., 1H), 6.98 (d, J = 7.7 Hz, 2H), 6.17 (br. s., 1H), 2.36 (s, 5H), 2.21 (d, J = 3.6 Hz, 2H), 1.82 - 1.74 (m, 2H), 1.70 - 1.62 (m, 2H) 128 CF H 591.3 R = CH S-isomer 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 129 CF H 591.3 R = CH S-isomer 130 CF H 568.2 8.47 (d, J = 1.9 Hz, 1H), R = CH S-isomer 7.61 - 7.54 (ab quartet, J = 8.8 Hz, 2H), 7.24 - 7.20 (ab quartet, J = 8.3 Hz, 2H), 7.19 - 7.15 (ab quartet, J = 8.3 Hz, 2H), 7.04 - 6.98 (m, 2H), 6.93 - 6.87 (m, 1H), 4.09 (t, J = 6.1 Hz, 2H), 3.75 (d, J = 17.1 Hz, 1H), 3.51 (d, J = 16.8 Hz, 1H), 2.46 - 2.34 (m, 2H), 2.32 (s, 3H), 2.12 - 2.01 (m, 3H). 131 CF H 581.3 R = CH S-isomer 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 N 13 132 CF H 570.2 R = CH S-isomer OMe 13 133 CF F 643.1 8.07 (br. s., 1H), 7.63 (d, J = R = CH Rac 8.8 Hz, 2H), 7.24 - 7.15 (m, 6H), 6.96 (d, J = 9.1 Hz, 2H), 6.76 (d, J = 8.8 Hz, 2H), 4.09 - 3.97 (m, 2H), 3.75 (s, 3H), 2.39 - 2.25 (m, 5H), 2.12 - 2.02 (m, 2H) 134 CF F 680.9 R = CH Rac N 135 CF F 626.9 R = CH F C N 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 N 13 136 CF H 594.1 N R = CH S-isomer 137 CF H 661.1 R = CH S-isomer O OCF 138 CF F CN 422.8 R = CH N OMe 13 140 CF F 643.9 R = CH F C O 141 CF F 604.9 R = CH F C O N 11 142 CF H 560.1 9.17 (s, 1H), 7.59 (d, J = 8.8 N R = F S-isomer Hz, 2H), 7.07 - 7.00 (m, 2H), R = OCH F C O 6.95 (t, J = 8.5 Hz, 1H), 6.69 (dd, J = 8.8, 2.5 Hz, 1H), 6.63 (dd, J = 12.7, 2.5 Hz, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 1H), 4.09 (t, J = 6.1 Hz, 2H), 3.90 - 3.81 (m, 1H), 3.76 (s, 3H), 3.74 - 3.66 (m, 1H), 2.47 - 2.30 (m, 2H), 2.12 - 1.94 (m, 2H).
N 13 143 CF H 542.1 9.05 (s, 1H), 7.60 (d, J = 8.8 N R = OCH S-isomer Hz, 2H), 7.04 (d, J = 9.1 Hz, F C O 2H), 6.97 - 6.88 (m, 1H), 6.86 - 6.76 (m, 1H), 4.09 (t, J = 6.1 Hz, 2H), 3.92 - 3.70 (m, 2H), 3.75 (S, 3H), 2.45 - 2.29 (m, 2H), 2.10 - 1.94 (m, 2H). 144 CF H CN 535.1 7.65 (br. s., 2H), 7.53 (d, J = R = OCHF S-isomer 9.1 Hz, 2H), 7.32 - 7.24 (m, F C O 2H), 7.05 - 6.98 (m, 2H), 6.96 (t, J = 75 Hz, 1H), 4.14 - 3.97 (m, 2H), 3.73 (d, J = 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 3.3 Hz, 2H), 2.42 - 2.27 (m, 2H), 2.09 - 1.96 (m, 2H). 145 CF H CN 513.1 7.64 (d, J = 9.1 Hz, 2H), 7.52 S-isomer (d, J = 9.1 Hz, 2H), 7.03 (d, J OCH CH F C O = 9.1 Hz, 4H), 4.12 (d, J = 6.9 Hz, 2H), 4.08 - 4.03 (m, 2H), 3.82 - 3.58 (m, 2H), 2.45 - 2.26 (m, 2H), 2.08 - 1.95 (m, 2H), 1.41 (t, J = 6.9 Hz, 3H). 12 13 146 CF H CN 519.1 8.15 (d, J = 1.7 Hz, 1H), 8.01 R and R S-isomer - 7.96 (m, 2H), 7.93 (d, J = F C O 8.0 Hz, 1H), 7.67 - 7.54 (m, 5H), 7.06 - 7.00 (m, 2H), 4.08 (s, 2H), 3.93 - 3.77 (m, 2H), 2.48 - 2.25 (m, 2H), 2.13 - 1.92 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 147 CF H CN 469.1 7.55 (d, J = 8.8 Hz, 2H), 7.52 R = CH S-isomer - 7.49 (m, 2H), 7.33 (d, J = 8.0 Hz, 2H), 7.04 - 6.99 (m, 2H), 4.24 (t, J = 6.1 Hz, 2H), 3.77 - 3.66 (m, 2H), 2.75 - 2.63 (m, 2H), 2.41 (s, 3H).
OMe 11 148 CF H 621.2 7.49 - 7.41 (m, 2H), 7.35 (d, R = CH S-isomer J = 8.0 Hz, 2H), 7.03 - 6.65 R = CH F C O (m, 7H), 4.07 - 3.99 (m, 2H), 3.71 (s, 3H), 3.51 - 3.42 (m, 1H), 3.28 – 3.14 (m, 1H), 2.38 - 2.21 (m, 8H), 2.11 - 1.99 (m, 2H).
N 13 149 CF H 578.1 9.09 (s, 1H), 7.61 (d, J = R = OCHF S-isomer 8.8 Hz, 2H), 7.08 - 7.03 (m, F C O 6H), 6.97 – 6.68 (m, 1H), 4.09 (t, J = 6.1 Hz, 2H), 3.92 (d, J = 17.9 Hz, 1H), 3.77 (d, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 J = 17.9 Hz, 1H), 2.43 - 2.31 (m, 2H), 2.10 - 1.98 (m, 2H).
N 13 150 CF F 582.2 7.54 (d, J = 8.8 Hz, 2H), R = CH S-isomer 7.23 - 7.11 (m, 4H), 7.03 - F C O 6.94 (m, 2H), 6.71 (s, 1H), 4.06 (t, J = 6.1 Hz, 2H), 3.98 (s, 1H), 3.71 (d, J = 17.1 Hz, 1H), 3.47 (d, J = 16.8 Hz, 1H), 2.45 - 2.31 (m, 2H), 2.29 (d, J = 6.3 Hz, 6H), 2.12 - 1.96 (m, 2H).
N 13 151 CF H 568.2 R = CH S-isomer F C O N 13 152 CF H 568.2 R = CH S-isomer F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 N 13 153 CF H 579.2 R = CH S-isomer N F C O N 13 154 CF H 583.1 7.58 - 7.49 (ab quartet, J = N R = CH S-isomer 8.8 Hz, 2H), 7.20 (d, J = 8.3 F C O Hz, 2H), 7.15 (d, J = 8.3 Hz, 2H), 7.03 - 6.92 (m, 2H), 4.06 (t, J = 6.1 Hz, 2H), 3.72 (d, J = 17.3 Hz, 1H), 3.49 (d, J = 17.1 Hz, 1H), 2.55 (s, 3H), 2.43 - 2.33 (m, 2H), 2.30 (s, 3H), 2.09 - 1.97 (m, 2H).
H OMe 13 155 CH H 478.4 R = CH N 11 156 CF H 540.1 N R = CH S-isomer N 13 R = CH F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 157 CF H 582.2 R = CH S-isomer F C O 158 CF H 613.2 R = CH S-isomer N N F C O N 13 159 CF F 562.2 R = CH F C O 160 CF F CN 519.2 R = CH F C O OMe 13 161 CH H 464.3 R = CH OMe 13 162 CF F 628.9 R = CH Rac N 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 OMe 13 163 CF F 656.9 8.05 (s, 1H), 7.61 (d, J = 8.8 R = CH Rac N Hz, 2H), 7.25 - 7.16 (m, 6H), F C O 7.02 - 6.94 (m, 2H), 6.82 - 6.73 (m, 2H), 6.60 (br. s., 1H), 4.01 (td, J = 6.1, 1.4 Hz, 2H), 3.75 (s, 3H), 2.35 (s, 3H), 2.24 - 2.10 (m, 2H), 1.93 - 1.85 (m, 2H), 1.83 - 1.74 (m, 2H), 1.67 (br. s., OMe 13 164 CF F 671.0 R = CH Rac N F 13 165 CF H 561.2 7.59 (d, J = 8.8 Hz, 2H), R = CH S-isomer 7.42 - 7.36 (m, 2H), 7.35 - 7.28 (m, 4H), 7.20 - 7.16 (m, 3H), 7.06 - 6.99 (m, 4H), 6.98 - 6.93 (m, 2H), 3.66 (d, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 J = 17.3 Hz, 1H), 3.52 (d, J = 17.3 Hz, 1H), 2.30 (s, 3H).
H OMe 13 166 CH H 492.2 R = CH 167 CF H 620.2 8.38 (d, J = 1.7 Hz, 1H), R = OHCF S-isomer 7.61 (s, 1H), 7.51 (d, J = 8.8 F C O Hz, 2H), 7.34 - 7.24 (m, 2H), 7.13 - 7.04 (ab quartet, J = 8.8 Hz, 2H), 6.99 - 6.92 (m, 2H), 6.86 (s, 1H), 4.26 (br. s., 1H), 4.05 (t, J = 5.9 Hz, 2H), 3.69 (d, J = 17.1 Hz, 1H), 3.42 (d, J = 17.1 Hz, 1H), 2.41 - 2.27 (m, 2H), 2.10 - 1.97 (m, 2H).
Cl 13 168 CF H 611.2 R = CH S-isomer F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 169 CF H 611.2 R = CH S-isomer F C O Cl F 13 170 CF H 629.2 R = CH S-isomer N F C O 171 CF H 629.3 R = CH S-isomer N F C O 172 CF H 584.2 8.37 (d, J = 1.7 Hz, 1H), R = OCH S-isomer 7.50 (d, J = 8.8 Hz, 2H), F C O 7.28 - 7.17 (m, 2H), 6.99 - 6.91 (m, 2H), 6.89 - 6.82 (m, 3H), 4.05 (t, J = 6.1 Hz, 2H), 3.78 (s, 3H), 3.68 (d, J = 16.8 Hz, 1H), 3.42 (d, J = 17.1 Hz, 1H), 2.45 - 2.23 (m, 2H), 2.10 - 1.98 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 N OMe 13 173 CH H 618.4 7.34 (d, J = 8.0 Hz, 2H), R = CH Rac 7.26 (d, J = 9.1 Hz, 2H), 7.15 (d, J = 8.0 Hz, 2H), 6.79 (d, J = 9.1 Hz, 2H), 3.76 (s, 3H), 3.43 - 3.37 (d, J = 17.5 Hz, 1H), 3.22 (t, J = 7.1 Hz, 2H), 2.86 - 2.80 (d, J = 17.2 Hz, 1H), 2.32 (s, 3H), 1.53 (s, 3H), 1.31 - 1.21 (m, 28H), 0.88 (t, J = 7.1 Hz, OMe 13 174 CH H 506.4 R = CH OMe 13 175 CH H 512.4 R = CH 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 176 CF H 598.2 7.54 (d, J = 8.8 Hz, 2H), R = OCH S-isomer 7.32 - 7.23 (m, 2H), 6.97 (d, F C O J = 8.8 Hz, 2H), 6.88 (d, J = 8.8 Hz, 2H), 6.73 (s, 1H), 4.06 (t, J = 6.1 Hz, 2H), 3.76 (s, 3H), 3.71 (d, J = 17.1 Hz, 1H), 3.49 (d, J = 17.1 Hz, 1H), 2.45 - 2.32 (m, 2H), 2.31 - 2.26 (m, 3H), 2.08 - 1.97 (m, 2H).
H 13 177 CH H CN 382.3 R = CH H OMe 13 178 CH H 526.4 R = CH 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 Br 13 179 CF H 648.2 7.54 (d, J = 8.8 Hz, 2H), R = CH S-isomer 7.23 - 7.11 (m, 4H), 7.03 - F C O 6.93 (m, 3H), 4.06 (t, J = 6.1 Hz, 2H), 3.72 (d, J = 16.9 Hz, 1H), 3.48 (d, J = 17.2 Hz, 1H), 2.44 - 2.32 (m, 2H), 2.30 (s, 3H), 2.08 - 1.97 (m, 2H). 180 CF H 674.3 R = CH S-isomer F C O OMe 13 181 CF F 547.0 R = CH 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 NH 13 182 CF H 510.3 R = CH N 13 183 CF H 554.3 7.57 - 7.48 (m, 2H), 7.13 - 3 R = CH Rac 7.05 (m, 2H), 7.02 - 6.94 (m, F C O S-isomer 2H), 6.88 - 6.77 (m, 2H), 4.07 - 3.99 (m, 2H), 2.29 (s, 3H), 2.19 - 2.07 (m, 2H), 1.89 - 1.78 (m, 2H), 1.69 - 1.55 (m, 4H) 184 CF H CN 511.1 7.46 (dd, J = 12.1, 8.5 Hz, 3 R = CH S-isomer 4H), 7.31 (d, J = 8.3 Hz, F C O 2H), 6.95 (d, J = 9.1 Hz, 2H), 4.32 - 4.22 (m, 1H), 4.00 (s, 2H), 3.67 - 3.55 (m, 1H), 2.41 (s, 3H), 2.23 - 1.99 (m, 2H), 1.88 - 1.76 (m, 2H), 1.70 - 1.50 (m, 5H) 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 185 CF H 678.3 3 R = CH S-isomer F C O 186 CF H 540.3 3 R = CH S-isomer F C O 187 CF H 598.3 7.57 - 7.50 (m, J = 8.8 Hz, 3 R = CH S-isomer 2H), 7.21 - 7.12 (m, 4H), F C O 7.01 - 6.94 (m, 2H), 6.48 (s, O 1H), 4.06 (t, J = 6.1 Hz, 2H), 3.95 (s, 3H), 3.71 (d, J = 17.1 Hz, 1H), 3.47 (d, J = 17.1 Hz, 1H), 2.44 - 2.32 (m, 2H), 2.30 (s, 3H), 2.07 - 1.98 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 OMe 13 188 CH H 450.1 3 R = CH Rac N OMe 13 189 CH H 484.1 3 R = CH Rac N 190 CF H 662.3 3 R = CH S-isomer F C O O OMe 13 191 CH H 520.2 3 R = CH Rac N 192 CF H 526.3 3 R = CH S-isomer F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 193 CF H 542.1 H NMR (500 MHz, CDCl : 3 R = OCH 3 S-isomer MeOD (1:1)) δ 7.65 (s, 1H), F C O 7.54 (d, J = 8.4 Hz, 2H), 6.98 (d, J = 8.6 Hz, 2H), 6.93 (d, J = 8.4 Hz, 2H), 6.79 (d, J = 8.4 Hz, 2H), 4.07 (t, J = 6.0 Hz, 2H), 3.77 (s, 3H), 3.69 (d, J = 17.7 Hz, 1H), 3.60 (d, J = 17.7 Hz, 1H), 2.41 - 2.28 (m, 2H), 2.06 (dq, J = 12.1, 6.3 Hz, 2H).
NH 13 194 CF H 432.3 7.21 (d, J = 7.9 Hz, 2H), N R = CH Rac 7.08 (d, J = 8.1 Hz, 2H), 6.25 (s, 1H), 3.39 (d, J = 18.5 Hz, 1H), 3.34 (d, J = 18.9 Hz, 1H), 2.39 (s, 3H), 2.23 (t, J = 7.2 Hz, 2H), 1.55 - 1.47 (m, 2H), 1.40 - 1.23 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 (m, 6H), 0.87 (t, J = 6.9 Hz, 3H). 195 CF H 460.4 7.21 (d, J = 7.9 Hz, 2H), 7.08 3 R = CH Rac (d, J = 8.1 Hz, 2H), 6.32 (s, 1H), 3.37 (AB quartet, J = 18.7 Hz, 2H), 2.39 (s, 3H), 2.23 (t, J = 7.0 Hz, 2H), 1.51 (quin, J = 7.2 Hz, 2H), 1.39 - 1.20 (m, 10H), 0.91 - 0.84 (m, 3H). 196 CF H 460.4 7.17 (d, J = 7.9 Hz, 2H), 3 R = CH R-isomer 7.04 (d, J = 8.1 Hz, 2H), 6.74 (s, 1H), 3.43 - 3.32 (m, 2H), 2.37 (s, 3H), 2.23 (t, J = 7.0 Hz, 2H), 1.55 - 1.47 (m, 2H), 1.37 - 1.22 (m, 10H). 197 CF H 388.3 7.20 (d, J = 7.9 Hz, 2H), 3 R = CH S-isomer 7.07 (d, J = 8.1 Hz, 2H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 6.34 (s, 1H), 3.38, 3.32 (ABq, J = 18.9 Hz, 2H), 2.39 (s, 3H), 1.28 (tt, J = 8.3, 5.0 Hz, 1H), 0.89 - 0.83 (m, 2H), 0.76 - 0.71 (m, 2H). 198 CF H 452.3 7.30 - 7.23 (m, 2H), 7.22 - 3 R = CH S-isomer 7.13 (m, 5H), 6.99 (d, J = 8.1 Hz, 2H), 6.57 (s, 1H), 3.33 (d, J = 18.9 Hz, 1H), 3.26 (d, J = 18.9 Hz, 1H), 2.85 - 2.80 (m, 2H), 2.57 - 2.53 (m, 2H), 2.38 (s, 3H) NH 13 199 CF H 418.3 7.19 (d, J = 7.9 Hz, 2H), 3 R = CH S-isomer 7.06 (d, J = 8.1 Hz, 2H), 6.68 (s, 1H), 3.38, 3.35 (ABq, J = 18.9 Hz, 2H), 2.37 (s, 3H), 2.24 (t, J = 7.4 Hz, 2H), 1.63 (dquin, J = 13.4, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 6.7 Hz, 1H), 1.41 (q, J = 7.3 Hz, 2H), 0.88 (d, J = 6.6 Hz, OCF 13 200 CF H 3 663.2 9.63 (s, 1H), 7.62 (s, 1H), 3 R = CH Rac 7.54 (s, 1H), 7.50 (d, J = 8.9 Hz, 2H), 7.22 (d, J = 8.1 Hz, 2H), 7.18 (d, J = 8.1 Hz, 2H), 7.13 (d, J = 8.5 Hz, 2H), 6.87 (s, 1H), 4.13 (t, J = 7.2 Hz, 2H), 3.48 (d, J = 18.4 Hz, 1H), 3.25 (d, J = 18.4 Hz, 1H), 2.42 - 2.31 (m, 3H), 2.08 - 2.03 (m, 2H), 1.96 - 1.85 (m, 2H), 1.62 - 1.53 (m, 2H), 1.41 - 1.31 (m, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 OHCF 13 201 CF H 2 645.3 10.16 (s, 1H), 9.00 (s, 1H), 3 R = CH Rac N 7.99 (s, 1H), 7.70 (s, 1H), 7.51 (d, J = 8.8 Hz, 2H), 7.39 (d, J = 8.0 Hz, 2H), 7.19 (d, J = 8.3 Hz, 2H), 7.12 (t, J = 74.3 Hz, 1H), 7.09 (d, J = 8.8 Hz, 2H), 4.11 (t, J = 6.9 Hz, 2H), 3.41-3.37 (d, J = 17.4 Hz, 1H), 3.35 - 3.31 (d, J = 17.4 Hz, 1H), 2.28 (s, 3H), 2.23 - 2.17 (m, 2H), 1.85 - 1.75 (m, 2H), 1.52 - 1.44 (m, 2H), 1.31 - 1.22 (m, 2H).
NH 13 202 CF3 H 556.3 HNMR (500 MHz, DMSO- N R = S-isomer d ) δ 9.57 (s, 1H), 7.64 (d, J CH OCH 6 F C O = 8.8 Hz, 2H), 7.20 (d, J = 8.1 Hz, 2H), 7.04 (d, J = 8.8 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 Hz, 2H), 6.99 (d, J = 7.9 Hz, 2H), 4.36 (s, 2H), 4.09 (t, J = 6.1 Hz, 2H), 3.75 (d, J = 17.5 Hz, 1H), 3.69 (d, J = 17.5 Hz, 1H), 3.27 (s, 3H), 2.48 - 2.37 (m, 2H), 2.01 - 1.89 (m, 2H) 203 CF H R = F 574.3 S-isomer F C O OCH CH 204 CF H 512.1 H NMR (500 MHz, 3 R = CH S-isomer MeOD:CDCl (1:1)) δ 7.61 (s, 2H), 7.55 (d, J = 8.6 Hz, 2H), 6.97 (dd, J = 10.4, 8.3 Hz, 4H), 6.84 (d, J = 8.1 Hz, 2H), 4.24 (t, J = 6.4 Hz, 2H), 3.71 (d, J = 17.2 Hz, 1H), 3.50 (d, J = 17.2 Hz, 1H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 3.08 - 3.03 (m, 4H), 2.67 (qt, J = 10.8, 6.3 Hz, 2H), 1.78 (dq, J = 11.3, 5.5 Hz, 4H), 1.69 (q, J = 5.9 Hz, 2H).
NH 13 205 CF3 H 578.1 H NMR (500 MHz, CDCl3: N R = OCHF S-isomer MeOD (1:1)) δ 7.61 (s, 2H), F C O 7.54 (d, J = 8.6 Hz, 2H), 7.05 - 6.96 (m, 5H), 6.66 (t, J = 73.4 Hz, 1H), 4.07 (t, J = 6.0 Hz, 2H), 3.70 (d, J = 17.9 Hz, 1H), 3.57 (d, J = 17.8 Hz, 1H), 2.41 - 2.28 (m, 2H), 2.11 - 2.02 (m, 2H).
NH 13 206 CF H 498.1 H NMR (500 MHz, MeOD- N R = CH S-isomer CDCl (1:1)) δ 7.60 - 7.57 F C O (m, 2H), 7.07 (d, J = 8.6 Hz, 2H), 6.86 (d, J = 8.0 Hz, 1H), 4.48 (q, J = 8.2 Hz, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 1H), 3.70 (d, J = 17.8 Hz, 1H), 3.58 (d, J = 17.8 Hz, 1H), 2.30 (s, 1H). 207 CF H 562.1 H NMR (500 MHz, CDCl : 3 R R = 3 S-isomer MeOD (1:1)) δ 7.81 - 7.73 F C O (m, 2H), 7.72 (d, J = 8.6 Hz, 1H), 7.61 (s, 1H), 7.60 - 7.55 (m, 3H), 7.53 - 7.46 (m, 2H), 7.01 (d, J = 8.8 Hz, 2H), 6.98 (dd, J = 8.5, 1.8 Hz, 1H), 4.08 (t, J = 6.0 Hz, 2H), 3.83 (d, J = 17.8 Hz, 1H), 3.70 (d, J = 17.8 Hz, 1H), 2.41 - 2.28 (m, 2H), 2.11 - 2.01 (m, 2H). 208 CF H 556.1 H NMR (500 MHz, CDCl3: 3 R = S-isomer MeOD (1:1)) δ 7.62 (s, 1H), OCH CH F C O 7.54 (d, J = 8.5 Hz, 2H), 6.98 (d, J = 8.9 Hz, 2H), 6.91 (d, J 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 = 8.7 Hz, 2H), 6.77 (d, J = 8.8 Hz, 2H), 4.07 (t, J = 6.0 Hz, 2H), 4.00 (q, J = 7.0 Hz, 2H), 3.68 (d, J = 17.8 Hz, 1H), 3.59 (d, J = 17.7 Hz, 1H), 2.40 - 2.28 (m, 2H), 2.10 - 2.01 (m, 2H), 1.38 (t, J = 7.0 Hz, 3H). 209 CF H 570.2 H NMR (500 MHz, DMSO- 3 R R = S-isomer d ) δ 9.51 (s, 1H), 7.61 (d, J F C O = 8.4 Hz, 2H), 7.01 (d, J = 8.8 Hz, 2H), 6.70 (d, J = 8.4 Hz, 1H), 6.57 (d, J = 2.2 Hz, 1H), 6.41 (dd, J = 8.5, 2.2 Hz, 1H), 4.25 - 4.14 (m, 4H), 4.06 (t, J = 6.2 Hz, 2H), 3.68 (d, J = 18.2 Hz, 1H), 3.63 (d, J = 18.5 Hz, 1H), 2.42 (ddd, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 J = 16.4, 8.0, 5.1 Hz, 2H), 1.93 (dq, J = 12.3, 6.4 Hz, 2H). 210 CF H 566.1 H NMR (500 MHz, DMSO- 3 R R = S-isomer d6) δ 9.42 (s, 1H), 7.60 (d, J F C O = 8.5 Hz, 2H), 7.01 (d, J = 8.6 Hz, 2H), 6.84 (d, J = 8.0 Hz, 1H), 6.80 (s, 1H), 6.56 (d, J = 7.7 Hz, 1H), 4.06 (t, J = 6.1 Hz, 2H), 3.67 (d, J = 17.8 Hz, 1H), 3.60 (d, J = 17.9 Hz, 1H), 2.61 (s, 2H), 2.56 (s, 2H), 2.42 (ddd, J = 11.4, 8.3, 5.2 Hz, 2H), 1.98 - 1.88 (m, 2H), 1.65 (p, J = 2.8 Hz, 4H). 211 CF H 556.2 H NMR (500 MHz, DMSO- 3 R R = S-isomer d ) δ 9.37 (s, 1H), 7.60 (d, J F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 O = 8.5 Hz, 2H), 7.01 (d, J = 8.6 Hz, 2H), 6.75 (d, J = 8.2 Hz, 1H), 6.57 (s, 1H), 6.48 (d, J = 8.1 Hz, 1H), 5.97 (d, J = 3.1 Hz, 2H), 4.06 (t, J = 6.1 Hz, 2H), 3.65 (d, J = 17.6 Hz, 1H), 3.58 (d, J = 17.7 Hz, 1H), 2.46 - 2.35 (m, 2H), 1.93 (dd, J = 9.9, 5.4 Hz, 2H).
NH 12 212 CF H 582.3 H NMR (500 MHz, DMSO- 3 R R = S-isomer d ) δ 9.30 (s, 1H), 7.56 (d, J F C O = 8.4 Hz, 2H), 6.99 (d, J = 8.7 Hz, 2H), 6.71 (d, J = 8.2 R = OCH Hz, 1H), 6.62 (d, J = 8.4 Hz, 1H), 4.12 - 4.01 (m, 2H), 3.70 (s, 3H), 3.67 (m, 1H), 3.60 (d, J = 17.4 Hz, 1H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 2.67 - 2.55 (m, 2H), 2.46 - 2.35 (m, 2H), 2.31 - 2.07 (m, 2H), 1.97 - 1.91 (m, 2H), 1.81 - 1.69 (m, 2H).
NH 13 213 CHF2 H 508.3 H NMR (500 MHz, DMSO- N R = CH S-isomer d ) δ 9.10 (s, 1H), 7.52 (d, J F C O = 8.5 Hz, 2H), 7.07 (d, J = 7.8 Hz, 2H), 7.02 (d, J = 9.0 Hz, 2H), 6.88 (d, J = 7.9 Hz, 2H), 6.33 (t, J = 55.0 Hz, 1H), 4.07 (t, J = 6.2 Hz, 2H), 3.57 (d, J = 17.9 Hz, 1H), 3.41 (d, J = 18.2 Hz, 1H), 2.44 (ddd, J = 11.4, 8.1, 5.2 Hz, 2H), 1.99 - 1.91 (m, 2H).
OMe 13 214 H H 539.4 H NMR (500 MHz, DMSO- R = CH R-isomer N d ) δ 9.97 (s, 1H), 8.06 (s, F C O 1H), 7.42 (d, J = 8.6 Hz, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 2H), 7.36 (dd, J = 10.3, 8.2 Hz, 4H), 7.13 (d, J = 7.9 Hz, 2H), 6.96 (d, J = 8.5 Hz, 2H), 6.85 (d, J = 9.0 Hz, 2H), 4.82 - 4.73 (m, 1H), 4.05 (t, J = 6.2 Hz, 2H), 3.83 - 3.72 (m, 1H), 3.71 (s, 3H), 2.88 (qd, J = 17.0, 7.6 Hz, 1H), 2.48 - 2.35 (m, 2H), 2.25 (s, 3H), 1.94 (dq, J = 12.5, 6.4 Hz, 2H). 215 CF H CN 513.1 H NMR (500 MHz, MeOD) R = CH S-isomer δ 7.61 (s, 1H), 7.58 (s, 1H), R = OCH F C O 7.54 - 7.41 (m, 1H), 7.30 (dd, J = 8.4, 2.0 Hz, 1H), 7.22 (s, 1H), 7.03 (d, J = 9.4 Hz, 1H), 6.96 (d, J = 8.9 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 4.32 (s, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 2H), 4.10 - 4.03 (m, 2H), 3.86 (s, 2H), 3.67 - 3.55 (m, 1H), 2.40 - 2.27 (m, 2H), 2.27 - 2.23 (m, 1H), 2.22 (s, 2H), 2.10 - 2.00 (m, 2H) 216 CF H CN 529.1 3 R = OCH S-isomer R = OCH F C O 217 CF H CN 497.1 H NMR (500 MHz, MeOD) 3 R = CH S-isomer δ 7.61 (s, 1H), 7.49 - 7.43 R = CH F C O (m, 2H), 7.35 - 7.23 (m, 3H), 6.99 - 6.94 (m, 2H), 4.32 (s, 2H), 4.06 (t, J = 5.9 Hz, 2H), 3.61 (s, 2H), 2.38 - 2.33 (m, 2H), 2.31 - 2.28 (m, 1H), 2.09 - 2.01 (m, 2H) 218 CF H CN 517.1 H NMR (500 MHz, MeOD) 3 R = F S-isomer δ 7.49 - 7.43 (m, 3H), 7.40 R = OCH F C O (dd, J = 11.9, 2.0 Hz, 1H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 7.14 (t, J = 8.4 Hz, 1H), 7.00 - 6.93 (m, 2H), 4.32 (s, 2H), 4.05 (t, J = 5.9 Hz, 2H), 3.96 (s, 3H), 3.61 (d, J = 3.0 Hz, 2H), 2.39 - 2.27 (m, 2H), 2.09 - 2.01 (m, 2H) 219 CF H CN 501.1 H NMR (500 MHz, MeOD) 3 R = F S-isomer δ 7.49 - 7.43 (m, 2H), 7.39 - R = CH F C O 7.32 (m, 1H), 7.30 - 7.21 (m, 2H), 7.00 - 6.93 (m, 2H), 4.29 (s, 1H), 4.08 - 4.04 (m, 2H), 3.60 (d, J = 2.0 Hz, 2H), 2.35 (d, J = 5.4 Hz, 1H), 2.32 - 2.28 (m, 1H), 2.09 - 2.00 (m, 2H) 220 CF H CN 521.0 H NMR (500 MHz, MeOD) 3 R = F S-isomer 13 δ 7.48 - 7.43 (m, 2H), 7.36 - R = Cl F C O 7.28 (m, 3H), 7.00 - 6.95 (m, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 2H), 4.07 (t, J = 5.9 Hz, 2H), 3.59 (q, J = 1.0 Hz, 2H), 2.40 - 2.28 (m, 2H), 2.10 - 2.02 (m, 2H) 221 CF3 H CN 517.1 H NMR (500 MHz, MeOD) R = CH S-isomer δ 7.61 (s, 1H), 7.58 (s, 1H), R = Cl F C O 7.54 - 7.41 (m, 1H), 7.30 (dd, J = 8.4, 2.0 Hz, 1H), 7.22 (s, 1H), 7.03 (d, J = 9.4 Hz, 1H), 6.96 (d, J = 8.9 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 4.32 (s, 2H), 4.10 - 4.03 (m, 2H), 3.86 (s, 2H), 3.67 - 3.55 (m, 1H), 2.40 - 2.27 (m, 2H), 2.27 - 2.23 (m, 1H), 2.22 (s, 2H), 2.10 - 2.00 (m, 2H) 222 CF H CN 539.0 3 R = Cl S-isomer R = Cl F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 223 CF H CN 532.8 3 R = Cl S-isomer R = OCH F C O 224 CF H CN 493.9 3 R = CN S-isomer F C O 225 CF H CN 562.1 3 R = S-isomer NHSO CH F C O 226 CF H CN 547.1 H NMR (500 MHz, MeOD) 3 R = Cl S-isomer 13 δ 7.61 - 7.58 (m, 1H), 7.44 F C O (d, J = 8.4 Hz, 2H), 7.06 - OCH CH 7.00 (m, 2H), 7.00 - 6.95 (m, 2H), 6.89 (dd, J = 8.4, 2.5 Hz, 1H), 4.11 - 4.04 (m, 4H), 3.66 (d, J = 18.3 Hz, 1H), 3.43 (d, J = 18.3 Hz, 1H), 2.40 - 2.28 (m, 2H), 2.10 - 2.02 (m, 2H), 1.45 - 1.39 (m, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 227 CF H CN 471.2 3 R = CH OMe 13 228 CF H 609.2 7.86 (s, 1H), 7.66 (s, 1H), 3 R = CH Rac 7.37 (d, J = 8.25 Hz, 2H), 7.26 (d, J = 9.08 Hz, 2H), 7.21 (d, J = 7.98 Hz, 2H), 6.82 (d, J = 9.08 Hz, 2H), 4.18 (t, J = 6.88 Hz, 2H), 3.76 (s, 3H), 3.58 (d, J = 17.33 Hz, 1H), 3.35 (d, J = 17.33 Hz, 1H), 2.34 (s, 3H), 2.13 (m, 2H), 1.89 (m, 2H), 1.57 (m, 2H), 1.35 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 OMe 13 229 CF H 595.2 3 R = CH Rac N 230 CF H CN 434.0 3 R = CH 231 CF H 581.4 H NMR (500 MHz, DMSO- 3 R = CH S-isomer d ) δ 10.43 (s, 1H), 9.23 (s, F C O 1H), 7.54 (d, J = 8.80 Hz, 2H), 7.48 (d, J = 1.65 Hz, 1H), 7.29 (d, J = 7.98 Hz, 2H), 7.15 (d, J = 7.98 Hz, 2H), 7.00 (d, J = 8.80 Hz, 2H), 6.34 (d, J = 1.93 Hz, 1H), 4.07 (t, J = 6.05 Hz, 2H), 3.67 (s, 3H), 3.48 (d, J 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 = 17.06 Hz, 1H), 3.39 (d, J = 17.06 Hz, 1H), 2.43 (m, 2H), 2.27 (s, 3H), 1.95 (m, 2H).
OMe 13 232 CF H 609.1 8.13 (d, J = 9.63 Hz, 1H), 3 R = CH S-isomer N N 7.54 (d, J = 8.80 Hz, 2H), F C O 7.26 (d, J = 8.25 Hz, 2H), 7.23 (d, J = 9.63 Hz, 1H), 7.17 (d, J = 7.99 Hz, 2H), 6.99 (d, J = 9.08 Hz, 2H), 4.07 (t, J = 6.05 Hz, 2H), 4.01 (s, 3H), 3.67 (d, J = 17.33 Hz, 1H), 3.53 (d, J = 17.06 Hz, 1H), 2.37 (m, 2H), 2.29 (s, 3H), 2.03 (m, 2H). 233 CF H R = CH 479.8 S-isomer 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 234 CF H 589.9 7.54 (d, J = 8.3 Hz, 2H), 7.43 3 R = Br S-isomer (d, J = 8.5 Hz, 2H), 6.97 (dd, F C O J = 8.7, 2.3 Hz, 4H), 6.85 (br. s., 1H), 4.04 (t, J = 5.8 Hz, 2H), 3.69 - 3.59 (m, 1H), 3.56 - 3.46 (m, 1H), 2.40 - 2.26 (m, 2H), 2.08 (dd, J = 9.4, 6.1 Hz, 2H). 235 CF H H NMR (500 MHz, DMSO) S-isomer δ 9.55 (br. s., 1H), 7.62 (d, J F C O = 8.8 Hz, 2H), 7.02 (d, J = 8.8 Hz, 2H), 6.98 - 6.92 (m, 2H), 6.87 (d, J = 8.3 Hz, 2H), 4.07 (t, J = 6.1 Hz, 2H), 3.80 - 3.61 (m, 2H), 2.46 - 2.29 (m, 2H), 2.00 - 1.90 (m, 2H), 1.89 - 1.79 (m, 1H), 1.00 - 0.88 (m, 2H), 0.69 - 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 0.54 (m, 2H). 236 CF H 578.0 H NMR (500MHz, DMSO) S-isomer δ 9.42 (br. s., 1H), 8.47 (d, J F C O = 2.5 Hz, 1H), 7.73 (d, J = 1.1 Hz, 1H), 7.69 (d, J = 8.8 Hz, 2H), 7.64 (d, J = 8.5 Hz, 2H), 7.12 (d, J = 8.5 Hz, 2H), 7.03 (d, J = 8.8 Hz, 2H), 6.53 (d, J = 1.9 Hz, 1H), 4.08 (t, J = 6.2 Hz, 2H), 3.78 - 3.63 (m, 2H), 2.46 - 2.34 (m, 2H), 2.02 - 1.86 (m, 2H).
NH 13 237 CF H 554.0 H NMR (500MHz, DMSO) 3 R R = S-isomer O δ 9.42 (br. s., 1H), 7.62 (d, J F C O = 8.5 Hz, 2H), 7.02 (d, J = 8.8 Hz, 2H), 6.97 (s, 1H), 6.68 (d, J = 8.5 Hz, 1H), 6.59 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 (d, J = 8.3 Hz, 1H), 4.50 (t, J = 8.7 Hz, 2H), 4.07 (t, J = 6.2 Hz, 2H), 3.65 (br. s., 2H), 3.06 (t, J = 8.7 Hz, 2H), 2.47 - 2.34 (m, 2H), 2.00 - 1.89 (m, 2H). 238 CF H 555.1 H NMR (500 MHz, DMSO) S-isomer R = δ 9.41 (br. s., 1H), 7.61 (d, J F C O = 8.3 Hz, 2H), 7.01 (d, J = 8.5 Hz, 2H), 6.84 (d, J = 8.5 Hz, 2H), 6.52 (d, J = 8.8 Hz, 2H), 4.06 (t, J = 5.9 Hz, 2H), 3.75 - 3.55 (m, 2H), 2.89 (s, 6H), 2.42 (dd, J = 16.5, 11.0 Hz, 2H), 2.02 - 1.84 (m, 2H).
H 13 239 CF H 593.3 H NMR (400 MHz, MeOD) 3 R = CH S-isomer δ 7.52 (d, J = 8.8 Hz, 2H), F C O 7.24 - 7.15 (m, 4H), 6.96 (d, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 J = 8.8 Hz, 2H), 4.06 (t, J = 6.1 Hz, 2H), 3.97 (s, 2H), 3.67 (d, J = 16.9 Hz, 1H), 3.43 (d, J = 16.9 Hz, 1H), 2.93 (s, 3H), 2.44 - 2.28 (m, 5H), 2.09 - 1.96 (m, 2H). 240 CF H 514.3 H NMR (400 MHz, MeOD) 3 R = OCH S-isomer δ 7.65 (d, J = 9.0 Hz, 2H), F C O 7.13 - 7.08 (m, 2H), 6.97 - 6.93 (m, 2H), 6.84 - 6.78 (m, 2H), 4.57 (q, J = 8.4 Hz, 2H), 3.75 (s, 3H), 3.72 (d, J = 2.4 Hz, 2H).
NH 13 241 CF H 528.3 H NMR (400 MHz, MeOD) 3 R = OCH S-isomer δ 7.61 (d, J = 8.8 Hz, 2H), F C N 7.03 (d, J = 9.0 Hz, 2H), 6.95 (d, J = 8.8 Hz, 2H), 6.81 (d, J = 8.8 Hz, 2H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 4.25 (t, J = 6.1 Hz, 2H), 3.76 (s, 3H), 3.71 (d, J = 1.5 Hz, 2H), 2.77 - 2.63 (m, 2H). 242 CF H 582.2 H NMR (400 MHz, MeOD) 3 R = OCF S-isomer δ 7.62 (d, J = 8.8 Hz, 2H), F C N 7.23 - 7.17 (m, 2H), 7.14 - 7.09 (m, 2H), 7.08 - 7.02 (m, 2H), 4.26 (t, J = 6.2 Hz, 2H), 3.83 - 3.66 (m, 2H), 2.77 - 2.63 (m, 2H).
NH 13 243 CF3 H 596.3 H NMR (400 MHz, MeOD) N R = OCF S-isomer δ 7.61 (d, J = 8.8 Hz, 2H), F C O 7.22 - 7.17 (m, 2H), 7.15 - 7.09 (m, 2H), 7.06 - 7.00 (m, 2H), 4.08 (t, J = 6.1 Hz, 2H), 3.82 - 3.65 (m, 2H), 2.44 - 2.30 (m, 2H), 2.09 - 1.99 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 244 CF H 556.3 H NMR (400 MHz, MeOD) 3 R = OCH S-isomer δ 7.58 (d, J = 8.8 Hz, 2H), F C O 7.00 (d, J = 9.0 Hz, 2H), 6.95 (d, J = 9.0 Hz, 2H), 6.81 (d, J = 8.8 Hz, 2H), 4.04 (t, J = 6.1 Hz, 2H), 3.76 (s, 3H), 3.70 (d, J = 1.3 Hz, 2H), 2.31 - 2.17 (m, 2H), 1.92 - 1.83 (m, 2H), 1.80 - 1.70 (m, 2H). 245 CF H 592.1 H NMR (400 MHz, MeOD) 3 R = S-isomer OCHCF δ 7.59 (d, J = 8.6 Hz, 2H), F C O 7.05 (s, 4H), 7.02 (d, J = 9.0 Hz, 2H), 6.87 (t, J = 74.6 Hz, 1H), 4.04 (t, J = 6.1 Hz, 2H), 3.74, 3.70 (ABq, J = 18.0 Hz, 2H), 2.31 - 2.17 (m, 2H), 1.92 - 1.84 (m, 2H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 1.80 - 1.70 (m, 2H) 246 CF H 582.3 H NMR (400 MHz, MeOD) R = CH S-isomer N δ 8.18 (s, 1H), 7.56 (d, J = F C O N 8.8 Hz, 2H), 7.25 - 7.19 (m, 2H), 7.11 (d, J = 7.9 Hz, 2H), 6.98 (d, J = 9.0 Hz, 2H), 4.09 (s, 3H), 4.07 - 4.03 (m, 2H), 3.71 (d, J = 16.9 Hz, 1H), 3.48 (d, J = 16.9 Hz, 1H), 2.44 - 2.31 (m, 2H), 2.28 (s, 3H), 2.08 - 1.98 (m, 2H). 247 CF H 570.1 H NMR (400 MHz, MeOD) 3 R = OCH S-isomer δ 7.57 (d, J = 8.8 Hz, 2H), F C O 7.03 - 6.92 (m, 4H), 6.84 - 6.79 (m, 2H), 4.02 (t, J = 6.3 Hz, 2H), 3.76 (s, 3H), 3.70 (d, J = 1.5 Hz, 2H), 2.25 - 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 2.11 (m, 2H), 1.87 - 1.77 (m, 2H), 1.69 - 1.53 (m, 4H). 248 CF H 606.3 H NMR (400 MHz, MeOD) 3 R = S-isomer OCHCF δ 7.61 (d, J = 8.8 Hz, 2H), F C O 7.08 (s, 4H), 7.05 - 7.01 (m, J = 9.0 Hz, 2H), 6.90 (t, J = 73.7 Hz, 1H), 4.05 (t, J = 6.2 Hz, 2H), 3.76, 3.73 (ABq, J = 18.5 Hz, 2H), 2.28 - 2.14 (m, 2H), 1.89 - 1.81 (m, 2H), 1.72 - 1.56 (m, 4H) F 13 249 CF H 595.3 H NMR (400 MHz, MeOD) 3 R = CH S-isomer N δ 7.62 - 7.53 (m, 3H), 7.51 - F C O O 7.45 (m, 1H), 7.27 - 7.09 (m, 6H), 7.01 - 6.95 (m, 2H), 4.07 (t, J = 6.1 Hz, 2H), 3.71 (d, J = 16.9 Hz, 1H), 3.48 (d, J = 16.9 Hz, 1H), 2.43 - 2.29 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 (m, 5H), 2.08 - 1.98 (m, 2H).
O 13 250 CF H 543.4 8.35 (br. s., 1H), 7.44 (d, J = R = CH S-isomer 8.8 Hz, 2H), 7.18 - 7.13 (m, F C O 2H), 7.11 - 7.06 (m, 2H), 6.96 - 6.90 (m, 2H), 6.30 (s, 1H), 4.03 (t, J = 5.9 Hz, 2H), 3.61 (d, J = 17.2 Hz, 1H), 3.38 (d, J = 17.4 Hz, 1H), 2.41 - 2.25 (m, 8H), 2.11 - 2.02 (m, 2H).
H 13 251 CF H 645.4 H NMR (400 MHz, MeOD) 3 R = S-isomer OCHCF δ 7.52 (d, J = 8.8 Hz, 2H), F C O 7.35 (d, J = 8.8 Hz, 2H), 7.12 (d, J = 8.8 Hz, 2H), 6.97 (d, J = 9.0 Hz, 2H), 7.04 - 6.63 (m, 1H), 4.06 (t, J = 6.1 Hz, 2H), 3.98 (s, 2H), 3.69 (d, J = 16.9 Hz, 1H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 3.45 (d, J = 16.9 Hz, 1H), 2.93 (s, 3H), 2.44 - 2.29 (m, 2H), 2.09 - 1.98 (m, 2H).
H 13 252 CF H 623.3 H NMR (400 MHz, MeOD) 3 R = OCH S-isomer δ 7.51 (d, J = 8.8 Hz, 2H), F C O 7.30 (d, J = 9.0 Hz, 2H), 6.98 - 6.88 (m, 4H), 4.02 (t, J = 6.1 Hz, 2H), 3.99 (s, 2H), 3.79 (s, 3H), 3.66 (d, J = 16.9 Hz, 1H), 3.44 (d, J = 16.7 Hz, 1H), 2.97 (s, 3H), 2.30 - 2.16 (m, 2H), 1.90 - 1.81 (m, 2H), 1.79 - 1.69 (m, 2H). 253 CF H 545.4 H NMR (400 MHz, MeOD) 3 R = CH S-isomer δ 7.56 (d, J = 8.8 Hz, 2H), F C O 7.22 - 7.17 (m, 4H), 7.03 - 6.96 (m, 2H), 4.09 (t, J = 6.1 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 Hz, 2H), 4.02 (q, J = 6.8 Hz, 1H), 3.66 (d, J = 17.2 Hz, 1H), 3.47 (d, J = 16.9 Hz, 1H), 2.46 - 2.32 (m, 5H), 2.10 - 2.00 (m, 2H), 1.19 (d, J = 6.8 Hz, 3H).
H H 13 254 CF H 596.2 H NMR (400 MHz, MeOD) 3 R = S-isomer δ 7.53 (d, J = 8.8 Hz, 2H), OCHCF F C O 7.39 - 7.33 (m, 2H), 7.11 (d, J = 8.6 Hz, 2H), 7.00 - 6.95 (m, 2H), 7.02 - 6.63 (m, 1H), 4.06 (t, J = 6.1 Hz, 2H), 3.65 (d, J = 16.9 Hz, 1H), 3.42 (d, J = 16.7 Hz, 1H), 2.96 (q, J = 7.3 Hz, 2H), 2.43 - 2.28 (m, 2H), 2.07 - 1.97 (m, 2H), 0.92 (t, J = 7.3 Hz, 3H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 H 13 255 CF H 545.3 H NMR (400 MHz, MeOD) 3 R = CH S-isomer δ 7.53 (d, J = 8.8 Hz, 2H), F C O 7.29 - 7.22 (m, 2H), 7.21 - 7.14 (m, 2H), 6.97 (d, J = 9.0 Hz, 2H), 4.06 (t, J = 6.2 Hz, 2H), 3.66 (d, J = 16.9 Hz, 1H), 3.45 (d, J = 16.7 Hz, 1H), 3.07 (q, J = 7.2 Hz, 2H), 2.45 - 2.29 (m, 5H), 2.02 (dd, J = 10.2, 5.8 Hz, 2H), 1.06 (t, J = 7.2 Hz, 3H). 256 CF H CN 501.1 H NMR (400 MHz, MeOD) 3 R = F S-isomer 13 δ 1.96 - 2.13 (m, 2 H) 2.24 - R = CH F C O 2.39 (m, 2 H) 2.41 (s, 3 H) 3.49 - 3.68 (m, 2 H) 3.75 (s, 1 H) 4.06 (t, J = 5.95 Hz, 2 H) 4.33 (s, 1 H) 6.97 (d, J = 1.00 Hz, 2 H) 7.00 - 7.14 (m, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 2 H) 7.24 (t, J = 7.68 Hz, 1 H) 7.45 (d, J = 1.00 Hz, 2 H) 7.61 (s, 1 H) 257 CF H CN R = 541.5 S-isomer NHCO2Me F C O 258 CF H CN R = Cl 533.1 S-isomer R = OCH F C O 12 1 259 CF H CN R = Cl [M-H] H NMR (500 MHz, MeOD) S-isomer R = CH 515.0 3 δ 2.05 (m, J = 9.90, 5.90 F C O Hz, 2 H) 2.22 - 2.41 (m, 2 H) 2.24 - 2.42 (m, 2 H) 2.43 (s, 2 H) 3.62 (br. s., 1 H) 4.06 (t, J = 5.95 Hz, 2 H) 4.29 - 4.48 (m, 2 H) 6.98 (d, J = 8.92 Hz, 1 H) 7.40 (br. s., 1 H) 7.47 (d, J = 8.42 Hz, 1 H) 7.54 (s, 1 H) 7.63 (br. s., 1 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 13 1 260 CF H CN R = 513.1 H NMR (500 MHz, MeOD) S-isomer CH OCH 2 3 δ 1.97 - 2.11 (m, 2 H) 2.33 F C O (m, J = 5.40 Hz, 2 H) 3.43 (s, 3 H) 3.63 (s, 2 H) 4.06 (t, J = 5.95 Hz, 2 H) 4.53 (s, 2 H) 6.97 (d, J = 1.00 Hz, 2 H) 7.47 (d, J = 8.42 Hz, 4 H) 7.55 (d, J = 7.93 Hz, 2 H) 7.60 (s, 1 H) 261 CF3 H CN R = F 571.1 S-isomer R = OCF F C O 12 1 262 CF H CN R = OCH 513.1 H NMR (500 MHz, MeOD) S-isomer R = CH 3 δ 1.95 - 2.12 (m, 2 H) 2.25 F C O (s, 3 H) 2.27 - 2.44 (m, 2 H) 3.63 (d, J = 2.48 Hz, 2 H) 3.87 (s, 3 H) 4.05 (t, J = 5.95 Hz, 2 H) 6.97 (m, J = 8.92 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 Hz, 2 H) 7.08 (d, J = 1.49 Hz, 1 H) 7.24 (s, 1 H) 7.47 (m, J = 8.92 Hz, 2 H) 7.60 (s, 1 H) 12 1 263 CF3 H CN R = Cl 547.1 H NMR (500 MHz, MeOD) S-isomer R = δ 1.49 (t, J = 6.94 Hz, 3 H) F C O OCH CH 2 3 1.95 - 2.12 (m, 2 H) 2.23 - 2.41 (m, 2 H) 3.60 (s, 2 H) 4.05 (t, J = 5.95 Hz, 2 H) 4.19 (q, J = 6.90 Hz, 2 H) 6.96 (d, J = 9.41 Hz, 2 H) Ĉ栕栕{栖Ӊ7栗栗*䡣桤뎑䝃ęĈ 䠄栅뎑䝃栕栕{栖栗*ēĈ䠄栅 뎑䝃栖紫* ㍨笽 쥨㜄 ㍨笽 ᔌ ᘀ ᔔ 쥨㜄㜄 䔀煮⋿ 55 (dd, J = 1.00 Hz, 1 H) 7.60 (s, 1 H) 12 1 264 CF H CN R = F [M-H] H NMR (500 MHz, MeOD) S-isomer R = 529.1 δ 1.47 (t, J = 7.18 Hz, 3 H) F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 OCH CH 1.95 - 2.12 (m, 2 H) 2.33 (s, 2 H) 4.06 (t, J = 5.95 Hz, 2 H) 4.19 (q, J = 6.90 Hz, 2 H) 6.97 (d, J = 8.92 Hz, 2 H) 7.12 (t, J = 1.00 Hz, 1 H) 7.37 - 7.53 (m, 4 H) 7.62 (s, 1 H) 13 1 265 CF H R = CH 607.1 H NMR (500 MHz, MeOD) S-isomer δ 7.88 (dd, J = 8.0, 1.4 Hz, F C O H 1H), 7.55 (d, J = 8.8 Hz, 2H), 7.25 (d, J = 8.0 Hz, 2H), 7.16 (d, J = 8.0 Hz, 2H), 7.05 - 6.97 (m, 3H), 6.89 (d, J = 7.4 Hz, 1H), 6.85 - 6.80 (m, 1H), 4.07 (t, J = 6.1 Hz, 2H), 3.68 (s, 3H), 3.63 (d, J = 17.3 Hz, 1H), 3.51 (d, J = 17.3 Hz, 1H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 2.43 - 2.32 (m, 2H), 2.31 (s, 3 H), 2.07 - 1.98 (m, 2H). 13 1 266 CF H R = CH 544.1 H NMR (500 MHz, MeOD) S-isomer δ 7.50 (dd, J = 12.7, 2.5 Hz, F C O 1H), 7.45 - 7.38 (m, 1H), 7.18 (t, J = 8.7 Hz, 1H), 7.12 - 7.06 (m, 2H), 6.89 (d, J = 8.2 Hz, 2H), 4.16 (t, J = 6.1 Hz, 2H), 3.74 (d, J = 17.8 Hz, 1H), 3.68 (d, J = 17.8 Hz, 1H), , 2.52 - 2.32 (m, 2H), 2.29 (s, 3H), 2.13 - 1.98 (m, 2H). 267 CF H R = CH 526.2 F C O S-isomer 13 1 F NH 268 CF H R = CH 544.2 HNMR (500 MHz, DMSO- S-isomer d ) δ 9.58 (s, 1H), 7.59 (t, J = 9.2 Hz, 1H), 7.09 (d, J = 8.0 F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 Hz, 2H), 6.99 – 6.92 (m, 2H), 6.89 (d, J = 8.1 Hz, 2H), 4.11 (t, J = 6.1 Hz, 2H), 3.81 (d, J = 17.7 Hz, 1H), 3.73 (d, J = 17.8 Hz, 1H), 2.49 – 2.30 (m, 2H), 2.25 (s, 3H), 2.03 – 1.78 (m, 2H). 13 1 269 CF H CN R = CH 483.2 HNMR (400 MHz, MeOD) F C O S-isomer δ 8.18 (s, 1H), 7.56 (d, J = 8.8 Hz, 2H), 7.25 - 7.19 (m, 2H), 7.11 (d, J = 7.9 Hz, 2H), 6.98 (d, J = 9.0 Hz, 2H), 4.09 (s, 3H), 4.07 - 4.03 (m, 2H), 3.71 (d, J = 16.9 Hz, 1H), 3.48 (d, J = 16.9 Hz, 1H), 2.44 - 2.31 (m, 2H), 2.28 (s, 3H), 2.08 - 1.98 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 13 1 270 CF H R = CH 554.1 H NMR (400 MHz, MeOD) R-isomer δ 7.51 (d, J = 8.8 Hz, 2H), F C O 7.30 (d, J = 9.0 Hz, 2H), 6.98 - 6.88 (m, 4H), 4.02 (t, J = 6.1 Hz, 2H), 3.99 (s, 2H), 3.79 (s, 3H), 3.66 (d, J = 16.9 Hz, 1H), 3.44 (d, J = 16.7 Hz, 1H), 2.97 (s, 3H), 2.30 - 2.16 (m, 2H), 1.90 - 1.81 (m, 2H), 1.79 - 1.69 (m, 2H). 13 1 271 CF H R = CH CF 594.1 H NMR (500 MHz, 1:1 3 2 3 S-isomer MeOD/CDCl ) δ 7.52 (d, J = F C O 8.8 Hz, 2H), 7.20 (d, J = 8.0 Hz, 2H), 6.96 (dd, J = 8.4, 2.1 Hz, 4H), 4.04 (t, J = 6.1 Hz, 2H), 3.73 - 3.64 (m, 1H), 3.60 - 3.49 (m, 1H), 3.35 (d, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 J = 10.7 Hz, 2H), 2.39 - 2.23 (m, 2H), 2.13 - 1.91 (m, 2H). 272 CF H R = CH 540.3 S-isomer N OMe 273 CF H R = CH 608.2 1.98 – 2.11 (m, 2H), 2.29 (s, S-isomer 3H), 2.33 - 2.49 (m, 2H), 3.71 - 3.47 (m, 2H), 3.91 (s, 3H), 4.20 - 4.02 (m, 2H), 6.95 – 7.03 (m, 2H), 7.15 (d, J = 8.3 Hz, 2H), 7.26 (d, J = 8.0 Hz, 2H), 7.53 (d, J = 8.8 Hz, 2H), 7.86 (d, J = 1.4 Hz, 1H), 8.76 (d, J = 1.4 Hz, 1H). 13 1 274 CF H R = OCHF 624.1 H NMR (500 MHz, DMSO- S-isomer d ) δ 9.63 (s, 1H), 7.58 (t, J F C O = 9.3 Hz, 1H), 7.43 – 6.98 (m, 5H), 6.92 (d, J = 12.1 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 Hz, 2H), 4.03 (t, J = 6.4 Hz, 2H), 3.80 (d, J = 17.8 Hz, 1H), 3.76 (d, J = 17.9 Hz, 1H), 2.37 – 2.14 (m, 2H), 1.83 – 1.66 (m, 2H), 1.63 – 1.32 (m, 4H). 13 1 275 CF H R = OCHF 630.2 H NMR (400 MHz, MeOD) S-isomer δ 8.75 (br. s., 2H), 7.87 (br.
F C O s., 2H), 7.57 (d, J = 8.6 Hz, 2H), 7.36 (d, J = 8.6 Hz, 2H), 7.12 (d, J = 8.6 Hz, 2H), 7.04 - 6.62 (m, 3H), 4.08 (t, J = 5.8 Hz, 2H), 3.75 (br. s., 1H), 3.52 (d, J = 16.9 Hz, 1H), 2.45 - 2.29 (m, 2H), 2.09 - 1.99 (m, 2H). 12 13 1 276 CF H R R = 608.2 H NMR (500 MHz, MeOD) S-isomer δ 7.83 – 7.66 (m, 3H), 7.61 F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 (s, 2H), 7.56 – 7.43 (m, 3H), 7.01 (d, J = 8.6 Hz, 1H), 6.85 – 6.67 (m, 2H), 4.05 (d, J = 18.0 Hz, 1H), 4.01 (t, J = 6.4 Hz, 2H), 3.76 (d, J = 17.9 Hz, 1H), 2.05-2.20 (m, 2H), 1.91 – 1.72 (m, 2H), 1.74 – 1.45 (m, 4H). 277 CF H 567.1 H NMR (500 MHz, 1:1 S-isomer CDCl :MeOD) δ 7.54 (d, J = F C O 8.4 Hz, 2H), 7.16 (d, J = 7.9 Hz, 2H), 6.99 (d, J = 8.9 Hz, 2H), 6.90 (d, J = 8.4 Hz, 2H), 4.07 (t, J = 5.9 Hz, 2H), 3.79 - 3.66 (m, 1H), 3.63 - 3.50 (m, 1H), 2.49 (d, J = 6.9 Hz, 2H), 2.40 - 2.25 (m, 2H), 2.13 - 1.97 (m, 2H), 0.97 - 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 0.82 (m, 1H), 0.58 - 0.45 (m, 2H), 0.16 (q, J = 5.0 Hz, 2H). 13 1 278 CF H R = CH 558.2 H NMR (500 MHz, 1:1 S-isomer CDCl :MeOD) δ 7.43 (t, J = 9.17 Hz, 1H), 7.02 (d, J = 7.93 Hz, 2H), 6.87 (d, J = 7.93 Hz, 2H), 6.75 (dd, J = 2.48, 8.92 Hz, 1H), 6.71 (dd, J = 2.23, 14.61 Hz, 1H), 3.99 (t, J = 5.95 Hz, 2H), 3.87 (d, J = 17.83 Hz, 1H), 3.61 (d, J = 17.83 Hz, 1H), 2.26 (s, 3H), 2.11-2.21 (m, 2H), 1.80-1.89 (m, 2H), 1.69-1.78 (m, 2H). 279 CF H R = CH 573.2 7.79 (br. s., 1H), 7.46 - 7.38 S-isomer (m, 7H), 7.22 - 7.17 (m, 2H), 7.15 - 7.10 (m, 2H), 7.02 (d, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 J = 9.0 Hz, 2H), 6.69 (br. s., 1H), 5.07 (s, 2H), 3.82 (br. s., 2H), 3.63 (d, J = 17.2 Hz, 1H), 3.37 (d, J = 17.2 Hz, 1H), 2.95 (s, 3H), 2.36 (s, 3H). 280 CF H R = OCHF 630.2 S-isomer F C O 13 1 281 CF H R = CH 582.2 H NMR (500MHz, MeOD) S-isomer δ 7.56 (d, J = 8.92 Hz, 2H), F C O H 7.30 (d, J = 8.42 Hz, 2H), 7.10 (d, J = 7.93 Hz, 2H), 6.97 (d, J = 8.92 Hz, 2H), 6.37 (s, 1H), 4.06 (m, 2H), 3.59 (d, J = 17.83 Hz, 1H), 3.43 (d, J = 17.34 Hz, 1H), 2.32 (m, 2H), 2.30 (s, 3H), 2.22 (s, 3H), 2.06 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 13 1 282 CF H R = CH 651.2 H NMR (500MHz, MeOD) S-isomer O δ 7.61 (s, 1H), 7.47 (d, J = F C O 8.4 Hz, 2H), 7.15 (d, J = 5.9 Hz, 4H), 6.93 (d, J = 8.9 Hz, 2H), 4.33 (br. s., 1H), 4.07 - 4.02 (m, 2H), 4.02 - 3.94 (m, 4H), 3.67 - 3.56 (m, 1H), 3.37 - 3.34 (m, 1H), 2.94 - 2.78 (m, 2H), 2.33 (s, 5H), 2.09 - 2.00 (m, 2H), 1.22 (d, J = 1.5 Hz, 6H). 13 1 283 CF H R = 602.2 H NMR (500 MHz, DMSO- S-isomer OCH CH d ) δ 9.39 (s, 1H), 7.56 (t, J = 2 3 6 F C O 9.3 Hz, 1H), 6.92 (t, J = 8.6 Hz, 4H), 6.81 (d, J = 8.8 Hz, 2H), 4.03 (t, J = 6.4 Hz, 2H), 3.99 (q, J = 6.9 Hz , 2H), 3.82 (d, J = 17.3 Hz, 1H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 3.69 (d, J = 17.7 Hz, 1H), 2.34 – 2.15 (m, 2H), 1.85 – 1.70 (m, 2H), 1.65 – 1.42 (m, 4H), 1.29 (t, J = 6.9 Hz, 3H). 13 1 284 CF3 H R = CH2CF3 640.1 H NMR (500 MHz, DMSO- S-isomer d ) δ 9.36 (w, 1H), 7.59 (t, J F C O = 9.3 Hz, 1H), 7.20 (d, J = 7.9 Hz, 2H), 7.07 – 6.74 (m, 4H), 4.03 (t, J = 6.4 Hz, 2H), 3.76 (d, J = 17.4 Hz, 1H), 3.70 (d, J = 17.7 Hz, 1H), 3.58 (dd, J = 23.3, 11.7 Hz, 2H), 2.33 – 2.16 (m, 2H), 1.86 – 1.68 (m, 2H), 1.545- 1.60 (m, 4H). 13 1 285 CF H R = CH 484.1 H NMR (500 MHz, DMSO- S-isomer d ) δ 9.58 - 9.48 (m, 1H), 7.65 - 7.55 (m, 2H), 7.07 (d, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 J = 8.0 Hz, 2H), 7.04 - 6.97 (m, 2H), 6.94 - 6.85 (m, 2H), 4.06 (s, 2H), 3.69 (s, 2H), 2.24 (s, 3H), 1.63 (q, J = 6.6 Hz, 2H), 1.35 - 1.08 (m, 1H), 0.94 - 0.77 (m, 1H), 0.51 - 0.38 (m, 2H), 0.16 - 0.06 (m, 11 1 286 CF H R = F 544.2 H NMR (500 MHz, DMSO- S-isomer R = CH 3 d ) δ 7.62 (d, J = 8.80 Hz, F C O 2H), 7.02 (d, J = 8.80 Hz, 2H), 7.01 (m, 1H), 6.98 (t, J = 7.98 Hz, 1H), 6.93 (d, J = 11.55 Hz, 1H), 4.08 (t, J = 6.93 Hz, 2H), 3.68 (d, J = 18.16 Hz, 1H), 3.62 (d, J = 17.88 Hz, 1H), 2.42 (m, 2H), 2.28 (s, 3H), 1.95 (m, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 13 1 287 CF H R = CH 568.2 H NMR (500 MHz, DMSO- S-isomer d ) δ 9.47 (s, 1H), 7.49 (s, F C O 1H), 7.45 (d, J = 8.80 Hz, 1H), 7.07 (d, J = 7.98 Hz, 2H), 6.98 (d, J = 8.80 Hz, 1H), 6.89 (d, J = 8.25 Hz, 2H), 4.00 (t, J = 6.19 Hz, 2H), 3.66 (s, 2H), 2.24 (s, 3H), 2.20-2.32 (m, 2H), 2.17 (s, 3H), 1.71-1.80 (m, 2H), 1.47-1.61 (m, 4H). 13 1 288 CF H R = CH 520.0 H NMR (500 MHz, DMSO- S-isomer d ) δ 9.67 (s, 1H), 8.15 (d, J = 4.1 Hz, 1H), 7.55 (d, J = 8.8 Hz, 2H), 7.05 (d, J = 4.1 Hz, 1H), 6.98 (d, J = 8.8 Hz, 2H), 4.03 (t, J = 6.2 Hz, 2H), 3.98 (d, J = 17.6 Hz, 1H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 3.62 (d, J = 17.3 Hz, 1H), 2.45 - 2.33 (m, 2H), 2.31 - 2.23 (m, 1H), 1.94 - 1.79 (m, 2H), 1.23 - 1.11 (m, 2H), 0.87 - 0.77 (m, 2H). 289 CF H R = CH 444.1 7.17 (d, J = 7.9 Hz, 2H), S-isomer 7.05 (d, J = 8.4 Hz, 2H), 6.84 (s, 1H), 3.39 (AB quartet, J = 18.7 Hz, 2H), 2.36 (s, 3H), 2.13 (d, J = 6.6 Hz, 2H), 1.77 - 1.60 (m, 5H), 1.55 - 1.42 (m, 1H), 1.29 - 0.88 (m, 5H). 290 CF H R = CH 468.1 7.36 - 7.27 (m, 5H), 7.18 (s, S-isomer 1H), 7.13 (d, J = 7.9 Hz, 2H), 7.01 (d, J = 8.1 Hz, 2H), 4.57 (AB quartet, J = 11.7 Hz, 2H), 4.22 (s, 2H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 3.39 (AB quartet, J = 18.9 Hz, 2H), 2.34 (s, 3H). 291 CF H R = CH 430.1 7.19 (d, J = 7.9 Hz, 2H), S-isomer 7.06 (d, J = 8.1 Hz, 2H), 6.59 (s, 1H), 3.38 (AB quartet, J = 18.9 Hz, 2H), 2.49 - 2.41 (m, 1H), 2.38 (s, 3H), 1.81 - 1.72 (m, 2H), 1.65 (dd, J = 5.9, 2.4 Hz, 2H), 1.54 - 1.40 (m, J = 12.5, 8.9, 8.9, 8.9 Hz, 3H), 1.38 - 1.28 (m, 3H). 292 CF H R = CH 394.5 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 293 CF H R = CH 432.1 7.19 (d, J = 7.9 Hz, 2H), S-isomer 7.06 (d, J = 8.1 Hz, 2H), 6.67 (s, 1H), 3.38 (AB quartet, J = 18.9 Hz, 2H), 2.37 (s, 3H), 2.23 (t, J = 7.0 Hz, 2H), 1.51 (quin, J = 7.3 Hz, 2H), 1.40 - 1.22 (m, 6H), 0.87 (t, J = 6.8 Hz, 3H). 294 CF H R = OCH 468.1 1H NMR (500 MHz, MeOD) S-isomer δ 7.31 - 7.15 (m, 5H), 6.96 (d, J = 8.9 Hz, 2H), 6.81 (d, J = 8.9 Hz, 2H), 3.79 (s, 3H), 3.34 (AB quartet, J = 18.3 Hz, 2H, partially overlapping with solvent), 2.85 (t, J = 7.2 Hz, 2H), 2.58 (t, J = 7.4 Hz, 2H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 295 CF H R = OCH 434.1 1H NMR (500 MHz, MeOD) S-isomer δ 7.01 (d, J = 8.9 Hz, 2H), 6.78 (d, J = 8.9 Hz, 2H), 3.77 (s, 3H), 3.40 (s, 2H), 2.29 (t, J = 7.4 Hz, 2H), 1.69 (dt, J = 13.5, 6.9 Hz, 1H), 1.45 (q, J = 7.4 Hz, 2H), 0.91 (dd, J = 6.9, 2.0 Hz, 6H). 11 1 296 CF H R = F 625.2 H NMR (500 MHz, DMSO- S-isomer R = CH 3 d ) δ 7.55 (d, J = 8.80 Hz, F C O 2H), 7.31 (d, J = 9.08 Hz, 2H), 7.13 (m, 1H), 7.06 (d, J = 11.83 Hz, 1H), 7.00 (d, J = 8.8 Hz, 2H), 6.94 (d, J = 7.70 Hz, 1H), 6.80 (d, J = 9.08 Hz, 2H), 4.08 (m, 2H), 3.70 (m, 2H), 3.68 (s, 3H), 2.43 (m, 2H), 2.27 (s, 3H), 1.95 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 (m, 2H). 297 CF H O R = CH 608.1 7.43 (d, J = 8.6 Hz, 2H), S-isomer 7.20 - 7.16 (m, 2H), 7.15 - F C O O 7.10 (m, 2H), 6.93 (d, J = 8.8 Hz, 2H), 6.57 (s, 1H), .10 (br. s., 1H), 4.03 (t, J = .9 Hz, 2H), 3.72 (br. s., 2H), 3.62 (d, J = 17.6 Hz, 1H), 3.38 (d, J = 17.8 Hz, 1H), 2.80 - 2.74 (m, 3H), 2.40 - 2.24 (m, 5H), 2.12 - 2.02 (m, 298 CF H R = CH 499.1 7.74 (br. s., 1H), 7.26 - 7.20 S-isomer (m, 4H), 5.88 (s, 1H), 3.84 (s, 2H), 3.29 (ABq, J = 17.6 Hz, 2H), 2.95 (s, 3H), 2.37 (s, 3H), 2.23 (t, J = 7.0 Hz, 2H), 1.57 - 1.48 (m, 2H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 1.41 - 1.25 (m, 6H), 0.89 (t, J = 6.8 Hz, 3H) 299 CF H R = CH 578.1 9.08 (br. s., 1H), 8.77 (d, J = S-isomer 4.2 Hz, 1H), 8.37 (br. s., 1H), F C O 8.25 (br. s., 1H), 7.65 (br. s., 1H), 7.46 (d, J = 8.8 Hz, 2H), 7.19 - 7.11 (m, 4H), 6.95 (d, J = 9.0 Hz, 2H), 6.88 (s, 1H), 4.04 (t, J = 5.9 Hz, 2H), 3.66 (d, J = 17.6 Hz, 1H), 3.43 (d, J = 17.4 Hz, 1H), 2.40 - 2.25 (m, 5H), 2.12 - 2.03 (m, 2H). 13 1 300 CF H R = CH 484.2 H NMR (400 MHz, MeOD) S-isomer δ 9.00 (br. s., 1H), 8.79 (dd, J = 5.3, 1.3 Hz, 1H), 8.46 (d, J = 7.3 Hz, 1H), 7.78 (dd, J = 8.0, 5.4 Hz, 1H), 7.33 (d, J 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 = 8.1 Hz, 2H), 7.21 (d, J = 7.9 Hz, 2H), 3.38 (s, 2H), 2.35 - 2.27 (m, 5H), 1.60 - 1.50 (m, 2H), 1.48 - 1.38 (m, 2H), 1.35 - 1.27 (m, 4H), 0.93 - 0.87 (m, 3H). 13 1 301 CF H R = OCHF 624.2 H NMR (500 MHz, DMSO- F 3 2 S-isomer d ) δ 9.53 (s, 1H), 7.65 (dd, J F C O = 13.0, 2.0 Hz, 1H), 7.49 (d, J = 7.4 Hz, 1H), 7.46 – 6.88 (m, 6H), 4.11 (t, J = 6.3 Hz, 2H), 3.77 (d, J = 17.9 Hz, 1H), 3.72 (d, J = 17.9 Hz, 1H), 2.36 – 2.08 (m, 2H), 1.88 – 1.71 (m, 2H), 1.67 – 1.34 (m, 4H). 13 1 302 CF H R = CH 525.0 HNMR (400 MHz, MeOD) S-isomer δ 7.50 (d, J = 8.8 Hz, 2H), 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 7.23 - 7.15 (m, 4H), 6.93 (d, J = 9.0 Hz, 2H), 4.00 - 3.92 (m, 4H), 3.66 (d, J = 17.2 Hz, 1H), 3.43 (d, J = 16.9 Hz, 1H), 2.93 (s, 3H), 2.32 (s, 3H), 1.79 (sxt, J = 7.0 Hz, 2H), 1.04 (t, J = 7.4 Hz, 3H). 303 CF H R = CH 572.3 F 3 N 3 S-isomer F C O 12 1 304 CF H R R13 = 608.3 HNMR (500 MHz, DMSO- S-isomer d ) δ 9.57 (br. s., 1H), 7.84 F C O (dd, J = 7.84, 13.34 Hz, 2H), 7.71-7.77 (m, 2H), 7.67 (d, J = 12.93 Hz, 1H), 7.47-7.57 (m, 3H), 7.26 (t, J = 8.94 Hz, 1H), 6.93 (d, J = 8.80 Hz, 1H), 5.56 (d, J = 7.70 Hz, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 1H), 4.10 (t, J = 6.19 Hz, 2H), 3.79-3.92 (m, 2H), 2.20-2.33 (m, 2H), 1.68-1.83 (m, 2H), 1.45-1.65 (m, 4H). 13 1 305 F CF3 H R = 602.3 HNMR (500 MHz, DMSO- S-isomer OCH CH 2 3 d ) δ 9.38-9.54 (m, 1H), 7.63 F C O (d, J = 13.20 Hz, 1H), 7.46 (d, J = 8.80 Hz, 1H), 7.24 (t, J = 8.80 Hz, 1H), 6.96 (d, J = 8.53 Hz, 2H), 6.79 (d, J = 8.53 Hz, 2H), 4.09 (t, J = 6.33 Hz, 2H), 3.98 (q, J = 6.88 Hz, 2H), 2.20-2.34 (m, 2H), 1.77 (quin, J = 6.81 Hz, 2H), 1.46-1.61 (m, 4H), 1.28 (t, J = 7.02 Hz, 3H). 13 1 306 CF H R = CH CF 640.2 HNMR (500 MHz, DMSO- F 3 2 3 S-isomer d ) δ 9.56 (br. s., 1H), 7.65 F C O 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 (d, J = 12.93 Hz, 1H), 7.49 (d, J = 8.80 Hz, 1H), 7.20- 7.28 (m, 3H), 7.03 (d, J = 7.98 Hz, 2H), 4.10 (t, J = 6.33 Hz, 2H), 3.71-3.81 (m, 2H), 3.61 (q, J = 11.55 Hz, 2H), 2.20-2.33 (m, 2H), 1.78 (quin, J = 6.74 Hz, 2H), 1.44-1.61 (m, 4H) 307 CF H R = OCHF 484.1 7.19 (d, J = 8.8 Hz, 2H), S-isomer 7.12 (d, J = 8.6 Hz, 2H), 6.78 (br. s., 1H), 6.56 (t, J = 73.1 Hz, 1H), 3.37 (AB quartet, J = 18.9 Hz, 2H), 2.24 (t, J = 7.0 Hz, 2H), 1.52 (quin, J = 7.3 Hz, 2H), 1.41 - 1.21 (m, 6H), 0.87 (t, J = 6.8 Hz, 3H). 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 308 CF H R = 462.1 7.10 (d, J = 9.0 Hz, 2H), S-isomer OCH CH 6.85 (d, J = 8.8 Hz, 2H), 6.72 (s, 1H), 4.04 (q, J = 7.0 Hz, 2H), 3.37 (AB quartet, J = 18.9 Hz, 2H), 2.22 (t, J = 7.2 Hz, 2H), 1.51 (quin, J = 7.3 Hz, 2H), 1.42 (t, J = 7.0 Hz, 3H), 1.39 - 1.22 (m, 6H), 0.87 (t, J = 6.6 Hz, 3H). 12 1 309 CF H R R13 = 612.1 HNMR (500 MHz, DMSO- S-isomer d ) δ 9.52 (br. s., 1H), 7.63 F C O (d, J = 13.20 Hz, 1H), 7.46 (d, J = 8.53 Hz, 1H), 7.25 (t, J = 8.94 Hz, 1H), 6.88 (d, J = 7.98 Hz, 1H), 6.83 (s, 1H), 6.58 (d, J = 7.98 Hz, 1H), 4.09 (t, J = 6.19 Hz, 2H), 3.64-3.75 (m, 2H), 2.54-2.67 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 (m, 4H), 2.20-2.34 (m, 2H), 1.77 (quin, J = 6.74 Hz, 2H), 1.67 (br. s., 4H), 1.44-1.61 (m, 4H). 13 1 310 CF3 H R = CH3 569.0 HNMR (400 MHz, MeOD) S-isomer δ 7.56 (d, J = 8.8 Hz, 2H), F C O 7.26 - 7.21 (m, 2H), 7.18 - 7.12 (m, 2H), 6.99 (d, J = 9.0 Hz, 2H), 4.07 (t, J = 6.2 Hz, 2H), 3.73 (d, J = 17.2 Hz, 1H), 3.51 (d, J = 16.9 Hz, 1H), 2.43 - 2.32 (m, 2H), 2.29 (s, 3H), 2.08 - 1.98 (m, 2H). 13 1 311 CF H R = CH 475.1 HNMR (400 MHz, MeOD) S-isomer δ 7.34 (d, J = 8.1 Hz, 2H), 7.19 (d, J = 7.9 Hz, 2H), 3.37 (s, 2H), 2.34 - 2.26 (m, 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 5H), 1.53 (d, J = 7.3 Hz, 2H), 1.42 (br. s., 2H), 1.34 - 1.25 (m, 4H), 0.92 - 0.86 (m, 3H). 312 CF3 H R = 529.4 7.73 (br. s., 1H), 7.31 (d, J = S-isomer OCH CH 8.6 Hz, 2H), 6.92 (d, J = 8.8 Hz, 2H), 5.91 (s, 1H), 4.06 (q, J = 7.0 Hz, 2H), 3.86 (s, 2H), 3.30, 3.28 (ABq, J = 18.3 Hz, 2H), 2.99 (s, 3H), 2.23 (t, J = 7.0 Hz, 2H), 1.52 (quin, J = 7.3 Hz, 2H), 1.43 (t, J = 6.9 Hz, 3H), 1.41 - 1.22 (m, 7H), 0.89 (t, J = 6.9 Hz, 2H). 313 CF H R = OCHF 551.0 7.85 (br. s., 1H), 7.38 (d, J = S-isomer 8.6 Hz, 2H), 7.17 (d, J = 8.6 Hz, 2H), 6.55 (t, J = 72.4 11841-PCT INFORMAL 3 4 1 1 2 6 11 15 Example R = R [M+H] HNMR (400 MHz, CDCl ) R R R R -R 3 Hz, 1H), 5.97 (s, 1H), 3.84 (s, 2H), 3.30, 3.28 (ABq, J = 17.8 Hz, 2H), 2.89 (s, 3H), 2.24 (t, J = 7.0 Hz, 2H), 1.52 (dt, J = 14.6, 7.2 Hz, 2H), 1.43 - 1.23 (m, 6H), 0.89 (t, J = 6.8 Hz, 3H). 314 CF H R = CH 540.3 3 N 3 S-isomer F C O 11841-PCT The term 'comprising' as used in this specification and claims means 'consisting at least in part of'. When interpreting statements in this specification and claims which include 'comprising'; other features besides the features prefaced by this term in each statement can also be present. Related terms such as 'comprise' and 'comprised' are to be interpreted in a similar manner. 11841-PCT

Claims (44)

What we claim is:
1. A compound of Formula (I): 12 14 5 or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof, wherein: designates a single or double bond; x and y can be both a single bond; when x is a double bond, then y is a single 4 16 5 bond and R and R are absent; when y is a double bond, then x is a single bond and R 10 and R are absent; R is independently selected from the group consisting of: -CONH(C alkyl), 4-18 -CONHC haloalkyl, -CONH(CH ) Ph, -CONHCH COC alkyl, 2-8 2 1-8 2 2-8 -(CH ) -(C carbocycle substituted with 0-2 R and 0-2 R ), -(CH ) -(5- to 2 m 3-10 2 m 6-membered heteroaryl comprising: carbon atoms and 1-4 heteroatoms selected from N, e b g 15 NR , O and S; wherein said heteroaryl is substituted with 0-1 R and 0-2 R ), and a C hydrocarbon chain substituted with 0-3 R ; wherein said hydrocarbon chain may be 1-12 straight or branched, saturated or unsaturated; R is independently selected from the group consisting of: C alkyl, C cycloalkyl, and C haloalkyl; 3-4 1-4 20 R is independently selected from the group consisting of: H, F, Cl, C alkyl and R and R are independently selected from the group consisting of: H, F, Cl, and C alkyl; when x is a single bond, R and R may be combined with the carbon atom to 25 which they are attached to form a 3- to 6-membered carbocycle; 11841-PCT R is independently selected from the group consisting of: H, halo, C alkyl, NO , R , -(CH ) -(X) -(CH ) R , NH , -CONH(C alkyl), -NHCOX SO R , 2 2 n t 2 m 2 1-6 1 2 j j j -NHCOCH PO(OEt) , -NHCOCOR , -NHCOCH(OH)R , -NHCOCH COR , 2 2 2 j f j -NHCONHR , and -OCONR R ; 5 X is independently selected from the group consisting of: O, S, NH, CONH, and NHCO; X is independently C hydrocarbon chain optionally substituted with C alkyl 1 1-4 1-4 or C cycloalkyl; when y is a single bond, R and R may be combined with the carbon atom to 10 which they are attached to form a 3- to 6-membered carbocycle; 11 12 13 14 15 R , R , R , R and R are independently selected from the group consisting of: H, halo, C alkyl substituted with 0-2 R , C alkoxy, C haloalkyl, 1-4 1-4 1-4 f j j j C haloalkoxy, -(CH ) -C cycloalkyl, CN, NR R , OR , SR , NHCO (C alkyl), 1-4 2 m 3-6 2 1-4 NHSO (C alkyl), and a 4- to 6-membered heterocycle comprising: carbon atoms and 2 1-4 15 1-4 heteroatoms selected from N, NR , O, and S; 11 12 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5- to 6-membered carbocyclic ring or a 5- to 6-membered heterocyclic ring comprising: carbon atoms and 1-3 heteroatoms selected from N, NR , O, and S; 12 13 20 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5- to 6-membered carbocyclic ring or a 5- to 6-membered heterocyclic ring comprising: carbon atoms and 1-3 heteroatoms selected from N, NR , O, and S; R is independently selected from the group consisting of: H and C alkyl; 25 R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkoxy, C haloalkyl, C haloalkoxy, N(C alkyl) , 1-6 1-6 1-6 1-4 2 -(CH ) -(X) -(CH ) R , and -(CH ) -(CH O) -(CH ) R ; 2 n t 2 m 2 n 2 m 2 n R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, C alkylthio, 1-10 1-10 1-10 1-10 1-10 11841-PCT C haloalkylthio, N(C alkyl) , -CONH(CH ) H, -O(CH ) O(C alkyl), R , 1-10 1-4 2 2 4-20 2 s 1-6 -(CH ) -(X) -(CH ) R , and -(CH ) -(CH O) -(CH ) R ; 2 n t 2 m 2 n 2 m 2 n R is, at each occurrence, independently selected from the group consisting of: C cycloalkyl substituted with 0-2 R , C cycloalkenyl substituted with 0-2 R , 3-6 3-6 5 -(CH ) -(phenyl substituted with 0-3 R ), and a 5- to 6-membered heterocycle comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S; wherein said heterocycle is substituted with 0-2 R ; R is, at each occurrence, independently selected from the group consisting of: halo, OH, CN, NO , C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, tetrazolyl, 2 1-4 1-4 1-4 1-4 10 OBn and phenyl substituted with 0-2 R ; R is, at each occurrence, independently selected from the group consisting of: H, C alkyl, C haloalkyl, benzyl optionally substituted with C alkoxy, CO(C alkyl) 1-8 1-8 1-4 1-4 and COBn; R is, at each occurrence, independently selected from the group consisting of: H 15 and C alkyl; g h i R , R and R are, at each occurrence, independently selected from the group consisting of: halo, C alkyl, C alkoxy, C haloalkyl, and C haloalkoxy; 1-4 1-4 1-4 1-4 R is, at each occurrence, independently selected from the group consisting of: 20 C alkyl, C cycloalkyl and phenyl; n, at each occurrence, is independently 0 or 1; m, at each occurrence, is independently 0, 1, 2, 3, or 4 s, at each occurrence, is independently 1, 2, or 3; and 25 t, at each occurrence, is independently 0 or 1; provided that the following compound is excluded: Me H
2. A compound according to claim 1, wherein: 11841-PCT R is independently selected from the group consisting of: -CONHC alkyl, 4-18 -CONH(CH ) Ph, C alkyl substituted with 0-2 R , C alkenyl substituted with 2 1-8 1-12 1-12 a a b 0-2 R , C alkynyl substituted with 0-2 R , -(CH ) -(phenyl substituted with 0-1 R 1-12 2 m and 0-2 R ), -(CH ) -(C cycloalkyl substituted with 0-1 R ), and -(CH ) -(5- to 2 m 3-6 2 m 5 6-membered heteroaryl substituted with 0-1 R and 0-2 R ), wherein said heteroaryl is selected from: pyridyl, oxazolyl, thiazolyl and .
3. A compound according to claim 1 or claim 2, wherein: 11 15 R and R are independently selected from the group consisting of: H, C 10 alkyl and halo; 12 14 R and R are independently selected from the group consisting of: H, halo, C alkyl and C alkoxy; and 1-4 1-4 R is independently selected from the group consisting of: H, halo, C alkyl substituted with 0-1 R , C alkoxy, C haloalkyl, C haloalkoxy, -(CH ) -C 1-4 1-4 1-4 2 m 3-4 f j j 15 cycloalkyl, CN, NR R , SR , NHCO (C alkyl), NHSO (C alkyl), and a 4- to 2 1-4 2 1-4 6-membered heterocycle comprising: carbon atoms and 1-4 heteroatoms selected from N, NR , O, and S.
4. A compound according to any one of claims 1 to 3, wherein the compound is of 20 Formula (II): (II) or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
5. A compound according to any one of claims 1 to 4, wherein: 11841-PCT R is independently selected from the group consisting of: C alkyl, C cycloalkyl, -CONHC alkyl, -CONHC haloalkyl, -CONH(CH ) Ph, 3-6 4-18 2-8 2 1-8 -(CH ) -(phenyl substituted with 1 R and 0-2 R ), and a 5- to 6-membered heteroaryl substituted with 0-1 R and 0-2 R , wherein said heteroaryl is selected from: pyridyl, 5 oxazolyl, thiazolyl and ; R is independently selected from the group consisting of: C alkyl and C haloalkyl; R is independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: H and F; 10 R is independently selected from the group consisting of: NH , -CONH(C alkyl), R , -(CH ) -(X) -(CH ) R , -NHCO(CH )SO (C alkyl), 1-6 2 n t 2 m 2 2 1-4 -NHCOCH PO(OEt) , -NHCOCO(C alkyl), -NHCOCH(OH)(C alkyl), 2 2 1-4 1-4 -NHCOCH CO(C alkyl), -NHCONH(C alkyl), and -OCONH(C alkyl); 2 1-4 1-4 1-4 11 15 R and R are independently selected from the group consisting of: H, C 15 alkyl and halo; 12 14 R and R are independently selected from the group consisting of: H, halo, C alkyl and C alkoxy; 1-4 1-4 R is independently selected from the group consisting of: H, halo, C alkyl substituted with 0-1 C alkoxy, C alkoxy, C haloalkyl, C haloalkoxy, 1-4 1-4 1-4 1-4 20 -(CH ) -C cycloalkyl, CN, N(C alkyl) , NHCO (C alkyl), NHSO (C alkyl), 2 m 3-4 1-4 2 2 1-4 2 1-4 pyrazolyl, and morpholinyl; 12 13 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5- to 6-membered carbocyclic ring or a 5- to 6-membered heterocyclic ring comprising: carbon atoms and 1-3 heteroatoms selected from N, NR , 25 O, and S; R is, at each occurrence, independently selected from the group consisting of: halo, OH, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) O(C 1-8 1-8 1-8 1-10 2 s 1-6 alkyl), 11841-PCT N(C alkyl) , -CONH(CH ) H, -(CH ) (C cycloalkyl), -(CH ) (C 1-4 2 2 6-20 2 m 3-6 2 m 4-6 cycloalkenyl), -O(CH ) (C cycloalkyl), 4-C alkoxy-Ph, -O(CH ) Ph, morpholinyl, 2 m 3-6 1-4 2 m pyridyl, 2-C alkoxy-pyridinyl, pyrimidinyl, pyrazinyl, and -O-pyrimidinyl; R is, at each occurrence, independently selected from the group consisting of: 5 halo, C alkyl, C alkoxy, C haloalkyl, and C haloalkoxy; 1-4 1-4 1-4 1-4 m, at each occurrence, is independently 0, 1, 2 or 3; and s, at each occurrence, is independently 1, 2, or 3. 10
6. A compound according to any one of claims 1 to 5, wherein: R is independently selected from the group consisting of: C alkyl, -CONHC alkyl, -CONH(CH ) Ph, and ; 4-18 2 1-8 R is independently selected from the group consisting of: NH , -CONH(C alkyl), 2 1-6 c c c -NHCOCH PO(OEt) , -NHCO(CH )SO (C alkyl), R , OR , -CONHR , and 2 2 2 2 1-4 15 -NHCOR ; R is independently selected from the group consisting of: H, halo, C alkyl and C alkoxy; R is independently selected from the group consisting of: H, halo, C alkyl substituted with 0-1 C alkoxy, C alkoxy, C haloalkyl, C haloalkoxy, 1-4 1-4 1-4 1-4 20 -(CH ) -C cycloalkyl, CN, N(C alkyl) , NHCO (C alkyl), NHSO (C alkyl), 2 m 3-4 1-4 2 2 1-4 2 1-4 pyrazolyl, and morpholinyl; 12 13 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5- to 6-membered carbocyclic ring or a 5- to 6-membered saturated heterocyclic ring comprising: carbon atoms and 1-2 oxygen atoms; 25 R is independently selected from the group consisting of: H and C alkoxy; R is, at each occurrence, independently selected from the group consisting of: halo, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, -O(CH ) O(C alkyl), 1-6 1-6 1-6 1-10 2 s 1-6 -CONH(CH ) H, -(CH ) (C cycloalkyl), -(CH ) (C cycloalkenyl), 2 6-20 2 m 3-6 2 m 4-6 11841-PCT -O(CH ) (C cycloalkyl), phenoxy, benzoxy, morpholinyl, 2-C alkoxy-pyridinyl, 2 m 3-6 1-4 pyrimidinyl, pyrazinyl and -O-pyrimidinyl; and R is, at each occurrence, independently selected from the group consisting of: C cycloalkyl substituted with 0-2 R , -(CH ) -(phenyl substituted with 0-3 R ), and a 3-6 2 m 5 heteroaryl selected from: oxazolyl, isoxazolyl, thiazolyl, pyrazolyl, imidazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, and pyrazinyl; wherein said heteroaryl is substituted with 0-2 R .
7. A compound according to any one of claims 1 to 6, wherein: 10 R is independently selected from the group consisting of: CF and Me; R is independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: H and F; R is independently selected from the group consisting of: NH , -CONHMe, OPh, -CONH(cyclopropyl), -CONH(cyclobutyl), -CONH(cyclopentyl), -CONH(cyclohexyl), 15 -CONHPh, -CONH(4-F-Ph), -CONH(2-Cl-Ph), -CONH(4-Cl-Ph), -CONH(4-Me-Ph), -CONH(4-OH-Ph), -CONH(3-OMe-Ph), -CONH(4-OMe-Ph), -CONH(4-CF -Ph), -CONH(4-OCF -Ph), -CONH(1-Me-pyrazolyl), -CONH(4-(1H-tetrazolyl)-Ph), -CONH(4-(2H-tetrazolyl)-Ph), -CONH(3-FMe-Ph), -CONH(3-FOMe-Ph), -CONH(CH ) Ph, -CONH(5-OMe-pyridyl), -CONH(6-OMe-pyridyl), 20 -CONH(5-OMe-pyrazinyl), -CONH(6-OMe-pyridazinyl), -NHCO(CH )SO Me, -NHCOPh, -NHCO(2-Me-Ph), -NHCO(3-Me-Ph), -NHCO(4-Me-Ph), -NHCO(2-Cl-Ph), -NHCO(3-Cl-Ph), -NHCO(2-ClF-Ph), -NHCO(2-ClF-Ph), -NHCO(isoxazolyl), -NHCO(3-Me-isoxazolyl), -NHCO(4-Me-isoxazolyl), -NHCO(3-OMe-isoxazolyl), -NHCO(3-Br-isoxazolyl), 25 -NHCO(3-(2-Cl-Ph)-isoxazolyl), -NHCO(3-(3-F-Ph)-isoxazolyl), -NHCO(3-OBn-isoxazolyl), 1H-imidazolyl, -NHCO(5-Me-1,3,4-oxadiazolyl), -NHCO(1-Me-1,2,3-triazolyl), -NHCO(6-OMe-pyridyl), -NHCO(6-Cl-pyridazinyl), 5-CF -1,3,4-oxadiazolyl, 1H-tetrazolyl, 1H-tetrazolyl, and 2H-tetrazolyl; 11841-PCT 11 15 R and R are independently selected from the group consisting of: H, Me, F, and Cl; R is independently selected from the group consisting of: H, F, Cl, Me and OMe; 5 R is independently selected from the group consisting of: H, F, Cl, Br, Me, OMe, OEt, CH OMe, CF , CH CF , OCHF , OCF , CN, N(Me) , cyclopropyl and 2 3 2 3 2 3 2 cyclopropylmethyl; 12 13 alternatively, R and R , together with the carbon atoms to which they are attached, combine to form a 5- to 6-membered carbocyclic ring or a 5- to 6-membered 10 saturated heterocyclic ring comprising: carbon atoms and 1-2 oxygen atoms; R is H; R is, at each occurrence, independently selected from the group consisting of: n-pentyl, methoxy, n-butoxy, i-butoxy, i-pentoxy, -O(CH ) CF , -O(CH ) CF CF , 2 1-6 3 2 1-4 2 3 -CONH(CH ) H, cyclopropyl, cyclopentenyl, cyclohexenyl, 2 6-20 15 -O(CH ) (cyclopentyl), phenoxy, benzoxy, pyrimidinyl, pyrazinyl and -O-pyrimidinyl; and R is F.
8. A compound according to Claim 4 or Claim 5, wherein: 20 R is ; R is independently selected from CF and CH ; 6 c c c R is independently selected from: R , -CONHR , -NHCOR , and -NHCOCH SO (C alkyl); 2 2 1-4 R is independently selected from: -O(CH ) CF , -O(CH ) CF CF , 2 1-6 3 2 1-4 2 3 25 -CONH(CH ) H, cyclopentenyl, cyclohexenyl, -O(CH ) (cyclopentyl), 2 6-20 2 2 phenoxy, benzoxy, pyrimidinyl, pyrazinyl and -O-pyrimidinyl; R is, at each occurrence, independently selected from the group consisting of: -(CH ) -(phenyl substituted with 0-3 R ), and a heteroaryl selected from: oxazolyl, 11841-PCT isoxazolyl, pyrazolyl, imidazolyl, oxadiazolyl, triazolyl, tetrazolyl, pyridyl, and pyrazinyl; wherein said heteroaryl is substituted with 0-2 R ; and R is, at each occurrence, independently selected from the group consisting of: halo, OH, CN, C alkyl, C alkoxy, C haloalkyl, C haloalkoxy, tetrazolyl and 1-4 1-4 1-4 1-4 5 OBn.
9. A compound according to any one of claims 1 to 6, or 8, wherein: R is independently selected from the group consisting of: H, halo, C alkyl substituted with 0-1 C alkoxy, C alkoxy, C haloalkyl, C haloalkoxy, CN or 1-4 1-4 1-4 1-4 10 C cycloalkyl.
10. A compound according to any one of claims 1 to 5, 8, or 9, wherein: R is independently . 15
11. A compound according to any one of claims 1 to 10, wherein: R is independently selected from: -O(CH ) CF , and -O(CH ) CF CF . 2 1-6 3 2 1-4 2 3
12. A compound according to any one of claims 1 to 5, wherein: R is independently 5-membered nitrogen heteroaryl.
13. A compound according to any one of claims 1 to 12, wherein: R is independently: 1H-imidazolyl, 1H-tetrazolyl, 1H-tetrazolyl, or 2H-tetrazolyl. 25
14. A compound according to claim 1, wherein the compound is selected from: 3-(1H-Tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)- 5,6-dihydropyridin-2(1H)-one; (S)(1H-Tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one; 11841-PCT (R)(1H-Tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one; N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoro- methyl)-1,2,5,6-tetrahydropyridinecarboxamide; 5 (R)-N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoro-methyl)-1,2,5,6-tetrahydropyridinecarboxamide; (S)-N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoro-methyl)-1,2,5,6-tetrahydropyridinecarboxamide; (S)(2H-Tetrazolyl)p-tolyl(4-(6,6,6-trifluorohexyloxy)phenyl) 10 (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one; 3-(2-Ethyl-2H-tetrazolyl)p-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)-5,6-dihydropyridin-2(1H)-one; 4-p-Tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)(5-(trifluoromethyl)- 1,3,4-oxadiazolyl)-5,6-dihydropyridin-2(1H)-one; 15 6-Methyloxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)-N-(4-(trifluoromethoxy)- phenyl)-1,2,5,6-tetrahydropyridinecarboxamide; 3-(2H-Tetrazolyl)p-tolyl(trifluoromethyl)(1-(5,5,5-trifluoropentyl)-1H- pyrazolyl)-5,6-dihydropyridin-2(1H)-one; 3-Nitrop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)-5,6- 20 dihydropyridin-2(1H)-one; N-(4-Methoxyphenyl)oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) (trifluoromethyl)piperidinecarboxamide; compounds of formula (IIa) 12 14 11 15 (IIa) 1 2 6 wherein R , R and R are the substituents set out in the table below, and 11 15 wherein R to R are hydrogen, unless otherwise noted in the table below: 1 2 6 11 15 R R R R -R Rac CH R = CH N 13 Rac CF R = CH F 13 Rac CF R = CH F 13 Rac CF R = CH F 13 Rac CF R = CH F 13 Rac CF R = CH Rac CF R = CH F 13 Rac CF R = CH N O H F 13 Rac CF R = CH Rac CF R = CH F C O 1 2 6 11 15 R R R R -R Rac CF O R = CH F C O Rac CF R = CH F C O Rac CF R = CH F C O Rac CF R = CH F C O Rac CF R = CH F C O H Rac CF R = CH N Cl F C O Rac CF Cl R = CH F C O Rac CF H R = CH F C O Rac CF CH R = CH F C O Rac CF R = CH O CF F C O 1 2 6 11 15 R R R R -R Rac CF O R = CH O CH F C O Rac CF CH R = CH F C O Rac CF CF R = CH F C O Rac CF R = CH S-isomer F C O Rac CF O R = CH O CH F C O Rac CF H R = CH F C O Rac CF H OCH CH F C O Rac CF H OCHF F C O S-isomer CF O R = CH O CF F C O S-isomer CF H R = F R = OCH F C O 1 2 6 11 15 R R R R -R Rac CF H R = CH F C O Rac CF H All H F C O Rac CF H R = CF F C O Rac CF H R = Cl F C O Rac CF H R = OCH F C O Rac CF H R = Cl F C O N -(4-Methoxyphenyl)methyloxop-tolyl-N -(4,4,4-trifluorobutyl)-1,2,3,6- tetrahydropyridine-2,5-dicarboxamide; N-(4-Cyanophenyl)-5,5-difluorooxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl) 5 (trifluoromethyl)-1,2,5,6-tetrahydropyridinecarboxamide; (S)Aminop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)-5,6- dihydropyridin-2(1H)-one; (S)Methyl-N-(2-oxop-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)- 1,2,5,6-tetrahydropyridinyl)benzamide; 10 (S)Phenoxyp-tolyl(4-(4,4,4-trifluorobutoxy)phenyl)(trifluoromethyl)-5,6- dihydropyridin-2(1H)-one; and compounds of formula (II) 12 14 11 15 (II) 1 2 3 4 6 wherein R , R , R , R and R are the substituents set out in the table below, and 1 15 wherein R to R are hydrogen, unless otherwise noted in the table below: 1 2 6 11 15 R = R R R R R -R N OMe S-isomer CF H 13 3 R = CH F C O S-isomer CF H 13 3 R = CH F C O Rac CF H 13 3 R = CH S-isomer CF H 13 3 R = CH F C O S-isomer CF H 13 3 R = CH F C O S-isomer CF H R = CH F C O NH 13 Rac CF H N R = CH F C O 1 2 6 11 15 R = R R R R R -R NH 13 Rac CF H R = CH NH 13 Rac CF H N R = CH O 13 S-isomer CF H R = CH F C O O 13 S-isomer CF H R = CH F C O S-isomer CF H R = CH F C O H OH 13 S-isomer CF H R = CH F C O H O 13 S-isomer CF H R = CH F C O Rac CF F R = CH F C O NH 13 Rac CF H N R = CH NH 13 Rac CF H N R = CH 1 2 6 11 15 R = R R R R R -R NH 13 Rac CF H R = CH S-isomer CF H R = CH S-isomer CF H R = CH S-isomer CF H R = CH S-isomer CF H R = CH N 13 S-isomer CF H R = CH OMe 13 Rac CF F R = CH Rac CF F R = CH Rac CF F R = CH F C N N 13 S-isomer CF H N R = CH 1 2 6 11 15 R = R R R R R -R S-isomer CF H R = CH O OCF N OMe 13 Rac CF F R = CH F C O Rac CF F R = CH F C O N 11 S-isomer CF H N R = F F C O N 13 S-isomer CF H N R = F C O OMe 11 S-isomer CF H R = CH R = CH F C O N 13 S-isomer CF H N R = OCHF F C O N 13 S-isomer CF F O R = CH F C O N 13 S-isomer CF H R = CH F C O N 13 S-isomer CF H R = CH F C O N 13 S-isomer CF H R = CH F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H R = CH F C O H OMe 13 Rac CH H R = CH N 11 S-isomer CF H R = CH R = CH F C O S-isomer CF H R = CH F C O Cl 13 S-isomer CF H R = CH F C O N 13 Rac CF F R = CH F C O H OMe 13 Rac CH H R = CH OMe 13 Rac CF F R = CH OMe 13 Rac CF F R = CH F C O OMe 13 Rac CF F R = CH 1 2 6 11 15 R = R R R R R -R F 13 S-isomer CF H R = CH H OMe 13 Rac CH H R = CH S-isomer CF H OHCF F C O Cl 13 S-isomer CF H R = CH F C O S-isomer CF H R = CH F C O Cl F 13 S-isomer CF H R = CH F C O Cl 13 S-isomer CF H R = CH F C O S-isomer CF H F C O N OMe 13 Rac CH H R = CH H OMe 13 Rac CH H R = CH 1 2 6 11 15 R = R R R R R -R OMe 13 Rac CH H R = CH S-isomer CF H N OCH F C O H OMe 13 Rac CH H R = CH Br 13 S-isomer CF H R = CH F C O S-isomer CF H R = CH F C O OMe 13 Rac CF F R = CH NH 13 Rac CF H N R = CH N 13 Rac CF H 3 R = CH S-isomer F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H 3 R = CH F C O S-isomer CF H N R = CH F C O S-isomer CF H 3 R = CH F C O H OMe 13 Rac CH H 3 R = CH OMe 13 Rac CH H 3 R = CH S-isomer CF H 3 R = CH F C O O OMe 13 Rac CH H 3 R = CH N 13 S-isomer CF H N R = CH F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H 3 R = F C O Rac CF H 3 R = CH Rac CF H 3 R = CH NH 13 R-isomer CF H 3 R = CH NH 13 S-isomer CF H N R = CH S-isomer CF H 3 R = CH S-isomer CF H 3 R = CH OCF 13 Rac CF H 3 3 R = CH OHCF 13 Rac CF H 2 3 R = CH NH 13 S-isomer CF H 3 R = CH2OCH F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H R = F F C O OCH CH NH 13 S-isomer CF3 H R = CH F C N S-isomer CF H 3 R = OCHF F C O S-isomer CF H 3 R = CH F C O NH 13 S-isomer CF3 H R R = F C O S-isomer CF H 3 R = OCH CH F C O NH 13 S-isomer CF3 H R R = F C O S-isomer CF H 3 R R = F C O S-isomer CF H 3 R R = F C O S-isomer CF H 3 R R = F C O 1 2 6 11 15 R = R R R R R -R R = OCH NH 13 S-isomer CHF2 H R = CH F C O OMe 13 R-isomer H H R = CH F C O OMe 13 Rac CF H 3 R = CH OMe 13 Rac CF3 H R = CH S-isomer CF H 3 R = CH F C O OMe 13 S-isomer CF H 3 R = CH F C O S-isomer CF H R = CH S-isomer CF H 3 R = Br F C O S-isomer CF H N 13 F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H 3 R = F C O N S-isomer CF H 3 R R = F C O NH 13 S-isomer CF3 H F C O H 13 S-isomer CF H 3 R = CH F C O S-isomer CF H 3 R = F C O S-isomer CF H 3 R = F C N S-isomer CF H 3 R = OCF NH 13 S-isomer CF3 H R = OCF F C O S-isomer CF H 3 R = F C O S-isomer CF H 3 R = OCHCF F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H 3 R = CH F C O S-isomer CF H 3 R = F C O S-isomer CF H 3 R = OCHCF F C O F 13 S-isomer CF H 3 R = CH F C O S-isomer CF H 3 R = CH F C O H 13 S-isomer CF H 3 R = OCHCF F C O H 13 S-isomer CF H 3 R = F C O S-isomer CF H 3 R = CH F C O H H 13 S-isomer CF H OCHCF F C O H 13 S-isomer CF H 3 R = CH F C O S-isomer CF H R = CH F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H R = CH F C O S-isomer CF H R = CH F C O N S-isomer CF H R = CH F C O R-isomer CF H R = CH F C O S-isomer CF H R = CH CF F C O S-isomer CF H R = CH N OMe S-isomer CF H R = CH S-isomer CF H R = OCHF F C O S-isomer CF H R = OCHF F C O 12 13 S-isomer CF H R R = F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H R = F C O S-isomer CF H R = CH S-isomer CF H R = CH S-isomer CF3 H R = OCHF F C O S-isomer CF H R = CH F C O S-isomer CF H R = CH F C O S-isomer CF3 H R = OCH CH F C O S-isomer CF H R = CH CF F C O S-isomer CF H R = CH S-isomer CF H R = F R = CH F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H R = CH F C O S-isomer CF H R = CH S-isomer CF H R = CH S-isomer CF H R = CH S-isomer CF H R = CH Rac CF H R = CH S-isomer CF H R = CH S-isomer CF H R = OCH S-isomer CF H R = OCH S-isomer CF H R = F R = CH F C O 1 2 6 11 15 R = R R R R R -R S-isomer CF H O R = CH F C O S-isomer CF H R = CH S-isomer CF H R = CH F C O S-isomer CF H R = CH S-isomer CF H R = OCHF F C O S-isomer CF H R = CH S-isomer CF H R = CH F 3 3 F C O S-isomer CF H R R13 = F C O S-isomer CF H R = OCH CH F C O S-isomer CF H R = CH CF F C O S-isomer CF H R = OCHF 1 2 6 11 15 R = R R R R R -R S-isomer CF H R = OCH CH S-isomer CF H R R13 = F C O S-isomer CF H R = CH F C O S-isomer CF3 H R = CH3 S-isomer CF H R = OCH CH S-isomer CF H R = OCHF S-isomer CF H R = CH F C O or a stereoisomer, a tautomer, a pharmaceutically acceptable salt, or a solvate thereof.
15. A compound according to claim 1, wherein the compound has the following 5 formula: or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof.
16. A compound according to claim 15, wherein the compound has the following formula: 5 or a tautomer, or a pharmaceutically acceptable salt thereof.
17. A compound according to claim 15, wherein the compound is:
18. A pharmaceutically acceptable salt of the compound according to claim 15 or claim 16.
19. A compound according to claim 1, wherein the compound has the following 15 formula: or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof.
20. A compound according to claim 19, wherein the compound has the following 5 formula: or a tautomer, or a pharmaceutically acceptable salt thereof.
21. A compound according to claim 19, wherein the compound is: 10 .
22. A compound according to claim 1, wherein the compound has the following formula: 15 or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof.
23. A compound according to claim 22, wherein the compound has the following formula: or a tautomer, or a pharmaceutically acceptable salt thereof.
24. A compound according to claim 22, wherein the compound is:
25. A compound according to claim 1, wherein the compound has the following 10 formula: or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof.
26. A compound according to claim 25, wherein the compound has the following 15 formula: or a tautomer, or a pharmaceutically acceptable salt thereof.
27. A compound according to claim 25, wherein the compound is:
28. A compound according to claim 1, wherein the compound has the following formula: 10 or a stereoisomer, a tautomer, or a pharmaceutically acceptable salt thereof.
29. A compound according to claim 28, wherein the compound has the following formula: or a tautomer, or a pharmaceutically acceptable salt thereof.
30. A compound according to claim 28, wherein the compound is:
31. A pharmaceutical composition, comprising: a pharmaceutically acceptable carrier and a compound of any one of claims 1 to 30, a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof, optionally in combination 10 simultaneously, separately or sequentially with one or more additional therapeutic agents.
32. The pharmaceutical composition according to claim 31, further comprising one or more additional therapeutic agents selected from: anti-diabetic agents, anti-hyperglycemic agents, anti-hyperinsulinemic agents, anti-retinopathic agents, 15 anti-neuropathic agents, anti-nephropathic agents, anti-atherosclerotic agents, anti-ischemic agents, anti-hypertensive agents, anti-obesity agents, anti-dyslipidemic agents, anti-hyperlipidemic agents, anti-hypertriglyceridemic agents, anti-hypercholesterolemic agents, anti-restenotic agents, lipid lowering agents, anorectic agents, and appetite suppressants.
33. The pharmaceutical composition according to claim 31, further comprising one or more other suitable therapeutic agents selected from: a dipeptidyl peptidase-IV inhibitor, a sodium-glucose transporter-2 inhibitor and a 11b-HSD-1 inhibitor. 5
34. A compound of any one of claims 1 to 30, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof, for use in therapy.
35. A compound of any one of claims 1 to 30, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof, for use in preventing, modulating or 10 treating diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, nonalcoholic fatty liver disease (NAFLD), retinopathy, neuropathy, nephropathy, delayed wound healing, atherosclerosis and its sequelae, abnormal heart function, myocardial ischemia, stroke, Metabolic Syndrome, hypertension, obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, 15 hypercholesterolemia, low high-density lipoprotein (HDL), high low-density lipoprotein (LDL), non-cardiac ischemia, lipid disorders, or glaucoma.
36. A compound for use according to claim 35, wherein the NAFLD is nonalcoholic steatohepatitis (NASH).
37. A compound for use according to any one of claims 34 to 36, wherein the compound is for use simultaneously, separately or sequentially with one or more additional therapeutic agents. 25
38. Use of a compound of any one of claims 1 to 30, or a stereoisomer, a tautomer, a pharmaceutically acceptable salt or a solvate thereof, for the manufacture of a medicament for preventing, modulating or treating diabetes, hyperglycemia, impaired glucose tolerance, gestational diabetes, insulin resistance, hyperinsulinemia, nonalcoholic fatty liver disease (NAFLD), retinopathy, neuropathy, nephropathy, delayed wound 30 healing, atherosclerosis and its sequelae, abnormal heart function, myocardial ischemia, stroke, Metabolic Syndrome, hypertension, obesity, dyslipidemia, hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, low high-density lipoprotein (HDL), high low-density lipoprotein (LDL), non-cardiac ischemia, lipid disorders, or glaucoma.
39. Use according to claim 38, wherein the NAFLD is nonalcoholic steatohepatitis 5 (NASH).
40. Use according to claim 38 or 39, wherein the medicament is for use simultaneously, separately or sequentially with one or more additional therapeutic agents. 10
41. A compound of any one of claims 1 to 30 substantially as herein described with reference to any example thereof.
42. A pharmaceutical composition of any one of claims 31 to 33 substantially as herein described with reference to any example thereof.
43. A compound of any one of claims 1 to 30 for use according to any one of claims 34 to 37 substantially as herein described with reference to any example thereof.
44. Use of any one of claims 38 to 40 substantially as herein described with reference 20 to any example thereof.
NZ626751A 2011-12-02 2012-11-30 Aryl dihydropyridinones and piperidinones as mgat2 inhibitors NZ626751B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161566039P 2011-12-02 2011-12-02
US61/566,039 2011-12-02
US13/688,584 2012-11-29
US13/688,584 US8791091B2 (en) 2011-12-02 2012-11-29 Aryl dihydropyridinone and piperidinone MGAT2 inhibitors
PCT/US2012/067173 WO2013082345A1 (en) 2011-12-02 2012-11-30 Aryl dihydropyridinone and piperidinone as mgat2 inhibitors

Publications (2)

Publication Number Publication Date
NZ626751A NZ626751A (en) 2016-07-29
NZ626751B2 true NZ626751B2 (en) 2016-11-01

Family

ID=

Similar Documents

Publication Publication Date Title
US10966967B2 (en) Aryl dihydropyridinones and piperidinone MGAT2 inhibitors
EP3114121B1 (en) Dihydropyridinone mgat2 inhibitors for use in the treatment of metabolic disorders
US9822074B2 (en) Dihydropyridinone MGAT2 inhibitors
CA2941951C (en) Tetrazolone-substituted dihydropyridinone mgat2 inhibitors
NZ626751B2 (en) Aryl dihydropyridinones and piperidinones as mgat2 inhibitors