NZ614776B2 - Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith - Google Patents

Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith Download PDF

Info

Publication number
NZ614776B2
NZ614776B2 NZ614776A NZ61477612A NZ614776B2 NZ 614776 B2 NZ614776 B2 NZ 614776B2 NZ 614776 A NZ614776 A NZ 614776A NZ 61477612 A NZ61477612 A NZ 61477612A NZ 614776 B2 NZ614776 B2 NZ 614776B2
Authority
NZ
New Zealand
Prior art keywords
alkyl
substituted
compound
cycloalkyl
mmol
Prior art date
Application number
NZ614776A
Other versions
NZ614776A (en
Inventor
Brydon L Bennett
Mercedes Delgado
Jan Elsner
Paul Erdman
Robert Hilgraf
Laurie Ann Lebrun
Meg Mccarrick
Mehran F Moghaddam
Mark A Nagy
Stephen Norris
Original Assignee
Signal Pharmaceuticals Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Signal Pharmaceuticals Llc filed Critical Signal Pharmaceuticals Llc
Priority claimed from PCT/US2012/034349 external-priority patent/WO2012145569A1/en
Publication of NZ614776A publication Critical patent/NZ614776A/en
Publication of NZ614776B2 publication Critical patent/NZ614776B2/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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular 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
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/38One sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • 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
    • C07D401/12Heterocyclic 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
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D405/02Heterocyclic 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 two hetero rings
    • C07D405/12Heterocyclic 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 two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

Provided are substituted diaminocarboxamide pyrimidine compounds, of general formula (I), and substituted diaminocarbonitrile pyrimidine compounds, of general formula (IB), where the variables are as defined in the specification. Examples of the compounds include 4-(isopropylamino)-2-((1R,4R)-4-(2,2,2-trifluoroethoxycyclohexylamino)pyrimidine-5-carboxamide and 2-((1R,4S)-4-ethoxycyclohexylamino)-4-((1S,3R)-3-hydroxycyclohexyl-amino)pyrimidine-5-carbonitrile. The compounds are JNK1/JNK2 inhibitors. The compounds may be useful in the treatment of liver fibrotic disorders such as non-alcoholic steatohepatitis, steatosis, cirrhosis, primary sclerosing cholangitis, primary biliary cirrhosis, hepatitis, hepatocellular carcinoma, or liver fibrosis coincident with chronic or repeated alcohol ingestion, with infection, with liver transplant, or with drug induced liver injury. ,2-trifluoroethoxycyclohexylamino)pyrimidine-5-carboxamide and 2-((1R,4S)-4-ethoxycyclohexylamino)-4-((1S,3R)-3-hydroxycyclohexyl-amino)pyrimidine-5-carbonitrile. The compounds are JNK1/JNK2 inhibitors. The compounds may be useful in the treatment of liver fibrotic disorders such as non-alcoholic steatohepatitis, steatosis, cirrhosis, primary sclerosing cholangitis, primary biliary cirrhosis, hepatitis, hepatocellular carcinoma, or liver fibrosis coincident with chronic or repeated alcohol ingestion, with infection, with liver transplant, or with drug induced liver injury.

Description

SUBSTITUTED DIAMINOCARBOXAMIDE AND DIAMINOCARBONITRILE PYRIMIDINES, COMPOSITIONS F, AND METHODS OF ENT THEREWITH Provided herein are certain diaminocarboxamide and diaminocarbonitrile pyrimidine compounds, compositions comprising an effective amount of such compounds, and methods for treating or preventing liver f1brotic disorders or a condition treatable or preventable by inhibition of a JNK pathway, comprising stering an ive amount of such diaminocarboxamide and diaminocarbonitrile pyrimidine compounds to a subject in need thereof.
OUND The connection between abnormal protein phosphorylation and the cause or consequence of diseases has been known for over 20 years. Accordingly, protein kinases have become a very important group of drug targets. [See Cohen, Nature, 1:309-315 (2002), Gaestel et al. Carr.Med.Chem.14: 23 (2007); Grimminger et al. Nat. Rev.
Drag Disc. 9(12):956-970 (2010)]. Various protein kinase inhibitors have been used clinically in the treatment of a wide variety of diseases, such as cancer and chronic inflammatory diseases, including rheumatoid arthritis and psoriasis. [See Cohen, EW. J.
Biochem. 268:5001-5010 (2001); Protein Kinase Inhibitors for the ent of Disease: The Promise and the Problems, Handbook ofExperimental Pharmacology, Springer Berlin Heidelberg, 167 (2005)].
JNK is a tously expressed serine/threonine kinase belonging, er with ERK (extracellular-regulated kinase) and p38, to the family of mitogen-activated protein s (MAPKs). [Kyriakis JM, Sci. STKE e1 (2000); Whitmarsh AJ, et al.
Sci. STKE (1):pe1 (1999); Schramek H, News Physio]. Sci.17:62-7 (2002); Ichijo H, Oncogene 18(45):6087-93 (1999)]. MAPKs are important mediators of signal transduction from the cell surface to the nucleus, using phosphorylation cascades to te a coordinated response by a cell to an external stimulus by phosphorylation of ed intracellular proteins, including transcription factors. Additionally, JNK also phosphorylates non-nuclear proteins, for example, IRS-1, and Bcl-2 family members.
ATI—25 14175v1 [Davis R], Trends Biochem. Sci. 9(11):470-473 (1994); Seger R et al., FASEB J.; 9(9):726-35 (1995); Fanger GR et al., Curr. Opin. Genet. Dev.; 7(1):67-74 (1997)].
The mitogen activated protein (MAP) s ipate in the uction of signals to the nucleus of the cell in response to extracellular stimuli. Examples ofMAP kinases from the ERK p38 and JNK isoforms include but are not limited to, mitogen- activated n kinase 1 (ERK2), mitogen-activated protein kinase 8 (JNKl), mitogen- activated protein kinase 9 (MAPK9 or JNK2), mitogen-activated protein kinase 10 (MAPK10 or JNK3) and mitogen-activated protein kinase 14 (MAPK14 or p3 8alpha).
MAP kinases are a family of proline-directed /threonine kinases that mediate signal transduction from ellular receptors or heat shock, osmotic stress, reactive oxidant species (ROS) or UV radiation. [See Sridhar et al., Pharmaceutical Research, 17: 11 1345- 1353 (2000)]. MAP kinases are activated via the phosphorylation of theonine and tyrosine by upstream dual-specificity protein kinases, including MK and MEKK kinases. Cell proliferation and differentiation have been shown to be under the regulatory control of multiple MAP kinase cascades. [See Sridhar et al., Pharmaceutical Research, 17:11 1345- 1353 (2000)]. As such, the MAP kinase pathway plays critical roles in a number of disease states. For e, s in activities ofMAP kinases have been shown to lead to aberrant cell proliferation and carcinogenesis. [See Hu et al., Cell Growth Difi’er. -200 (2000); and Das et al., Breast Cancer Res. Treat. 40: 141 (1996)]. Moreover, MAP kinase activity has also been ated in insulin resistance ated with type-2 diabetes [See Virkamaki et al., J. Clin. Invest. 103:931-943 (1999)] and obesity. s in insulin resistance may have a direct impact on the metabolism of glucose and lipid in the liver contributing to the development of steatosis that may progress to liver fibrosis rie et al. Science Translational Medicine 2(60):1-7 (2010)].
Steatosis may p in the presence of either saturated or unsaturated free fatty acids (FFA). FFA promote robust JNK activation in liver and excessive concentrations of FFA may lead to hepatocyte apoptosis. It has been reported that JNK2-/— mice are partially protected from steatosis and apoptosis by saturated FFA (e.g. stearic acid) but not by unsaturated FFA [Malhi et al. J.Bi0l. Chem. 281 :12093-12101 (2006)].
JNK1-/— mice were not protected from FFA induced injury. The role of JNK1 and JNK2 has been studied in CDAA-fed mice that progressed from steatosis to hepatitis to ATI—25 14175v1 WO 45569 hepatic fibrosis [Kodama et al., Gastroenterology 137:1467-1477 (2009)]. While both JNKl-/— and JNK2-/— mice developed steatosis, the JNKl-/— mice, but not JNK2-/— mice, were remarkably resistant to progression to tis and fibrosis. Chimeric mice with — deletion restricted to bone marrow cells were similarly resistant to hepatitis and fibrosis implicating the activated Kupffer cell as a key trigger for e progression beyond steatosis. Indeed, JNKl-/— macrophages do not express IL-1, IL-6, TNF and NO in response to LPS [Sanchez-Tillo et al., JBiol Chem. 282(17): 73 (2007)], and Kupffer cells derived fiom JNKl -/- mice or fiom wild-type mice and treated with JNK inhibitor SP600125 display reduced TNF, IL-6, and IL-1 expression in response to LPS [Kodama et al., Gastroenterology 137: 1467-1477 (2009)].
The ation of the intricacy of protein kinase ys and the complexity of the relationship and interaction among and between the various protein kinases and kinase pathways ghts the importance of developing pharmaceutical agents capable of acting as n kinase modulators, regulators or inhibitors that have ial activity on multiple kinases or multiple kinase pathways. Accordingly, there remains a need for new kinase modulators, and in particular JNK modulators.
Citation or identification of any reference in Section 2 of this application is not to be construed as an admission that the reference is prior art to the t application.
SUMMARY Provided herein are compounds having the following formula (I): N \ NH2 JL , HN N NH R1 R2 and pharmaceutically acceptable salts, tautomers, isotopologues, stereoisomers and prodrugs thereof, wherein R1, and R2 are as defined herein.
Provided herein are compounds having the following formula (1B): ATI—2514l75vl H[\l N NH R3 R4 (13) and pharmaceutically acceptable salts, tautomers, isotopologues, isomers and prodrugs thereof, wherein R3 and R4 are as defined herein.
A compound of formula (I) or formula (IE) or a ceutically acceptable salt, tautomer, isotopologue, stereoisomer or prodrug thereof (each being ed to herein as an “Diaminopyrimidine Compound”) is useful for treating or ting liver f1brotic disorders, or diabetes and/or metabolic syndrome leading to liver f1brotic ers, as described herein. In another , a Diaminopyrimidine Compound is useful for treating or preventing a condition treatable or preventable by inhibition of a JNK pathway, as described herein.
In one aspect, provided herein are Diaminopyrimidine Compounds as bed in the instant disclosure, such as, for example, in Tables 1, 2 and 3.
In one aspect, provided herein are pharmaceutical itions comprising an effective amount of a Diaminopyrimidine Compound as described herein, and a pharmaceutically acceptable carrier, excipient or vehicle. In some embodiments the pharmaceutical composition is suitable for oral, parenteral, mucosal, transdermal or topical administration.
In one aspect, provided herein are methods for treating or preventing liver f1brotic disorders, such as non-alcoholic steatohepatitis, steatosis, cirrhosis, primary sclerosing cholangitis, primary biliary cirrhosis, hepatitis, hepatocellular oma, and liver fibrosis coincident with chronic or repeated l ingestion, with infection, with liver lant, or with drug d liver injury, comprising administering to a subject in need thereof an effective amount of a Diaminopyrimidine Compound as described herein; and a pharmaceutically acceptable carrier, excipient or vehicle. In another aspect, provided herein are methods for treating or preventing diabetes or lic syndrome leading to liver f1brotic disorders, such as non-alcoholic steatohepatitis, steatosis, tis, or cirrhosis, comprising administering to a subject in need thereof an effective amount of a Diaminopyrimidine Compound. Also provided herein are methods for treating or ATI—2514l75vl preventing a condition treatable or table by inhibition of a JNK y, comprising administering to a t in need thereof an effective amount of a Diaminopyrimidine Compound as described herein; and a pharmaceutically acceptable carrier, excipient or vehicle. [0013a] In another aspect, provided herein is the use of a compound as described herein in the manufacture of a medicament for the treatment of a liver fibrotic disorder, or diabetes or metabolic syndrome leading to a liver fibrotic er.
In one aspect, provided herein are methods for ting a kinase, for example JNK1, JNK2 or both, in a cell sing said kinase, comprising contacting said cell with an effective amount of a Diaminopyrimidine Compound as described herein. [0014a] In another aspect provided herein is the use of a compound as described herein in the manufacture of a ment for inhibiting a JNK1 or JNK2 kinase.
In another aspect provided herein are methods for preparing Diaminopyrimidine Compounds as bed herein.
The present embodiments can be understood more fully by reference to the detailed description and examples, which are intended to exemplify non-limiting embodiments.
ED DESCRIPTION TIONS An ” group is a saturated, partially saturated, or unsaturated straight chain or branched non-cyclic hydrocarbon having from 1 to 10 carbon atoms, typically from 1 to 8 carbons or, in some embodiments, from 1 to 6, 1 to 4, or 2 to 6 or carbon atoms. Representative alkyl groups include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl and -n-hexyl; while saturated branched alkyls include -isopropyl, utyl, tyl, -tert-butyl, -isopentyl, -neopentyl, tert-pentyl, methylpentyl, methylpentyl, methylpentyl, -2,3-dimethylbutyl and the like. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, allyl, -CH=CH(CH3), -CH=C(CH3)2, -C(CH3)=CH2, -C(CH3)=CH(CH3), -C(CH2CH3)=CH2, -C≡CH, -C≡C(CH3), -C≡C(CH2CH3), CH, -CH2C≡C(CH3) and -CH2C≡C(CH7CH3), among others. An alkyl group can be tuted or unsubstituted. When the alkyl groups described herein are said to be “substituted,” they may be substituted with any substituent or substituents as those found in the exemplary compounds and embodiments disclosed herein, as well as halogen (chloro, iodo, bromo, or fluoro); alkyl; hydroxyl; alkoxy; alkoxyalkyl; amino; alkylamino; carboxy; nitro; cyano; thiol; thioether; imine; imide; e; guanidine; enamine; aminocarbonyl; acylamino; phosphonato; phosphine; thiocarbonyl; sulfonyl; sulfone; WO 45569 sulfonamide; ketone; de; ester; urea; ne; oxime; hydroxyl amine; alkoxyamine; aralkoxyamine; e; hydrazine; ide; hydrazone; azide; isocyanate; isothiocyanate; e; thiocyanate; B(OH)2, or O(alkyl)aminocarbonyl.
A “cycloalkyl” group is a saturated, partially saturated, or unsaturated cyclic alkyl group of from 3 to 10 carbon atoms haVing a single cyclic ring or multiple condensed or bridged rings which can be optionally substituted with from 1 to 3 alkyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, whereas in other ments the number of ring carbon atoms ranges from 3 to 5, 3 to 6, or 3 to 7.
Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, l-methylcyclopropyl, 2-methylcyclopentyl, 2-methylcyclooctyl, and the like, or multiple or bridged ring structures such as l-bicyclo[l . ntyl, bicyclo[2.l.l]hexyl, bicyclo[2.2. l]heptyl, bicyclo[2.2.2]octyl, adamantyl and the like. Examples of unsaturared cycloalkyl groups include cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, hexadienyl, among others. A cycloalkyl group can be substituted or unsubstituted. Such substituted cycloalkyl groups include, by way of example, cyclohexanol and the like.
An “aryl” group is an aromatic carbocyclic group of from 6 to 14 carbon atoms haVing a single ring (e.g., phenyl) or multiple sed rings (e.g., naphthyl or anthryl). In some embodiments, aryl groups contain 6-l4 carbons, and in others from 6 to 12 or even 6 to 10 carbon atoms in the ring portions of the groups. Particular aryls include phenyl, biphenyl, naphthyl and the like. An aryl group can be substituted or unsubstituted.
The phrase “aryl groups” also includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like).
A “heteroaryl” group is an aryl ring system haVing one to four heteroatoms as ring atoms in a aromatic ring system, wherein the remainder of the atoms are carbon atoms. In some embodiments, heteroaryl groups n 3 to 6 ring atoms, and in others from 6 to 9 or even 6 to 10 atoms in the ring portions of the groups. Suitable heteroatoms include oxygen, sulfur and en. In certain embodiments, the heteroaryl ring system is monocyclic or bicyclic. Non-limiting examples include but are not limited to, groups such as pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, ATI—ZS 14175Vl isoxazolyl, benzisoxazolyl (e.g., benzo[d]isoxazolyl), thiazolyl, pyrolyl, pyridazinyl, pyrimidyl, pyrazinyl, thiophenyl, benzothiophenyl, furanyl, benzofuranyl, indolyl (e.g., indolylonyl or isoindolin-l-onyl), azaindolyl (pyrrolopyridyl or lH-pyrrolo[2,3- b]pyridyl), lyl, benzimidazolyl (e.g., lH-benzo[d]imidazolyl), imidazopyridyl (e.g., azabenzimidazolyl or lH-imidazo[4,5-b]pyridyl), pyrazolopyridyl, triazolopyridyl, benzotriazolyl (e. g., lH-benzo[d][l,2,3]triazolyl), benzoxazolyl (e. g., benzo[d]oxazolyl), hiazolyl, benzothiadiazolyl, isoxazolopyridyl, phthalenyl, purinyl, xanthinyl, adeninyl, yl, quinolinyl, isoquinolinyl (e. g., 3,4-dihydroisoquinolin-l(2H)-onyl), tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl groups.
A “heterocyclyl” is an aromatic (also referred to as heteroaryl) or non- aromatic cycloalkyl in which one to four of the ring carbon atoms are independently replaced with a heteroatom from the group consisting of O, S and N. In some embodiments, heterocyclyl groups include 3 tolO ring members, Whereas other such groups have 3 to 5, 3 to 6, or 3 to 8 ring members. Heterocyclyls can also be bonded to other groups at any ring atom (i.e., at any carbon atom or heteroatom of the heterocyclic ring). A heterocycloalkyl group can be substituted or unsubstituted. Heterocyclyl groups encompass unsaturated, partially saturated and ted ring systems, such as, for example, imidazolyl, olinyl and imidazolidinyl (e. g., imidazolidinone or imidazolidin-2,4-dionyl) groups. The phrase heterocyclyl includes fused ring species, ing those comprising fiased aromatic and non-aromatic groups, such as, for example, l-and 2-aminotetraline, benzotriazolyl (e. g., lH-benzo[d][l,2,3]triazolyl), benzimidazolyl (e.g., lH-benzo[d]imidazolyl), 2,3-dihydrobenzo[l,4]dioxinyl, and benzo[l,3]dioxolyl. The phrase also includes bridged polycyclic ring systems ning a heteroatom such as, but not limited to, quinuclidyl. Representative examples of a heterocyclyl group include, but are not d to, aziridinyl, azetidinyl, azepanyl, yl, idyl, imidazolidinyl (e. g., olidinonyl or imidazolidin-2,4-dionyl), pyrazolidinyl, thiazolidinyl, ydrothiophenyl, tetrahydrofuranyl, yl, furanyl, thiophenyl, pyrrolyl, pyrrolinyl, imidazolyl, imidazolinyl, pyrazolyl, pyrazolinyl, triazolyl, tetrazolyl, oxazolyl, olyl, benzisoxazolyl (e. g., benzo[d]isoxazolyl), thiazolyl, thiazolinyl, isothiazolyl, thiadiazolyl, oxadiazolyl, piperidyl, piperazinyl (e. g., piperazinonyl), morpholinyl, thiomorpholinyl, tetrahydropyranyl (e.g., tetrahydro-2H-pyranyl), tetrahydrothiopyranyl, oxathianyl, dioxyl, ATI—ZS 14175Vl dithianyl, pyranyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, nyl, dihydropyridyl, dihydrodithiinyl, dihydrodithionyl, l,4-dioxaspiro[4.5]decanyl, homopiperazinyl, quinuclidyl, indolyl (e.g. , indolylonyl or isoindolin- l -onyl), indolinyl, isoindolyl, isoindolinyl, azaindolyl (pyrrolopyridyl or lH-pyrrolo[2,3-b]pyridyl), indazolyl, indolizinyl, benzotriazolyl (e. g. lH-benzo[d][l,2,3]triazolyl), benzimidazolyl (e.g., zo[d]imidazolyl or lH-benzo[d]imidazol-2(3H)—onyl), benzofilranyl, benzothiophenyl, benzothiazolyl, benzoxadiazolyl, benzoxazinyl, benzodithiinyl, benzoxathiinyl, benzothiazinyl, benzoxazolyl (i.e., benzo[d]oxazolyl), benzothiazolyl, benzothiadiazolyl, benzo[l,3]dioxolyl, pyrazolopyridyl (for example, lH-pyrazolo[3,4- b]pyridyl, lH-pyrazolo[4,3-b]pyridyl), opyridyl (e. g., zimidazolyl or dazo[4,5-b]pyridyl), triazolopyridyl, isoxazolopyridyl, purinyl, nyl, adeninyl, guaninyl, quinolinyl, isoquinolinyl (e. g., 3,4-dihydroisoquinolin-l(2H)-onyl), quinolizinyl, quinoxalinyl, quinazolinyl, cinnolinyl, azinyl, naphthyridinyl, pteridinyl, thianaphthalenyl, dihydrobenzothiazinyl, dihydrobenzofuranyl, dihydroindolyl, dihydrobenzodioxinyl, tetrahydroindolyl, tetrahydroindazolyl, tetrahydrobenzimidazolyl, tetrahydrobenzotriazolyl, tetrahydropyrrolopyridyl, tetrahydropyrazolopyridyl, tetrahydroimidazopyridyl, tetrahydrotriazolopyridyl, ydropyrimidin-2(lH)-one and tetrahydroquinolinyl groups. Representative non-aromatic heterocyclyl groups do not include fused ring species that comprise a fused aromatic group. Examples of non- ic heterocyclyl groups include aziridinyl, azetidinyl, azepanyl, pyrrolidyl, imidazolidinyl (e.g., imidazolidinonyl or imidazolidin-2,4-dionyl), pyrazolidinyl, thiazolidinyl, tetrahydrothiophenyl, tetrahydrofilranyl, piperidyl, piperazinyl (e. g., zinonyl), morpholinyl, thiomorpholinyl, tetrahydropyranyl (e.g., tetrahydro- 2H-pyranyl), tetrahydrothiopyranyl, oxathianyl, dithianyl, l,4-dioxaspiro[4.5]decanyl, homopiperazinyl, quinuclidyl, or tetrahydropyrimidin-2(lH)—one. Representative substituted heterocyclyl groups may be mono-substituted or substituted more than once, such as, but not limited to, pyridyl or morpholinyl groups, which are 2-, 3-, 4-, 5-, or 6-substituted, or disubstituted with various substituents such as those listed below.
A “cycloalkylalkyl” group is a radical of the formula: -cycloalkyl, wherein alkyl and cycloalkyl are as defined above. Substituted lkylalkyl groups may be substituted at the alkyl, the cycloalkyl, or both the alkyl and the cycloalkyl ATI—2514l75v1 portions of the group. Representative cycloalkylalkyl groups include but are not limited to cyclopropyl, methylcyclobutyl, cyclopentyl, methylcyclohexyl, ethylcyclopropyl, ethylcyclobutyl, yclopentyl, ethylcyclohexyl, propylcyclopentyl, propylcyclohexyl and the like.
An “aralkyl” group is a radical of the formula: -alkyl-aryl, wherein alkyl and aryl are defined above. Substituted aralkyl groups may be substituted at the alkyl, the aryl, or both the alkyl and the aryl portions of the group. Representative aralkyl groups e but are not d to benzyl and phenethyl groups and fused (cycloalkylaryl)alkyl groups such as 4-ethyl-indanyl.
An “heterocyclylalkyl” group is a radical of the formula: - heterocyclyl, wherein alkyl and cyclyl are defined above. Substituted heterocyclylalkyl groups may be substituted at the alkyl, the heterocyclyl, or both the alkyl and the heterocyclyl portions of the group. Representative heterocylylalkyl groups include but are not limited to 4-ethyl-morpholinyl, 4-propylmorpholinyl, 2-yl methyl, furan-3 -yl methyl, pyridin-3 -yl methyl, ydrofidranyl ethyl, and indolyl propyl.
A “halogen” is fluorine, chlorine, bromine or iodine.
A “hydroxyalkyl” group is an alkyl group as bed above substituted with one or more hydroxy groups.
An “alkoxy” group is -O-(alkyl), wherein alkyl is defined above.
An “alkoxyalkyl” group is -(alkyl)-O-(alkyl), wherein alkyl is defined above.
An “amino” group is a radical of the formula: -NH2.
An “alkylamino” group is a radical of the formula: -NH-alkyl or —N(alkyl)2, wherein each alkyl is independently as defined above. [003 l] A “carboxy” group is a radical of the formula: -C(O)OH.
An “aminocarbonyl” group is a radical of the formula: -C(O)N(R#)2, -C(O)NH(R#) or -C(O)NH2, wherein each R# is independently a substituted or unsubstituted alkyl, cycloalkyl, aryl, aralkyl, heterocyclyl or heterocyclyl group as defined herein.
An “acylamino” group is a radical of the formula: -NHC(O)( R#) or -N(alkyl)C(O)(R#), wherein each alkyl and R# are independently as defined above.
ATI—2514l75v1 A “sulfonylamino” group is a radical of the formula: -NHSOZ(R#) or -N(alkyl)SOz(R#), wherein each alkyl and R# are defined above.
A “urea” group is a radical of the formula: -N(alkyl)C(O)N(R#)2, -N(a1kyl)C(O)NH(R#), —N(alkyl)C(O)NH2, -NHC(O)N(R#)2, -NHC(O)NH(R#), or -NH(CO)NHR#, wherein each alkyl and R# are independently as defined above.
When the groups described , with the exception of alkyl group, are said to be “substituted,” they may be substituted with any appropriate substituent or substituents. Illustrative examples of substituents are those found in the exemplary compounds and embodiments disclosed , as well as halogen (chloro, iodo, bromo, or fluoro); alkyl; hydroxyl; alkoxy; alkyl; amino; alkylamino; carboxy; nitro; cyano; thiol; her; imine; imide; amidine; guanidine; enamine; aminocarbonyl; acylamino; phosphonato; phosphine; rbonyl; sulfonyl; sulfone; sulfonamide; ketone; aldehyde; ester; urea; urethane; oxime; hydroxyl amine; alkoxyamine; aralkoxyamine; N—oxide; hydrazine; hydrazide; hydrazone; azide; isocyanate; isothiocyanate; cyanate; thiocyanate; oxygen (=0); B(OH)2, O(alkyl)aminocarbonyl; lkyl, which may be monocyclic or fused or non-fused polycyclic (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or a heterocyclyl, which may be monocyclic or fused or non-fused polycyclic (e. g., idyl, piperidyl, piperazinyl, morpholinyl, or thiazinyl); monocyclic or fused or non-fused polycyclic aryl or aryl (e.g., phenyl, yl, pyrrolyl, indolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, lyl, tetrazolyl, pyrazolyl, pyridyl, quinolinyl, isoquinolinyl, acridinyl, pyrazinyl, pyridazinyl, pyrimidyl, benzimidazolyl, benzothiophenyl, or benzofuranyl) aryloxy; aralkyloxy; heterocyclyloxy; and heterocyclyl alkoxy. [003 7] As used herein, the term “Diaminopyrimidine Compound” refers to compounds of a (I) and formula (1B) as well as to further embodiments provided . In one ment, a “Diaminopyrimidine Compound” is a compound set forth in Tables 1, 2 and 3. The term “Diaminopyrimidine Compound” includes pharmaceutically acceptable salts, tautomers, isotopologues, stereoisomers, and prodrugs of the compounds provided herein.
As used herein, the term “pharmaceutically acceptable salt(s)” refers to a salt prepared from a ceutically acceptable non-toxic acid or base including an ATI—2514l75v1 nic acid and base and an organic acid and base. Suitable ceutically acceptable base addition salts of the compounds of formula (I) include, but are not limited to metallic salts made from aluminum, calcium, m, magnesium, potassium, sodium and zinc or organic salts made from lysine, N,N’-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, ine (N-methylglucamine ) and procaine. Suitable non-toxic acids include, but are not limited to, inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, , filmaric, furoic, galacturonic, gluconic, glucuronic, glutamic, glycolic, romic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phenylacetic, phosphoric, propionic, salicylic, stearic, succinic, ilic, sulfilric, ic acid, and p- toluenesulfonic acid. Specific non-toxic acids include hloric, romic, maleic, phosphoric, sulfiaric, and methanesulfonic acids. Examples of c salts thus include hydrochloride and mesylate salts. Others are well-known in the art, see for example, Remington ’s Pharmaceutical Sciences, 18th eds., Mack Publishing, Easton PA (1990) or Remington: The Science and Practice ofPharmacy, l9th eds., Mack Publishing, Easton PA (1 995).
As used herein and unless otherwise indicated, the term “prodrug” means a Diaminopyrimidine nd that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound, particularly a Diaminopyrimidine Compound or a compound of formula (1). Examples of prodrugs include, but are not limited to, derivatives and lites of a Diaminopyrimidine Compound that e biohydrolyzable moieties such as biohydrolyzable amides, rolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. In certain embodiments, prodrugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid. The carboxylate esters are iently formed by esterifying any of the carboxylic acid moieties present on the molecule. Prodrugs can typically be prepared using well-known methods, such as those described by Burger ’s Medicinal Chemistry and Drug Discovery 6th ed. (Donald J. Abraham ed., 2001, Wiley) ATI—2514l75v1 and Design and Application ofProdrugs (H. Bundgaard ed., 1985, Harwood Academic Publishers Gmih).
As used herein and unless otherwise indicated, the term “stereoisomer” or “stereomerically pure” means one stereoisomer of a Diaminopyrimidine Compound that is substantially free of other stereoisomers of that compound. For example, a stereomerically pure nd having one chiral center will be substantially free of the opposite enantiomer of the nd. A stereomerically pure compound having two chiral centers will be ntially free of other reomers of the compound. A typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, or r than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound. The Diaminopyrimidine nds can have chiral centers and can occur as racemates, individual enantiomers or diastereomers, and mixtures thereof. All such isomeric forms are included within the embodiments disclosed herein, including mixtures thereof.
The use of stereomerically pure forms of such opyrimidine Compounds, as well as the use of mixtures of those forms, are encompassed by the embodiments disclosed herein. For e, mixtures comprising equal or unequal amounts of the enantiomers of a particular Diaminopyrimidine Compound may be used in methods and compositions disclosed herein. These isomers may be asymmetrically synthesized or ed using standard techniques such as chiral columns or chiral resolving agents. See, e. g., Jacques, J et al., Enantiomers, tes and Resolutions (Wiley-Interscience, New York, 1981); Wilen, S. H., et al., edron 33 :2725 (1977); Eliel, E. L., Stereochemistry ofCarbon Compounds (McGraw-Hill, NY, 1962); and Wilen, S. H., Tables ofResolving Agents and Optical Resolutions p. 268 (EL. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN, 1972).
ATI—25 14175v1 WO 45569 It should also be noted the Diaminopyrimidine Compounds can include E and Z isomers, or a mixture thereof, and cis and trans isomers or a mixture f. In certain embodiments, the Diaminopyrimidine Compounds are isolated as either the E or Z isomer. In other embodiments, the Diaminopyrimidine nds are a mixture of the E and Z isomers.
"Tautomers" refers to isomeric forms of a compound that are in equilibrium with each other. The concentrations of the isomeric forms will depend on the environment the nd is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution. For e, in aqueous solution, pyrazoles may exhibit the ing isomeric forms, which are referred to as tautomers of each other: IN\ IN / N\ | As readily understood by one skilled in the art, a wide variety of functional groups and other stuctures may exhibit tautomerism and all tautomers of compounds of formula (I) are within the scope of the present invention.
It should also be noted the Diaminopyrimidine Compounds can n unnatural proportions of atomic isotopes at one or more of the atoms. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (1251), sulfur-35 (358), or carbon-l4 (14C), or may be ically enriched, such as with deuterium (2H), carbon-l3 (13C), or nitrogen-15 (UN). As used herein, an “isotopologue” is an isotopically ed compound. The term “isotopically enriched” refers to an atom having an isotopic composition other than the natural isotopic composition of that atom. “Isotopically enriched” may also refer to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom. The term “isotopic composition” refers to the amount of each isotope present for a given atom. Radiolabeled and isotopically encriched compounds are useful as therapeutic agents, e.g., cancer and inflammation therapeutic agents, research reagents, e.g., binding assay reagents, and diagnostic , e. g., in vivo imaging agents.
All isotopic variations of the Diaminopyrimidine Compounds as described , r radioactive or not, are intended to be encompassed within the scope of the embodiments ATI—2514l75v1 provided herein. In some embodiments, there are provided isotopologues of the Diaminopyrimidine Compounds, for example, the isotopologues are deuterium, carbon-l3, or en-15 enriched Diaminopyrimidine Compounds.
“INK” means a protein or an isoform thereof expressed by a INKl, INK2, or INK3 gene (Gupta, S., Barrett, T., Whitmarsh, A.I., Cavanagh, I ., Sluss, H.K., Derijard, B. and Davis, RI. The EMBO J. 0-2770 (1996)).
“Treating” as used herein, means an alleviation, in whole or in part, of a disorder, disease or condition, or one or more of the symptoms associated with a disorder, disease, or condition, or g or halting of further progression or worsening of those symptoms, or alleviating or eradicating the cause(s) of the disorder, disease, or condition itself. In one embodiment, the er is a liver f1brotic disorder, such as non-alcoholic steatohepatitis, steatosis (i.e. fatty , cirrhosis, y sclerosing cholangitis, primary biliary cirrhosis, hepatitis, hepatocellular carcinoma, or liver fibrosis coincident with chronic or repeated alcohol ingestion (alcoholic hepatitis), with infection (e.g. viral ion such as HCV), with liver transplant, or with drug induced liver injury (e.g. acetaminophen ty). In some embodiments, “treating” means an alleviation, in whole or in part, of a disorder, disease or condition, or symptoms associated with es or metabolic syndrome leading to liver fibrotic disorders, such as non-alcoholic steatohepatitis, sis (i.e. fatty liver), hepatitis or sis, or a g, or halting of further progression or ing of those symptoms. In one embodiment, the symptom is jaundice. In another embodiment, “treating” means and alleviation, in whole or in part, of a er, disease or condition, or symptoms ated with a condition, treatable or preventable by inhibition of a INK y.
“Preventing” as used herein, means a method of delaying and/or precluding the onset, recurrence or spread, in whole or in part, of a disorder, disease or condition; barring a subject from acquiring a disorder, disease, or condition; or reducing a subject’s risk of acquiring a disorder, disease, or condition. In one embodiment, the disorder is a liver f1brotic disorder, or diabetes or metabolic me leading to liver fibrotic disorders, as described herein, or symptoms thereof In another, the disorder is a condition, treatable or preventable by inhibition of a INK pathway.
ATI—2514l75vl The term “effective amount” in connection with a Diaminopyrimidine Compound means an amount capable of treating or preventing a disorder, disease or condition, or symptoms thereof, disclosed herein.
The term “subject” includes an , including, but not limited to, an animal such a cow, monkey, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit or guinea pig, in one ment a mammal, in another embodiment a human. In one embodiment, a subject is a human having or at risk for having liver fibrotic disorders or diabetes or metabolic syndrome leading to liver fibrotic disorders, or a condition, treatable or preventable by inhibition of a JNK pathway, or a symptom thereof DIAMINOPYRIMIDINE COMPOUNDS Provided herein are compounds having the following formula (I): N \ NH2 HN N NH R1 R2 and pharmaceutically acceptable salts, ers, stereoisomers, enantiomers, isotopologues, and prodrugs thereof, wherein: R1 is tuted or unsubstituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted non-aromatic cyclyl, substituted or unsubstituted ycloalkyl, or substituted or unsubstituted alkylheterocyclyl, provided that R1 is not l-aminocyclohexyl; and R2 is substituted or tituted C1_g alkyl, substituted or tituted saturated cycloalkyl, substituted or unsubstituted ycloalkyl, or substituted or unsubstituted non-aromatic heterocyclyl.
In one embodiment, the compound of formula (I) is not: 2-(2-aminoethylamino)(methylamino)pyrimidinecarboxamide ATI—2514l75v1 H N1*N H H 2-(2-arnin0pr0py1arnino)(cyclohexylamino)pyrirnidinecarboxarnide vwN1“H NOH 2-(2-arninooxoethy1amino)(cyclohexylamino)pyrirnidine carboxarnide H2N\n/\NJ\N/[\ll/YLNHZH NOH minoethylamino)—4-(cyc10hexy1arnino)pyrirnidinecarb0xamide H2N\/\NlYNHZ/ N NO H H (S)(2-aminopropylamino)(cyclobutylamino)pyrimidine carboxarnide ATI—ZS 14175V1 (R)(l -aminomethyloxobutanylamino)(cyclobutylamino)- pyrimidine-S-carboxamide N N/ NH \N / N NH or 2-(l -acetylpiperidinylamino)(cyclopropylamino)pyrimidinecarboxamide A O )LN/YkNHZ/ M N N H In one embodiment, R1 is not HZND\; for example, :e‘ :0 so ~30 H2N HZN‘“ H2N or HZN‘“ 9 9 , _ In some embodiments of compounds of formula (I), R1 is a branched C1_g alkyl, for example, R1 is isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, ylpentyl, or entyl. In others, R1 is isopropyl, tyl, isobutyl, tert-butyl, 2,3-dimethylbutyl, isopentyl, 2-methylpentyl, neopentyl, tert-pentyl, or 3-methylpentyl. In , R1 is a substituted or unsubstituted cycloalkyl, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[l.1.l]pentyl, or bicyclo[2.2.2]octyl. In others, the cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, ATI—ZS 14175Vl cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[l . l . l]pentyl, bicyclo[2.2. l ]heptyl, or bicyclo[2.2.2]octyl. In some such embodiments, the cycloalkyl is tuted with one or more halogen, -(C1_4 alkyl), -NR2, -(C0_3 alkyl)OR, -NRC(O)R’, ’, R2, -C(O)OR’, or -NRS(O)2R’, n each R is ndently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated. For example, the cycloalkyl is substituted with one or more methyl, ethyl, t-butyl, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, H2F, -OCH2CF3, -CH20H, -CH20CH3, )20H, -NH2, -NH(CH3), -NHC(O)CH3, -C(O)NHCH3, (CH3)2, or -NHSOZCH3. In other embodiments, the cycloalkyl is substituted with one or more halogen, -CF3, -(C1_4 alkyl), -(C1_6 cycloalkyl), -NR2, -(C0_3 alkyl)OR, -(C0_3 alkyl)OR”, -NRC(O)R’, -C(O)R’, -C(O)NR2, -C(O)OR’, or -NRS(O)2R’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally ated, each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R” is independently a C1_6 cycloalkyl, wherein the cycloalkyl is optionally fluorinated. For example, the cycloalkyl is substituted with one or more methyl, ethyl, t-butyl, ropyl, -CF3, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, -OCH2CH2F, -OCH2CF3, -O(cyclopropyl), -CH20H, -CH20CH3, -C(CH3)20H, -NH2, -NH(CH3), -NHC(O)CH3, -C(O)NHCH3, -C(O)N(CH3)2, or -NHSOZCH3. In some embodiments, R1 is a substituted or tituted non-aromatic cyclyl, for example, pyrrolidinyl, tetrahydropyranyl, l,4-dioxaspiro[4.5]decanyl, or piperidyl. In others, the non-aromatic heterocyclyl is oxetanyl, pyrrolidinyl, tetrahydropyranyl, l,4-dioxaspiro[4.5]decanyl, or piperidyl. In some such embodiments, the piperidyl is substituted with —C(O)R’, or -C(O)OR’, wherein R’ is C1_4 alkyl, wherein the alkyl is optionally fluorinated. In yet other embodiments, R1 is a substituted or unsubstituted alkylcycloalkyl, for e, (C1_3 alkyl)cyclopropyl, (C1_3 cyclobutyl, (C1_3 alkyl)cyclopentyl, or (C1_3 alkyl)cyclohexyl. In some such embodiments, R1 is -(CH2)cyclopropyl, —(CH2)cyclobutyl, —(CH2)cyclopentyl, -(CH2)cyclohexyl, -CH(CH3)cyclopropyl, —CH(CH3)cyclobutyl, -CH(CH3)cyclopentyl, or -CH(CH3)cyclohexyl. In , R1 is —(CH2)cyclopropyl, —CH(CH3)cyclopropyl, -CH(CH3)cyclobutyl,—CH(CH3)cyclohexyl, or -C(CH3)2cyclopropyl. In still other embodiments, R1 is a substituted or unsubstituted alkylheterocyclyl, for example, ATI—ZS 14175Vl -(C1_4 alkyl)tetrahydrofuranyl, -(C1_4 alkyl)dioxolanyl, (C1_4 alkyl)furanyl, (C1_3 thiophenyl, or -(C1_3 all<yl)pyridyl.
In some embodiments of compounds of formula (I), R1 is selected from branched C1-8 alkyl, nfifiee :98 Q3 “93ija JMQ wherein R3, is halogen, -CF3, -(C1_4 alkyl), -(C1_6 lkyl), -NR2, -(C0_3 alkyl)OR, -(C0_3 alkyl)OR”, -NRC(O)R’, -C(O)R’, -C(O)NR2, -C(O)OR’, or -NRS(O)2R’; R4, is —C(O)R’, or -C(O)OR’; each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally ated; each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated; each R” is independently a C1_6 cycloalkyl wherein the cycloalkyl is optionally fluorinated; and n is 0-2.
In some such embodiments, R3 is methyl, ethyl, t-butyl, cyclopropyl, -CF3, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, -OCH2CH2F, -OCH2CF3, -O(cyclopropyl), -CH20H, -CH20CH3, -C(CH3)20H, -NH2, -NH(CH3), -NHC(O)CH3, -C(O)NHCH3, -C(O)N(CH3)2, or -NHSOZCH3. [005 7] In some embodiments, R2 is a substituted or unsubstituted C1_g alkyl, for e, , ethyl, n-propyl, isopropyl, n-butyl, tyl, isobutyl, tert-butyl, tert-pentyl, isopentyl, or 2-methylpentyl. In other embodiments, R2 is methyl, ethyl, n-propyl, isopropyl, l, sec-butyl, isobutyl, tert-butyl, 3-methylbutyl, methylbutyl, 3,3-dimethylbutyl, 2,3,3-trimethylbutyl, tert-pentyl, isopentyl, 3-pentyl, ATI—2514l75Vl 3-methylpentyl, 2-methylpentyl, or 2,4-dimethylpentyl. In some such embodiments, R2 is substituted with one or more -(C1_4 alkyl), -(C0_3 alkyl)OR, -C(O)NR2 or -NRCOR’, wherein each R is ndently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, n the alkyl is optionally fluorinated. For example, R2 is substituted with one or more -OH, or -CH3. In other embodiments, R2 is substituted with one or more -OH, -OCH3, or -CH3. In some embodiments, R2 is a substituted or unsubstituted cycloalkyl, for example, R2 is cyclopropyl, utyl, cyclopentyl, cyclohexyl, cycloheptyl, or cyclooctyl. In some such embodiments, R2 is tuted with one or more -(C1_4 , -(C0_3 alkyl)OR, -(C0_3 C(O)NR2, -NR2, or -NRCOR’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is ally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated. In other embodiments, R2 is cyclopropyl, utyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, o[l.l.l]pentyl, bicyclo[2. 1.1]hexyl, or bicyclo[2.2.l]heptyl. In some such embodiments, R2 is substituted with one or more -CF3, -(C1_4 , -(C1_6 cycloalkyl), -(C0_3 alkyl)OR, -(C0_3 alkyl)C(O)NR2, -NR2, or -NRCOR’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated, and wherein the cycloalkyl is optionally ated. In some such embodiments, R2 is substituted with one or more methyl, ethyl, isopropyl, -CH20H, -OH, -OCH3, -OCH2CH3, H2, - NHC(O)CH3, or -NHC(O)CH2CH3. In others, R2 is substituted with one or more methyl, ethyl, isopropyl, -cyclopropyl, -CF3, -CH20H, -OH, -OCH3, -OCH2CH3, -C(O)NH2, - NHC(O)CH3, or -NHC(O)CH2CH3. In some embodiments, R2 is a tuted or unsubstituted alkylcycloalkyl, for example, a substituted or unsubstituted (C1_3 alkyl)cyclopropyl, (C1_3 alkyl)cyclobutyl, (C1_3 alkyl)cyclopentyl, or (C1_3 alkyl)cyclohexyl. For example, R2 is —(CH2)cyclopropyl, -(CH2)cyclobutyl, -CH(CH3)cyclopropyl, -CH(CH3)cyclobutyl, -CH(CH2CH3)cyclopropyl, -C(CH3)2cyclopropyl, or —CH2CH2cyclobutyl. In some embodiments, R2 is a substituted or unsubstituted non-aromatic heterocyclyl, for example, tetrahydrofuranyl, tetrahydropyranyl, piperidyl, piperidinonyl or l,4-dioxaspiro[4.5]decanyl. In others, R2 is oxetanyl, tetrahydrofilranyl, tetrahydropyranyl, piperidyl, piperidinonyl or ATI—ZS 14175Vl l,4-dioxaspiro[4.5]decanyl. In some such embodiments, R2 is substituted with one or more -(C1_4 alkyl), -(C0_3 alkyl)OR, or -C(O)R’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated.
In some embodiments of compounds of formula (I), R2 is a substituted or unsubstituted C1_g alkyl, JEFF“ l \ / !}L¢ a'fiL E >R5n 9 k 9 R5.“ 9 QR5H R6' 0 N/ >11; 0 :71 ‘1’.
\R5'n 2.
,Q/ i or 5- R5'n’ /”7.A R5n. ’ R R5; “ wherein R5, IS —CF3, -(C1_4 alkyl), -(C1_6 cycloalkyl), -(C0_3 alkyl)OR, -(C0_3 alkyl)C(O)NR2, -NR2, or -NRCOR’, wherein the cycloalkyl is ally fluorinated; R6, is H, or -C(O)R’; each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated; each R’ is independently C1_4 alkyl, wherein the alkyl is ally ated; and n is 0-2.
In some such embodiments, R5, is , ethyl, isopropyl, -cyclopropyl, -CF3, , -OH, -OCH3, -OCH2CH3, -C(O)NH2, )CH3, or -NHC(O)CH2CH3.
In some embodiments, R6’ is H or H3, —C(O)CH2CH3, —C(O)CH(CH3)2, or -C(O)CH2CH(CH3)2.
In some such embodiments of R2, R1 is a cycloalkyl, optionally substituted with one or more halogen, -CF3, -(C1_4 alkyl), -(C1_6 cycloalkyl), -NR2, -(C0_3 alkyl)OR, -(C0_3 alkyl)OR”, -NRC(O)R’, -C(O)R’, -C(O)NR2, -C(O)OR’, or -NRS(O)2R’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, each ATI—ZS 14175Vl R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R” is independently a C1_6 cycloalkyl., wherein the lkyl is optionally fluorinated.
Further embodiments ed herein include combinations of one or more of the particular embodiments set forth above.
Representative compounds of formula (I) are set forth in Table l.
In some embodiments, the compound is selected from Table 2. ed herein are compounds having the following formula (1B): HN N NH R3 R4 (13) and pharmaceutically acceptable salts, tautomers, stereoisomers, enantiomers, isotopologues, and prodrugs thereof, wherein: R3 is substituted or unsubstituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted omatic heterocyclyl, substituted or unsubstituted alkylcycloalkyl, or substituted or unsubstituted alkyl-(non—aromatic heterocyclyl); and R4 is substituted or tituted C1_g alkyl, tuted or unsubstituted cycloalkyl, or substituted or unsubstituted non-aromatic heterocyclyl.
In one embodiment, the compound is not 4-(isopentylamino)(3 -(2-methylpiperidin- l -yl)propylamino)pyrimidine- -carbonitrile WNAN/I A)\ H H (28,2'S)—dimethyl 2,2'-(5-cyanopyrimidine-2,4-diyl)bis(azanediyl)- bis(4-methylpentanoate) ATI—2514l75v1 HN)\ “Y/ N I N/ ”\V 0\ Mo\ 0 S)—diethy1 2,2'-(5-cyanopyrimidine-2,4-diy1)bis(azanediyl)— bis(3-methy1butanoate) \/O 'I NY|[NAN/ O WO 4-(cycloheptylamino)—2-(3-(2-methy1piperidin-1 -y1)propy1amino)- pyrimidine-5 -carbonitrile 4-(4-methy1cyclohexylamino)(3-(2-methy1piperidiny1)propy1amino)— pyrimidine-5 -carbonitrile OCANXN/I N H H ' 01‘ 2-(3-(diethy1amino)propy1amino)—4-(4-methy1cyclohexylamino)pyrimidine- -carbonitrile jMMAN/| M In one embodiment of nds of formula (1B), R3 is a branched C1_g alkyl, for example, R3 is isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, ATI—ZS 14175V1 2-methylpentyl, or tert-pentyl. In other embodiments, R3 is isopropyl, sec-butyl, isobutyl, tert-butyl, 2,3-dimethylbutyl, isopentyl, 2-methylpentyl, tyl, tert-pentyl, or ylpentyl. In some embodiments, R3 is tert-butyl. In other ments, R3 is a substituted or unsubstituted cycloalkyl, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[l.l.l]pentyl, or bicyclo[2.2.2]octyl. In some such embodiments, R3 is cyclopropyl, cyclobutyl, cyclopentyl, exyl, cycloheptyl, cyclooctyl, bicyclo[l.l.l]pentyl, bicyclo[2.2. l]heptyl, or bicyclo[2.2.2]octyl. In some embodiments, R3 is cyclobutyl or cyclohexyl. In some embodiments, the cycloalkyl is substituted with one or more halogen, -CF3, -(C1_4 alkyl), -(C1_6 cycloalkyl), -NR2, -(C0_3 alkyl)OR, -(C0_3 OR”, -NRC(O)R’, -C(O)R’, -(C0_3 alkyl)C(O)NR2, -C(O)OR’, or -NRS(O)2R’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R” is independently a C1_6 cycloalkyl, n the cycloalkyl is optionally fluorinated. For example, the cycloalkyl is tuted with one or more methyl, ethyl, t-butyl, cyclopropyl, -CF3, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, -OCH2CH2F, F3, -O(cyclopropyl), , -CH20CH3, -C(CH3)20H, -NH2, -NH(CH3), -NHC(O)CH3, -C(CH3)2C(O)N(CH3)2, -C(O)NHCH3, -C(O)N(CH3)2, or -NHSOZCH3. In other embodiments, the cycloalkyl is substituted with one or more halogen, -(C1_4 , -NR2, -(C0_3 alkyl)OR, -NRC(O)R’, -C(O)R’, -(C0_3 alkyl)C(O)NR2, -C(O)OR’, or -NRS(O)2R’, wherein each R is ndently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated. For example, the cycloalkyl is substituted with one or more methyl, ethyl, t-butyl, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, -OCH2CH2F, -OCH2CF3, -O(cyclopropyl), -CH20H, -CH20CH3, -C(CH3)20H, -NH2, -NH(CH3), -NHC(O)CH3, )2C(O)N(CH3)2, -C(O)NHCH3, -C(O)N(CH3)2, or CH3. In some embodiments, the cycloalkyl is substituted with one or more methyl, -F, -OH, -OCH3, -OCH2CH3, -OCHF2, -OCH2CF3, -NH(CH3), )CH3, -C(O)N(CH3)2, or -C(CH3)2C(O)N(CH3)2. In yet other embodiments, R3 is a substituted or unsubstituted non-aromatic heterocyclyl, for example, idinyl, tetrahydrofilranyl, tetrahydropyranyl, l,4-dioxaspiro[4.5]decanyl, or piperidyl. In some embodiments, the non-aromatic heterocyclyl is oxetanyl, tetrahydrofuranyl, ATI—2514l75vl ydropyranyl, piperidyl, piperidinonyl or l,4-dioxaspiro[4.5]decanyl. In some embodiments, R3 is tetrahydropyranyl. In some embodiments, the piperidyl is substituted with —C(O)R’, or -C(O)OR’, wherein R’ is C1_4 alkyl, n the alkyl is optionally fluorinated. In some embodiments, R3 is a substituted or unsubstituted alkylcycloalkyl, for example, (C1_3 alkyl)cyclopropyl, (C1_3 alkyl)cyclobutyl, (C1_3 alkyl)cyclopentyl, or (C1_3 alkyl)cyclohexyl. In some embodiments, R3 is -(CH2)cyclopropyl, -(CH2)cyclobutyl, -(CH2)cyclopentyl, -(CH2)cyclohexyl, -CH(CH3)cyclopropyl, -CH(CH3)cyclobutyl, -CH(CH3)cyclopentyl, or 3)cyclohexyl. In others, R3 is cyclopropyl, -CH(CH3)cyclopropyl, -CH(CH3)cyclobutyl, 3)cyclohexyl, or -C(CH3)2cyclopropyl. In some embodiments, R3 is a substituted or unsubstituted alkyl- (non-aromatic heterocyclyl), for example, -(C1_4 alkyl)tetrahydrofuranyl, or -(C1_4 alkyl)dioxolanyl.
In some embodiments of nds of formula (1B), R3 is selected from branched C1_g alkyl, J?\:nLW5%;5 '2 ‘7 E‘s/Q C 0 is R33” £1 \RS'; 3, \RSn 39 JR3; ’ R% wherein R3, is halogen, -CF3, -(C1_4 , -(C1_6 cycloalkyl), -NR2, -(C0_3 alkyl)OR, -(C0_3 alkyl)OR”, -NRC(O)R’, -C(O)R’, -C(O)NR2, -C(O)OR’, or )2R’; R4, is —C(O)R’, or -C(O)OR’; each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated; each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated; each R” is independently a C1_6 cycloalkyl, wherein the cycloalkyl is optionally fluorinated; and n is 0-2.
ATI—ZS 14175Vl In some embodiments, R3 is methyl, ethyl, t-butyl, cyclopropyl -CF3, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, -OCH2CH2F, -OCH2CF3, -O(cyclopropyl), -CH20H, -CH20CH3, -C(CH3)20H, -NH2, -NH(CH3), )CH3, -C(O)NHCH3, -C(O)N(CH3)2, or -NHSOZCH3.
In some embodiments of compounds of formula (IB), R3 is ed from branched C1_g alkyl, 1,11 Q fix 0 0 ’,QR3”, / RBIn , ELL \RB'n 21L \RB'n or ’ ’ 3'1LJ L \RB. , R3'n R3, is n, -(C1_4 alkyl), -NR2, -(C0_3 alkyl)OR, -NRC(O)R’, -C(O)R’, -(C0_3 alkyl)C(O)NR2, -C(O)OR’, or )2R’; each R is independently H, or C1_4 alkyl, n the alkyl is ally fluorinated, each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated; n is 0-2.
In some such embodiments, R3, is methyl, ethyl, t-butyl, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, -OCH2CH2F, -OCH2CF3, -CH20H, -CH20CH3, -C(CH3)20H, -NH2, -NH(CH3), -NHC(O)CH3, -C(CH3)2C(O)N(CH3)2, -C(O)NHCH3, -C(O)N(CH3)2, or CH3 In some embodiments of compounds of formula (IB), R4 is a substituted or tituted C1_g alkyl, for example, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, isopentyl, 2-methylpentyl, or tert-pentyl. In some embodiments, R4 is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, 3-methylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 2,3,3-trimethylbutyl, tert-pentyl, isopentyl, 3-pentyl, 3-methylpentyl, 2-methylpentyl, or 2,4-dimethylpentyl. In some embodiments, R4 is isopropyl, isobutyl, isopentyl, tert-butyl, or tert-pentyl. In some such embodiments, R4 is tuted with one or more -(C1_4 alkyl), -(C0_3 alkyl)OR, -C(O)NR2 or ’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated. For example, R4 is substituted with one or more -OH, or -CH3. In ATI—2514l75vl some other embodiments, R4 is a substituted or unsubstituted cycloalkyl, for example, ropyl, utyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[l.l.l]pentyl, bicyclo[2. l.l]hexyl, or bicyclo[2.2.l]heptyl. In some embodiments, R4 is cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or o[l . l .l]pentyl. In some ments, R4 is substituted with one or more halogen, -CF3, -(C1_4 alkyl), -(C1_6 cycloalkyl), -(C0_3 alkyl)OR, -(C0_3 alkyl)C(O)NR2, -NR2, or -NRCOR’, n each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated, and wherein the lkyl is optionally fluorinated. For example, R4 is substituted with one or more methyl, ethyl, isopropyl, -cyclopropyl, -CF3, -CH20H, -OH, -OCH3, -OCH2CH3, -C(O)NH2, -NHC(O)CH3, or -NHC(O)CH2CH3. In some embodiments, R4 is substituted with one or more halogen, -(C1_4 alkyl), -(C0_3 alkyl)OR, -(C0_3 alkyl)C(O)NR2, -NR2, or -NRCOR’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated. In some such embodiments, R4 is tuted with one or more F, methyl, ethyl, isopropyl, -CH20H, -OH, -OCH3, -OCH2CH3, -OCHF2, -OCH2CF3, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -CH2C(O)NH2, -CH2C(O)NHCH3, O)N(CH3)2, -CH(CH3)C(O)NH2, -CH(CH3)C(O)NHCH3, -CH(CH3)C(O)N(CH3)2, -C(CH3)2C(0)NH2, ) HCH3, -C(CH3) 2C(O)N(CH3)2, -NH(CH3), -NHC(O)CH3, or -NHC(O)CH2CH3. In some embodiments, R4 is substituted with one or more methyl, ethyl, -F, -CH20H, or -OH. In some embodiments, R4 is a substituted or tituted non-aromatic cyclyl, for example, yl, tetrahydrofuranyl, tetrahydropyranyl, piperidyl, piperidinonyl or l,4-dioxaspiro[4.5]decanyl. In some embodiments, R4 is tetrahydrofuranyl or tetrahydropyranyl. In some such embodiments, R4 is substituted with one or more -(C1_4 alkyl), -(C0_3 alkyl)OR, or -C(O)R’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated.
ATI—2514l75vl In some embodiments of compounds of a (IB), R4 is a substituted or unsubstituted C1_g alkyl, wherein R5, is —CF3, -(C1_4 alkyl), -(C1_6 cycloalkyl), -(C0_3 alkyl)OR, -(C0_3 C(O)NR2, -NR2, or -NRCOR’, wherein the cycloalkyl is optionally fluorinated; R6, is H, or -C(O)R’; each R is independently H, or C1_4 alkyl, wherein the alkyl is ally fluorinated; each R’ is independently C1_4 alkyl, wherein the alkyl is ally fluorinated; and n is 0-2.
In some such embodiments, R5, is methyl, ethyl, isopropyl, propyl, -CF3, -CH20H, -OH, -OCH3, -OCH2CH3, -C(O)NH2, -NHC(O)CH3, or -NHC(O)CH2CH3.
In some embodiments, R6’ is H or —C(O)CH3, —C(O)CH2CH3, —C(O)CH(CH3)2, or -C(O)CH2CH(CH3)2.
In some embodiments of compounds of formula (IB), R4 is a substituted or unsubstituted C1_g alkyl, 21L \ . O \ a :51 a a n \R5.n R5 E O § L \Rsln or ‘71, ’ R5 wherein R5, is halogen, -(C1_4 alkyl), -(C0_3 alkyl)OR, -(C0_3 alkyl)C(O)NR2, -NR2, or _ 28 _ ATI—2514l75vl -NRCOR’; each R is independently H, or C1_4 alkyl, n the alkyl is optionally fluorinated; each R’ is ndently C1_4 alkyl, wherein the alkyl is optionally fluorinated; n is 0-2.
In some such embodiments, R5, is F, methyl, ethyl, isopropyl, -CH20H, -OH, -OCH3, -OCH2CH3, -OCHF2, -OCH2CF3, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -CH2C(O)NH2, -CH2C(O)NHCH3, -CH2C(O)N(CH3)2, -CH(CH3)C(O)NH2, -CH(CH3)C(O)NHCH3, -CH(CH3)C(O)N(CH3)2, -C(CH3)2C(O)NH2, -C(CH3) 2C(O)NHCH3, -C(CH3) 2C(O)N(CH3)2, -NH(CH3), -NHC(O)CH3, or -NHC(O)CH2CH3. In some embodiments, R5’ is F, -OH, -OCH3, -OCH2CH3, -OCHF2, -OCH2CF3, -NH(CH3), -NHC(O)CH3, -C(O)N(CH3)2, -C(CH3)2C(O)N(CH3)2.
In some embodiments, wherein R4 is a substituted or tituted cycloalkyl, R3 is a cycloalkyl, optionally substituted with one or more halogen, -CF3, -(C1_4 alkyl), -(C1_6 cycloalkyl), -NR2, -(C0_3 OR, -(C0_3 alkyl)OR”, -NRC(O)R’, -C(O)R’, -C(O)NR2, -C(O)OR’, or -NRS(O)2R’, wherein each R is independently H, or C1_4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1_4 alkyl, wherein the alkyl is optionally fluorinated and each R” is independently a C1_6 cycloalkyl, n the lkyl is optionally fluorinated. r embodiments provided herein include combinations of one or more of the particular embodiments set forth above.
Representative compounds of formula (IB) are set forth in Table 3.
Diaminopyrimidine nds set forth in Table 1, Table 2, and Table 3 were tested in the INK inhibitor assays described herein and were found to have activity as JNK inhibitors. In one ment, the Diaminopyrimidine Compound is a compound as described , wherein the compound at a concentration of 10 uM inhibits JNK1 by at least about 50% or more.
ATI—2514l75v1 METHODS FOR MAKING DIAMINOPYRIMIDINE COMPOUNDS The Diaminopyrimidine nds can be made using conventional organic syntheses and commercially available starting materials. By way of e and not limitation, Diaminopyrimidine Compounds of formula (I) can be prepared as outlined in Schemes 1-9 shown below as well as in the examples set forth . It should be noted that one skilled in the art would know how to modify the procedures set forth in the illustrative schemes and examples to arrive at the desired ts. 0 o o o X X SkN/NYO’R—» NYINL lYNN lYN/ CI SkN/ NH 8 N/ NH 8 N NH éx éx R2 éx R2 éx R2 0 0 R‘NJ\N/1 NYN m RX iYN/ NH \5 N N“ H éz (5)m R2 As shown in Scheme 1, compounds of formula (I), wherein R1 and R2 are as defined herein, can be prepared starting from an appropriately derivatized 4-chloro alkylthiopyrimidine carboxylate ester (wherein each Rx is independently a C1_2 alkyl), by treatment with RZNHZ at elevated ature (for example 60-80 c’C) in an organic solvent (for example, ethanol, methanol, isopropanol, THF, NMP, DMF, DMSO, or dioxane), in the presence of a base (for example, DIEA, TEA, N—methylmorpholine, l,8-diazabicyclo[5.4.0]undecene, cesium carbonate, sodium ate, sodium bicarbonate, potassium carbonate, or potassium phosphate). Hydrolysis of the ester is achieved by treatment with an aqueous base, such as for example s sodium hydroxide, potassium hydroxide, or lithium hydroxide, in a cosolvent such as ethanol, methanol, isopropanol, THF, or dioxane. Amide formation is accomplished by reaction with NH4Cl in the ce of a coupling agent (such as, for example, HATU, CDI, HBTU, EDC optionally in combination with HOBt, or ethyl chloroformate), in an organic solvent, such as NMP, DMF, DMSO, dioxane, THF, DCM, or chloroform, in the presence of a base (such as DIEA, TEA, or potassium carbonate). Oxidation of the hiol ATI—2514l75v1 moiety is achieved by ent in an organic solvent (such as, for example, acetone, DCM, NMP, DMF, or chloroform) with an oxidant such as mCPBA, oxone, hydrogen peroxide, or 3-phenyl(phenylsulfonyl)-l ,2-oxaziridine. The resulting e of e (m = l) and sulfoxide (m = 2) is treated at elevated temperature (for example, 80-100 C’C) with RlNHz in a solvent (such as, for example, e, DMSO, NMP, DMF, THF, or water) in the presence of an organic base, such as DIEA, TEA, N—methylmorpholine, or 1,8-diazabicyclo[5.4.0]undecene, to afford the compounds of formula (I). o o o o ,R" ,R" N \ o’ N \ OH N\ 0 Nasr N\ 0 R‘NHz —> A —> A AY A / / N N CI N/ CI CI N/ S’RX HIT] § HIT] § R1 Rx R1 Rx N \ o’ NH4C| § N Cl 0 N \ NH 0 0 if 2 HN N/ RZNHz s N \ NH2 N \ NH2 -1 .X JL / _ JL / R R Rx / HN N NH HN N fi’ R1 R2 R1 (om Selle—r1162 Alternatively, compounds of formula (I) can be prepared as shown in Scheme 2 (wherein R1, R2 and RK are as defined above). Treatment of a 2,4-dichloropyrimidinecarboxylate alkyl ester with NaSRX in an organic solvent (for example THF, DCM, or dioxane) in the presence of a catalyst (for example, triethylbenzylammonium chloride, tetrabutylammonium chloride, or tetrabutylammonium bromide), under cooling (for example, at -10 CC) affords a mixture of thioalkyl derivatives. uent treatment with RINHZ at elevated temperature (e.g. 80 CC) in an organic solvent (for e, dioxane, THF, NMP, DMF, DMSO, l, methanol, or isopropanol), in the presence of a base (for example, DIEA, TEA, N—methylmorpholine, azabicyclo[5.4.0]undecene, cesium carbonate, sodium carbonate, sodium bicarbonate, potassium carbonate, or potassium phosphate) incorporates the R1 sidechain.
Hydrolysis of the alkyl ester, coupling with NH4Cl and oxidation as above, provides a mixture of sulfone (m = l) and sulfoxide (m = 2) derivatives, which upon ent with RZNHZ in a solvent (such as dioxane, NMP, DMF, DMSO, THF, or water), in the presence _ 31 _ ATI—2514l75v1 of an organic base (such as DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, or N—methylmorpholine), at elevated temperature (e.g. 80- 110 oC) provides the compounds of formula (I). o o o X X )L / —2’C|)\N/RZNH RlNH NH HN N/ NH —> HNAN/ NH + o 1\ 0’ 0 N N/ CI N \ NH2 H JL / HN N NH R1 R2 Scheme3 Compounds of a (I) can also be prepared as shown in Scheme 3.
Treatment of 2,4-dichloropyrimidinecarboxylate alkyl ester with RZNHZ in a solvent (for example, ethyl ether, THF, DCM, toluene, or methyl tert-butyl ether) in the presence of a base (for example, DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, N—methylmorpholine, sodium bicarbonate, cesium carbonate, or potassium phosphate) at low temperature (for example, -70 CC) provides uction of the R2 sidechain.
Separation of the mixture of regioisomers, and hydrogenation of the remaining chlorine in a sample allows for eric assignment of the R2 sidechain incorporation. The desired regioisomer compound is then fithher derivatized. Subsequent treatment with RINHZ in an c solvent (for example, THF, NMP, DMF, DMSO, dioxane, ethanol, methanol, or panol) in the presence of a base (for example, DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, N—methylmorpholine, sodium carbonate, sodium bicarbonate, cesium carbonate, or potassium phosphate) at elevated temperature (for example, 70 oC) es introduction of the R1 sidechain. Hydrolysis of the alkyl ester and coupling with NH4Cl, as , provides the compounds of formula (I).
ATI—2514l75vl /N /N /N / / / R NH22 N \ R NH21 N \ N \ H202 \ A / R1 )L / )L / ‘ CI N NH N N NH R1\NJ\N/NI/jfLNHZNH CI N CI II?2 H F32 H (I) éz RZ‘MJLN/ CI Scheme 4 An alternative method to the sis of compounds of formula (I) is shown in Scheme 4. Treatment of 2,4-dichloropyrimidinecarbonitrile with RZNHZ in an organic solvent (for example, ethanol, ol, isopropanol or THF) in the presence of a base (for example, DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, N-methylmorpholine, sodium carbonate, sodium onate, cesium carbonate, or potassium phosphate) at low temperature (for example, -60 OC) es introduction of the R2 sidechain. As before, hydrogenation of the remaining chlorine allows for regioisomeric assignment of the R2 incorporation. Subsequent treatment with RINHZ in an organic solvent (for example, l-butanol, THF, NMP, DMF, DMSO, dioxane, ethanol, methanol, or isopropanol) in the presence of a base (for example, cesium ate, ium carbonate, sodium carbonate, sodium bicarbonate, ium phosphate, l,8-diazabicyclo[5 .4.0]undecene, N-methylmorpholine, DIEA, or TEA) at elevated temperature (for example, 120 CC) provides introduction of the R1 sidechain (providing in some instances when R1 = R3 and R2 = R4, compounds of formula (IB)). Conversion of the e moiety with, for example, catalytic peroxide, in the presence of a strong aqueous base, such as sodium hydroxide or potassium hydroxide, in a t, such as, for example, DMSO, NMP, DMF, ethanol or methanol, provides the compounds of formula (I). o o O /N / N / / \ \ N \ NH i\ RZNH N \ N NH2 N NH2 RlNH 2 41/ —»|/ —»Rx\x/ —»2 A/ s N/ N Cl 8 N NH 177 N NH .3: N NH HN NH éx lIRX R2 RX R2 (mm R2 R1 R2 Scheme5 0085 Yet r method for the synthesis of com ounds of formulaP I is shown in Scheme 5. Treatment of 4-chloro(alkylthio)pyrimidinecarbonitrile with RlNHgin an organic solvent (for example, n-butanol, NMP, DMF, DMSO, dioxane, or ethanol) in ATI—2514l75v1 the presence of a base (such as, for example, DIEA, TEA, N—methylmorpholine, l,8-diazabicyclo[5.4.0]undecene, potassium carbonate, sodium carbonate, sodium bicarbonate, cesium carbonate, or ium phosphate), at a temperature between 50 oC and 90 oC) introduces the R1 sidechain. Conversion of the carbonitrile to the amide, for example by treatment with peroxide (H202), in a a solvent (such as, for example, DMSO, NMP, DMF, ethanol, or methanol), in the presence of a base (such as sodium ide or potassium hydroxide), and oxidation, (for example using mCPBA, oxone, hydrogen peroxide, or yl(phenylsulfonyl)-l ,2-oxaziridine, in a solvent such as DCM, NMP, DMF, or DMA), provides the mixture of e (m=l) and sulfoxide (m = 2) as before, which can be treated with RZNHZ in a solvent such as (dioxane, DMSO, NMP, DMF, THF, or n-butanol) in the ce of a base (such as DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, or ylmorpholine), optionally at elevated temperature (for e, between room temperature and 130 0C) to provide compounds of formula (I).
B N N\ rR1NH2 N\Br N\// N\ NH2 A / —’ I —> / I )L / Cl N s HN N s )x / HN N s . I HN N s .1 IX RX R1 RX F3 ng R R /N /N / / N \ RZNHZ N \ )L / RX —’ )L / HN N T3 HN N NH R1 (O)m R1 i R2 o o 1%“.2R NH2 l\ x HN N/ NH HN N/ s’R R1 R2 R1 (0)m Scheme6 Compounds of formula (I) can also be obtained as shown in Scheme 6.
Treament of 5-bromochloro(alkylthio)pyrimidine with RINHZ in an organic solvent (for example, n-butanol, NMP, DMF, DMSO, dioxane, or ethanol) in the presence of a base (such as, for example, DIEA, TEA, ylmorpholine, l,8-diazabicyclo- [5.4.0]undecene, potassium carbonate, sodium carbonate, sodium bicarbonate, cesium carbonate, or potassium phosphate), at a temperature between 80 oC and 100 oC, _ 34 _ ATI—2514l75vl introduces the R1 sidechain. Introduction of the itrile moiety is achieved by treatment with zinc and dicyanozinc in a solvent (such as DMF, DMSO, NMP, or DMA) in the presence of a catalyst such as Pd(O), at elevated temperature (for example, n 80 oC — 100 oC). As before, conversion to the amide moiety by treatment with peroxide, followed by oxidation to the sulfone/sulfoxide, and treatment with RZNHZ provides the compounds of formula (1). Alternatively, the alkylthio moiety is oxidized first, followed by introduction of the R2 sidechain, (providing in some instances when R1 = R3 and R2 = R4 compounds of formula (IB)), and conversion to the amide moiety, provides compounds of formula (I).
Alternatively, opyrimidine Compounds of Formula (1B), wherein R3 and R4 are as defined herein, can be made as ed in Schemes 7, 8 and 9 shown below, as well as in the examples set forth herein. It should be noted that one skilled in the art would know how to modify the procedures set forth in the illustrative schemes and examples to arrive at the desired product. if!»R4—»”“21/ R—W AT RiMJLN/ CI Scheme 7 As shown in Scheme 7, treatment of 2,4-dichloropyrimidinecarbonitrile with R4NH2 in an organic t (for example, ethanol, methanol, isopropanol or THF) in the presence of a base (for example, DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, N—methylmorpholine, sodium carbonate, sodium bicarbonate, cesium carbonate, or potassium phosphate) provides introduction of the R4 sidechain. Subsequent treatment with R3NH2 in an organic t (for example, l-butanol, THF, NMP, DMF, DMSO, e, ethanol, methanol, or isopropanol) in the ce of a base (for example, cesium carbonate, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium phosphate, l,8-diazabicyclo[5.4.0]undecene, N—methylmorpholine, DIEA, or TEA) at elevated temperature (for example, 50 0C to 90 oC) provides compounds of a (1B).
ATI—2514l75v1 ell/TNR3NH2 HNJNL/ij R4NH2 T (”3) Scheme 8 Alternatively, the R3 substituent is introduced first, followed by introduction of the R4 substituent by essentially the same methods, as shown in Scheme 8.
In some embodiments, coupling with R4NH2 is med at a temperature n room temperature and 110 CC.
WNW /:£/’Omi —’ 3* RX\3* *N/ RxSiN/ NH RScheme 9 (GmN/:E:N LNW HNN \UB) In a third approach, compounds of formula (IB) can be prepared starting from an appropriately derivatized 4-chloroalkylthiopyrimidine-carbonitrile (wherein each Rx is independently a C1_2 alkyl), by treatment with R4NH2 at ed temperature (for example 50 CC - 90 CC) in an organic solvent (for example, n-butanol, NMP, DMF, DMSO, or dioxane), in the presence of a base (for example, DIEA, TEA, N-methylmorpholine, l,8-diazabicyclo[5.4.0]undecene, potassium carbonate, sodium ate, sodium bicarbonate, cesium carbonate or potassium phosphate). Oxidation of the alkylthiol moiety is achieved by treatment in an organic solvent (such as, for example, DCM, NMP, DMF, or DMA) with an oxidant such as mCPBA, oxone, hydrogen de, or 3-phenyl(phenylsulfonyl)-l,2-oxaziridine. The resulting mixture of sulfone (m = l) and sulfoxide (m = 2) is treated at room temperature or elevated temperature (for e, CC - 110 CC) with R3NH2 in a solvent (such as, for example, dioxane, DMSO, NMP, DMF, THF, or n-butanol) in the presence of an c base, such as DIEA, TEA, 1,8- diazabicyclo[5.4.0]undecene, N-methylmorpholine, to afford the compounds of formula (1B).
ATI—2514l75vl In one aspect, provided herein are methods for preparing a compound of formula (I): )LN/YLNHZ/ HN N NH {21 R2 the methods comprising contacting a compound of formula (la) RKSJLNY N/ NH || ' (O)m R2 (1a) 9 with RINHZ in a solvent, in the presence of an organic base, wherein: R1 is substituted or unsubstituted C1_g alkyl, substituted or unsubstituted cycloalkyl, tuted or unsubstituted non-aromatic heterocyclyl, substituted or unsubstituted alkylcycloalkyl, or substituted or unsubstituted alkylheterocyclyl, provided that R1 is not l-aminocyclohexyl; R2 is substituted or unsubstituted C1_g alkyl, substituted or unsubstituted saturated cycloalkyl, substituted or unsubstituted alkylcycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl; Rx is a C1_2 alkyl; and m is l or 2.
In one ment, the compound of formula (I) is not 2-(2-aminoethyl-amino)— hylamino)pyrimidinecarboxamide; 2-(2-aminopropylamino) hexylamino)pyrimidinecarboxamide; 2-(2-aminooxoethylamino) (cyclohexylamino)pyrimidinecarboxamide; 2—(2-aminoethylamino)—4- (cyclohexylamino)pyrimidinecarboxamide; (S)(2-aminopropylamino) butylamino)pyrimidinecarboxamide; (R)( l -aminomethyloxobutan ylamino)(cyclobutylamino)pyrimidinecarboxamide; lopentylamino) ATI—2514l75v1 2012/034349 (methylamino)pyrimidinecarboxamide; or 2-(l-acetylpiperidinylamino) (cyclopropylamino)pyrimidinecarboxamide.
In one embodiment, the solvent is dioxane, DMSO, NMP, DMF, THF, or water. In another, the base is DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, or N—methylmorpholine. In some ments, the contacting is performed at elevated temperature, for example, from about 80 °C to about 100 CC.
In some embodiments, the methods fithher comprise ing a compound of formula (Ia): Risk NYNH2 N/ NH || ' (O)m R2 (1a) 9 the methods comprising oxidizing a compound of formula (Ib) N \ NH2 )L / s N NH RX R2 (1b), in a solvent by treatment with an oxidant ed from mCPBA, oxone, hydrogen peroxide, or 3-phenyl(phenylsulfonyl)-l ,2-oxaziridine.
In one embodiment, the solvent is e, DCM, NMP, DMF, or chloroform. In some embodiments, the method is performed at a temperature between about 0 0C to about 20 0C.
In some embodiments, the methods fithher comprise preparing a compound of formula (Ib): N%NH2 )L / S N NH I I RX R2 (1b), ATI—2514l75vl the methods comprising ting a compound of formula (Ic) N \ OH )L / s N NH RX R2 (Ic), with NH4Cl, in the presence of a coupling agent and a base, in a solvent.
In some embodiments, the solvent is NMP, DMF, DMSO, dioxane, THF, DCM, or chloroform. In others, the coupling agent is HATU, CDI, HBTU, EDC/HOBt, or ethyl chloroformate, and the base is DIEA, TEA, or potassium carbonate.
In some embodiments, the methods fiarther comprise preparing a compound of formula (Ic) N \ OH )L / s N NH Flax R2 (10), the methods sing contacting a compound of formula (Id) N \ o’R )L / s N NH F'ax R2 (1d), with an aqueous base, in a cosolvent.
In some embodiments, the aqueous base is aqueous sodium hydroxide, ium hydroxide, or lithium ide. In other embodiments, the cosolvent is ethanol, methanol, isopropanol, THF, or dioxane.
ATI—ZS 14175Vl In some embodiments, the methods fithher comprise preparing a compound of a (Id) N \ o’RX S N NH RX Fla? (1d), the methods comprising contacting a compound of formula (Ie) (16), with RZNHZ in an organic solvent, in the ce of a base.
In some embodiments, the organic solvent is ethanol, methanol, isopropanol, THF, NMP, DMF, DMSO, or dioxane. In others, the base is DIEA, TEA, N—methylmorpholine, 1,8-diazabicyclo[5.4.0]undecene, cesium carbonate, sodium carbonate, sodium bicarbonate, potassium carbonate, or potassium phosphate. In some embodiments, the contacting is performed at ed temperature, for example, from about 60 0C to about 80 0C.
In some ments, the methods fiarther comprise preparing a compound of a (Ib) N \ NH2 )L / s N NH RX R2 (1b), the methods comprising contacting a compound of formula (If) s N NH RX R2 ATI—ZS 14175v1 with peroxide, in the presence of a base, in a solvent.
In some embodiments, the solvent is DMSO, NMP, DMF, l, or ol.
In others, the base is sodium ide or potassium hydroxide.
In some embodiments, the methods fiarther comprise preparing a compound of formula (If) s N NH RX R2 (If), the methods comprising contacting a compound of formula (Ig) A/NY S N CI (1g) with RZNHZ in an organic solvent, in the presence of a base.
In some ments, the c solvent is n-butanol, NMP, DMF, DMSO, dioxane, or ethanol. In others, the base is DIEA, TEA, N—methylmorpholine, 1,8-diazabicyclo[5.4.0]undecene, potassium ate, sodium carbonate, sodium bicarbonate, cesium carbonate, or potassium phosphate. In some embodiments, the contacting is performed at elevated temperature, for example, from about 50 0C to about 90 0C.
Further provided are methods for preparing a compound of formula (I): N \ NH2 )L / HN N NH IIRl R2 the methods comprising contacting a compound of formula (11a) HNAN/N1“fig R1 (O)m _ 41 _ ATI—ZS 14175v1 (Ha), with RZNHZ in a solvent, in the presence of an c base, wherein: R1 is substituted or tituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or tituted non-aromatic heterocyclyl, substituted or unsubstituted ycloalkyl, or substituted or unsubstituted alkylheterocyclyl, provided that R1 is not 1-aminocyclohexyl; R2 is substituted or unsubstituted C1_g alkyl, substituted or unsubstituted ted cycloalkyl, substituted or unsubstituted alkylcycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl; Rx is a C1_2 alkyl; and m is 1 or 2.
In one embodiment, the compound of formula (I) is not 2-(2-aminoethyl- amino)(methylamino)pyrimidinecarboxamide; 2-(2-aminopropylamino) (cyclohexylamino)pyrimidinecarboxamide; 2-(2-aminooxoethylamino) hexylamino)pyrimidinecarboxamide; 2—(2-aminoethylamino)—4- (cyclohexylamino)pyrimidinecarboxamide; (S)(2-aminopropylamino) (cyclobutylamino)pyrimidinecarboxamide; (R)( 1 -aminomethyloxobutan o)(cyclobutylamino)pyrimidinecarboxamide; 4-(cyclopentylamino) lamino)pyrimidinecarboxamide; or 2-(1-acetylpiperidinylamino) (cyclopropylamino)pyrimidinecarboxamide.
In one embodiment, the solvent is dioxane, DMSO, NMP, DMF, THF, or water. In another, the base is DIEA, TEA, 1,8-diazabicyclo[5.4.0]undecene, or N—methylmorpholine. In some embodiments, the contacting is performed at elevated temperature, for example, from about 80 °C to about 110 CC.
In some embodiments, the methods fiarther comprise preparing a compound of formula (11a): 11*:52 HN N S/ {21 (51“ _ 42 _ ATI—ZS 14175v1 (113'): the methods comprising oxidizing a compound of formula (11b) (11b), in a solvent by treatment with an oxidant selected from mCPBA, oxone,hydrogen peroxide, or 3-phenyl(phenylsulfonyl)-l ,2-oxaziridine.
In one embodiment, the solvent is acetone, DCM, NMP, DMF, or chloroform. In some embodiments, the ing is performed at low temperature, for example, at about 0 °C.
In some embodiments, the methods r comprise preparing a compound of formula (11b): (11b), the methods comprising contacting a compound of formula (11c) (11c), with NH4Cl in the ce of a coupling agent and a base, in a solvent.
In some embodiments, the solvent is NMP, DMF, DMSO, dioxane, THF, DCM, or chloroform. In , the coupling agent is HATU, CDI, HBTU, EDC/HOBt, or ethyl chloroformate, and the base is DIEA, TEA, or potassium carbonate.
ATI—2514l75v1 In some embodiments, the s filrther comprise preparing a compound of formula (IIc) (11C): the methods comprising contacting a compound of formula (IId) (IId), with an aqueous base, in a cosolvent.
In some embodiments, the aqueous base is aqueous sodium hydroxide, potassium hydroxide, or lithium hydroxide. In other embodiments, the cosolvent is ethanol, methanol, isopropanol, THF, or dioxane.
In some embodiments, the methods filrther comprise preparing a compound of formula (11d) N \ o’R )L / HI}! N § R1 RX (11d), the methods sing ting a compound of a (He) N \ o’R CI N § (116), with RINHZ in an organic solvent, in the ce of an organic base.
ATI—ZS 14175Vl In one embodiment, the organic solvent is dioxane, THF, NMP, DMF, DMSO, ethanol, methanol, or isopropanol. In another, the base is DIEA, TEA, N—methylmorpholine, 1,8-diazabicyclo[5.4.0]undecene, cesium carbonate, sodium carbonate, sodium bicarbonate, potassium carbonate, or ium phosphate. In some embodiments, the contacting is performed at elevated temperature, for example, from about 80 0C to about 100 0C.
In some ments, the s r comprise ing a compound of formula (IIe) (116): the s comprising contacting a compound of formula (IIf) (Hf), with NaSRX in an organic solvent in the presence of a catalyst.
In one embodiment, the solvent is THF, DCM, or dioxane. In one embodiment, the catalyst is triethylbenzylammonium chloride, tetrabutylammonium de, or tetrabutylammonium bromide. In some embodiments, the contacting is performed under cooling for example, about -10 0C.
In some embodiments, the methods fiarther comprise preparing a compound of formula (11b): )LN/YLNHZ/ HI}! N s R1 RX (11b), _ 45 _ ATI—ZS 14175v1 the methods comprising contacting a compound of formula (IIg) JL / ”Y/ N HN N S F'z1 F'zx (Hg), with peroxide, in the presence of a base, in a solvent.
In some embodiments, the solvent is DMSO, NMP, DMF, ethanol, or methanol. In others, the base is sodium hydroxide or potassium hydroxide.
In some embodiments, the methods fiarther comprise preparing a compound of a (IIg) )L / ”Y/ N HN N § R1 RX (11g), the methods comprising ting a nd of a (IIh) HN N 8 E21 F'ex (IIh), with zinc and dicyanozinc, in the presence of a catalyst, in a solvent.
In some embodiments, the st is Pd(0). In some embodiments, the solvent is DMF, DMSO, NMP, or DMA. In others, the contacting is performed at elevated temperature, for example at a temperature between about 80 oC and about 100 0C.
In some embodiments, the methods filrther comprise preparing a compound of formula (IIh) )L / HN N S IIRl F'ex (11h), _ 46 _ ATI—25 14175v1 the methods comprising contacting a compound of formula (Hi) CI N 8 (IIi) with RINHZ, in the ce of a base, in an organic solvent.
In some embodiments, the organic solvent is n-butanol, NMP, DMF, DMSO, e, or ethanol. In , the base is DIEA, TEA, N-methylmorpholine, l,8-diazabicyclo-[5.4.0]undecene, potassium carbonate, sodium ate, sodium bicarbonate, cesium carbonate, or potassium phosphate. In some embodiments, the contacting is performed at elevated temperature, for example at a temperature between about 80 oC and about 100 0C.
Also provided are methods for preparing a compound of formula (I): N \ NH2 )L / HN N NH R1 R2 the methods comprising contacting a compound of formula (IIIa) )LN/\fko’H/ HN N NH R1 R2 (IIIa), with with NH4Cl, in the presence of a coupling agent and a base, in a solvent, wherein: R1 is substituted or tituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or tituted non-aromatic heterocyclyl, substituted or unsubstituted alkylcycloalkyl, or substituted or unsubstituted alkylheterocyclyl, ed that R1 is not l-aminocyclohexyl; and ATI—2514l75v1 R2 is substituted or unsubstituted C1_g alkyl, substituted or unsubstituted saturated cycloalkyl, tuted or tituted alkylcycloalkyl, tuted or unsubstituted non-aromatic cyclyl.
In one ment, the compound of formula (I) is not 2-(2-aminoethyl- amino)(methylamino)pyrimidinecarboxamide; 2-(2-aminopropylamino) (cyclohexylamino)pyrimidinecarboxamide; 2-(2-aminooxoethylamino) (cyclohexylamino)pyrimidinecarboxamide; 2-(2-aminoethylamino) (cyclohexylamino)pyrimidinecarboxamide; (S)(2-aminopropylamino) butylamino)pyrimidinecarboxamide; (R)( 1 -aminomethyloxobutan ylamino)(cyclobutylamino)pyrimidinecarboxamide; 4-(cyclopentylamino) (methylamino)pyrimidinecarboxamide; or 2-(1-acetylpiperidinylamino) (cyclopropylamino)pyrimidinecarboxamide.
In some ments, the solvent is NMP, DMF, DMSO, dioxane, THF, DCM, or chloroform. In others, the coupling agent is HATU, CDI, HBTU, EDC/HOBt, or ethyl chloroformate, and the base is DIEA, TEA, or potassium carbonate.
In some such embodiments, the methods further comprise preparing a compound of formula (Ma): N \ o’H )L / HN N NH F'ai F'az (IIIa), the methods comprising ting a compound of formula (IIIb) )L / HN N NH FIR] R2 (IIIb), with an aqueous base, in a cosolvent, wherein Rx is a C1_2 alkyl.
In some embodiments the aqueous base is aqueous sodium hydroxide, potassium hydroxide, or lithium hydroxide. In other embodiments, the cosolvent is ethanol, methanol, isopropanol, THF, or dioxane.
ATI—25 14175V1 In some embodiments, the method further comprise preparing a compound of formula (IIIb): N \ o’R )L / HN N NH Flil R2 (IIIb), the method comprising contacting a compound of formula (IIIc) N \ o’R CI N NH F'az (IIIc), with RINHZ in the presence of an organic base, in an organic solvent.
In some ments, the organic t is THF, NMP, DMF, DMSO, e, ethanol, methanol, or isopropanol. In others the organic base is DIEA, TEA, l,8-diazabicyclo[5.4.0]undecene, N—methylmorpholine, sodium carbonate, sodium bicarbonate, cesium ate, or ium phosphate. In some embodiments, the contacting is performed at elevated temperature, for example, about 70 0C.
In some embodiments, the methods fiarther comprise ing a compound of formula (IIIc) N \ o’R CI N NH (IIIc), the methods comprising contacting a compound of formula (111d) N\ o’R CI N CI (111d) with RZNHZ in the presence of an organic base in a solvent.
ATI—2514l75v1 In some embodiments, the solvent is ethyl ether, THF, DCM, toluene, or methyl tert-butyl ether. In others, the base is DIEA, TEA, 1,8-diazabicyclo[5.4.0]undec ene, N—methylmorpholine, sodium bicarbonate, cesium carbonate, or potassium ate.
In some embodiments the contacting is performed at low temperature, for example, about -70 ° C.
Also provided are methods for preparing a compound of formula (I): )LN/YLNHZ/ HN N NH R1 R2 the methods comprising contacting a compound of formula (IVa) RLMJLN/ El: (IVa), with catalytic peroxide, in the presence of a strong s base, in a solvent, wherein: R1 is substituted or tituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or tituted non-aromatic heterocyclyl, substituted or unsubstituted alkylcycloalkyl, or tuted or unsubstituted alkylheterocyclyl, ed that R1 is not 1-aminocyclohexyl; and R2 is substituted or unsubstituted C1_g alkyl, substituted or unsubstituted saturated cycloalkyl, substituted or unsubstituted alkylcycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl.
In one embodiment, the compound of formula (I) is not 2-(2-aminoethyl- amino)(methylamino)pyrimidinecarboxamide; 2-(2-aminopropylamino) (cyclohexylamino)pyrimidinecarboxamide; 2-(2-aminooxoethylamino) (cyclohexylamino)pyrimidinecarboxamide; minoethylamino)—4- (cyclohexylamino)pyrimidinecarboxamide; (S)(2-aminopropylamino) (cyclobutylamino)pyrimidinecarboxamide, (R)(1-aminomethyloxobutan ATI—ZS 14175v1 ylamino)(cyclobutylamino)pyrimidinecarboxamide; 4-(cyclopentylamino) lamino)pyrimidinecarboxamide; or 2-( 1 -acetylpiperidinylamino) (cyclopropylamino)pyrimidinecarboxamide.
In some embodiments, the solvent is DMSO, NMP, DMF, ethanol or methanol. In others, the strong aqueous base is such as sodium hydroxide or potassium ide.
In some such ments, the methods further comprise preparing a compound of formula (IVa): R \NAN/1 ”Y (IVa), the methods comprising contacting a compound of formula (IVb) CI N NH F'az (IVb), with RINHZ in the presence of a base, in an organic solvent.
] In some embodiments, the organic solvent is 1-butanol, THF, NMP, DMF, DMSO, dioxane, ethanol, methanol, or isopropanol. In others the base is cesium carbonate, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium phosphate, 1,8-diazabicyclo[5.4.0]undecene, ylmorpholine, DIEA, or TEA. In some embodiments, the contacting is performed at elevated temperature, for example, about 120 CC.
In some such embodiments, the methods further comprise preparing a compound of a (IVb): CI N NH (IVb), ATI—ZS 14175v1 WO 45569 the methods comprising contacting a compound of formula (IVc) CI N CI (IVc), with RZNHZ in the presence of a base, in an organic solvent.
In some embodiments, the organic solvent is ethanol, methanol, isopropanol or THF. In others the base is DIEA, TEA, 1,8-diazabicyclo[5.4.0]undec ene, N—methylmorpholine, sodium carbonate, sodium bicarbonate, cesium carbonate, or potassium phosphate. In some ments, the contacting is performed at low temperature, for example, about -60 0C.
In some embodiments, the s fiarther comprise preparing a compound of formula (IVa): JL / HN N NH II?1 R2 (IVa), the methods comprising contacting a compound of formula (IVd) HNJLN/ RX Isl.” R1 (O)m (IVd), with RZNHZ in the presence of a base, in an organic t.
In some embodiments, the organic solvent is DCM, NMP, DMF, or DMA.
In others, the base is DIEA, TEA, 1,8-diazabicyclo[5.4.0]undecene, or N—methylmorpholine. In some embodiments, the contacting is performed at a temperature between room temperature and about 130 0C.
ATI—ZS 14175v1 ] In some embodiments, the methods filrther comprise preparing a compound of a (IVd): HNAN/I S’Rx II?1 (5% (IVd), the methods comprising oxidizing a compound of formula (Hg) JL / HN N § R1 RX (Hg), in a solvent by treatment with an oxidant selected from mCPBA, oxone,hydrogen peroxide, or 3-phenyl(phenylsulfonyl)-l ,2-oxaziridine.
In one embodiment, the solvent is DCM, NMP, DMF, or DMA. In some embodiments, the oxidizing is performed at a temperature from about 0 °C to about room temperature.
Also provided are methods for preparing a compound of a (IB) )L / HN N NH R3 PE“ (113), the methods comprising contacting a nd of a (Va) CI N NH Fla“ (Va), with R3NH2 in the presence of a base, in an organic solvent, wherein R3 is tuted or unsubstituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl, substituted or unsubstituted alkylcycloalkyl, or substituted or unsubstituted alkyl-(non—aromatic heterocyclyl); and _ 53 _ ATI—2514l75vl R4 is substituted or unsubstituted C1_g alkyl, tuted or unsubstituted cycloalkyl, or substituted or unsubstituted non-aromatic heterocyclyl.
] In one embodiment, the compound is not 4-(isopentylamino)(3-(2- methylpiperidin- l -yl)propylamino)pyrimidine-5 -carbonitrile; (ZS ,2'S)—dimethyl 2,2'-(5 - cyanopyrimidine-2,4-diyl)bis(azanediyl)bis(4-methylpentanoate); (28,2'S)-diethyl 2,2'-(5- cyanopyrimidine-2,4-diyl)bis(azanediyl)bis(3-methylbutanoate); 4-(cycloheptylamino) (3 -(2-methylpiperidin- l -yl)propylamino)pyrimidine-5 nitrile; 4-(4-methylcyclo- hexylamino)(3-(2-methylpiperidin- l -yl)propylamino)pyrimidine-5 -carbonitrile; or 2-(3-(diethylamino)propylamino)(4-methylcyclohexylamino)-pyrimidinecarbonitrile.
In some embodiments, the organic solvent is l-butanol, THF, NMP, DMF, DMSO, dioxane, ethanol, methanol, or isopropanol. In others the base is cesium carbonate, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium phosphate, l,8-diazabicyclo[5.4.0]undecene, N—methylmorpholine, DIEA, or TEA. In some embodiments, the contacting is med at elevated temperature, for example, from about 50 0C to about 90 0C.
] In some such ments, the methods further comprise preparing a compound of formula (Va): (V3) 9 the methods comprising contacting a compound of formula (Vb) CI N CI (Vb), with R4NH2 in the presence of a base, in an organic solvent.
] In some embodiments, the organic t is ethanol, methanol, isopropanol or THF. In others the base is DIEA, TEA, 1,8-diazabicyclo[5.4.0]undec ene, N—methylmorpholine, sodium carbonate, sodium bicarbonate, cesium carbonate, or ATI—2514l75v1 potassium phosphate. In some embodiments, the contacting is performed at low temperature, for example, from about -70 °C to about 20 CC.
Also provided are methods for preparing a nd of formula (IB) JL / HN N NH R3 PE“ (13), the methods comprising contacting a nd of a (VIa) HN N CI (VIa), with R4NH2 in the presence of a base, in an organic solvent, wherein R3 is substituted or unsubstituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl, substituted or tituted alkylcycloalkyl, or substituted or unsubstituted alkyl-(non—aromatic heterocyclyl); and R4 is substituted or tituted C1_g alkyl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted non-aromatic heterocyclyl.
In one embodiment, the compound is not 4-(isopenty1amino)(3-(2- methylpiperidinyl)propylamino)pyrimidine-5 -carbonitrile; (ZS ,2'S)—dimethyl 2,2'-(5 - yrimidine-2,4-diyl)bis(azanediyl)bis(4-methylpentanoate); (28,2'S)-diethyl 2,2'-(5- yrimidine-2,4-diyl)bis(azanediyl)bis(3-methylbutanoate); 4-(cycloheptylamino) (3 thylpiperidinyl)propylamino)pyrimidine-5 -carbonitrile; 4-(4-methylcyclohexylamino )(3-(2-methylpiperidinyl)propylamino)pyrimidine-5 -carbonitrile; or 2-(3-(diethylamino)propylamino)(4-methylcyclohexylamino)-pyrimidinecarbonitrile.
In some embodiments, the c solvent is 1-butanol, NMP, DMF, DMSO, or dioxane. In others the base is cesium carbonate, potassium carbonate, sodium carbonate, sodium bicarbonate, potassium phosphate, 1,8-diazabicyclo[5 .4.0]undecene, N—methylmorpholine, DIEA, or TEA. In some embodiments, the contacting is performed at elevated temperature, for example, from about 25 °C to about 110 CC.
ATI—25 14175v1 In some such embodiments, the methods further comprise preparing a compound of formula (VIa): HN N CI (VIa), the methods comprising ting a compound of formula (Vb) CI N CI (Vb), with R3NH2 in the presence of a base, in an organic solvent.
In some such embodiments, the organic solvent is ethanol, methanol, isopropanol or THF. In others the base is DIEA, TEA, 1,8-diazabicyclo[5.4.0]undec ene, N-methylmorpholine, sodium carbonate, sodium bicarbonate, cesium carbonate, or potassium phosphate. In some embodiments, the contacting is performed at low ature, for e, from about -70 °C to about 20 CC.
Provided are methods for ing a compound of formula (IB) )L / HN N NH R3 PE“ (113), the methods comprising contacting a compound of formula (VIIa) /x | (O)m R4 (VIIa), with R3NH2 in the presence of a base, in an organic solvent, wherein R3 is substituted or tituted C1_g alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl, substituted or unsubstituted ATI—25 14175v1 alkylcycloalkyl, or substituted or unsubstituted alkyl-(non-aromatic heterocyclyl); R4 is substituted or unsubstituted C1_g alkyl, tuted or unsubstituted cycloalkyl, or substituted or unsubstituted non-aromatic heterocyclyl; Rx is C1_2 alkyl and m is 1 or 2.
In one embodiment, the nd is not 4-(isopentylamino)(3-(2- methylpiperidinyl)propylamino)pyrimidine-5 -carbonitrile; (ZS ,2'S)—dimethyl 2,2'-(5 - cyanopyrimidine-2,4-diyl)bis(azanediyl)bis(4-methylpentanoate); (28,2'S)-diethyl 2,2'-(5- cyanopyrimidine-2,4-diyl)bis(azanediyl)bis(3-methylbutanoate); 4-(cycloheptylamino) (3 -(2-methylpiperidinyl)propylamino)pyrimidine-5 -carbonitrile; 4-(4-methylcyclohexylamino )(3-(2-methylpiperidin- 1 opylamino)pyrimidine-5 -carbonitrile; or 2-(3-(diethylamino)propylamino)(4-methylcyclohexylamino)-pyrimidinecarbonitrile.
In one embodiment, the solvent is dioxane, DMSO, NMP, DMF, THF, or n-butanol. In another, the base is DIEA, TEA, 1,8-diazabicyclo[5.4.0]undecene, N—methylmorpholine. In some embodiments, the contacting is performed at elevated temperature, for example, from about 25 °C to about 110 CC.
In some ments, the s fithher comprise preparing a compound of formula (VIIa): R\S)\N/ ”Y/ N x | (O)m [Q4 (VIIa), the methods comprising oxidizing a compound of formula (VIIb) (VIIb), in a solvent by treatment with an t selected from mCPBA, oxone, en peroxide, or 3-phenyl(phenylsulfonyl)-1,2-oxaziridine.
ATI—25 14175v1 In one ment, the t is DCM, NMP, DMF, or DMA. In some embodiments, the oxidizing is performed at low temperature, for example, about 0 °C.
In some embodiments, the methods further comprise preparing a compound of formula (VIIb) (VIIb), the methods comprising contacting a compound of formula (VIIc) RX‘SJLN/ CI (VIIc), with R4NH2 in an organic solvent, in the presence of a base.
In one embodiment, the organic t is n-butanol, NMP, DMF, DMSO, or dioxane. In another, the base is DIEA, TEA, N-methylmorpholine, 1,8-diazabicyclo[5.4.0]undecene, potassium carbonate, sodium carbonate, sodium bicarbonate, cesium ate or potassium phosphate. In some embodiments, the contacting is performed at elevated temperature, for e, from about 50 °C to about 90 OC.
METHODS OF USE The Diaminopyrimidine Compounds have utility as pharmaceuticals to treat, prevent or improve conditions in animals or humans. Further, the Diaminopyrimidine Compounds are active against n kinases, particularly JNKl and/or JNK2. ingly, provided herein are many uses of the Diaminopyrimidine Compounds, including the ent or prevention of those diseases set forth below. The methods provided herein comprise the administration of an effective amount of one or more Diaminopyrimidine Compound(s) to a subject in need thereof In one aspect provided herein are methods of inhibiting a kinase in a cell sing said kinase, comprising contacting said cell with an effective amount of a ATI—ZS 14175v1 2012/034349 Diaminopyrimidine Compound. In one embodiment the kinase is JNKl, JNKZ, or mutants or isoforms thereof, or a combination thereof. For example, the opyrimidine Compound is a compound from Table 1, 2 or 3.
In another aspect provided herein are methods for treating or preventing liver f1brotic disorders, such as non-alcoholic steatohepatitis, steatosis (i.e. fatty , cirrhosis, primary sclerosing cholangitis, primary biliary cirrhosis, hepatitis, hepatocellular carcinoma, and liver fibrosis coincident with chronic or ed alcohol ingestion (alcoholic hepatitis), with ion (e.g. viral infection such as HCV), with liver transplant, or with drug induced liver injury (e.g. inophen toxicity), comprising administering to a subject in need thereof an effective amount of a Diaminopyrimidine Compound. In some such s, provided herein are methods for treating or preventing diabetes or metabolic me leading to liver fibrotic disorders, such as non-alcoholic steatohepatitis, steatosis (i.e. fatty liver), sis, primary sclerosing cholangitis, primary biliary cirrhosis, and hepatitis, sing administering to a t in need thereof an effective amount of a Diaminopyrimidine Compound.
In r aspect provided herein are methods for treating or preventing one ore more disorders selected from interstitial pulmonary fibrosis, systemic sclerosis, derma, chronic allograft nephropathy, antibody mediated rejection, or lupus, comprising administering to a subject in need thereof an effective amount of a Diaminopyrimidine Compound. In some such embodiments, the lupus is lupus erythematosus (such as d lupus erythematosus, or cutaneous lupus erythematosus) or ic lupus.
In another aspect provided herein are methods for treating or preventing conditions treatable or preventable by inhibition of INK1 and/or JNK2, the method comprising administering to a subject in need thereof an ive amount of a Diaminopyrimidine Compound. Examples of such conditions include rheumatoid arthritis; rheumatoid spondylitis; osteoarthritis; asthma, bronchitis; allergic rhinitis; chronic obstructive pulmonary disease; cystic fibrosis; inflammatory bowel disease; irritable bowel me; mucous s; ulcerative colitis; Crohn's disease; Huntington's disease; hepatitis; pancreatitis; nephritis; multiple sclerosis; lupus erythematosus; Type II diabetes; obesity; atherosclerosis; restenosis following angioplasty; left ventricular ATI—ZS 14175v1 rophy; myocardial infarction; stroke; ischemic damages of heart, lung, gut, kidney, liver, pancreas, spleen and brain; acute or chronic organ transplant rejection; preservation of the organ for transplantation; organ failure or loss of limb (e.g., including, but not limited to, that resulting from ischemia-reperfusion injury, trauma, gross bodily injury, car accident, crush injury or lant failure); graft versus host disease; endotoxin shock; multiple organ failure; psoriasis; burn from exposure to fire, chemicals or radiation; eczema; dermatitis; skin graft; ia; ischemic conditions ated with surgery or traumatic injury (e.g., vehicle accident, gunshot wound or limb crush); epilepsy; Alzheimer's disease; Parkinson's disease; immunological response to bacterial or viral infection; ia; angiogenic and proliferative diseases; solid tumor; and cancers of a variety of tissues such as colon, rectum, prostate, liver, lung, bronchus, pancreas, brain, head, neck, stomach, skin, , cervix, blood, larynx, esophagus, mouth, pharynx, y bladder, ovary or uterine.
PHARMACEUTICAL COMPOSITIONS AND ROUTES OF ADMINISTRATION The opyrimidine Compounds can be administered to a t orally, topically or parenterally in the conventional form of preparations, such as capsules, microcapsules, tablets, granules, powder, troches, pills, suppositories, injections, suspensions, syrups, patches, creams, lotions, ointments, gels, sprays, solutions and emulsions. Suitable formulations can be ed by methods commonly employed using conventional, c or inorganic additives, such as an excipient (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate or calcium carbonate), a binder (e.g, cellulose, methylcellulose, ymethylcellulose, polypropylpyrrolidone, nylpyrrolidone, n, gum arabic, polyethyleneglycol, sucrose or starch), a disintegrator (e.g, starch, carboxymethylcellulose, hydroxypropylstarch, low tuted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate or calcium citrate), a lubricant (e.g., magnesium stearate, light anhydrous silicic acid, talc or sodium lauryl sulfate), a ng agent (e.g., citric acid, menthol, glycine or orange powder), a preservative (e. g, sodium benzoate, sodium bisulf1te, methylparaben or propylparaben), a stabilizer (e.g., citric acid, sodium citrate or _ 60 _ ATI—2514l75v1 2012/034349 acetic acid), a suspending agent (e.g, methylcellulose, polyvinyl pyrroliclone or aluminum stearate), a dispersing agent (e.g., hydroxypropylmethylcellulose), a diluent (e.g., water), and base wax (e.g., cocoa butter, white petrolatum or polyethylene glycol). The ive amount of the Diaminopyrimidine nds in the pharmaceutical composition may be at a level that will exercise the desired effect; for example, about 0.005 mg/kg of a subject’s body weight to about 10 mg/kg of a subject’s body weight in unit dosage for both oral and parenteral administration.
The dose of a Diaminopyrimidine Compound to be administered to a subject is rather widely le and can be subject to the nt of a health-care practitioner. In general, the Diaminopyrimidine Compounds can be administered one to four times a day in a dose of about 0.005 mg/kg of a subject’s body weight to about mg/kg of a subject’s body weight in a subject, but the above dosage may be properly varied depending on the age, body weight and medical condition of the subject and the type of stration. In one embodiment, the dose is about 0.01 mg/kg of a subject’s body weight to about 5 mg/kg of a subject’s body weight, about 0.05 mg/kg of a subject’s body weight to about 1 mg/kg of a subject’s body weight, about 0.1 mg/kg of a subject’s body weight to about 0.75 mg/kg of a subject’s body weight or about 0.25 mg/kg of a subject’s body weight to about 0.5 mg/kg of a subject’s body weight. In one embodiment, one dose is given per day. In any given case, the amount of the Diaminopyrimidine Compound administered will depend on such factors as the solubility of the active component, the formulation used and the route of administration. In one embodiment, application of a topical concentration provides intracellular exposures or concentrations of about 0.01 — 10 uM.
In another embodiment, ed herein are methods for the treatment or prevention of a disease or disorder comprising the stration of about 0.375 mg/day to about 750 , about 0.75 mg/day to about 375 mg/day, about 3.75 mg/day to about 75 mg/day, about 7.5 mg/day to about 55 mg/day or about 18 mg/day to about 37 mg/day of a Diaminopyrimidine Compound to a subject in need thereof.
In another embodiment, provided herein are methods for the treatment or tion of a e or disorder comprising the administration of about 1 mg/day to about 1200 mg/day, about 10 mg/day to about 1200 mg/day, about 100 mg/day to about ATI—2514l75vl 1200 mg/day, about 400 mg/day to about 1200 mg/day, about 600 mg/day to about 1200 mg/day, about 400 mg/day to about 800 mg/day or about 600 mg/day to about 800 mg/day of a Diaminopyrimidine Compound to a subject in need thereof. In a particular embodiment, the methods sed herein comprise the administration of 400 mg/day, 600 mg/day or 800 mg/day of a Diaminopyrimidine Compound to a t in need thereof.
] In another ment, provided herein are unit dosage formulations that comprise between about 1 mg and 200 mg, about 35 mg and about 1400 mg, about 125 mg and about 1000 mg, about 250 mg and about 1000 mg, or about 500 mg and about 1000 mg of a Diaminopyrimidine Compound.
In a particular embodiment, provided herein are unit dosage ations comprising about 100 mg or 400 mg of a Diaminopyrimidine Compound.
In another embodiment, provided herein are unit dosage formulations that comprise 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 35 mg, 50 mg, 70 mg, 100 mg, 125 mg, 140 mg, 175 mg, 200 mg, 250 mg, 280 mg, 350 mg, 500 mg, 560 mg, 700 mg, 750 mg, 1000 mg or 1400 mg of a Diaminopyrimidine Compound.
A Diaminopyrimidine Compound can be administered once, twice, three, four or more times daily. In a particular embodiment, doses of 600 mg or less are administered as a once daily dose and doses of more than 600 mg are administered twice daily in an amount equal to one half of the total daily dose.
A Diaminopyrimidine Compound can be administered orally for reasons of convenience. In one embodiment, when stered orally, a Diaminopyrimidine Compound is administered with a meal and water. In another embodiment, the Diaminopyrimidine Compound is dispersed in water or juice (e.g., apple juice or orange juice) and administered orally as a suspension.
The Diaminopyrimidine Compound can also be administered intradermally, intramuscularly, intraperitoneally, percutaneously, intravenously, subcutaneously, intranasally, ally, sublingually, intracerebrally, aginally, transdermally, rectally, mucosally, by tion, or topically to the ears, nose, eyes, or skin. The mode of administration is left to the discretion of the health-care practitioner, and can depend in- part upon the site of the medical condition.
ATI—25 14175v1 In one embodiment, provided herein are capsules containing a Diaminopyrimidine Compound without an additional carrier, excipient or vehicle.
In another embodiment, provided herein are compositions comprising an effective amount of a Diaminopyrimidine Compound and a pharmaceutically able carrier or vehicle, wherein a pharmaceutically acceptable r or vehicle can comprise an excipient, t, or a e thereof. In one embodiment, the composition is a pharmaceutical composition.
The itions can be in the form of tablets, chewable tablets, es, solutions, parenteral solutions, troches, suppositories and suspensions and the like.
Compositions can be formulated to contain a daily dose, or a convenient fraction of a daily dose, in a dosage unit, which may be a single tablet or capsule or convenient volume of a liquid. In one ment, the ons are prepared from water-soluble salts, such as the hydrochloride salt. In general, all of the compositions are prepared according to known s in pharmaceutical chemistry. Capsules can be prepared by mixing a Diaminopyrimidine Compound with a le carrier or diluent and filling the proper amount of the mixture in capsules. The usual rs and diluents include, but are not limited to, inert powdered substances such as starch ofmany different kinds, powdered cellulose, especially crystalline and microcrystalline cellulose, sugars such as fructose, mannitol and sucrose, grain flours and similar edible powders.
] Tablets can be prepared by direct compression, by wet granulation, or by dry granulation. Their ations usually incorporate diluents, binders, lubricants and disintegrators as well as the compound. Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, nic salts such as sodium chloride and powdered sugar. Powdered cellulose derivatives are also useful. Typical tablet binders are substances such as starch, n and sugars such as lactose, fructose, glucose and the like. Natural and synthetic gums are also convenient, including acacia, alginates, methylcellulose, polyvinylpyrrolidine and the like.
Polyethylene glycol, ethylcellulose and waxes can also serve as binders.
A lubricant might be necessary in a tablet formulation to t the tablet and s from sticking in the dye. The lubricant can be chosen from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable ATI—ZS 14175v1 oils. Tablet disintegrators are substances that swell when wetted to break up the tablet and e the compound. They include starches, clays, celluloses, algins and gums. More particularly, corn and potato starches, methylcellulose, agar, bentonite, wood cellulose, powdered natural sponge, -exchange resins, alginic acid, guar gum, citrus pulp and carboxymethyl cellulose, for example, can be used as well as sodium lauryl sulfate.
Tablets can be coated with sugar as a flavor and sealant, or with film-forming protecting agents to modify the dissolution properties of the tablet. The compositions can also be formulated as chewable tablets, for example, by using substances such as mannitol in the formulation.
When it is desired to administer a Diaminopyrimidine Compound as a suppository, typical bases can be used. Cocoa butter is a traditional itory base, which can be modified by addition of waxes to raise its melting point slightly. Water- miscible suppository bases comprising, particularly, polyethylene glycols of various molecular weights are in wide use.
] The effect of the opyrimidine Compound can be delayed or prolonged by proper formulation. For example, a slowly soluble pellet of the Diaminopyrimidine nd can be prepared and incorporated in a tablet or capsule, or as a slow-release implantable device. The technique also includes making pellets of several ent dissolution rates and filling capsules with a mixture of the pellets.
Tablets or capsules can be coated with a film that resists dissolution for a predictable period of time. Even the parenteral preparations can be made long-acting, by dissolving or suspending the Diaminopyrimidine nd in oily or emulsified vehicles that allow it to disperse slowly in the serum.
EXAMPLES The following Examples are presented by way of ration, not limitation.
Compounds are named using the automatic name ting tool provided in Chemdraw Ultra 9.0 (Cambridgesoft), which generates systematic names for chemical structures, with support for the Cahn-Ingold-Prelog rules for stereochemistry. One skilled in the art can modify the procedures set forth in the illustrative es to arrive at the desired products.
ATI—ZS 14175v1 [001 84] Abbreviations used: DCM Dichloromethane DEA D1ethylamine EDC Ethyl-(N ’ ,N ’ hylamino)propylcarbodiimide hydrochloride HATU zabenzotriazol- l -yl)-N,N,N',N'- tetramethyluronium hexafluorophosphate HOBt l -Hydroxybenzotriazole HPLC High performance liquid chromatography HTRF Homogeneous time resolved fluorescence LCMS Liquid chromatography mass spectrometry mCPBA Meta-chloroperoxybenzoic acid MS Mass spectrometry N-methylpyrrolidone Nuclear magnetic resonance Supercritical fluid chromatography O-benzotriazol- l -yl-N,N,N',N'-tetra- methyluronium tetrafluoroborate Triethylamine Trifluoracetic acid Tetrahydrofuran Thin layer tography ATI—2514l75vl COMPOUND SYNTHESIS e 1: 4-((1R,3R)Hydr0xycyclohexylamin0)((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarboxamide 4-Chlor0((1r,4r)—4-methoxycyclohexylamino)pyrimidine—5- carbonitrile. 2,4-Dichloro-pyrimidinecarbonitrile (1.2 g, 6.93 mmol) in anhydrous ethanol (10 mL) and (1r,4r)methoxycyclohexanamine (893 mg, 6.93 mmol) in anhydrous ethanol (10 mL) were mixed at -60 CC, then DIEA (1.34 g, 10.4 mmol) was added drop wise. The mixture was stirred at -60 0C for 1.5 h, then at room temperature overnight. Volatile fractions were d and the residue was purified on silica gel (eluting with 9.1%-25% ethyl acetate in petroleum ether and .7% methanol in DCM) to give the 2 isomers (identified as described below), namely 4-chloro((1r,4r) methoxycyclohexylamino)-pyrimidinecarbonitrile (560 mg, 2.10 mmol, yield 30%) as a white solid and 2-chloro((1r,4r)methoxycyclohexylamino)pyrimidinecarbonitrile (227 mg, 0.85 mmol, yield 12%) as a white solid. MS(ESI): m/z 266.9 [M+l]+.
] Identification of the two isomers was achieved by characterization of the dehalogenated intermediate. Dehalogenation for the two fractions was achieved as follows. 4-Chloro((1r,4r)methoxycyclohexylamino)pyrimidinecarbonitrile (50 mg, 0.18 mmol) and Raney nickel in a ent of THF (10 mL) and aqueous ammonia (1 mL) was stirred at room temperature under a hydrogen balloon overnight.
The mixture was d and the filtrate was concentrated and purified by reverse-phase preparatory HPLC (10% to 40% acetonitrile + 0.005% ammonia solution) to give -(aminomethyl)-N-((1r,4r)methoxycyclohexyl)pyrimidinamine (25 mg, 0.10 mmol, yield 58%). 1H NMR (CDC13 300 MHz): 5 ppm 8.23 (s, 2H), 4.96 (d, J=10.0 Hz, 1H), 3.85-3.73 (m, 1H), 3.69 (s, 2H), 3.34 (s, 3H), .11 (m, 1H), .04 (m, 4H), 1.44-1.34 (m, 4H); MS(ESI): m/Z 236.9 [M+1]+. [001 87] Similarly, 2-chloro((1r,4r)methoxycyclohexylamino)pyrimidine carbonitrile (50 mg, 0.18 mmol) and Raney nickel in a cosolvent of THF (10 mL) and ATI—25 14175v1 2012/034349 aqueous ammonia (1 mL) was stirred at room temperature under a hydrogen balloon overnight. The mixture was filtered and the filtrate was concentrated and purified by reverse-phase preparatory HPLC (10% to 40% acetonitrile + 0.005% a on) to give 5-(aminomethyl)-N-((1r,4r)methoxycyclohexyl)pyrimidinamine (30 mg, 0.12 mmol, yield 66%). 1H NMR (CDC13 300 MHz): 8 ppm 8.46 (s, 1H), 7.84 (s, 1H), 7.25 (s, 1H), 4.00-3.93 (m, 1H), 3.79 (s, 2H), 3.34 (s, 3H), 3.23-3.14 (m, 1H), 2.14-2.02 (m, 4H), 1.48-1.22 (m, 4H); MS(ESI): m/z 236.9 [M+1]+.
B. 4-((1R,3R)—3-Hydr0xycyclohexylamin0)—2-((1r,4R)—4—meth0xy- cyclohexylamin0)pyrimidinecarb0nitrile. A mixture of 4-chloro((1r,4r) methoxycyclohexylamino)pyrimidinecarbonitrile (236 mg, 0.88 mmol), (1R,3R)aminocyclohexanol (151 mg, 1.32 mmol; prepared as described in Tetrahedron: Asymmetry 15:2051—2056 (2004)) and cesium carbonate (573 mg, 1.76 mmol) in anhydrous nol (20 mL) was stirred at 120 CC under nitrogen for 3 h.
The mixture was partitioned between water and DCM. The organic layers were combined, concentrated and purified on silica gel (eluting with 16%-50% ethyl e in petroleum ether and 3.2%-4.7% methanol in DCM) to give 4-((1R,3R)—3-hydroxycyclohexylamino)- ,4R)—4-methoxycyclohexylamino)-pyrimidinecarbonitrile (140 mg, 0.40 mmol, yield 46%) as a white solid. MS(ESI): m/z 346.0 [M+1]+.
C. 4-((1R,3R)Hydr0xycyclohexylamin0)((1r,4R)—4-meth0xy- cyclohexylamin0)pyrimidine—5-carboxamide. 4-((1R,3R)Hydroxycyclohexylamino)- 2-((1r,4R)—4-methoxycyclohexylamino)pyrimidinecarbonitrile (140 mg, 0.40 mmol) was dissolved in DMSO (10 mL) and ten drops of ted aqueous sodium hydroxide solution and ten drops of aqueous hydrogen peroxide solution (30%) were added at room temperature. The reaction mixture was stirred at 50 CC for 6 h. The reaction mixture was partitioned between water and DCM-isopropanol (5:1). The c layers were combined, concentrated and d on silica gel (eluting with 50%-75% ethyl acetate in petroleum ether and 4.76%-9. 1% methanol in DCM) to give 4-((1R,3R) hydroxycyclohexylamino)((1r,4R)methoxycyclohexylamino)-pyrimidine carboxamide (103 mg, 0.28 mmol, yield 70%) as a white solid. 1H NMR (DMSO-d6 400 MHz): 8 ppm 9.00 (d, J=5.2 Hz, 1H), 8.33 (s, 1H), 7.54 (brs, 1H), 7.00 (s, 1H), 6.81 ATI—25 14175v1 (s, 1H), 4.47 (s, 1H), 4.28 (s, 1H), 3.81 (s, 1H), 3.63 (s, 1H), 3.22 (s, 3H), 3.08 (s, 1H), 1.98-1.54 (m, 8H), 1.41—1.14 (m, 7H); MS(ESI): m/Z 363.9 [M+1]+.
Example 2: 4-((1S,3S)—3-Hydr0xycyclohexylamin0)((1r,4S)—4- ycyclohexylamin0)pyrimidinecarboxamide A. 4-((1 S,3 S)—3-Hydr0xycyclohexylamin0)((1r,4S)—4-meth0xy- cyclohexylamin0)pyrimidinecarb0nitrile. A mixture of 4-chloro((1r,4r) methoxycyclohexylamino)pyrimidinecarbonitrile (236 mg, 0.88 mmol, synthesis described herein), (1S,3S)aminocyclohexanol (209 mg, 1.33 mmol; ed as described in Tetrahedron: Asymmetry 15: 2051—2056 (2004)) and cesium carbonate (573 mg, 1.76 mmol) in anhydrous n-butanol (20 mL) was stirred at 120 CC under nitrogen for 3 h. The mixture was ioned between water and DCM. The c layers were combined, concentrated and purified on silica gel (eluting with 16%-50% ethyl acetate in petroleum ether and 3.2%-4.7% methanol in DCM) to give ,3S)—3- hydroxycyclohexylamino)((1r,4S)methoxycyclohexylamino)pyrimidine carbonitrile (140 mg, 0.40 mmol, yield 46%) as a white solid. MS(ESI): m/Z 346.1 [M+l]+.
B. 4-((1 S,3 S)—3-Hydr0xycyclohexylamin0)((1r,4S)—4-meth0xy- exylamin0)pyrimidinecarboxamide. 4-((1S,3S)Hydroxycyclohexylamino)- 2-((1r,4S)methoxycyclohexylamino)pyrimidinecarbonitrile (140 mg, 0.40 mmol) was dissolved in DMSO (10 mL) and ten drops of saturated aqueous sodium hydroxide solution and ten drops of aqueous hydrogen peroxide solution (30%) were added at room temperature. The reaction mixture was stirred at 50 CC for 6 h. The reaction mixture was partitioned between water and DCM-isopropanol (5:1). The organic layers were ed, concentrated and purified on silica gel (eluting with 50%-75% ethyl acetate in petroleum ether and 4.76%-9.1% methanol in DCM) to give 4-((lS,3S)—3- hydroxycyclohexylamino)((1r,4S)methoxycyclohexylamino)pyrimidine carboxamide (102 mg, 0.28 mmol, yield 70%) as a white solid. 1H NMR d6 ATI—2514l75v1 400 MHZ): 8 ppm 9.01 (d, J=6.0 HZ, 1H), 8.34 (s, 1H), 7.56 (brs, 1H), 7.02 (d, J=4.8 HZ, 1H), 6.82 (s, 1H), 4.48 (s, 1H), 4.29 (s, 1H), 3.81 (s, 1H), 3.64 (s, 1H), 3.23 (s, 3H), 3.08 (s, 1H), 2.01-1.55 (m, 8H), 1.41-1.15 (m, 7H); MS(ESI): m/z 363.9 [M+1]+.
Example 3: 2-((1r,4S)Eth0xycyclohexylamin0)((1S,ZS) hydroxycyclopentylamin0)pyrimidinecarboxamide V0 N/ l NH2 ” N 1ng A. tert-Butyl (1r,4r)—4-hydr0xycyclohexylcarbamate. To a suspension of )aminocyclohexanol hydrochloride (25 g, 217 mmol) in e (200 mL) was added a solution of sodium hydroxide (8.7 g, 217 mmol) in water (150 mL) at room temperature, followed by addition of di-tert-butyl-dicarbonate (47 g, 270 mmol). The ing mixture was stirred at room temperature for 15 h. When the ng material was ed, the volatiles were removed and the residue was diluted with water. The solid was collected by filtration and dried to give tert-butyl (1r,4r)hydroxycyclohexyl- carbamate as a white solid (33 g, 0.15 mol, yield 92%). 1H NMR dg, 300 MHZ): 8 ppm 6.65 (d, J=7.5 HZ, 1H), 4.48 (d, J=3.0 HZ, 1H), 3.14 (s, 1H), 1.77 (m, 4H), 1.37 (s, 9H), 1.14 (m, 4H).
B. utyl (1r,4r)—4-eth0xycyclohexylcarbamate. To a solution of tert- butyl (1r,4r)hydroxycyclohexylcarbamate (20 g, 93 mmol) in ous THF (100 mL) was added sodium hydride (4 g, 100 mmol, 60% in mineral oil) in portions under nitrogen at 0 CC. The mixture was warmed to room temperature and stirred for 30 min. A solution of iodoethane (16 g, 102 mmol) in anhydrous THF (50 mL) was added dropwise to the mixture at 0 OC and the resulting mixture was heated to 60 0C for 15 h. After being cooled to room temperature, the mixture was poured into ice-water (100 mL). The aqueous mixture was extracted with ethyl acetate (100 mL X 3), and the organic phase was washed with brine, dried over sodium sulfate and filtered. The filtrate was concentrated in vacuo.
The residue was purified on silica gel column (eluting with 5% to 30% ethyl acetate in petroleum ether) to give tert-butyl (1r,4r)ethoxycyclohexylcarbamate (3.7 g, 15 mmol, yield 16%). 1H NMR (DMSO-dg, 400 MHZ): 8 ppm 6.70 (d, J=7.6 HZ, 1H), 3.42 (q, ATI—25 14175v1 J=7.2 Hz, 2H), 3.11-3.17 (m, 2H), 1.92 (m, 2H), 1.74 (m, 2H), 1.37 (s, 9H), 1.21 (m, 4H), 1.07 (t, J=7.2 Hz, 3H).
C. (1r,4r)—4-Eth0xycyclohexanamine hydrochloride. A solution of tert- butyl (1r,4r)ethoxycyclohexylcarbamate (3.7 g, 15 mmol) in methanolic hydrochloride solution (2 M, 20 mL) was stirred at room temperature for 2 h. The reaction mixture was evaporated under reduced re to give (1r,4r)ethoxycyclohexanamine hydrochloride (2.7 g, yield 100%).
D. (1S,2S)Aminocyclopentanol hydrochloride. To a on of (1S,2S)—2-(benzyloxy)cyclopentanamine (4.5 g, 22 mmol) and hloric acid (0.5 mL) in methanol (100 mL) was added 10 wt.% palladium on activated carbon (500 mg). The mixture was stirred at room temperature under hydrogen atmosphere (40 psi) for 24 h.
The catalyst was filtered off and the e was concentrated under reduced pressure to give (1S,2S)—2-aminocyclopentanol hydrochloride as a white solid (2.14 g, 15 mmol, yield 93%).
E. Ethyl 4-((1 S,2S)hydroxycyclopentylamin0)(methylthio)— dine-S-carboxylate. A e of ethyl 4-chloro(methylthio)pyrimidine carboxylate (1.4 g, 6.0 mmol), (1S,2S)aminocyclopentanol hydrochloride (0.7 g, 6.9 mmol) and DIEA (1.0 g) in ethanol (20 mL) was heated at 60 0C for 15 h. After being cooled to room temperature, the reaction solution was concentrated and the residue was purified on silica gel column (eluting with 0~10% ethyl acetate in petroleum ether) to give ethyl 4-((1S,2S)hydroxycyclopentylamino)(methylthio)pyrimidinecarboxylate (1.3 g, 4.37 mmol, yield 73%) as a white solid. MS (ESI): m/z 298.5 [M+1]+.
F. 4-((1 S,2S)Hydr0xycyclopentylamin0)(methylthio)pyrimidine— -carb0xylic acid. To a solution of ethyl ,2S)—2-hydroxycyclopentylamino) (methylthio)pyrimidinecarboxylate (1.3 g, 4.37 mmol) in l (15 mL) was added aqueous sodium hydroxide solution (15 mL, 2 N), and the mixture was stirred for 1 h at room temperature. When the starting al was consumed, the reaction mixture was neutralized with saturated aqueous citric acid solution. The solids were collected by filtration and dried to give 4-((1S,2S)hydroxycyclopentylamino) (methylthio)pyrimidinecarboxylic acid (1.0 g, 3.7 mmol, yield 85%) as a white solid.
MS (ESI): m/Z 270.6 [M+1]+.
ATI—2514175vl G. 4-((1 S,2S)Hydr0xycyclopentylamin0)(methylthio)pyrimidine- -carb0xamide. The e of 4-((1S,2S)—2-hydroxycyclopentylamino)(methylthio)- pyrimidinecarboxylic acid (1.0 g, 3.7 mmol), ammonium chloride (0.989 g, 18.5 mmol), HATU (2.25 g, 5.92 mmol), DIEA (2.39 g, 18.5 mmol) and l-HOBt (0.80 g, .92 mmol) in DMF (10 mL) was d at room temperature for 2 h. The reaction mixture was diluted with water (15 mL), and the aqueous mixture was extracted with ethyl acetate (20 mL X 3). The combined organic layers were washed with brine, dried over sodium sulfate and filtered. The filtrate was ated in vacuo. The residue was purified on silica gel column (eluting with 30~50% ethyl e in petroleum ether) to give 4-((1 S,2S)—2-hydroxycyclopentylamino)(methylthio)pyramidinecarboxamide (800 mg, 2.98 mmol, yield 81%) as a white solid. MS (ESI): m/z 269.4 [M+1]+.
H. 4-((1 S,2S)Hydr0xycyclopentylamin0)(methylsulf0nyl)— pyrimidine—5-carb0xamide. To a mixture of 4-((1S,2S)—2-hydroxycyclopentylamino) (methylthio)pyrimidinecarboxamide (800 mg, 2.98 mmol) in acetone (10 mL) was added a solution of potassium peroxymonosulfate (4.66 g, 7.45 mmol) in water (10 mL), and the resulting mixture was stirred at room temperature for 2 h. When the starting material was consumed, the reaction mixture was partitioned n ethyl acetate (20 mL) and water (15 mL). The aqueous layer was extracted with ethyl acetate (20 mL X 3). The combined organic layers were washed with brine, dried over sodium sulfate and filtered. The filtrate was evaporated in vacuo to give 4-((lS,2S)—2- hydroxycyclopentylamino)(methylsulfonyl)pyrimidinecarboxamide (700 mg, 2.3 mmol, yield 78%) as a solid. MS (ESI): m/z 301.2 [M+1]+.
I. ,4S)Ethoxycyclohexylamino)—4-((1S,2S)hydr0xycyclopentylamin0 )pyrimidine—5-carboxamide. A solution of 4-((1S,2S) hydroxycyclopentylamino)(methylsulfonyl)pyrimidinecarboxamide (700 mg, 2.3 mmol), (1r,4r)ethoxycyclohexanamine hydrochloride (668 mg, 4.66 mmol) and DIEA (600 mg, 4.66 mmol) in dioxane (10 mL) was heated at 80 0C for 15 h. After being cooled to room temperature, the on mixture was concentrated and the residue was dissolved in ethyl acetate (20 mL). The organic solution was washed with water and brine, dried over sodium sulfate and filtered. The e was evaporated in vacuo. The residue was purified by a e-phase preparatory HPLC (40-75% acetonitrile + 0.05% ATI—2514l75vl ammonium hydroxide in water, 7.5 min.) to give 2-((1r,4S)ethoxycyclohexylamino) ((1S,2S)hydroxycyclopentylamino)pyrimidinecarboxamide (320 mg, 0.88 mmol, yield 38%, mp. l26.0~l26.6°C) as a solid. 1H NMR (DMSO-dg, 400 MHz): 5 ppm 9.02-8.93 (m, 1H), 8.32 (d, J=9.2 Hz, 1H), 7.60-7.51 (m, 1H), 7.04 (d, J=7.2 Hz, 1H), 6.88 (d, J=7.2 Hz, 1H), 4.82 (s, 1H), 3.99-3.89 (m, 3H), 3.42 (q, J=7.2 Hz, 2H), 3.15 (s, J=6.0 Hz, 1H), 2.10-2.06 (m, 1H), 1.97-1.77 (m, 5H), 1.66-1.60 (m, 2H), 1.46 (s, 1H), 1.28-1.18 (m, 5H), 1.06 (t, J=7.2 Hz, 3H); MS (ESI): m/z 364.3 [M+l]+.
Example 4: 4-((1R,3S)—3-Hydr0xycyclohexylamin0)((1R,4R)—4- (methylcarbamoyl)cyclohexylamin0)pyrimidine—5-carboxamide O 0 We 11*"’N N/ NH ‘\gOH A. Ethyl ,3S)—3-hydr0xycyclohexylamin0)(methylthi0)— dine-S-carboxylate. Ethyl 4-chloro(methylthio)pyrimidinecarboxylate (4.38 g, 18.82 mmol) and (1S,3R)aminocyclohexanol (2.276 g, 19.76 mmol; prepared as described in Tetrahedron: Asymmetry 1—2056 (2004)) were dissolved in ethanol (75 mL) before adding DIEA (4.93 ml, 28.2 mmol) and g to 60 CC. After 2 hrs, LCMS showed the desired t mass was the dominant peak formed. The reaction was removed from heat and concentrated. The crude material was purified on a 340G SNAP Biotage column (20-100% ethyl acetate in hexane) to give ethyl ,3S)hydroxycyclohexylamino )(methylthio)pyrimidinecarboxylate (5 g, 16.06 mmol, 85 % yield) as a white foam; MS (ESI) m/z 312.1 [M+1]+ B. 4-((1R,3S)—3-Hydr0xycyclohexylamin0)(methylthio)pyrimidine—5- carboxylic acid. Ethyl 4-((1R,3 S)hydroxycyclohexylamino)(methylthio)- pyrimidinecarboxylate (5 g, 16.06 mmol) was dissolved in ethanol (50 mL) before adding sodium hydroxide solution (2 M in water, 20 mL, 40.0 mmol) and stirring at room temperature. After 30 min, LCMS shows mostly desired product mass. The reaction mixture was neutralized by addition of saturated aqueous citric acid on. The resulting precipitate was filtered and dried to give 4-((1R,3S)hydroxycyclohexylamino) _ 72 _ ATI—2514l75vl (methylthio)pyrimidinecarboxylic acid (4.5 g, 15.88 mmol, 99 % yield) as a white solid; 1H NMR (400 MHz, DMSO-d6) 5 ppm 13.21 (br. s., 1H), 8.44 - 8.54 (m, 2H), 4.74 (d, J=3.90 Hz, 1H), 3.97 - 4.12 (m, 1H), 3.56 (d, J=3.12 Hz, 1H), 2.46 (s, 3H), 2.05 - 2.16 (m, 1H), 1.84 (d, J=10.15 Hz, 1H), 1.65 - 1.80 (m, 2H), 1.11 - 1.36 (m, 4H); MS (ESI) m/z 284.1 [M+1]+ C. 4-((1R,3S)—3-Hydr0xycyclohexylamin0)(methylthi0)pyrimidine amide. 4-((1R,3 S)Hydroxycyclohexylamino)(methylthio)pyrimidine-5 - carboxylic acid (4.5 g, 15.88 mmol) and HATU (9.06 g, 23.82 mmol) were dissolved in DMF (75 mL) and allowed to stir for 5 min at room temperature before adding ammonium chloride (4.25 g, 79 mmol) and DIEA (13.87 mL, 79 mmol). The on was allowed to stir at room temperature ght. After LCMS showed the d product mass as the dominant peak, the reaction was partitioned between water and ethyl acetate. The organic layer was washed once with brine before drying over sodium sulfate, filtering, and sing. After drying, 4-((1R,3 S)hydroxycyclohexylamino)(methylthio)- pyrimidinecarboxamide (4.19 g, 14.84 mmol, 93 % yield) was obtained as an off-white solid; MS (ESI) m/z 283.2 [M+1]+ D. 4-((1R,3S)Hydroxycyclohexylamin0)(methylsulf0nyl)— pyrimidine—5-carb0xamide. 4-((1R,3S)Hydroxycyclohexylamino)(methylthio)- pyrimidinecarboxamide (302 mg, 1.070 mmol) was suspended in DCM (10 mL) and acetone (10 mL). mCPBA (479 mg, 2.139 mmol) was then added and the reaction was stirred at room temperature. After 90 min, LCMS showed the desired product mass as the dominant peak. The reaction was quenched by addition of 10 mL of 10% aqueous sodium lfate solution. After stirring for 5 minutes, the reaction was partitioned between ethyl acetate and water. The organic layer was then washed with saturated sodium bicarbonate and brine. The combined s layers were washed with ethyl acetate three times before combining the organic layers, drying over sodium sulfate, filtering, and condensing. After drying, 4-((1R,3 S)hydroxycyclohexylamino)(methylsulfonyl)- pyrimidinecarboxamide (259 mg, 0.824 mmol, 77 % yield) was obtained as a white solid ; MS (ESI) m/z 315.2 [M+1]+ E. (1R,4R)—4-Amino-N-methylcyclohexanecarboxamide. tert-Butyl (1R,4R)(methylcarbamoyl)cyclohexylcarbamate (0.411 g, 1.603 mmol) was dissolved ATI—2514l75vl WO 45569 in DCM (5 mL), treated with TFA (5.56 mL, 72.1 mmol) and stirred at room temperature for 1 h. After that the solvent was reduced in vacuo, the residue ved in acetonitrile and loaded on a Phenomenex Strada-X-C solid phase extraction column and flushed with 300 mL of acetonitrile. The desired compound was released from the column using 2 M ammonia in methanol. The on containing the desired compound was reduced in vacuo to afford (1R,4R)amino-N-methylcyclohexanecarboxamide (0.230 g, 1.475 mmol, 92 % yield); MS (ESI) m/Z 157.0 [M+1]+.
F. 4-((1R,3S)—3-Hydr0xycyclohexylamino)—2-((1R,4R)—4-(methyl- carbamoyl)cyclohexylamino)pyrimidine—5-carboxamide. 4-((1R,3S)Hydroxy- cyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (0.231 g, 0.736 mmol), (1R,4R)amino-N-methylcyclohexanecarboxamide (0.230 g, 1.472 mmol), DIEA (0.514 mL, 2.94 mmol), and DMSO (4 mL) were combined and heated at 100 CC for 2 h.
The solvent was evaporated under d pressure, the residue dissolved in a minimum amount of methanol, loaded on a silica gel column and purified (0-15% ammonia saturated methanol in DCM) to afford 4-((1R,3S)—3-hydroxycyclohexylamino)((1R,4R) (methylcarbamoyl)-cyclohexylamino)pyrimidinecarboxamide (0.106 g, 0.271 mmol, 36.9 % yield) as a white powder; MS (ESI) m/Z 391.2 [M+1]+; 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.10 (br. s., 1H), 8.33 (s, 1H), 7.68 (br. s., 1H), 6.92 - 7.29 (m, 3H), 4.67 (br. s., 1H), 3.87 (br. s., 1H), 3.41 = 4.30 Hz, 3H), 1.66 - 3.71 (m, 2H), 2.55 (d, J - 2.16 (m, 9H), 1.00 - 1.47 (m, 8H).
Example 5: 4-((1R,3S)—3-Hydr0xycyclohexylamino)—2-((1r,4R)—4- lamin0)cyclohexylamin0)pyrimidinecarboxamide hydrochloride / N \ NH2 ., )L / ’N N NH A. tert-Butyl )—4-(5-carbamoyl—4-((1R,3S)—3-hydr0xycyclohexyl— amin0)pyrimidinylamin0)cyclohexyl(methyl)carbamate. 4-((1R,3S)Hydroxycyclohexylamino )(methylsulfonyl)pyrimidinecarboxamide (0.397 g, 1.264 mmol; synthesis described herein), tert-butyl (1R,4R)aminocyclohexyl-(methyl)carbamate _ 74 _ ATI—25 14175v1 (0.577 g, 2.53 mmol), DIEA (0.883 mL, 5.05 mmol), and DMSO (4 mL) were combined and heated at 100 0C for 2 h. The solvent was evaporated under reduced pressure, the e dissolved in a minimum amount of methanol, loaded on a silica gel column and purified (0-l5% ammonia saturated methanol in DCM) to afford tert-butyl (lR,4r)—4-(5- carbamoyl(( l R,3 S)hydroxycyclohexylamino)pyrimidinylamino)cyclohexyl- (methyl)carbamate (0.210 g, 0.455 mmol, 36 % yield) as a white powder; MS (ESI) m/z 463.3 [M+1]+.
B. 4-((1R,3S)Hydr0xycyclohexylamino)—2-((1R,4R)—4-(methyl- amino)cyclohexylamin0)pyrimidine—5-carboxamide hydrochloride. tert-Butyl (lR,4r)- 4-(5-carbamoyl((lR,3S)hydroxycyclohexylamino)pyrimidinylamino)- cyclohexyl(methyl)carbamate (0.150 g, 0.324 mmol) was dissolved in DCM (2 mL) and TFA (2 mL, 26.9 mmol) was added drop wise. The solution was d at room temperature for l h and then concentrated under reduced pressure. The residue was purified via preparative HPLC (5-40% acetonitrile/water, 20mL/min.) to afford the title compound as the corresponding TFA-salt. The fractions containing the desired compound were concentrated under reduced pressure, the residue dissolved in methanol and hydrogen chloride (4N in dioxane; 3 mL) was added. The resulting solution was concentrated under reduced pressure and this ure was ed twice to afford 4-((lR,3S)hydroxy- cyclohexylamino)((lr,4R)(methylamino)cyclohexylamino)pyrimidine amide hydrochloride (0.063 g, 0. 158 mmol, 48.7 % . MS (ESI) m/z 363.5 [M+l]+; 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.94 (br. s., 1H), 8.83 - 9.01 (m, 1H), 8.38 - 8.49 (m, 2H), 8.15 (br. s., 1H), 7.64 (br. s., 1H), 3.96 (br. s., 1H), 3.69 (d, J = 13.67 Hz, 2H), 3.49 (d, J = 11.32 Hz, 3H), 2.97 (br. s., 1H), 2.52 (br. s., 2H), 1.69 - 2.17 (m, 7H), 3(nn8H) ATI—2514l75vl Example 6: 2-(4,4-Diflu0r0cyclohexylamin0)—4-((1R,3S)—3- hydroxycyclohexylamin0)pyrimidinecarboxamide ] 4-((1R,3 S)—3-Hydroxycyclohexylamino)(methylsulfonyl)pyrimidine-5 - carboxamide (478 mg, 1.521 mmol; synthesis described herein), 4,4-difluorocyclo- hexanamine hydrochloride (522 mg, 3.04 mmol), DIEA (1.062 mL, 6.08 mmol), and DMSO (8 mL) were ed in a round bottom flask and heated at 100 CC for 1 h. The reaction was removed from heat and condensed in vacuo. The crude material was purified on a 100G SNAP Biotage column (2-12% ammonia saturated methanol in DCM). The peak fractions were combined and evaporated. After drying under high , 2-(4,4-difluorocyclohexylamino)((1R,3 S)hydroxycyclohexylamino)pyrimidine-5 - carboxamide (325 mg, 0.880 mmol, 58%) was obtained as a slightly yellow powder; 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.93 (d, J=7.42 Hz, 1H), 8.36 (s, 1H), 7.22 (br. s., 1H), 2.94 - 3.05 (m, 1H), 1.64 - 2.21 (m, 14H), 1.36 - 1.62 (m, 4H), 0.92 - 1.33 (m, 3H); MS (ESI) m/z 370.2 [M+1]+.
ATI—25 14175V1 Example 7: ,5R)—5-Hydr0xytetrahydr0-2H-pyranylamin0)((1r,4S) methoxycyclohexylamin0)pyrimidine—5-carb0xamide, 4-((3R,SS)hydroxytetrahydro-ZH-pyranylamin0 )—2—((1r,4R)—4-meth0xycyclohexylamino)pyrimidine— -carb0xamide, 4-((3S,5S)hydr0xytetrahydr0-2H-pyranylamin0)—2-((1r,4S) methoxycyclohexylamin0)pyrimidinecarboxamide, 4-((3R,5R)—5- hydr0xy-tetrahydro-ZH-pyranylamin0)—2-((1r,4R)—4- methoxycyclohexylamin0)pyrimidine—5-carb0xamide. o O o O / \ / U 1W ”U 1% ”U 1W U 1W ., / "I 'N . ., N NH "N)\N/ N NH NH ’N N/ NH n H i H a H (Quw oé-, 0 ’OH OH QOH A. 0xy-2H-pyran-3(6H)—0ne. Furanylmethanol (17.67 mL, 204 mmol) was dissolved in DCM (500 mL) and cooled to 0 0C before adding 3-chloroperoxybenzoic acid (68.5 g, 306 mmol) portion wise. The reaction was slowly allowed to warm to room temperature over 6 h, during which time solid m-chlorobenzoic acid precipitated from solution. The solution was cooled to -78 CC for 15 min and the solids were filtered off. The e was condensed to give a yellow solid. The crude material was purified on a 340G SNAP Biotage column (20-100% ethyl acetate in hexane). The peak fractions were combined and ated to give 6-hydroxy-2H-pyran- one (15.8 g, 138 mmol, 67.9 % yield) as a pale yellow oil; 1H NMR (400 MHz, CDC13)5 ppm 6.94 - 7.01 (m, 1H), 6.18 (d, J=10.93 Hz, 1H), 5.61 - 5.68 (m, 1H), 4.56 (s, 1H), 4.17 (s, 1H), 3.61 (br. s., 1H).
B. 6-(tert-Butyldimethylsilyloxy)—2H-pyran-3(6H)—0ne. 6-Hydroxy-2H- pyran-3(6H)-one (9 g, 79 mmol) was dissolved in DCM (250 mL), placed under a nitrogen atmosphere, and cooled to -78 oC. 2,6-Lutidine (13.78 mL, 118 mmol) was then added in one n, followed by slow addition of t-butyldimethylsilyltrifiuoromethanesulfonate (21.74 mL, 95 mmol). The reaction was allowed to slowly warm to 0 0C over 4 h. The on was quenched by addition of ~20 mL of water. The solution turned pale yellow upon quenching. After transferring to a separatory filnnel, the organic phase was washed successively with 10% citric acid and brine. The organic layer was then dried over sodium sulfate, filtered and condensed to give a yellow oil. The crude material was purified on a ATI—25 14175v1 340G SNAP Biotage column (0-20% ethyl acetate in hexane) to give 6-(tert- butyldimethylsilyloxy)-2H-pyran-3(6H)-one (9.90 g, 43.4 mmol, 55.0 % yield) as a yellow oil; 1H NMR (400 MHz, CDC13)5 ppm 6.87 (dd, J=10.54, 3.12 Hz, 1H), 6.08 (d, J=10.15 Hz, 1H), 5.53 (d, J=3.12 Hz, 1H), 4.51 (d, J=16.79 Hz, 1H), 4.08 (d, J=16.79 Hz, 1H), 0.92 (s, 9H), 0.17 (s, 6H).
C. 6-(tert—Butyldimethylsilyloxy)—3,6-dihydro-ZH-pyranol. To a cooled (-20 oC) solution of 6-(tert-butyldimethylsilyloxy)-2H-pyran-3(6H)-one (9.9 g, 43.4 mmol) and cerium(IH) chloride heptahydrate (16.15 g, 43.4 mmol) in methanol (173 mL) was added n wise sodium borohydride(1.640 g, 43.4 mmol). Intense bubbling was observed upon addition. After the e was stirred for 30 min at -20 0C, the reaction was quenched with acetone (~20 mL) and stirred at room temperature for 1 h.
Most of the volatile solvents were then evaporated. Brine was added and the slurry was transferred to a separatory . The mixture was extracted with DCM (thick emulsion formed) three times. The combined organic layers were dried over sodium sulfate, d and condensed to give crude 6-(tert-butyldimethylsilyloxy)-3,6-dihydro-2H-pyranol (5.6 g, 24.31 mmol, 56.1 % yield) as a tan colored oil; 1H NMR (400 MHz, CDClg) 5 ppm .95 (dd, 5, 2.34 Hz, 1H), 5.75 (d, 5 Hz, 1H), 5.25 (s, 1H), 4.15 (d, J=9.37 Hz, 1H), 3.72 - 3.82 (m, 2H), 1.74 (d, J=8.98 Hz, 1H), 0.86 - 0.95 (m, 9H), 0.13 (s, 6H).
D. 6-(tert—Butyldimethylsilyloxy)—3,6-dihydr0-2H-pyranyl e.
To a solution of 6-(tert-butyldimethylsilyloxy)-3,6-dihydro-2H-pyranol (13.3 g, 57.7 mmol) and TEA (16.09 mL, 115 mmol) in DCM (200 mL) was added acetic anhydride (27.2 mL, 289 mmol), and the mixture was stirred at room temperature ght. Methanol (~3 mL) was added and the mixture was stirred for 30 min at room temperature before adding water and transferring to a separatory funnel. The organic layer was washed with water and brine, dried over sodium sulfate, filtered and condensed. The crude material was purified on a 340G SNAP Biotage column (0-50% ethyl acetate in hexane) to give 6-(tert-butyldimethylsilyloxy)-3,6-dihydro-2H-pyranyl acetate (13.6 g, 49.9 mmol, 86 % yield) as a pale yellow oil; 1H NMR (400 MHz, CDClg) 5 ppm .84 - 5.88 (m, 2H), 5.26 - 5.28 (m, 1H), 5.20 - 5.25 (m, 1H), 3.84 (dd, J=6.25, 1.95 Hz, 2H), 2.08 (s, 3H), 0.91 (s, 9H), 0.13 (d, J=1.17 Hz, 6H).
ATI—25 14175v1 E. 3,6-Dihydr0-2H-pyranyl acetate. 6-(tert-Butyldimethylsilyloxy)- hydro-2H-pyranyl acetate (13.6 g, 49.9 mmol) was dissolved in DCM (250 mL), placed under a nitrogen atmosphere, and cooled to -30 oC (dry ice/acetone, until temperature was reached). Triethylsilane (15.95 mL, 100 mmol) was then added slowly via e, followed by drop wise addition of boron ride etherate (7.59 mL, 59.9 mmol). The reaction was kept under nitrogen and slowly allowed to warm. After 1 h, the on mixture was quenched by additions of saturated sodium onate.
After transferring to a separatory filnnel, the organic layer was washed with water and brine, dried over sodium sulfate, filtered and condensed. The crude material was purified on a 340G SNAP Biotage column (0-80% ethyl acetate in hexane). Evaporation of clean fractions gave hydro-2H-pyranyl acetate (5.5 g, 38.7 mmol, 77 % yield) as a colorless oil; 1H NMR (400 MHz, CDClg) 8 ppm 5.09 (dtt, J=5.73, 2.21, 2.21, 1.17, 1.17 Hz, 1H), 4.19 - 4.28 (m, 1H), 4.04 - 4.15 (m, 1H), 3.89 - 3.98 (m, 1H), 3.76 - 3.84 (m, 1H), 2.10 (s, 3H).
F. 3,6-Dihydr0-2H-pyranol. To a solution of 3,6-dihydro-2H-pyran yl acetate (5.5 g, 38.7 mmol) in methanol (130 mL) was added 10 drops of 25% sodium methoxide in ol. The solution was allowed to stir at room temperature. After min, TLC (10% ethyl e in hexane; permanganate stain) shows ~1 :1 ratio of starting material to product. An additional 10 drops of 25% sodium methoxide in methanol was added. After 30 min, TLC showed ~3:1 ratio of product to starting material.
An additional 10 drops of 25% sodium methoxide in methanol were added. After 30 more min, TLC showed complete conversion to product. Amberlist 15 was added and stirring was continued for 15 min before filtering off. Evaporation of the solvent gave hydro-2H-pyranol (3.8 g, 38.0 mmol, 98 % yield) as a colorless oil; 1H NMR (400 MHz,CDC13)8 ppm 5.90 - 6.02 (m, 2H), 4.19 (dt, J=2.83, 1.51 Hz, 1H), 4.15 (dt, J=2.83, 1.51 Hz, 1H), 4.09 (q, J=1.95 Hz, 1H), 4.05 (q, J=1.95 Hz, 1H), 3.96 - 4.00 (m, 1H), 1.92 (br. s., 1H).
G. 2H-Pyran-3(6H)—0ne. 3,6-Dihydro-2H-pyranol (2.44 g, 24.37 mmol) was dissolved in DCM (100 mL) and cooled to 0 0C before slowly adding Dess-Martin periodinane (10.34 g, 24.37 mmol). The reaction was allowed to slowly warm to room temperature over 5 h. The reaction was then filtered through celite. After ATI—25 14175v1 WO 45569 concentrating the filtrate, the crude al was purified on a 100G SNAP Biotage column (0-80% ethyl acetate in hexane) to give 2H-pyran-3(6H)-one (2.33 g, 23.75 mmol, 97 % yield) as a colorless oil; 1H NMR (400 MHz, CDClg) 5 ppm 7.07 - 7.17 (m, 1H), 6.15 - 6.25 (m, 1H), 4.39 (t, J=2.54 Hz, 2H), 4.19 (s, 2H).
H. Benzyl 5-0x0tetrahydr0-2H-pyranylcarbamate. 2H-pyran-3(6H)- one (2.33 g, 23.75 mmol), benzyl carbamate (4.31 g, 28.5 mmol), and DCM (2.375 mL) were added to a small vial and stirred usly. To the thick syrup was added bismuth(III) nitrate pentahydrate (1.728 g, 3.56 mmol). The vial was capped and the reaction was stirred vigorously overnight. The reaction was diluted with DCM and filtered through a pad of celite. The filtrate was condensed and purified on a 100G SNAP Biotage column (10-100% ethyl acetate in hexane) to give benzyl etrahydro-2H-pyran ylcarbamate (4.35 g, 17.45 mmol, 73.5 % yield) as a pale yellow oil; 1H NMR (400 MHz, CDCh)8pmn728-741Gm5HL520-528Gm1H)505-515&m2H)424-435 (m, 1H), 4.01 - 4.06 (m, 1H), 3.89 - 3.95 (m, 1H), 3.80 - 3.87 (m, 1H), 2.73 (d, J=5.47 Hz, 11D,264-271(nn1H) I. Benzyl 5-hydr0xytetrahydr0-2H-pyranylcarbamate. Benzyl 5- oxotetrahydro-2H-pyranylcarbamate (4.35 g, 17.45 mmol) and cerium(III) chloride heptahydrate (6.50 g, 17.45 mmol) were dissolved in methanol (100 mL) before cooling to 0 oC. Sodium borohydride (0.660 g, 17.45 mmol) was then added slowly. Intense bubbling was observed. The reaction was allowed to stir at 0 0C for 30 min before quenching by addition of acetone (~3 mL) and stirring at room temperature for additional min. The on was then evaporated to dryness. The material was ioned between DCM and water and the aqueous layer was washed with DCM (5X).
The organic layers were combined, dried over sodium sulfate, filtered, and condensed to give impure benzyl 5-hydroxytetrahydro-2H-pyranylcarbamate (2.5 g, 9.95 mmol, 57.0 % yield) as a white solid; 1H NMR (400 MHz, CDClg) 8 ppm 7.28 - 7.39 (m, 5H), .90 - 6.06 (m, 1H), 5.10 (d, J=4.29 Hz, 2H), 3.59 - 3.97 (m, 6H), 1.77 - 2.06 (m, 3H); MS (ESI) m/z 252.1 [M+1]+.
J. 5-Amin0tetrahydr0-2H-pyranol. Benzyl oxytetrahydro-2H- pyranylcarbamate (2.5 g, 9.95 mmol) was dissolved in DCM (20 mL) and methanol (20.00 mL) before loading catalyst (10% palladium on carbon) and g with a t-joint.
ATI—25 14175v1 The flask was evacuated and flushed with hydrogen (3X), ining a hydrogen atmosphere with a balloon. The reaction was allowed to stir at room temperature overnight. Once the al had all been converted to product (as determined by LCMS), the reaction was filtered through a pad of celite, washing ghly with DCM and methanol. The filtrate was condensed to give crude 5-aminotetrahydro-2H-pyranol (1.13 g, 9.65 mmol, 97 % yield) as tan gum. 1H NMR (400 MHz, DMSO-d6) 5 ppm 4.28 - 4.66 (m, 2H), 3.66 (ddd, J=10.93, 3.90, 1.56 Hz, 2H), 3.44 - 3.55 (m, 1H), 2.89 - 3.01 (m, 2H), 2.81 (s, 1H), 2.01 - 2.13 (m, 1H), 1.22 (d, J=12.10 Hz,1H).
K. Ethyl 2-((1r,4r)—4-meth0xycyclohexylamin0)(methylthi0)— pyrimidine-S-carboxylate. )Methoxycyclohexanamine (4.39 g, 34.0 mmol), ethyl 2-chloro(methylthio)pyrimidinecarboxylate (3.95 g, 16.98 mmol) DIEA (5.93 mL, 34.0 mmol) and 1,4-dioxane (100 mL) were combined and heated at 80 oC overnight.
The reaction was removed from heat and condensed. The crude material was purified on a Biotage (20-80% ethyl acetate in hexanes) to give ethyl ,4r) methoxycyclohexylamino)(methylthio)pyrimidinecarboxylate (3.1 g, 9.53 mmol, 56 % yield) as a pale yellow solid; 1H NMR (DMSO-d6 ,400 MHz) 8 ppm 8.60 (s, 0.35H), 8.54 (s, 0.65H), 8.05 (d, J=7.4 Hz, 0.64H), 7.87 (d, J=8.2 Hz, 0.36H), 4.22 (q, J=7.0 Hz, 2H), 3.69 - 3.90 (m, 1H), 3.23 (s, 3H), 3.00 - 3.20 (m, 1H), 2.40 (s, 2H), 2.38 (s, 1H), 1.79 - 2.12 (m, 4H), 1.09 - 1.44 ppm (m, 7H); MS (ESI) m/z 326.3 [M+1]+.
L. 2-((1r,4r)—4-Meth0xycyclohexylamin0)(methylthi0)pyrimidine carboxylic acid. Ethyl 2-((1r,4r)methoxycyclohexylamino)(methylthio)pyrimidine- -carboxylate (3.1 g, 9.53 mmol) was dissolved in ethanol (60 mL) before adding aqueous sodium hydroxide solution (2 M, 23.81 mL, 47.62 mmol) and ng at room temperature.
After 30 min, LCMS showed mostly d product mass. The reaction mixture was neutralized by addition of saturated aqueous citric acid solution. The resulting itate was filtered and dried to give 2-((1r,4r)methoxycyclohexylamino)(methylthio)- pyrimidinecarboxylic acid (2.76 g, 9.28 mmol, 97 % yield) as a white solid; 1H NMR (DMSO-d6 ,400 MHz) 8 ppm 12.59 (s, 1H), 8.57 (s, 0.38H), 8.50 (s, 0.62H), 7.94 (d, J=7.4 Hz, 0.62H), 7.77 (d, J=7.8 Hz, 0.38H), 3.70 - 3.90 (m, 1H), 3.23 (s, 3H), 3.02 - 3.18 (m, 1H), 2.37 (s, 2H), 2.36 (s., 1H), 1.83 - 2.09 (m, 4H), 1.11 - 1.46 ppm (m, 4H); MS (ESI) m/z 298.2 [M+1]+ ATI—25 14175v1 M. 2-((1r,4r)—4-Meth0xycyclohexylamino)(methylthio)pyrimidine—5- carboxamide. 2-((lr,4r)methoxycyclohexylamino)(methylthio)pyrimidine carboxylic acid (2.66 g, 8.94 mmol) and HATU (5.10 g, 13.42 mmol) were dissolved in DMF (35 mL) and allowed to stir for 5 minutes at room temperature before adding ammonium chloride (2.392 g, 44.7 mmol) and DIEA (7.81 mL, 44.7 mmol). The reaction was allowed to stir at room temperature for 30 min. LCMS shows the desired product mass as dominant and the reaction was partitioned between water and ethyl acetate. The organic layer was washed once with brine before drying over sodium sulfate, ng, and condensing. After drying, ,4r)methoxycyclohexylamino)(methylthio) dinecarboxamide (2.54 g, 8.57 mmol, 96 % yield) was obtained as an white solid; 1H NMR (DMSO-d6 ,400 MHz) 8 ppm 8.41 (s, 1H), 7.63 (d, J=7.0 Hz, 0.76H), 7.45 (d, J=7.0 Hz, 0.24H), 7.08 (br. s., 1H), 3.66 - 3.85 (m, 1H), 3.23 (s, 3H), 3.11 (br. s., 1H), 2.32 (s, 3H), 1.81 - 2.11 (m, 4H), 1.10 - 1.44 ppm (m, 4H); MS (ESI) m/z 283.2 [M+l]+ N. 2-((1r,4r)—4-Methoxycyclohexylamino)—4-(methylsulfinyl)- pyrimidine—5-carb0xamide. 2-((lr,4r)Methoxycyclohexylamino) (methylthio)pyrimidinecarboxamide (3 g, 10.12 mmol) was dissolved in NMP (30 mL).
At 0 CC, mCPBA (2.268 g, 10.12 mmol) was then added portion wise and the on was d while elevating the reaction temperature to room temperature slowly. After 30 min, the reaction mixture was diluted with water (200 mL), stirred for 10 min and the resulting solid was removed by filtration. The filtrate was concentrated in vacuo to afford ,4r)methoxycyclohexylamino)(methylsulf1nyl)pyrimidinecarboxamide as a clear, slightly yellow solution in NMP (~25mL). This crude product was used in the next step without further ation. MS (ESI) m/z 3 13.3 [M+l]+.
O. 4-((3S,5R)—5-Hydr0xytetrahydr0-2H—pyranylamin0)—2-((1r,4S) methoxycyclohexylamin0)pyrimidinecarb0xamide, 4-((3R,SS)hydroxy- tetrahydr0-2H-pyranylamin0)—2-((1r,4R)—4-meth0xycyclohexylamino)pyrimidine— -carb0xamide, 4-((3S,5S)hydroxytetrahydro-ZH-pyranylamin0)—2-((1r,4S) methoxycyclohexylamin0)pyrimidinecarb0xamide, 4-((3R,5R)—5-hydr0xy- tetrahydr0-2H-pyranylamin0)—2-((1r,4R)—4-meth0xycyclohexylamino)pyrimidine— 0xamide. To 2-((lr,4r)methoxycyclohexylamino)(methylsulf1nyl)- pyrimidinecarboxamide (3.1 g, 9.92 mmol) in NMP (25 mL)) was added crude ATI—2514l75vl otetrahydro-2H-pyranol (1.15 g, 9.82 mmol) as a solution in NMP (20 mL). To the suspension was added DIEA (8.57 mL, 49.1 mmol) and the reaction was heated to 100 oC overnight. Most of the NMP was removed by evaporation at 70 0C, the residue was diluted with DCM and purified on a 340G SNAP Biotage column (0-15% methanol in DCM over 2000 mL). The product fractions were combined and concentrated to give 2.8 g (78%) of material that was ~85% pure. The al was dissolved in DCM and repurified on a 340G SNAP Biotage column (2-15% ammonia saturated methanol in dichloromethane). The product fractions were combined and concentrated to afford a mixture of four products (two diastereomers and their corresponding enantiomers; 1.95 g), >99% pure. The crude material was separated by chiral SFC using an AD-H column to afford 4 compounds labeled PEAK 1 to PEAK 4, with PEAK 1 being the first eluting compound and PEAK 4 being the latest eluting compound.
] PEAK 1: 1H NMR (400 MHz, g) 8 ppm 8.41 (s, 1H), 4.38 (br. s., 1H), 3.53 - 3.79 (m, 4H), 3.24 (s, 3H), 2.87 - 2.98 (m, 3H), 1.89 - 2.07 (m, 4H), 1.65 - 1.78 (m, 1H), 1.19 - 1.43 (m, 4H); MS (ESI) m/z 366.3 [M+1]+.
PEAK 2: 1H NMR (400 MHz, DMSO-dg) 5 ppm 8.86 - 9.01 (m, 1H), 8.35 (br. s., 1H), 6.91 - 7.18 (m, 1H), 4.91 - 4.99 (m, 1H), 3.49 - 4.13 (m, 4H), 3.23 (s, 3H), 2.85 - 3.15 (m, 3H), 1.79 - 2.28 (m, 5H), 1.08 - 1.41 (m, 5H), 1.04 (d, J=6.25 Hz, 1H); MS (ESI) m/z 366.3 [M+1]+ .
PEAK 3; 1H NMR (400 MHz, DMSO-dg) 5 ppm 8.86 — 9.02 (m, 1H), 832-8400n,HD,688-7200n,HD,491-5000n,HD,349-4J5(nn5H)322(s 3H), 2.79 - 3.14 (m, 3H), 1.82 (s, 5H), 1.12 (t, J=7.22 Hz, 6H); MS (ESI) m/z 366.3 [M+1]+ .
PEAK 4: 1H NMR (400 MHz, DMSO-dg) 5 ppm 8.99 - 9.24 (m, 1H), 8.36 wrs,1H)679-720&m2H)472-494@m1HL417-446fim1H)343-3770n 4H), 3.23 (s, 4H), 3.03 - 3.13 (m, 1H), 2.73 (s, 1H), 1.98 (br. s., 6H), 1.03 - 1.35 (m, 5H); MS (ESI) m/z 366.3 .
ATI—25 14175v1 Example 8: 4-(3-Hydr0xymethylbutylamin0)—2-((1r,4r)—4- methoxycyclohexylamin0)pyrimidinecarboxamide N \ NH2 ., )L/ ’N N NH H >OH A. 2-((1r,4r)—4-Methoxycyclohexylamin0)(methylsulfinyl)- pyrimidine—5-carb0xamide. To a d colorless solution of 2-((1r,4r) methoxycyclohexylamino)(methy1thio)pyrimidinecarboxamide (0.200 g, 0.675 mmol; synthesis described herein) in NMP (2 mL) was added mCPBA (0.151 g, 0.675 mmol) at 0 oC portion wise. The reaction mixture was then stirred at room temperature for 2 h until completion of the reaction as indicated by LCMS. The reaction mixture was diluted with water (20 mL) and filtered. The filtrate was concentrated to afford 2-((1r,4r)methoxycyclohexylamino)(methy1su1finy1)pyrimidine carboxamide (0.211 g, 100% yield) as a white sticky solid which was used in the next step without further purification. MS (ESI) m/z 313.1 .
B. 4-(3-Hydr0xymethylbutylamino)—2-((1r,4r)—4-meth0xy-cyclo- hexylamin0)pyrimidine—5-carb0xamide. To a solution of 2-((1r,4r)methoxy- cyclohexylamino)(methylsulfiny1)pyrimidinecarboxamide (0.211 g, 0.675 mmol) and DIEA (0.236 mL, 1.351 mmol) in NMP (2 mL) was added 4-aminomethy1butan 01 (0.105 g, 1.013 mmol) at room ature. The reaction e was stirred for 2 h at 70 oC. Upon completion of the reaction as indicated by LCMS and TLC the reaction mixture was concentrated and purified by silica gel chromatography (0% - 15% ammonia saturated ol in DCM). The product fractions were combined and concentrated to afford 4-(3-hydroxymethy1buty1amino)((1r,4r)methoxycyclohexylamino)- dinecarboxamide (0.143 g, 60.2 % yield, 95.8% pure) as a pale yellow solid. 1H NMR (400 MHz, DMSO-dg) 8 ppm 8.81 - 9.03 (m, 1H), 8.32 (s, 1H), 6.75 - 7.06 (m, 2H), 4.28 (s, 1H), 3.63 - 3.77 (m, 1H), 3.39 - 3.52 (m, 2H), 3.22 (s, 3H), 3.02 - 3.11 (m, 1H), 1.81 - 2.06 (m, 4H), 1.57 - 1.67 (m, 2H), 1.21 - 1.34 (m, 2H), 1.16 (br. s., 1H), 1.13 (s, 8H). MS (ESI) m/z 352.4 [M+1]+.
ATI—25 14175V1 Example 9: 4-((1R,2S)(Hydroxymethyl)cyclopentylamino)—2-((1r,4R)—4- ycyclohexylamino)pyrimidine—5-carb0xamide, 4-((1 S,2R)—2- (Hydroxymethyl)cyclopentylamino)—2-((1r,4S) methoxycyclohexylamin0)pyrimidinecarboxamide o 0 ”MA“ NH 'I’MAN/ 1ng m 0% A. Ethyl 4-(cis—2-(hydroxymethyl)cyclopentylamino)—2-(methylthi0)- pyrimidine-S-carboxylate. To a stirred solution of ethyl 4-chloro(methylthio)- pyrimidinecarboxylate (5.00 g, 21.49 mmol) and cis-(2-aminocyclopentyl)-methanol (2.60 g, 22.56 mmol) in l (85 mL) was added DIEA (5.61 mL, 32.2 mmol) and heated to 60 0C for 2.5 h. Upon completion of reaction, as indicated by LCMS and TLC, the reaction was removed from heat and concentrated, then purified by silica gel chromatography (0-80% ethyl acetate/hexane). The desired product fractions were combined and concentrated to afford ethyl 4-(cis(hydroxymethyl)cyclopentylamino) (methylthio)pyrimidinecarboxylate (6.4 g, 96 % yield) as a white solid. MS (ESI) m/z 312.4 [M+1]+.
B. 4-(cis—2-(Hydr0xymethyl)cyclopentylamin0)(methylthi0)— pyrimidine-S-carboxylic acid. To a stirred solution of the ethyl 4-(cis(hydroxyl- methyl)cyclopentylamino)(methylthio)pyrimidinecarboxylate (6.4 g, 20.55 mmol) in ethanol (100 mL) was added aqueous sodium hydroxide solution (1 M, 51.4 mL, 51.4 mmol) at room temperature. The reaction mixture was stirred at room temperature for 2 h. Upon tion of reaction as indicated by LCMS and TLC, the reaction mixture was concentrated, diluted with water (100 mL), then lized while stirring with aqueous citric acid solution (2 M, 51.4 mL, 103 mmol). The resulting precipitate was ed, washed with water (2 x 50 mL) and dried to afford 4-(cis(hydroxymethyl)- entylamino)(methylthio)pyrimidinecarboxylic acid (5.6 g, 96 % yield) as a white solid. 1H NMR (400 MHz, DMSO-dg) 8 ppm 8 13.17 (br. s., 1H), 8.71 (d, J: 7.42 Hz, 1H), 8.50 (s, 1H), 4.49 - 4.60 (m, 2H), 3.40 - 3.46 (m, 1H), 3.32 - 3.39 (m, ATI—25 14175V1 2H), 2.47 (s, 3H), 2.22 (sxt, J: 6.87 Hz, 1H), 1.95 - 2.04 (m, 1H), 1.78 (qd, J= 4.49, 8.00 Hz, 1H), 1.66 - 1.72 (m, 1H), 1.49 - 1.61 (m, 2H). MS (ESI) m/z 284.3 [M+1]+.
C. (Hydr0xymethyl)cyclopentylamino)—2-(methylthi0)- pyrimidine—5-carb0xamide. A white suspension of the 4-(cis(hydroxymethyl)- cyclopentylamino)(methylthio)pyrimidinecarboxylic acid (5.6 g, 19.76 mmol) and HATU (11.27 g, 29.60 mmol) were dissolved in DMF (79 mL) and stirred for 5 min at room temperature before adding ammonium chloride (5.29 g, 99 mmol) and DIEA (17.26 mL, 99 mmol). The reaction mixture was stirred at room temperature for 1 h and then diluted with water (200 mL) and extracted with ethyl acetate (2 x 200 mL). The combined organic layers were washed with brine (100 mL), dried over sodium e, filtered, and concentrated. The residual DMF was removed by suspending the white suspension in 200 mL water and 200 mL hexanes. The biphasic mixture was stirred vigorously for 30 min, d and washed with hexanes to afford 4-(cis(hydroxyl- methyl)cyclopentylamino)(methylthio)pyrimidinecarboxamide (4.50 g, 81 % yield) as a white solid. MS (ESI) m/z 283.4 [M+1]+.
D. 4-(cis(Hydr0xymethyl)cyclopentylamin0)(methylsulf0nyl)— pyrimidine—5-carb0xamide. To a d suspension of the 4-(cis(hydroxymethyl)- cyclopentylamino)(methylthio)pyrimidinecarboxamide (3.5 g, 12.40 mmol) in DCM (62 mL) and acetone (62 mL) was added mCPBA (5.56 g, 24.79 mmol) and stirred at room ature for 1.5 h. The crude reaction mixture was quenched by addition of 75 mL of 10% aqueous sodium thiosulfate solution, stirred for 5 min, before ating most of the volatile solvents. The material was partitioned between ethyl acetate (200 mL) and water (200 mL), and the combined organic layers were washed with saturated sodium bicarbonate solution and then brine. The combined aqueous layers were then washed with ethyl acetate (3x), the c layers were combined, dried over sodium sulfate, filtered, and condensed. After drying under high vacuum, 4-(cis(hydroxymethyl)cyclopentyl- amino)(methylsulfonyl)pyrimidinecarboxamide (1.2 g, 3.82 mmol, 30.8 % yield) was ed as a white solid. MS (ESI) m/z 315.2 [M+1]+.
E. 4-((1R,2S)(Hydr0xymethyl)cyclopentylamin0)((1r,4R)—4- methoxycyclohexylamin0)pyrimidine—5-carboxamide, 4-((1S,2R)—2-(Hydr0xymethyl)— cyclopentylamino)—2-((1r,4S)methoxycyclohexylamin0)pyrimidine—5-carboxamide.
ATI—25 14175v1 To a stirred suspension of 4-(cis(hydroxymethyl)cyclopentylamino)(methyl- sulfonyl)pyrimidinecarboxamide (1.2 g, 3.82 mmol) and (1r,4r)methoxycyclo- mine hydrochloride (1.90 g, 11.46 mmol) in dioxane (40 mL) was added DIEA (4 mL, 22.92 mmol) and the resulting solution was stirred at 100 0C for 48 h. The crude reaction mixture was trated, and then purified by silica gel chromatography (0% - 15% ammonia saturated methanol in DCM). The product fractions were impure and the d product was precipitated and filtered from the combined fractions using DCM, methanol and water to afford the title compound as a mixture of two enantiomers (0.420 g, .3 % yield) as a white solid. The crude material was separated by chiral SFC using an AD-H column to afford 2 compounds labeled PEAK 1 and PEAK 2, with PEAK 1 being the first eluting nd and PEAK 2 being the second eluting compound.
PEAK 1. 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.04 - 9.21 (m, 1H), 8.31 - 8.39 (m, 1H), 6.87 - 7.12 (m, 2H), 4.33 - 4.46 (m, 2H), 3.57 - 3.80 (m, 1H), 3.35 - 3.44 (m, 1H), 3.25 - 3.31 (m, 1H), 3.23 (s, 3H), 3.08 (t, .1: 8.20 Hz, 1H), 2.09 - 2.23 (m, 1H), 2.00 (d,J= 11.71 Hz, 2H), 1.80 - 1.94 (m, 3H), 1.63 - 1.78 (m, 2H), 1.50 - 1.61 (m, 2H), 1.44 (br. s., 1H), 1.09 - 1.33 (m, 5H); MS (ESI) m/z 364.5 [M+1]+.
PEAK 2. 1H NMR (400 MHz, DMSO-dg) 5 ppm 9.03 - 9.18 (m, 1H), 8.34 (br. s., 1H), 6.87 - 7.11 (m, 2H), 4.32 - 4.48 (m, 2H), 3.58 - 3.77 (m, 1H), 3.35 - 3.45 (m, 1H), 3.28 (dd, .1: 7.03, 10.15 Hz, 1H), 3.22 (s, 3H), 3.03 - 3.13 (m, 1H), 2.09 - 2.25 (m, 1H), 2.00 (d, .1: 8.59 Hz, 2H), 1.81 - 1.95 (m, 3H), 1.63 - 1.77 (m, 2H), 1.51 - 1.60 (m, 2H), 1.37 - 1.47 (m, 1H), 1.09 - 1.33 (m, 5H); MS (ESI) m/z 364.5 [M+1]+.
ATI—25 14175V1 2012/034349 Example 10: 4-((1R,3R)—3-Hydr0xymethylcyclohexylamin0)((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarboxamide, 4-((1R,3S)Hydr0xy cyclohexylamin0)((1r,4R)—4-meth0xycyclohexylamin0)pyrimidine carboxamide o o ”U NYNN "’MAN/ NH ”U YNN”Mk“ NH ) (5% ’OH ’/ A. (1 S,3R)—3-(Dibenzylamin0)cyclohexanol. To a suspension of )- 3-aminocyclohexanol (5 g, 43.4 mmol; prepared as described in Tetrahedron: Asymmetry : 2051—2056 (2004)) and sodium bicarbonate (12.03 g,143 mmol) in ethanol (100 mL) was added omethyl)benzene (15.01 mL, 130 mmol) at room temperature. The reaction mixture was heated at 75 CC overnight. Upon completion of reaction as indicated by LCMS and TLC the reaction mixture was filtered and the filtrate was concentrated.
The residue was then dissolved in DCM (250 mL) and washed with aqueous sodium ide solution (1 N, 2 x 100 mL) and brine (1x 100 mL). The combined organic layers were dried over anhydrous ium sulfate, concentrated, and purified by silica gel chromatography (0% - 80% ethyl acetate in hexanes) to yield (lS,3R) (dibenzylamino)cyclohexanol (11.70g, 91%) as a thick yellow oil. 1H NMR (400 MHz, DMSO-dg) 5 ppm 7.26 - 7.37 (m, 8H), 7.17 - 7.22 (m, 2H), 4.56 (d, .1: 4.30 Hz, 1H), 3.57 (s, 4H), 3.16 - 3.26 (m, 1H), 2.41 (tt, J: 3.17, 11.86 Hz, 1H), 1.99 - 2.06 (m, 1H), 1.72 (d, .1: 8.20 Hz, 2H), 1.63 - 1.69 (m, 1H), 1.18 - 1.28 (m, 2H), 0.98 (t, 2H); MS (ESI) m/z 296.4 [M+1]+.
B. (R)—3-(Dibenzylamin0)cyclohexanone. Oxalyl chloride (2.70 mL, .9 mmol) was ved in dry DCM (150 mL) and cooled to -78 oC. DMSO (4.78 mL, 67.3 mmol) in dry DCM (20 mL) was added dropwise to the reaction mixture and the reaction was stirred for 15 min at -78 CC. Next, (1S,3R)—3-(dibenzylamino)cyclohexanol (8.287 g, 28.1 mmol) in dry DCM (100 mL) was added drop wise using an addition fiJnnel and the reaction mixture was stirred at -78 CC for 15 min. TEA (19.55 mL, 140 mmol) was then added and the reaction was stirred at -78 CC for 1 h. The dry-ice bath was _ 88 _ ATI—25 14175v1 removed and the reaction e was allowed to warm to room temperature and stirred overnight. The reaction mixture was washed with brine (100 mL), the organic layer was separated, dried over magnesium e and concentrated. Upon slow addition of diethyl ether to the residue the impurities precipitated, and were separated by filtration. Addition of diethyl ether to the filtrate yielded (R)—3-(dibenzylamino)cyclohexanone (6.05 g, 74%) as a white solid which was ted by filtration and dried. 1H NMR (400 MHz, DMSO-dg) 5 ppm 7.26 - 7.39 (m, 8H), 7.17 - 7.25 (m, 2H), 3.56 - 3.70 (m, 4H), 2.71 - 2.81 (m, 1H), 2.60 - 2.69 (m, 1H), 2.25 - 2.42 (m, 2H), 2.04 - 2.11 (m, 1H), 1.88 - 1.99 (m, 2H), 1.69 - 1.82 (m, 1H), 1.18 - 1.33 (m, 1H); MS (ESI) m/z 294.4 [M+1]+.
C. (3R)—3-(Dibenzylamin0)—l-methylcyclohexanol. To a clear ess solution of (R)(dibenzylamino)cyclohexanone (5.697g, 19.42 mmol) in dry diethyl ether (300 mL) was added a solution of 3 M methylmagnesium bromide (8.09 mL, 24.27 mmol) dropwise slowly at 0 CC. The reaction mixture was stirred at 0 CC for min and the ice-bath was then removed. The reaction e was stirred at room temperature for 2 h. Upon completion of the reaction, as indicated by LCMS and TLC, the cloudy solution was slowly poured into a saturated aqueous solution of ammonium chloride (250 mL). The ether layer was separated, and the aqueous layer was re-extracted with ether (250 mL). The combined ether layers were dried over magnesium sulfate trated and purified by silica gel chromatography (0% - 80% ethyl acetate in hexanes) to yield two diastereomeric isomers of (3R)(dibenzylamino) methylcyclohexanol (isomer 1 and isomer 2). Isomer 1 (2.704g, 45%) was obtained as a white solid and isomer 2 (1 .866g, 31%) as a colorless oil, which still contained a small amount of impurities. Isomer 2 was used without r ation in the next step.
Isomer 1: 1H NMR (400 MHz, DMSO-dg) 8 ppm 7.25 - 7.36 (m, 8H), 7.15 - 7.21 (m, 2H), 3.85 (s, 1H), 3.55 (s, 4H), 2.86 (tt, .1: 3.37, 12.06 Hz, 1H), 1.70 - 1.81 (m, 2H), 1.38 - 1.49 (m, 3H), 1.32 (t,J= 12.30 Hz, 1H), 1.15 - 1.27 (m, 2H), 1.12 (s, 3H); MS (ESI) m/z 310.4 [M+1]+.
Isomer 2: MS (ESI) m/z 310.4 [M+1]+.
] D. (3R)—3-Amin0-l-methylcyclohexanol. A solution of isomer 1 (2.704 g, 8.74 mmol) in ethanol (50 mL) was treated with palladium hydroxide on carbon and stirred under a balloon filled with en gas overnight. Upon completion of the ATI—25 14175v1 2012/034349 reaction as indicated by LCMS and TLC the reaction mixture was filtered through a pad of celite and the filtrate was concentrated to yield one diastereomeric isomer of (3R)—3- aminomethylcyclohexanol (isomer 3, 0.856g, 76%) as a thick yellow oil. Isomer 3: 1H NMR (400 MHz, DMSO-d6) 8 ppm 3.91 (br. s., 1H), 2.73 - 2.84 (m, 1H), 1.61 - 1.72 (m, 2H), 1.54 (tt, J: 3.66,13.13 Hz, 1H), 1.34 - 1.47 (m, 2H), 1.01 - 1.13 (m, 4H), 0.86 - 0.93 (m, 1H), 0.74 - 0.85 (m, 1H); MS (ESI) m/z 130.2 [M+1]] The same procedure as described above using isomer 2 (1.823 g, .89 mmol) in ethanol (25 mL) was utilized to access another diastereomeric isomer of (3R)aminomethylcyclohexanol (isomer 4, 0.162 g, 21%) containing a small amount of impurities. Isomer 4 was used without r purification in the next step.
E. 2-((1r,4r)—4-Meth0xycyclohexylamin0)—4-(methylsulfinyl)— dine—5-carb0xamide. To a stirred colorless solution of 2-((1r,4r) methoxycyclohexylamino)(methylthio)pyrimidinecarboxamide (0.250 g, 0.843 mmol; sis described ) in NMP (2 mL) was added mCPBA (0.189 g, 0.843 mmol) at 0 °C portion wise. The reaction mixture was then stirred at room temperature for 2 h until completion of the reaction as indicated by LCMS. The reaction mixture was diluted with water (20 mL) and filtered. The filtrate was concentrated to afford 2-((1r,4r)methoxycyclohexylamino)(methylsulfinyl)pyrimidine carboxamide (0.263 g, 100% yield) as a white sticky solid which was used in the next step without further purification. MS (ESI) m/z 313.1 [M+1] +.
] F. 4-((1R,3R)Hydr0xymethylcyclohexylamino)—2—((1r,4R)—4- methoxycyclohexylamin0)pyrimidine—5-carboxamide, 4-((1R,3S)Hydr0xy methylcyclohexylamin0)((1r,4R)—4-meth0xycyclohexylamin0)pyrimidine carboxamide. To a solution of 2-((1r,4r)methoxycyclohexylamino) (methylsulfinyl)pyrimidinecarboxamide (0.263 g, 0.842 mmol) and DIEA (0.294 mL, 1.684 mmol) in NMP (3 mL) was added isomer 3 (0.163 g, 1.263 mmol) at room temperature. The reaction mixture was stirred for 2 h at 70 CC. Upon completion of the reaction as indicated by LCMS and TLC the reaction mixture was concentrated and purified by silica gel chromatography (0% - 20% ammonia saturated methanol in DCM).
The product ons were combined and concentrated to afford one diastereomeric isomer of )hydroxymethylcyclohexylamino)((1r,4R)methoxycyclo- ATI—25 14175V1 hexylamino)pyrimidinecarboxamide r 5, 0.265 g, 83 % yield) as a white solid.
Isomer 5: 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.79 (d, .1: 7.42 Hz, 1H), 8.32 (s, 1H), 6.76 - 7.05 (m, 2H), 4.12 - 4.32 (m, 1H), 4.07 (s, 1H), 3.57 - 3.82 (m, 1H), 3.23 (s, 3H), 3.04 - 3.16 (m, 1H), 1.81 - 2.05 (m, 6H), 1.60 - 1.74 (m, 2H), 1.42 - 1.54 (m, 2H), 1.15 - 1.35 (m, 6H), 1.12 (s, 4H); MS (ESI) m/z 378.5 [M+1]+.
The same procedure as described above using isomer 4 (0.163 g, 1.263 mmol), 2-((1r,4r)methoxycyclohexylamino)(methylsulfinyl)pyrimidine carboxamide (0.263 g, 0.842 mmol) and DIEA (0.294 mL, 1.684 mmol) in NMP (3 mL) was utilized to access r diastereomeric isomer of 4-((1R)-3 -hydroxy methylcyclohexylamino)((1r,4R)methoxycyclohexylamino)pyrimidine carboxamide (isomer 6, 0.043 g, 14 % yield) as a white solid. Isomer 6: 1H NMR (400 MHz, DMSO-dg) 5 ppm 8.99 (d, .1: 4.69 Hz, 1H), 8.32 (s, 1H), 6.69 - 7.08 (m, 2H), 4.45 (s, 1H), 3.95 - 4.13 (m, 1H), 3.59 - 3.71 (m, 1H), 3.22 (s, 3H), 3.04 - 3.16 (m, 1H), 1.86 - 2.05 (m, 5H), 1.62 - 1.83 (m, 2H), 1.43 - 1.57 (m, 1H), 1.23 - 1.40 (m, 5H), 1.09 - 1.21 (m, 7H); MS (ESI) m/z 378.5 [M+1]+.
The stereochemistry of the individual isomers could be determined after repeating the sis using (1 S,3R)aminomethylcyclohexanol (prepared as described herein), which provided isomer 6, namely 4-((1R,3S)hydroxy methylcyclohexylamino)((1r,4R)methoxycyclohexylamino)pyrimidine carboxamide. Therefore, isomer 5 is 4-((1R,3R)hydroxymethylcyclohexylamino) ((1r,4R)methoxycyclohexylamino)pyrimidinecarboxamide.
Example 11: 2-((1r,4R)—4-Acetamid0cyclohexylamino)—4-((1R,3S) ycyclohexylamino)pyrimidine—5-carboxamide O 11*“ "’N N/ NH ‘\gOH A. utyl (1r,4r)—4—acetamid0cyclohexylcarbamate. To a stirring solution of tert-butyl (1r,4r)aminocyclohexylcarbamate (1.0 g, 4.67 mmol) in ethyl acetate (30 mL) was added acetic anhydride (0.485 mL, 5.13 mmol) in one portion. The _ 91 _ ATI—25 14175v1 resulting mixture was allowed to stir overnight at room temperature. Additional acetic anhydride (0.162 mL, 1.71 mmol) was added to the mixture and the mixture was stirred for an additional hour at room temperature. The reaction mixture was then diluted with an additional 50 mL of ethyl acetate and was washed with 50 mL each of a saturated aqueous sodium bicarbonate on and ted aqueous sodium chloride solution. The aqueous layers were combined and extracted with 50 mL ethyl acetate. The ed ethyl acetate layers were dried over anhydrous magnesium sulfate, filtered, and concentrated to a afford tert-butyl (1r,4r)acetamidocyclohexylcarbamate (1.04 g, 4.06 mmol, 87 % yield) as a solid that was used without further purification. 1H NMR (400 MHz, DMSO-d6) 5 ppm 766-7740n,HD,667-677@m1H)340(¢J=742HL1H)311-322@m1HL 1.65 - 1.82 (m, 7H), 1.37 (s, 9H), 1.16 (d, J=8.20 Hz, 4H). MS (ESI) m/z 201.2 [M-tBu]+.
B. N-((1r,4r)—4-Amin0cyclohexyl)acetamide 2,2,2-triflu0roacetate. To a stirring solution of tert—butyl (1r,4r)acetamidocyclohexylcarbamate (1.04 g, 4.06 mmol) in DCM (25 mL) was added TFA (25 mL) in one n. The resulting mixture was allowed to stir at room temperature for 90 min. The reaction mixture was concentrated to dryness. The resulting oil was triturated with diethyl ether to afford solids that were filtered, rinsed with additional diethyl ether and then dried in vacuo to afford N—((1r,4r) yclohexyl)acetamide 2,2,2-trifluoroacetate (1.07 g, 3.96 mmol, 98 % , which was used without fiarther purification. 1H NMR (400 MHz, 6) 5 ppm 7.82 (br. s., 3H), 3.42 (ddd, J=15.42, 7.61, 3.90 Hz, 1H), 2.89 - 3.04 (m, 1H), 1.87 - 1.95 (m, 2H), 1.78 - 1.85 (m, 2H), 1.77 (s, 3H), 1.29 - 1.42 (m, 2H), 1.11 - 1.25 (m, 2H). MS (ESI) m/z 157.0 [M+1]+.
C. ,4R)—4-acetamidocyclohexylamin0)((1R,3 S) ycyclohexylamino)pyrimidine—5-carb0xamide. To a stirring solution of 4-((1R,3 S)—3-hydroxycyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (190 mg, 0.604 mmol; synthesis described herein) and N-((1r,4r)aminocyclohexyl)- acetamide 2,2,2-trifluoroacetate (245 mg, 0.907 mmol) in DMSO (3 mL) was added DIEA (0.317 mL, 1.813 mmol). The resulting mixture was stirred at 100 CC overnight.
The crude reaction mixture was concentrated and then purified by silica gel cmpmmpgqmym45%nmmmmHnDCM)Themomwfihwmmswmeammmmhmd concentrated to afford ,4R)—4-acetamidocyclohexylamino)((1R,3S)hydroxy- ATI—25 14175V1 cyclohexylamino)pyrimidinecarboxamide (102 mg, 0.261 mmol, 43.2 % yield) as a solid. 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.29 (s, 1H), 7.69 (d, J=7.81 Hz, 1H), 4.60 (br. s., 1H), 3.36 - 3.62 (m, 2H), 2.00 - 2.17 (m, 1H), 1.60 - 1.97 (m, 10H), 0.93 - 1.35 (m, 10H). MS (ESI) m/z 391.2 [M+1]+.
Example 12A: 4-((1R,3S)Hydr0xycycloheptylamino)—2-((1r,4R)—4-meth0xycyclohexylamino )—pyrimidine—5-carboxamide, 4-((1R,3R)Hydr0xycycloheptylamin0 )((1r,4R)—4-methoxycyclohexylamin0)-pyrimidine—5-carb0xamide, 4-((1S,3R)—3-Hydr0xycycloheptylamino)—2-((1r,4S)methoxycyclohexylamino)— pyrimidine-S-carboxamide, 4-((1S,3S)Hydr0xycycloheptylamin0)((1r,4S) methoxycyclohexylamin0)-pyrimidine—5-carb0xamide I 0 I 0 ., / ., / ’M N NH ’M N NH | O | 0 0 o ’N N/ NH "INJ\N/I NH H : H ? A. tert-Butyl ycloheptylcarbamate. To a ng mixture of cyclohept—2 enone (26.96 g, 245.0 mmol) and tert-butyl ate (28.7 g, 245 mmol) in DCM (245 mL) was added bismuth nitrate pentahydrate (22.79 g, 47.0 mmol). The resulting mixture was stirred at room temperature overnight. The on was diluted reaction with ethyl acetate (500 mL) and water (300 mL) and the biphasic mixture was filtered through a bed of Celite. The bed of Celite was washed well with ethyl acetate and water, and layers of the filtrate separated. The organic layer was concentrated to an oil that was purified by silica gel chromatography (0-40% ethyl acetate in hexanes). Product containing fractions were concentrated to afford the title compound (31.09 g, 137 mmol, 55.9 % . 1H NMR (400 MHz, DMSO-d6) 8 ppm 6.92 (d, J=7.03 Hz, 1 H), 3.60 (d, J=8.59 Hz, 1 H), 2.55 - 2.69 (m, 1 H), 2.26 - 2.45 (m, 2 H), 1.69 - 1.93 ATI—25 14175V1 W0 2012/145569 (m, 3 H), 1.44 - 1.61 (m, 2 H), 1.37 (d, J=5.08 Hz, 11 H). MS (ESI) m/z 228.5 [M+H]+.
B. tert-Butyl (1R,3S)—3-hydr0xycycloheptylcarbamate, tert-butyl (1R,3R) hydroxycycloheptylcarbamate, utyl (1 S,3S)—3-hydr0xycycloheptylcarbamate, tert- butyl (1 S,3R)—3-hydr0xycycloheptylcarbamate. To a solution of tert-butyl 3-oxocycloheptyl- carbamate (31.09 g, 137 mmol) in methanol (454 mL) was added sodium borohydride (15.52 g, 410 mmol) portion-wise over ~ 10 minutes with mixing. The resulting solution was stirred for 2 h at room temperature and then diluted with water (200 mL). The methanol was removed in- vacuo and the ing aqueous e was diluted fiarther with 500 mL ethyl acetate and 100 mL saturated aqueous sodium bicarbonate. The layers were ted and the aqueous layer back-extracted with 2 x 1000 mL ethyl acetate. The combined organic layers were dried over anhydrous magnesium e, filtered and concentrated to an oil that was purified by silica gel chromatography (0-50% ethyl acetate in hexanes). Product containing fractions were concentrated to afford tert-butyl 3-hydroxycycloheptylcarbamate (21.1 g, 92 mmol, 67.3 % . MS (ESI) m/z 230.3 . The above two reactions were repeated starting with 9.52 g cycloheptenone and yielding 9.27 g of utyl oxycycloheptylcarbamate. The two batches of tert-butyl 3-hydroxycycloheptylcarbamate were then combined. 29.2 g of the combined material was separated into the 4 constituent stereoisomers (two diastereomers and their corresponding enantiomers) by preparative chiral SFC utilizing multiple injections over a series of 3 separate s. First column: ChiralPak IC-H, 250><30 mm I.D., isocratic 15% ethanol in CO2, 38 oC. Second column: ChiralCel OJ-H, 250><30 mm I.D., tic 10% isopropanol in CO2, 38 CC. Third column: ChiralPak AD-H, 250><50 mm I.D., isocratic 20% isopropanol in CO2, 38 oC. The separated isomers were characterized on an analytical scale Phenomenex Lux e-2 column, 250><4.6 mm I.D., isocratic 15% ethanol in CO2 (10 min run time) and labeled as Intermediate 1 to Intermediate 4.
Intermediate 1: 5.4 g (23.55 mmol, 18.5 % yield from SFC purification).
Retention time: 4.065 minutes.
Intermediate 2: 5.5 g (23.98 mmol, 18.8 % yield from SFC purification).
Retention time: 3.019 minutes.
Intermediate 3: 7.2 g (31.34 mmol, 24.6 % yield from SFC purification).
Retention time: 3.675 minutes.
Intermediate 4: 4.7 g (20.50 mmol, 16.1 % yield from SFC purification). _ 94 _ ATI—2514l75vl Retention time: 3.263 s.
C. (1S,3R)—3-Amin0cycloheptanol, (1R,3R)aminocycloheptanol, (IS,3S) aminocycloheptanol, (1R,3S)amin0cycloheptanol. 75 mg (0.327 mmol) of each of the materials corresponding to Intermediate 1 through Intermediate 4 from step B was separately dissolved in DCM (11.25 mL) and TFA (3.75 mL). Each reaction was allowed to stir overnight at t temperature. The solvent of each reaction was removed in vacuo and each was then dissolved in a mixture of 4N hloric acid in dioxane (5 mL) and methanol (5 mL). Each on was mixed overnight at ambient ature. Each of the 4 on mixtures (each containing an individual 3-aminocycloheptanol isomer) was concentrated under reduce pressure and used without further purification. MS (ESI) m/z 130.2 [M+H]+ for each. The nomenclature of Intermediate 1 to Intermediate 4 was maintained for tracking purposes.
D. 4-((1R,3S)Hydr0xycycloheptylamin0)((1r,4R)—4-meth0xycyclohexyl- amin0)pyrimidinecarboxamide, 4-((1R,3R)—3-hydr0xycycloheptylamino)—2-((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarb0xamide, ,3R)—3-hydr0xycycloheptylamino )—2-((1r,4R)—4-methoxycyclohexylamin0)pyrimidine—5-carb0xamide, 4-((1 S,3S) hydroxycycloheptylamin0)((1r,4R)—4-methoxycyclohexylamino)pyrimidine—5- carboxamide. To each of the concentrates (Intermediate 1 to Intermediate 4) from step C was added 2-((1r,4r)methoxycyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (53.7 mg, .1635 mmol), NMP (2 mL), and DIEA (0.286 mL, 1.635 mmol). The resulting 4 mixtures were stirred at 80 OC overnight. The reaction solutions were condensed separately under reduced pressure and the products purified by reverse-phase preparative HPLC (5-80% acetonitrile + 0.1% trifluoroacetic acid in water + 0.1% trifluoroacetic acid, over 30 min).
Fractions containing product were concentrated under reduced pressure. The resulting residues were tely redissolved in a methanol (5 mL), passed over an Varian StratoSpheres HCO3 resin SPE tube for TFA removal (0.9 mmol bicarbonate equiv), and then concentrated under reduced re to afford the title compounds as Peak 1 to Peak 4. The te isomers (Peak 1 to Peak 4) were characterized on an analytical scale ChiralPak AD-H column, 250 x 4.6 mm ID, isocratic 40% methanol + 0.1% diethylamine in C02 (10 minute run time).
PEAK 1: 10.3 mg (0.027 mmol); 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.02 (d, J=6.64 Hz, 1 H), 8.33 (s, 1 H), 7.04 (d, J=7.42 Hz, 1 H), 4.44 (br. s., 1 H), 4.19 (br. s., 1 H), 3.56 - 3.90 (m, 2 H), 3.23 (s, 3 H), 3.09 (br. s., 1 H), 1.04 - 2.11 (m, 18 H). MS (ESI) m/z 378.3 ATI—25 14175v1 [M+H]+. Retention time: 2.534 minutes.
] PEAK 2: 30.0 mg (0.079 mmol); 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.95 (d, J=6.25 Hz, 1 H), 8.33 (s, 1 H), 7.06 (d, J=7.03 Hz, 1 H), 4.46 (d, J=3.12 Hz, 1 H), 3.86 - 4.18 (m, 1 H), 3.48 - 3.81 (m, 2 H), 3.23 (s, 3 H), 3.09 (br. s., 1 H), 1.06 - 2.16 (m, 18 H). MS (ESI) m/z 378.5 [M+H]+. Retention time: 3.848 minutes.
PEAK 3: 22.0 mg (0.058 mmol); 1H NMR (400 MHz, DMSO-dg) 8 ppm 9.02 (d, J=7.03 Hz, 1 H), 8.33 (s, 1 H), 7.04 (d, J=7.03 Hz, 1 H), 4.45 (d, J=3.12 Hz, 1 H), 4.20 (br. s., 1 H), 3.56 - 3.91 (m, 2 H), 3.23 (s, 3 H), 3.09 (br. s., 1 H), 1.06 - 2.12 (m, 18 H). MS (ESI) m/z 378.5 [M+H]+. Retention time: 4.557 minutes.
] PEAK 4: 22.5 mg (0.060 mmol); 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.95 (d, J=6.64 Hz, 1 H), 8.33 (s, 1 H), 6.47 - 7.21 (m, 1 H), 4.46 (d, J=3.12 Hz, 1 H), 3.94 (br. s., 1 H), 3.73 (br. s., 2 H), 3.23 (s, 3 H), 3.09 (br. s., 1 H), 1.04 - 2.18 (m, 18 H). MS (ESI) m/z 378.3 [M+H]+. Retention time: 2.607 minutes.
From the chemical shift of the proton at ~ 4.2 ppm in Peak 1 and Peak 3 versus that of ~ 3.94 ppm in Peaks 2 and 4 and also of the proton at ~ 9.02 ppm in Peak 1 and Peak 3 versus that of ~ 8.95 ppm in Peaks 2 and 4 in the 1H NMR data given above, the following assignment was made: Peak 1 and Peak 3 are omers. Peak 2 and Peak 4 are enantiomers.
Alternatively, a chiral synthetic route was used, described below.
Example 12B: 4-((1R,3S)—3-hydr0xycycloheptylamino)—2-((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarboxamide A. Cyclohept-Z-enol. To a solution of cycloheptenone (10 g, 91 mmol) and cerium(III) chloride heptahydrate (33.8 g, 91 mmol) in methanol (45.5 mL) was added sodium borohydride (3.43 g, 91 mmol) portion-wise over 10 minutes period with cooling in a water bath.
The reaction mixture was then d for 2.5 hr at room ature. The reaction was quenched by addition of water (45 mL) and then was extracted with pentane (4 x 100 mL). The combined organic ts were dried over anhydrous magnesium sulfate, filtered, and concentrated under ATI—25 14175v1 reduced pressure to afford the title compound (8.30 g, 74.0 mmol, 82 % yield) as an oil that was used without filrther purification. 1H NMR (400 MHz, DMSO-d6) 8 ppm 5.49 - 5.71 (m, 2 H), 4.66 (d, J=4.29 Hz, 1 H), 4.05 - 4.20 (m, 1 H), 1.07 - 2.14 (m, 8 H).
B. Cyclohept-Z-enyl methyl carbonate. To a solution of cycloheptenol (8.3 g, 74.0 mmol) in DCM (227 ml) and pyridine (35.9 ml, 444 mmol), was added methyl formate (14.24 ml, 185 mmol) with cooling in an ice bath at such a rate that T < 10 CC.
Once addition was complete, the reaction was allowed to mix and warm to ambient temperature overnight. The reaction mixture was washed with 2 x 150 mL 1N aqueous hydrochloric acid.
The combined washes were back-extracted with 100 mL DCM. The combined DCM layers were then washed with 150 mL of an aqueous saturated sodium bicarbonate solution, dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure to afford the title compound (11.95 g, 70.2 mmol, 95 % yield) as an oil that was used without further purification. 1H NMR (400 MHz, DMSO-d6) 8 ppm 5.83 (dddd, J=11.81, 7.03, 5.17, 2.15 Hz, 1 H), 5.65 (d, J=11.71 Hz, 1 H), 5.17 (d, J=6.64 Hz, 1 H), 4.71 (d, J=4.30 Hz, 1 H), 3.69 (s, 2 H), 1.99 - 2.23 (m, 2 H), 1.79 - 1.94 (m, 2 H), 1.56 - 1.72 (m, 3 H), 1.34 (d, J=3.51 Hz, 1 H).
C. (Cyclohept-Z-enyl)is0indoline—1,3-di0ne. Phthalimide potassium salt (19.59 g, 106 mmol), allylpalladium chloride dimer (0.477 g, 1.322 mmol), (1S,2S)-(-)-1,2- diaminocyclohexane-N,N'-bis(2'-diphenylphosphinobenzoyl) (2.74 g, 3.97 mmol), exylammonium bromide (50.6 g, 116 mmol), and DCM (212 mL) were ed in a round-bottomed flask. The flask was flushed with nitrogen and then placed in a sonication bath for 10 minutes. Cycloheptenyl methyl carbonate (9.00 g, 52.9 mmol) was then added to the flask in one portion and the resulting mixture was stirred under a nitrogen atmosphere at ambient temperature ght. The reaction was quenched by on of 50 mL water and was then extracted with 3 x 100 mL diethyl ether. The combined c layers were dried over anhydrous magnesium sulfate, d, and concentrated to an oil that slowly solidified overnight. The solids were ated with methanol (50 mL) and filtered to afford 7.6 g of crude product. The crude solid product was recrystallized from methanol and dried in vacuo to afford the title compound (6.70 g, 27.8 mmol, 52.5 % yield) as a solid that was determined to have an % e.e. of 97.2 (the S isomer was ously prepared as a standard using the (1R,2R)-(-)-1,2- diaminocyclohexane-N,N'-bis(2'-diphenylphosphinobenzoyl) ligand) by analytical chiral SFC chromatography menex Lux Cellulose-4, 250 x 4.6 mm I.D., 5-50 % isopropanol in C02, ATI—2514175v1 13 minute gradient). 1H NMR (400 MHz, DMSO-dg) 8 ppm 7.41 - 8.22 (m, 4 H), 5.62 - 5.97 (m, 2 H), 4.77 (d, 2 Hz, 1 H), 2.01 - 2.29 (m, 3 H), 1.87 - 2.00 (m, 1 H), 1.62 - 1.78 (m, 2 H), 1.45 - 1.61 (m, 1 H), 1.17 - 1.32 (m, 1 H). MS (ESI) m/z 242.3 [M+1]+.
] D. 2-((1R,ZS,3S)Br0m0hydr0xycycloheptyl)is0indoline-1,3-di0ne. To a solution of (R)(Cycloheptenyl)isoindoline-1,3-dione (4.00 g, 16.58 mmol) in chloroform (40.0 ml) and ethanol (1.400 mL) was added N—bromosuccinimide (3.78 g, 21.22 mmol) as a solid over 5 minutes at room temperature. After addition was complete, the reaction mixture was stirred at room temperature overnight under a nitrogen atmosphere. An additional n of osuccinimide (1.9 g) and 1.4 mL of ethanol were then added and the mixture was allowed to continue to mix at room temperature overnight under a nitrogen atmosphere. A third portion ofN—bromosuccinimide (1.0 g) and 1.4 mL onal ethanol were added to the mixture and it was again mixed at room temperature for a third night under a nitrogen here. The reaction mixture was then concentrated to dryness. The resulting residue was triturated with mL fresh chloroform. The solids were d, rinsed with form, and the filtrate concentrated under reduced pressure to dryness. THF (40 mL) and 1N hydrochloric acid (aq.) (10 mL) were added to the concentrated filtrate and the resulting mixture was stirred at room temperature for 90 minutes. The solution was concentrated under reduced pressure to remove THF. The concentrate was diluted with 125 mL ethyl acetate and 75 mL of a 1:1 mixture of water:saturated s sodium bicarbonate. The layers were separated and the aqueous was back extracted with 75 mL ethyl acetate. The combined ethyl acetate layers were washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure to afford an oil that was purified by silica gel chromatography (0-30% ethyl acetate in hexanes).
Fractions containing the desired product were concentrated under reduced pressure to the title compound (3.9 g, 11.53 mmol, 69.6 % yield) as a solid. 1H NMR (400 MHz, DMSO-d6) 8 ppm 7.88 (d, J=7.42 Hz, 4 H), 5.48 (d, J=5.47 Hz, 1 H), 4.73 (dd, J=10.54, 6.25 Hz, 1 H), 4.24 - 4.38 (m, 1 H), 3.91 - 3.99 (m, 1 H), 2.32 (br. s., 1 H), 1.67 - 1.96 (m, 5 H), 1.40 - 1.60 (m, 2 H).
MS (ESI) m/z 320.1 [M-18]+. A COSY NMR ment and the 1H NMR data ed above were used to verify the bromohydrin regiochemistry. An NOE (via NOESY experiment) was observed between the protons on the amino group carbon and the hydroxyl group carbon. This verifles a cis relationship between these two fianctional groups (a trans relationship, lacking this spacial ity, would not show such an effect). _ 98 _ ATI—25 14175v1 E. 2-((1R,3S)Hydr0xycycloheptyl)is0indoline-1,3-dione. To a solution of 2-((1R,2S,3S)bromohydroxycyclohepty1)isoindoline-1,3-dione (4.2 g, 12.42 mmol) in toluene (69 mL) and methanol (6.9 mL) was added tributyltin hydride (4.34 mL, 16.14 mmol) via syringe over 10 minutes under nitrogen followed by zobis(2-methy1propanenitri1e) (0.204 g, 1.242 mmol) in one portion. The reaction was then heated to reflux overnight under a nitrogen atmosphere. The reaction mixture was concentrated to dryness under reduced pressure to afford a e that was purified by silica gel chromatography (0-50% ethyl acetate in hexanes). Fractions containing the desired product were concentrated under reduced pressure to afford the title compound (2.6 g, 10.03 mmol, 81 % yield) as a solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.72 - 7.98 (m, 4 H), 4.58 (d, J=4.29 Hz, 1 H), 4.14 (br. s., 1 H), 3.67 (dt, J=6.25, 3.90 Hz, 1 H), 2.24 - 2.40 (m, 1 H), 2.04 -2.19 (m, 1 H), 1.81 - 1.95 (m, 2 H), 1.39 - 1.80 (m, 6 H). MS (ESI) m/z 259.9 [M+H]+.
E. (1 3-Aminocycloheptanol. 2-((1R,3S)Hydroxycycloheptyl)- isoindoline-1,3-dione (1800 mg, 6.94 mmol) was dissolved in ol (86.80 mL). Hydrazine monohydrate (0.354 mL, 7.29 mmol) was added to the solution and the resulting mixture was stirred overnight at reflux. An onal 0.2 eq (0.067 mL) of hydrazine was added and the mixture was refluxed for a second night. The reaction mixture was concentrated to a volume of a few milliliters and filtered. The solids were rinsed with 50 mL each ofDCM and chloroform and the resulting filtrate concentrated under reduced pressure to afford the title compound (174 mg, 1.347 mmol, 19.40 % yield) as an oil that was used without fiarther purification. MS (ESI) m/z 130.2 [M+H]+.
F. 4-((1R,3S)Hydr0xycycloheptylamino)—2-((1r,4R)—4—meth0xycyclohexyl- amin0)pyrimidine—5-carb0xamide. (1S,3R)Aminocycloheptanol (87 mg, 0.672 mmol) and DIEA (0.235 mL, 1.344 mmol) were added to a solution of ,4r)methoxycyclohexyl- amino)(methylsulf1ny1)pyrimidinecarboxamide (105 mg, 0.336 mmol) in NMP (1 mL).
The resulting mixture was heated to 80 CC for 4 h and then allowed to cool to ambient temperature overnight. The on mixture was condensed under reduced re and the residue purified by e-phase ative HPLC (5-80% acetonitrile + 0.1% TFA in water + 0.1% TFA, over 30 min). Fractions containing product were concentrated under reduced pressure. The resulting residue was re-dissolved in a methanol (5 mL), passed over a Varian StratoSpheres HCO3 resin SPE tube for TFA removal (0.9 mmol bicarbonate equiv.), and then _ 99 _ ATI—25 14175v1 concentrated under reduced pressure. The residue was triturated with acetonitrile and concentrated under reduced pressure to afford the title compound (67 mg, 0.177 mmol, 52.8 % yield) as a solid. 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.96 (d, J=5.86 Hz, 1 H), 8.33 (s, 1 H), 7.00 - 7.16 (m, 1 H), 4.46 (d, J=3.90 Hz, 1 H), 3.85 - 4.09 (m, 1 H), 3.56 - 3.81 (m, 2 H), 3.23 (s, 3 H), 2.97 - 3.15 (m, 1 H), 1.71 - 2.14 (m, 7 H), 1.51 (d, J=10.15 Hz, 11H). MS (ESI) m/z 378.3 [M+H]+.
Analytical chiral SFC using a ChiralPak AD-H column (250 x 4.6 mm I.D., isocratic 40% ol + 0.1% diethylamine in C02, 10 minute run time) was used to establish the relationship between the single isomer afforded in this chiral route with that of the preVious preparation of all four stereoisomers. The material prepared in this chiral route was found to have an identical retention time (3.848 s) as Peak 2 in Example 12A thus: PEAK 2 = 4-((1R,3S)hydr0xycycloheptylamin0)((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarb0xamide.
Based on 1H NMR, Peak 2 and Peak 4 were found to be an enantiomeric pair in Example 12A, thus: PEAK 4 = 4-((1S,3R)—3-hydr0xycycloheptylamin0)((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarb0xamide.
] In addition, it follows that Intermediate 2 used to make Peak 2 in Example 12A can then be assigned as tert-Butyl (1R,3 S)hydroxycycloheptylcarbamate and Intermediate 4 used to make Peak 4 in e 12A can then be assigned as tert-Butyl (1S,3R)—3- hydroxycycloheptylcarbamate.
In order to identify the absolute stereochemistry of Intermediate 1 and Intermediate 3 (and thereby the absolute stereochemistry of Peak 1 and Peak 3) in e 12A the following experiment was performed.
] Mitsunobu reaction of tert-butyl (1R,3R)hydroxycycloheptyl—carbamate to afford tert-butyl )hydr0xycycloheptylcarbamate.
O NH ATI—25 14175V1 A. tert-Butyl (1R,3S)—3-hydr0xycycloheptylcarbamate. ediate 3 from step B in Example 12A, was dissolved in THF (31.700 mL). The mixture was cooled to 0 0C in an ice bath under a nitrogen atmosphere and chloroacetic acid (261 mg, 2.76 mmol) followed by triphenylphosphine (723 mg, 2.76 mmol) were both added in one portion. A diethyl azodicarboxylate solution (0.437 mL, 2.76 mmol) was added ise waiting until the disappearance of the yellow color before adding the next drop. Once addition was complete, the solution was stirred at 0 CC under a nitrogen here for 3 h. The reaction was concentrated under reduced pressure and the resulting oil purified by silica gel chromatography (0-40% ethyl acetate in hexanes). Pure product containing fractions were ed and concentrated under d re and then olved in methanol (16 mL). Sodium carbonate (186 mg, 1.755 mmol) was added to the solution and the resulting mixture was stirred at ambient temperature for 3 h. The reaction mixture was concentrated under reduced re to dryness.
The residue was diluted with 50 mL of a 1:1 mixture of water:saturated aqueous sodium bicarbonate and 75 mL DCM. The layers were separated and the aqueous back extracted with 50 mL DCM. The combined DCM layers were dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced re to afford a material that had an identical retention time by analytical SFC as Intermediate 2 from step B in Example 12A, or tert—butyl (1R,3S)hydroxycycloheptylcarbamate (417 mg, 1.818 mmol, 72.5 % yield) as a solid. 1H NMR (400 MHz, DMSO-dg) 8 ppm 6.63 - 6.87 (m, 1 H), 4.44 (d, J=3.90 Hz, 1 H), 3.47 - 3.71 (m, 1 H), 0.99 — 1.98 (m, 19 H). MS (ESI) m/z 260.1 .
Intermediate 3 from step B in Example 12A is therefore assigned as tert-butyl (1R,3R)hydroxycycloheptylcarbamate. Peak 3 from step D derived from Intermediate 3 in Example 12A is therefore assigned as 4-((1R,3R)hydroxycycloheptylamino)((1r,4R) methoxycyclohexylamino)pyrimidinecarboxamide.
Because Peak 1 and Peak 3 are enantiomers by 1H NMR as described in e 12A Peak 1 can be assigned as 4-((1S,3S)Hydroxycycloheptylamino)((1r,4S) methoxycyclohexylamino)pyrimidinecarboxamide. It follows that Intermediate 1 from step B in Example 12A that was used to prepare Peak 1 in step D of Example 12A can be assigned as tert-butyl (l S,3 S)hydroxycycloheptylcarbamate.
Example 13: 4-((1R,3S)—3-Hydr0xycyclohexylamin0)((1r,4R)—4-(2,2,2- trifluoroethoxy) cyclohexylamin0)pyrimidine—5-carboxamide, 4-((1R,3S) ATI—2514l75vl ycyclohexylamino)—2-((1s,4S)—4-(2,2,2-triflu0r0eth0xy)— cyclohexylamin0)pyrimidine—5-carb0xamide F o F o FFX/O’" l/YkNHZ F N/ Fflvo‘"’N)\N/hl/YLNHZ "’N NH NH H i H i g ® OH OH A. 4-((1R,3S)—3-Hydr0xycyclohexylamin0)((1r,4R)—4-(2,2,2-triflu0r0- ethoxy)cyclohexylamin0)pyrimidinecarb0xamide and ,3S)—3-hydr0xycyclo- hexylamin0)((1s,4S)—4-(2,2,2-triflu0r0ethoxy)cyclohexylamino)—pyrimidine carboxamide. To a stirring solution of 4-((1R,3S)—3-hydroxycyclohexyl-amino) (methylsulfonyl)pyrimidinecarboxamide (440 mg, 1.400 mmol; synthesis described herein) and bis(2,2,2-trifiuoroethoxy)-cyclohexanamine trihydrochloride (529 mg, 2.099 mmol) in DMSO (6.964 mL) was added DIEA (0.978 mL, 5.60 mmol). The resulting mixture was allowed to stir overnight at 100 CC. The crude reaction mixture was concentrated, purified by silica gel chromatography (0-10% methanol/dichloromethane), and then purified by reversed-phase silica gel chromatography (methanol/water with 0.1% formic acid modifier). The product containing fractions were lized and concentrated to afford 4-((1R,3 S)—3 -hydroxycyclohexylamino)(4-(2,2,2-trifiuoroethoxy)cyclohexyl- pyrimidinecarboxamide (135 mg, 0.313 mmol, 22.36 % yield) as a mixture of 2 stereoisomers. MS (ESI) m/z 432.2 [M+H]+. This mixture was separated using chiral fluid chromatography (AD-H colum) to afford 87.8 mg (0.203 mmol) of a single, faster eluting, stereoisomer (PEAK 1) and 24.2 mg (0.056 mmol) of a second single, slower eluting, stereoisomer (PEAK 2).
Subsequently the same reaction ce was repeated using (1r,4r) (2,2,2-trifluoroethoxy)cyclohexanamine, which was prepared as follows. (1r,4r)—4-(Benzylamin0)cyclohexanol. A mixture of (1r,4r)amino- cyclohexanol (230 g, 2 mol), benzaldehyde (212 g, 2 mol) and 4A Molecular Sieves in methanol (2 L) was refluxed under a nitrogen here for 3 h. The mixture was cooled using an ice-water bath, and sodium borohydride (72 g, 2 mol) was carefully added in small ns. After the completion of the addition, the reaction was stirred at room ATI—25 14175V1 temperature overnight. The solvent was evaporated, and the residue was partitioned between dichloromethane (2 L) and water (1 L). The dichloromethane layer was separated, washed with a saturated aqueous sodium de solution, and directly used in the next step.
Benzyl benzyl((1r,4r)—4-hydr0xycyclohexyl)carbamate. To the dichloromethane solution of (lr,4r)(benzylamino)cyclohexanol obtained in the previous step was added saturated aqueous sodium bicarbonate (1.5 L), and to this biphasic system was added benzyl chlorofomate (358.2 g, 2.1 mol) slowly at 0 0C. After completion of addition, the reaction was stirred at room temperature for another 1 h. Then the organic phase was separated and evaporated. After concentration, the residue was purified by silica gel chromatography (0-30% ethyl acetate in hexanes) to give the title nd as a colorless liquid (400 g, 1.18 mol, 59% yield for two steps). Both steps were repeated, to afford a total of 800 g of benzyl benzyl((1r,4r)hydroxycyclohexyl)carbamate.
Benzyl benzyl((1r,4r)—4-(2,2,2-triflu0roeth0xy)cyclohexyl)carbamate.
To a solution of benzyl benzyl((1r,4r)hydroxycyclohexyl)carbamate (400 g, 1.18 mol) in chloroform (2 L) was added a 50% aqueous on of fluoroboric acid (20 mL), and a stream of diazotrifluoroethane (freshly prepared by mixing trifluoroethaneamine hydrochloride (1.6 kg, 11.8 mol) and NaNOZ (814 g, 11.8 mol) in water (3 L) and then bubbling it into the reaction solution . The reaction was monitored by TLC, and after completion, a saturated aqueous solution of potassium carbonate (300 mL) was added. The chloroform layer was separated, concentrated under reduced pressure, and ed by column chromatography (0-10% ethyl acetate in hexanes) to afford benzyl benzyl ((1r,4r)(2,2,2-trifluoroethoxy)cyclohexyl)carbamate (192 g, 0.456 mol, 38% yield). This reaction was repeated, and another batch of benzyl benzyl ((lr,4r)(2,2,2- trifluoroethoxy)cyclohexyl)carbamate (185 g, 0.439 mol) was obtained. (1r,4r)—4-(2,2,2-Triflu0r0eth0xy)cyclohexanamine. To a solution of benzyl benzyl((1r,4r)(2,2,2-trifluoroethoxy)cyclohexyl)carbamate (377 g, 0.89 mol) in ethyl acetate (2 L) was added 20% palladium hydroxide on carbon (50 g), and the e was stirred under a 55 psi hydrogen atmosphere for 24 h at room temperature. The catalyst was d by tion, and the te was concentrated under reduced re. The resulting residue was dissolved in 2N hydrochloric acid (1.5 L) and the on was ATI—2514l75vl washed with methyl l ether (300 mL X 5). Solid potassium carbonate was added to adjust the pH to above 10. The product was ted into dichloromethane (500 mL x 5).
The ed organic phases were dried and concentrated under reduced pressure. The residue was purified by silica gel chromatography (10-30% methanol in dichloromethane) to afford (1r,4r)(2,2,2-trifluoroethoxy)cyclohexanamine (115 g, 0.584 mol, 66% yield): 1H NMR (400 MHz, DMSO-d6) 8 ppm 4.01 (q, J: 8.8 Hz, 2H), 3.40-3.33 (m, 1H), 2.56 - 2.53 (m, 1H), 1.94-1.92 (m, 2H), 1.75-1.72 (m, 2H), 1.24-1.15 (m, 2H), 1.08 - 0.97 (m, 2H); MS (ESI) m/z 198.2 [M+l]+.
Using the intermediate above, it was shown that the material obtained as PEAK 1 ponded to 4-((1R,3S)hydroxycyclohexylamino)((1r,4R)(2,2,2- trifluoroethoxy)cyclohexylamino)pyrimidinecarboxamide, while the material obtained as PEAK 2 coresponded to 4-((1R,3S)hydroxycyclohexylamino)((1s,4S)(2,2,2- trifluoroethoxy)cyclohexylamino)pyrimidinecarboxamide.
] PEAK 1: 4-((1R,3S)—3-hydr0xycyclohexylamino)—2-((1r,4R)—4-(2,2,2- trifluoroeth0xy)cyclohexylamin0)pyrimidine—5-carb0xamide. 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.90 (d, J=6.25 Hz, 1H), 8.34 (s, 1H), 7.46 - 7.52 (m, 1H), 7.07 - 7.17 (m, 1H), 6.91 (dd, J=8.59, 2.73 Hz, 1H), 4.60 - 4.76 (m, 1H), 4.02 (q, J=9.63 Hz, 2H), 3.39 — 3.96 (m, 4H), 0.92 — 2.24 (m, 16H). 19F NMR (376 MHz, DMSO-d6) 5 ppm -76.47 — 72.01 (m, 3 F). MS (ESI) m/z 432.0 [M+H]+.
PEAK 2: 4-((1R,3S)—3-hydroxycyclohexylamino)—2-((ls,4S)(2,2,2- trifluoroeth0xy)cyclohexylamin0)pyrimidine—5-carb0xamide. 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.90 (d, J=6.64 Hz, 1H), 8.34 (br. s., 1H), 6.79 - 7.50 (m, 2H), 4.64 (br. s., 1H), 4.00 — 4.12 (m, 2H), 3.42 (d, J=4.69 Hz, 4H), 0.91 — 2.25 (m, 16H). ”P NMR (376 MHz, DMSO-d6) 5 ppm -73.63 (t, J=9.47 Hz, 3 F). MS (ESI) m/z 432.0 [M+H]+. —104— ATI—25 14175Vl Example 14: 2-((1r,4R)—4-(Diflu0r0meth0xy)cyclohexylamin0)((1R,3S) hydroxycyclohexylamin0)pyrimidinecarboxamide FY. 1* F "’NJ\N/ NH A. Methyl 2-chlor0((1R,3S)—3-hydr0xycyclohexylamin0)pyrimidine- -carb0xylate. To a stirring solution of methyl 2,4-dichloropyrimidinecarboxylate (4.00 g, 19.32 mmol) in THF (85 mL) was added DIEA (3.37 mL, 19.32 mmol). The resulting mixture was cooled to -78 0C in a dry ice/acetone bath. A solution of (lS,3R)—3- aminocyclohexanol (2.448 g, 21.25 mmol; prepared as described in Tetrahedron: Asymmetry 15:205 l—2056(2004)) in 40 mL THF was added at such a rate that the temperature remained at -78 CC. The resulting mixture was d at -78 CC for 1 h. The reaction mixture was then concentrated to dryness and diluted with 200 mL ethyl e and 75 mL of a 1:1 mixture of water and an aqueous saturated sodium bicarbonate solution. The layers were separated and the s layer back-extracted with 100 mL ethyl acetate. The combined ethyl acetate layers were washed with 50 mL of a saturated aqueous sodium chloride solution, dried over ous magnesium sulfate, filtered and concentrated to an oil that solidified upon standing. The solid was triturated with diethyl ether (5 mL), d, rinsed with diethyl ether (5 mL) and dried in vacuo to afford methyl 2-chloro((1R,3 S)—3-hydroxycyclohexylamino)pyrimidinecarboxylate (3 .7 g, 12.95 mmol, 67% yield) that was used without further ation. MS (ESI) m/z 286.0 [M+1]+.
B. (1r,4r)(Dibenzylamin0)cyclohexanol. transAminocyclohexanol hydrochloride (25 g, 217 mmol) was suspended in acetonitrile (500 mL). Benzyl bromide (54.2 mL, 456 mmol) and ium carbonate (120 g, 868 mmol) were added to the suspension and the ing mixture was d Vigorously overnight at room temperature. The reaction mixture was filtered through a plug of celite and the plug was washed thoroughly with acetonitrile. The filtrate was condensed to give a white solid. The crude solids were purified by silica gel chromatography (0-60% ethyl acetate in hexanes).
ATI—2514175V1 Pure product containing fractions were combined and concentrated to afford (1r,4r) (dibenzylamino)cyclohexanol (25 g, 85 mmol, 39.0 % yield) as a white solid. 1H NMR (400 MHz, DMSO-dg) 5 ppm 7.26 - 7.36 (m, 8H), 7.16 - 7.22 (m, 2H), 4.43 (d, J=4.69 Hz, 1H), 3.55 (s, 4H), 3.27 - 3.33 (m, 1H), 2.28 - 2.40 (m, 1H), 1.69 - 1.88 (m, 4H), 1.40 (qd, J=12.56, 2.93 Hz, 2H), 0.89 - 1.05 (m, 2H); MS (ESI) m/z 296.3 .
C. (1r,4r)—N,N-Dibenzyl(diflu0romethoxy)cyclohexanamine. (1r,4r)- 4-(Dibenzylamino)cyclohexanol (5 g, 16.93 mmol) and copper(I) iodide (0.645 g, 3.39 mmol) were dissolved in 170 mL acetonitrile and heated to 45 0C under a en atmosphere. To this mixture was added a on of 2,2-difluoro(fluorosulfonyl)acetic acid (3.50 mL, 33.9 mmol) in 30 mL acetontrile over 10 min. Once addition was complete, the mixture was stirred at 45 CC under nitrogen atmosphere for 1 h. An additional portion of 2,2-difluoro(fluorosulfonyl)acetic acid (2.86 mL, 27.7 mmol) reagent in 30 mL acetonitirile was then added over 10 min. The reaction mixture was stirred for an additional 1 h at 45 CC under nitrogen atmosphere. le components were then removed via evaporation and the residue was diluted with 175 mL ethyl acetate and 75 mL of a 1:1 mixture of water and saturated aqueous sodium onate. The resulting biphasic mixture containing solids was filtered through a sintered glass Buchner funnel. The filtrate layers were separated and the s layer was extracted with 50 mL ethyl acetate. The combined ethyl acetate layers were washed with 50 mL of a 1:1 mixture of saturated sodium chloride and water, dried over anhydrous magnesium sulfate, filtered, and concentrated to an oil. The crude oil was purified by silica gel tography (0-30% ethyl acetate in hexanes). Product ning fractions were ed and concentrated to afford (1r,4r)-N,N-dibenzyl(difluoromethoxy)cyclohexanamine (3.12 g, 9.03 mmol, 53.4 % yield) as an oil that solidified to an off-white solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.26 - 7.38 (m, 8H), 7.17 - 7.23 (m, 2H), 6.66 (s, 1H), 3.97 (t, J=4.49 Hz, 1H), 3.57 (s, 4H), 2.36 - 2.47 (m, 1H), 1.98 (d, 4 Hz, 2H), 1.83 (d, J=12.10 Hz, 2H), 1.49 (d, J=12.89 Hz, 2H), 1.22 (d, J=12.49 Hz, 2H). ”P NMR (376 MHz, DMSO-d6) d ppm -79.28 (d, J=73.00 Hz, 2 F). MS(ESI) m/z 346.1 [M+1]+.
D. (1r,4r)—4-(Difluor0meth0xy)cyclohexanamine. (1r,4r)-N,N-Dibenzyl- 4-(difluoromethoxy)cyclohexanamine (1.6 g, 4.63 mmol) was dissolved in ethanol (23 mL) in a Parr shaker flask and 20 wt% palladium hydroxide on carbon (0.650 g, ATI—25 14175v1 0.926 mmol) was added. The container was evacuated, placed on a Parr shaker apparatus, and shaken under a 50 psi hydrogen atmosphere overnight. The palladium hydroxide was removed by filtration h a pre-wetted (with ethanol) bed of celite. The filter cake was rinsed well with ethanol. The e was trated to an oil that was dissolved in 100 mL ethyl acetate, dried over anhydrous ium sulfate, filtered, and concentrated to an oil that solidified, yielding (1r,4r)(difiuoromethoxy)cyclohexanamine (0.621 g, 3.76 mmol, 81 % yield). 1H NMR (400 MHz, DMSO-d6) 8 ppm 6.46 -6.93 (m, 1H), 3.96 (s, 1H), 2.52 - 2.59 (m, 1H), 1.84 - 1.95 (m, 2H), 1.68 - 1.79 (m, 2H), 1.29 - 1.43 (m, 2H), 1.01 — 1.14 (m, 2H). ”P NMR (376 MHz, DMSO-d6) 5 ppm -78.93 (d, J=91.00 Hz, 2 F).
MS(ESI) m/z 166.2 [M+1]+.
E. Methyl 2—((1r,4R)—4-(diflu0r0meth0xy)cyclohexylamin0)—4-((1R,3S)- 3 hydroxycyclohexylamin0)pyrimidine—5-carboxylate. To a stirring mixture of methyl 2-chloro((1R,3 S)—3-hydroxycyclohexylamino)pyrimidinecarboxylate (300 mg, 1.050 mmol) and (1r,4r)(difiuoromethoxy)cyclohexanamine (260 mg, 1.575 mmol) in DMSO (5mL) was added DIEA (0.550 mL, 3.15 mmol). The resulting mixture was stirred at 70 CC for 1 h. The crude on mixture was trated and then d by silica gel chromatography (0-60% ethyl e in s). The product containing fractions were combined and concentrated to afford methyl 2-((1r,4R)(difiuoro- methoxy)cyclohexylamino)((1R,3S)hydroxycyclohexylamino)pyrimidine carboxylate (165 mg, 0.398 mmol, 37.9 % yield). 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.37 - 8.49 (m, 1H), 8.06 (d, J=7.42 Hz, 1H), 7.22 - 7.55 (m, 1H), 6.49 - 6.94 (m, 1H), 4.67 - 4.76 (m, 1H), 3.75 - 4.10 (m, 2H), 3.72 (s, 3H), 3.43 - 3.69 (m, 2H), 1.63 - 2.24 (m, 8H), 1.08 - 1.54 (m, 8H). MS(ESI) m/z 415.5 [M+1]+.
F. 2-((1r,4R)—4-(Diflu0r0meth0xy)cyclohexylamin0)((1R,3S)-3 hydroxycyclohexylamino) pyrimidine-S-carboxylic acid. To a stirring solution of methyl 2-(( 1 r,4R)—4-(difiuoromethoxy)cyclohexylamino)(( 1 R,3 S)hydroxycyclo- hexylamino)-pyrimidinecarboxylate (170 mg, 0.410 mmol) in methanol (10 mL) was added an aqueous solution of sodium hydroxide (1 M, 3.075 mL, 3.075 mmol) in one portion. The resulting mixture was allowed to stir at 50 CC for 2 days. The reaction mixture was concentrated to remove methanol, was diluted with additional water (10 mL), and an aqueous solution of citric acid (2 M, 4.10 mL, 8.20 mmol) was slowly added. The ATI—25 14175v1 2012/034349 ing precipitate was stirred at room temperature for 30 min, filtered, and the solids then washed well with water. The solids were dried in vacuo to afford 2-((1r,4R) (difluoromethoxy)cyclohexylamino)((1R,3 S)hydroxycyclohexylamino)pyrimidine- -carboxylic acid (117 mg, 0.292 mmol, 71.2 % yield) as a solid that was used without further purification. MS (ESI) m/z 401.5 [M+1]+.
G. 2-((1r,4R)—4-(Difluorometh0xy)cyclohexylamin0)((1R,3S)—3- hydroxycyclohexylamino)pyrimidine—5-carb0xamide. To a stirring suspension of 2- ((1r,4R)(difluoromethoxy)cyclohexylamino)((1R,3 S)hydroxycyclohexylamino)- pyrimidinecarboxylic acid (117 mg, 0.292 mmol) and HATU (167 mg, 0.438 mmol) in DMF (2 mL) was added ammonium chloride (78 mg, 1.461 mmol) and DIEA (0.255 mL, 1.461 mmol). The resulting e was stirred at room temperature overnight. The crude reaction mixture was concentrated to remove the le components and then purified by silica gel chromatography (10-90% ethyl acetate (containing 10% ammonia saturated ol) in hexanes). The product fractions were combined and concentrated to afford 2-((1r,4R)(difluoromethoxy)cyclohexylamino)((1R,3 S)hydroxycyclohexyl- amino)pyrimidinecarboxamide (35.3 mg, 0.088 mmol, 30.2 % yield) as a solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.90 (d, J=6.64 Hz, 1H), 8.34 (br. s., 1H), 6.50 — 7.14 (m, 2H), 4.64 (br. s., 1H), 3.42 — 4.11 (m, 4H), 0.87 — 2.23 (m, 16H). ”P NMR (376 MHz, DMSO-d6) 5 ppm -78.98 (d, J=110.82 Hz, 2 F). MS (ESI) m/z 400.5 [M+1]+.
Example 15: 4-((1R,3S)—3-Meth0xycyclohexylamin0)((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarboxamide /O N\ NH2 .,)'\/ ’N N NH A. utyl (1R,3S)—3-hydr0xycyclohexylcarbamate. To a stirring solution of (1S,3R)aminocyclohexanol (1.0 g, 8.68 mmol; prepared as bed in Tetrahedron: Asymmetry 15:2051—2056 (2004)) in dioxane (8.04 mL) was added di-tert- butyl-dicarbonate (2.369 g, 10.85 mmol). The resulting mixture was stirred at room temperature for 18 h. The reaction was concentrated, sodium chloride (0.507 g, ATI—25 14175v1 WO 45569 8.68 mmol) was added, and the resulting solution was diluted with water (20 mL) and hexanes (20 mL). The suspension was stirred vigorously for 20 min, then filtered and the solids were rinsed with hexanes and dried under vacuum to afford tert-butyl )—3- hydroxycyclohexylcarbamate (1.7836 g, 8.28 mmol, 95 % yield) as an off-white solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 6.74 (d, J=8.20 Hz, 1H), 4.58 (d, J=4.29 Hz, 1H), 3.35 - 3.43 (m, 1H), 3.11 - 3.27 (m, 1H), 1.90 (d, J=11.32 Hz,1H),1.68 - 1.78 (m, 1H), 1.63 (d, J=3.12 Hz, 2H), 1.37 (s, 9H), 0.87 - 1.23 (m, 4H); MS (ESI) m/z 216.3 [M+1]+.
B. tert-Butyl (1R,3S)meth0xycyclohexylcarbamate. To a solution of tert-butyl (1R,3S)hydroxycyclohexylcarbamate (1.78 g, 8.27 mmol) in anhydrous THF (8 mL) was added sodium hydride (0.230 g, 9.09 mmol) in ns under nitrogen at 0 oC.
The mixture was warmed to room temperature and stirred for 30 min. A solution of iodomethane (0.824 mL, 13.23 mmol) in anhydrous THF (4 mL) was added drop wise to the mixture at 0 CC and the resulting mixture was stirred at room ature for 20 h.
The crude mixture was quenched with water (50 mL) and extracted with ethyl acetate (3 x 50 mL). The combined c layers were washed with brine, dried over sodium sulfate and filtered. The filtrate was concentrated in vacuo, and then purified by silica gel chromatography (0 — 50 % ethyl acetate in hexanes). The t fractions were combined and concentrated to afford tert-butyl (1R,3S)—3-methoxycyclohexylcarbamate (1.0686 g, 4.66 mmol, 56.4 % yield) as a white solid; MS (ESI) m/z 230.5 [M+1]+.
] C. (1R,3S)Meth0xycyclohexanamine trifluoroacetate. To a solution of tert-butyl (1R,3S)methoxycyclohexylcarbamate (1.0686 g, 4.66 mmol) in anhydrous DCM (18.64 mL) was added TFA (3.59 mL, 46.6 mmol) and the resulting solution was stirred at room temperature for 1 h. After that, the solvents were removed in vacuo, and the resulting residue was triturated with ethyl ether (2 x 50mL) to afford (1R,3S) methoxycyclohexanamine trifluoroacetate (1 . 133 g, 4.66 mmol, 100 % yield) as a white solid. The crude product was used without fiarther purification in the next step.
D. Ethyl 4-((1R,3S)meth0xycyclohexylamin0)—2-(methylthi0)- dine-S-carboxylate. To a stirring solution of ethyl 4-chloro(methylthio)- dinecarboxylate (1.084 g, 4.66 mmol) and (1R,3S)—3-methoxycyclohexanamine trifluoroacetate (1.133 g, 4.66 mmol) in ethanol (18.63 mL) was added DIEA (2.434 mL, 13.97 mmol). The on mixture was heated to 60 CC for 17 h. The reaction was ATI—25 14175v1 2012/034349 d from heat, cooled to room temperature and concentrated. The crude material was purified by silica gel chromatography (0-25% ethyl acetate in hexanes) to afford ethyl 4- ((lR,3S)methoxycyclohexylamino)(methylthio)pyrimidinecarboxylate (l .4284 g, 4.39 mmol, 94 % yield) as a clear oil. MS (ESI) m/z 326.2 [M+l]+.
E. 4-((1R,3S)Methoxycyclohexylamin0)(methylthi0)pyrimidine carboxylic acid. To a stirring solution of ethyl 4-((lR,3S)—3-methoxycyclohexylamino)- hylthio)pyrimidinecarboxylate (l .4284 g, 4.39 mmol) in ethanol (1254 mL) was added aqueous sodium hydroxide solution (1 N, 8.78 mL, 8.78 mmol) and the e was stirred for 2 h. The reaction was concentrated, diluted with water (50mL), and then neutralized while stirring with aqueous citric acid solution (2 M, 5.49 mL, 10.97 mmol).
The precipitate was filtered, then washed with water (2 x 50 mL) and dried to afford 4-(( l R,3 S)—3 -methoxycyclohexylamino)(methylthio)pyrimidinecarboxylic acid (1.1679 g, 3.93 mmol, 89% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 1322 (s, 1H), 8.65 (d, J=7.03 Hz, 1H), 8.51 (s, 1H), 4.07 - 4.24 (m, 1H), 3.29 - 3.33 (m, 1H), 3.26 (s, 3H), 2.46 (s, 3H), 2.15 (d, J=l2.10 Hz, 1H), 1.63 - 1.88 (m, 3H), 1.21 - 1.48 (m, 4H); MS (ESI) m/z 298.1 [M+1]+.
F. 4-((1R,3S)Meth0xycyclohexylamin0)(methylthio)pyrimidine—5- amide. 4-(( l R,3 S)—3 -Methoxycyclohexylamino)(methylthio)pyrimidine-5 - carboxylic acid (1.1662 g, 3.92 mmol) and HATU (2.237 g, 5.88 mmol) were stirred in DMF (7.84 mL) for 5 min at room temperature, then ammonium chloride (1 .049 g, 19.61 mmol) and DIEA (3.42 mL, 1961 mmol) were added while stirring. The reaction was allowed to stir at room temperature for l h. The reaction mixture was poured into water (100 mL) and ted with ethyl acetate (3 x 50mL). The combined organic layers were washed with water (50 mL), brine (25 mL), dried over sodium sulfate, filtered, and concentrated. The residual DMF was removed by suspending the oily residue in 100 mL water and 100 mL hexanes. The biphasic suspension was stirred Vigorously for 30 min, filtered and washed with hexanes to afford 4-((lR,3S)—3-methoxycyclohexylamino) (methylthio)pyrimidinecarboxamide (1.1081 g, 3.74 mmol, 95 % yield) as an ite solid. MS (ESI) m/z 397.2 [M+l]+.
G. 4-((1R,3S)Meth0xycyclohexylamin0)(methylsulf0nyl)— pyrimidine—5-carb0xamide. To a solution of 4-((lR,3S)—3-methoxycyclohexylamino) -llO- ATI—2514l75Vl (methylthio)pyrimidinecarboxamide (1.1081 g, 3.74 mmol) in DCM (18.69 mL) and acetone (18.69 mL) at room temperature was added mCPBA (1.676 g, 7.48 mmol) in five batches over 2 min and stirred at room temperature for 16 h. To the crude reaction mixture was added 10% s sodium thiosulfate solution (35 mL) and the mixture was stirred for 5 min before evaporation of the le solvents. The material was partitioned between ethyl acetate and water, and the organic layer was then washed with saturated aqueous sodium bicarbonate solution and then brine. The combined s layers were then washed with ethyl acetate (3x), organic layers were combined, dried over sodium sulfate, filtered, and condensed. After drying under high , 4-((1R,3S)methoxy- cyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (0.9887 g, 3.01 mmol, 81 % yield) was obtained as a white solid. MS (ESI) m/z 329.3 [M+l]+.
H. 4-((1R,3S)Meth0xycyclohexylamin0)((1r,4R)—4- methoxycyclohexylamin0)pyrimidine—5-carb0xamide. To a stirring solution of 4-((1R,3 ethoxycyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (0.3 g, 0.914 mmol) and (1r,4r)methoxycyclohexanamine (0.236 g, 1.827 mmol) in DMSO (0.914 mL) was added DIEA (0.637 mL, 3.65 mmol) and the reaction mixture was d at 100 CC for 17 h. The crude reaction e was concentrated and then purified by silica gel chromatography (0-10% methanol in ethyl acetate). The product fractions were combined and concentrated to afford 4-((1R,3 S)—3-methoxycyclohexylamino) ((1r,4R)methoxycyclohexylamino)-pyrimidinecarboxamide (0.155 g, 0.411 mmol, 44.9 % yield) as a white solid. 1H NMR (400 MHZ, DMSO-d6) 5 ppm 8.95 (d, J=7.42 Hz, 1H), 8.34 (s, 1H), 7.01 - 7.17 (m, 1H), 6.76 - 6.88 (m, 1H), 3.51 - 4.07 (m, 2H), 2.97 - 3.29 (m, 8H), 1.63 - 2.42 (m, 8H), 0.98 - 1.39 (m, 9H); MS (ESI) m/z 378.4 [M+l]+.
ATI—2514l75vl Example 16: 2-((1r,4R)—4-Hydroxymethylcyclohexylamino)((1R,3S)—3- hydroxycyclohexylamino)pyrimidine—5-carboxamide \‘O N \ NH2 ., 9L / ’N N NH A. )—4-Amino-l-methylcyclohexanol hydrochloride. A solution of (1r,4r)(dibenzylamino)methylcyclohexanol (15 .3 g, 49.4 mmol; prepared as described in PCT Int. Appl. publication WO2010/027500 ) in methanol (137 mL) and ethyl acetate (137 mL) in a 500 mL Parr vessel was degassed by bubbling nitrogen through the solution for 5 min. To the on was added palladium hydroxide on carbon (6.94 g, 4.94 mmol) and the vessel was placed on a Parr shaker for 6 days at 3 atm hydrogen. The vessel was removed from the Parr shaker and then the suspension was filtered through celite. The filtrate was concentrated, the residue was dissolved in 50 mL methanol, acidified with aqueous hydrochloric acid on (6 M, 9.06 mL, 54.4 mmol) and concentrated to afford (lr,4r)—4-amin0-l-methylcyclohexanol hydrochloride (8.31 g, 50.2 mmol, 101 % yield) as an off-white solid, which was used without further purification in the next step.
] B. utyl (1r,4r)—4-hydroxymethylcyclohexylcarbamate. To a stirring solution of (lr,4r)amino-l-methylcyclohexanol hydrochloride (8.31 g, 50.2 mmol) in an aqueous sodium hydroxide solution (1 N, 100 mL, 100 mmol) was added di-tert-butyl-dicarbonate (13.68 g, 62.7 mmol) and the reaction mixture was stirred at room temperature for 2 h. The reaction was concentrated and then purified by silica gel chromatography % ethyl acetate in hexanes). The product fractions were combined and concentrated. The material contained ~50% desired product as shown by LCMS, so the material was repurified twice on silica gel, using 0-50% ethyl e in hexanes as the mobile phase. The product fractions were combined and concentrated to afford tert-butyl (lr,4r)hydr0xymethylcyclohexylcarbamate (2.2 g, 9.59 mmol, 19.13 % yield) as a white solid. MS (ESI) m/z 230.5 [M+1]+. -ll2- ATI—2514l75vl C. (1r,4r)—4-Amin0-l-methylcyclohexanol trifluoroacetate. To a stirring solution of tert-butyl (1r,4r)hydroxymethylcyclohexylcarbamate (2.04 g, 8.90 mmol) in DCM (44 mL) was added TFA (13.71 mL, 178 mmol) and the solution was stirred at room temperature for 22 h. The reaction was concentrated, the residue was triturated twice with ethyl ether to afford (1r,4r)aminomethylcyclohexanol trifluoroacetate (2.106 g, 8.66 mmol, 97 % yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.73 (br. s., 3H), 4.39 (br. s., 1H), 3.05 (d, J=3.51 Hz, 1H), 1.27 - 1.97 (m, 8H), 1.11 (s, 3H); MS (ESI) m/z 130.2 [M+1]+.
D. 2-((1r,4R)—4-Hydr0xymethylcyclohexylamino)—4-((1R,3S) hydroxycyclohexylamino)pyrimidine—5-carb0xamide. To a stirring on of ,3 S)—3-hydroxycyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (0.432 g, 1.374 mmol; synthesis described herein) and )aminomethylcyclo- hexanol trifluoroacetate (0.435 g, 1.786 mmol) in dioxane (2.75 mL) was added DIEA (0.718 mL, 4.12 mmol) and the resulting solution was stirred at 110 °C for 23 h. The crude reaction mixture was concentrated and the residue was d by HPLC (20-100% methanol in water). The product fractions were combined, trated and the residue was triturated with acetonitrile. The resulting precipitate was collected to afford 2-((1r,4R)—4-hydroxymethylcyclohexylamino)((1R,3 S)hydroxycyclohexylamino )pyrimidinecarboxamide (0.0483 g, 0.133 mmol, 9.67 % yield) as an off white solid. 1H NMR (400 MHz, 6) 8 ppm 8.91 (d, J=6.64 Hz, 1H), 8.34 (br. s., 1H), 6.56 - 7.26 (m, 1H), 3.39 - 4.78 (m, 4H), 2.54 (s, 3H), 0.66 - 2.25 (m, 19H); MS (ESI) m/z 364.5 [M+1]+ ATI—25 14175V1 Example 17: 4-((1S,3S,4S)Hydr0xymethylcyclohexylamino)—2-((1r,4S) methoxy-cyclohexylamin0)pyrimidinecarboxamide; 4-((1R,3R,4R)—3-Hydr0xy methylcyclohexylamino)—2-((1r,4R)—4-methoxy-cyclohexylamin0)pyrimidine carboxamide; 4-((1S,3R,4R)—3-Hydr0xymethylcyclohexylamino)—2-((1r,4S) methoxy-cyclohexylamin0)pyrimidine—5-carboxamide; 4-((1R,3S,4S)Hydr0xy methylcyclohexylamino)—2-((1r,4R)—4-methoxy-cyclohexylamin0)pyrimidine carboxamide o o o “U 1%“ ”U 11* “O 11*~O o / ‘0 11*“\ / ll, ’N 'I N NH N NH I"NJ\N/ NH 'N N/ NH fl H : H H : ; .II 'OH : ‘OH : 'OH OH A. 2-((1s,4s)—4-Hydr0xy—4-methylcyclohexyl)is0indoline-1,3-di0ne. To a stirring on of (1s,4s)aminomethylcyclohexanol (10 g, 77 mmol; prepared as described in PCT Int. Appl. publication WO2010/027500) and potassium carbonate (18.72 g, 135 mmol) in water (155 mL) at 0 CC was added N—carbethoxyphthalimide (18.66 g, 85 mmol) and the on was stirred for 2 h at room temperature. The fine suspension was filtered, resuspended in 100 mL water, and refiltered to afford 2-((1s,4s)- 4-hydroxymethylcyclohexyl)isoindoline-1,3-dione (11.413 g, 44.0 mmol, 56.9 % yield) as a white powder. 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.79 — 7.88 (m, 4H), 4.11 (s, 1H), 3.94 (s, 1H), 2.51 - 2.56 (m, 1H), 2.44 - 2.49 (m, 1H), 1.56 - 1.70 (m, 2H), 1.33 - 1.47 (m, 4H), 1.13 (s, 3H); MS (ESI) m/z 260.5 .
B. 2-(4-Methylcyclohexenyl)is0indoline-1,3-di0ne. 2-((1s,4s) Hydroxymethylcyclohexyl)isoindoline-1,3-dione (4.6568 g, 17.96 mmol) and potassium hydrogen sulfate (4.89 g, 35.9 mmol) were ed and heated to 140 0C for min. No on was observed, so water (11.22 mL) and sulfuric acid (7.66 mL, 144 mmol) were added and the reaction mixture was heated to 100 0C for 2 h. The crude suspension was cooled to room temperature and poured into 200 mL crushed ice/water, stirred vigorously for 30 min, then filtered to afford the crude product as a white solid.
The white solid was purified by silica gel tography (0-10% ethyl acetate in hexanes), and the product ons were combined and concentrated to afford —114— ATI—25 14175V1 ethylcyclohexenyl)isoindoline-1,3-dione (3.33 g, 13.80 mmol, 77 % yield) as a white solid. MS (ESI) m/z 242.0 [M+1]+.
C. 4-Methylcyclohexenamine. To a stirring suspension of 2-(4-methylcyclohexenyl)isoindoline-1,3-dione (5.24 g, 21.72 mmol) in methanol (163 mL) and DCM (54.3 mL) was added hydrazine e (3.69 mL, 76 mmol) and the reaction was stirred for 15 h. To the crude sion was added 400 mL water, and the mixture was stirred for 10 min, then extracted with DCM (4 x 400mL). The combined organic layers were dried over sodium sulfate then concentrated to afford 4-methylcyclohexenamine (2.415 g, 21.72 mmol, 100 % yield) as a light yellow oil. MS (ESI) m/z 112.2 [M+1]+.
D. tert-Butyl 4-methylcyclohexenylcarbamate. To a stirring on of 4-methylcyclohexenamine (2.415 g, 21 .72 mmol) and TEA (3.33 mL, 23.89 mmol) in DCM (43.4 mL) at 0 CC was added di-tert-butyl-dicarbonate (7.11 g, 32.6 mmol). The resulting mixture was stirred for 3 days at room temperature. The crude reaction mixture was concentrated then purified by silica gel chromatography (0-10% ethyl acetate in hexanes). The product fractions were ed and concentrated to afford tert-butyl 4- methylcyclohexenylcarbamate (1.65 g, 7.81 mmol, 36.0 % yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 6.72 (d, J=7.42 Hz, 1H), 5.26 (br. s., 1H), 3.35 - 3.49 (m, 1H), 1.66 - 2.22 (m, 5H), 1.60 (s, 3H), 1.38 (s, 10H); MS (ESI) m/z 212.4 [M+1]+.
E. tert—Butyl 3-hydroxy—4-methylcyclohexylcarbamate. To a ng solution of tert-butyl 4-methylcyclohexenylcarbamate (1.5844 g, 7.50 mmol) in THF (94 mL) at 0 0C was added 1M borane THF complex (33.7 mL, 33.7 mmol). The solution was stirred at 0 CC for 1 h and then at room temperature for 2 h. The reaction was quenched very slowly with water (40.5 mL, 2249 mmol), diluted with ethanol (39.8 mL, 682 mmol) and basified with aqueous sodium hydroxide solution (5 N, 37.5 mL, 187 mmol). To the stirring biphasic mixture was slowly added hydrogen peroxide (38.3 mL, 375 mmol) and the ing mixture was heated to 45 0C for 20 h. The crude reaction was quenched with saturated aqueous sodium sulfite on (70 mL) and extracted with ethyl acetate (3 x 100mL). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated. The crude product was ATI—2514l75Vl d by silica gel chromatography % ethyl acetate in hexanes). The product fractions were combined and trated to afford a mixture of four isomers of tert-butyl 3-hydroxymethylcyclohexylcarbamate (trans relationship between 3-hydroxy and 4-methyl moieties) (0.9608 g, 4.19 mmol, 55.9 % yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 6.60 - 6.85 (m, 1H), 4.29 - 4.58 (m, 1H), 3.35 - 3.80 (m, 1H), 2.81 - 3.28 (m, 1H), 1.20 - 1.96 (m, 14H), 0.79 - 1.14 (m, 5H); MS (ESI) m/z 230.5 [M+1]+.
F. 5-Amin0-(trans)—2—methylcyclohexanol hydrochloride. To vigorously stirring methanol (10.47 mL) at 0 0C was added acetyl de (0.893 mL, 12.57 mmol).
The solution was stirred for 30min, then tert-butyl oxymethylcyclohexyl- carbamate (0.9608 g, 4.19 mmol) was added and stirring was continued for 22 h at room temperature. The crude reaction mixture was concentrated, and the residue was triturated with ethyl ether (2 x 50 mL) to afford 5-amino-(trans)methylcyclohexanol hydrochloride (0.694 g, 4.19 mmol, 100 % yield) as a white solid. MS (ESI) m/z 112.2 [003 17] G. 2-(trans—4-Methoxycyclohexylamin0)(methylsulfinyl)pyrimidine— -carb0xamide. To a solution of 2-((1r,4r)methoxycyclohexylamino)(methylthio)- pyrimidinecarboxamide (1.24 g, 4.18 mmol; synthesis described herein) in NMP (12.30 mL) at 0 °C was added mCPBA (0.938 g, 4.18 mmol) portion wise and the reaction mixture was stirred for 1 h, while allowing to reach room temperature slowly. The reaction mixture was diluted with water (120 mL), stirred for 10 min and the resulting solids were removed by ion. The filtrate was concentrated in vacuo at less than CC to afford 2-((1r,4r)methoxycyclohexylamino)(methylsulf1nyl)pyrimidine amide (1.307 g, 4.18 mmol, 100 % yield) as a white suspension in NMP (~10 mL).
This crude suspension was used in the next step without further purification. MS (ESI) m/z 313.5 [M+1]+.
H. 4-((1 S,3S,4S)—3-Hydr0xymethylcyclohexylamino)—2—((1r,4S)—4- methoxycyclohexylamin0)pyrimidine—5-carboxamide, 4-((1R,3R,4R)—3-hydr0xy methylcyclohexylamin0)((1r,4R)—4-meth0xycyclohexylamin0)pyrimidine carboxamide, 4-((1S,3R,4R)—3-hydroxymethylcyclohexylamin0)((1r,4S)—4- methoxycyclohexylamin0)pyrimidine—5-carboxamide, 4-((1R,3S,4S)—3-hydr0xy ATI—25 14175V1 methylcyclohexylamino)—2-((1r,4R)—4-meth0xycyclohexylamin0)pyrimidine carboxamide. To the crude on mixture of (2-((1r,4r)methoxycyclohexylamino)- 4-(methylsulfinyl)pyrimidinecarboxamide (1.31 g, 4.19 mmol) in NMP (10 mL) was added crude 5-amino-(trans)methylcyclohexanol hydrochloride (0.695 g, 4.19 mmol) as a solution in NMP (15.00 mL). To the sion was added DIEA (3.65 mL, .97 mmol) and the reaction was heated to 100 0C for 46 h. The NMP was d by ation at 70 0C, the residue was diluted with DCM and purified by silica gel tography (0-15% methanol in DCM). The t fractions were combined, concentrated, and then repurif1ed by silica gel chromatography (0-10% methanol in DCM).
The product fractions were combined and concentrated to afford a mixture of four products (two diastereomers and their corresponding enantiomers (1.665 g). NMR of mixture: 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.35 (d, J=5.45 Hz, 1H), 4.50 - 4.71 (m, 1H), 4.34 (br. s., 1H), 3.53 - 3.69 (m, 1H), 3.23 (s, 3H), 3.03 - 3.19 (m, 2H), 1.77 - 2.09 (m, 5H), 1.65 (br. s., 1H), 1.07 - 1.39 (m, 13H), 0.94 (d, J=6.49 Hz, 3H); MS (ESI) m/z 378.0 [M+1]+ The crude material was separated by chiral SFC using an AD-H column to afford 4 compounds labeled PEAK 1 to PEAK 4, with PEAK 1 being the first eluting compound and PEAK 4 being the latest eluting compound. Stereochemistry of Peaks 1 through 4 was determined by methods known to one of skill in the art.
PEAK 1: 4-((1S,3S,4S)Hydr0xymethylcyclohexylamino)—2- ((1r,4S)methoxycyclohexylamin0)pyrimidine—5-carb0xamide. 0.3482 g, 0.922 mmol, 22 % yield. 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.88 (d, J=6.64 Hz, 1H), 8.34 (br. s., 1H), 7.07 (d, J=7.03 Hz, 1H), 4.58 (d, J=5.08 Hz, 1H), 4.10 (q, J=5.08 Hz, 1H), 3.51 - 3.92 (m, 1H), 3.44 (dd, J=7.03, 5.08 Hz, 1H), 3.23 (s, 3H), 3.17 (d, J=5.47 Hz, 3H), 3.09 (br. s., 1H), 2.98 (br. s., 1H), 2.55 (d, J=7.03 Hz, 1H), 1.77 - 2.23 (m, 5H), 1.66 (d, J=12.49 Hz, 1H), 1.04 - 1.35 (m, 6H), 0.94 (d, J=5.86 Hz, 3H).
PEAK 2: 4-((1R,3R,4R)—3-hydroxymethylcyclohexylamino)—2- ((1r,4R)—4-methoxycyclohexylamin0)pyrimidine—5-carb0xamide. 0.4146 g, 1.098 mmol, 26.2 % yield. 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.88 (d, J=6.25 Hz, 1H), 8.34 (br. s., 1H), 7.07 (d, J=7.03 Hz, 1H), 4.57 (br. s., 1H), 4.11 (br. s., 1H), 3.50 - 3.67 (m, 1H), 3.23 (s, 2H), 3.17 (br. s., 2H), 3.03 - 3.13 (m, 1H), 2.94 - 3.02 (m, 1H), 2.89 (q, J=7.16 Hz, 4H), 1.60 - 2.22 (m, 4H), 0.87 - 1.37 (m, 13H).
ATI—25 14175V1 PEAK 3: 4-((1 S,3R,4R)—3-hydroxymethylcyclohexylamin0) ((1r,4R)—4-methoxycyclohexylamin0)pyrimidine—5-carb0xamide. 0.3145 g, 0.833 mmol, 19.9 % yield. 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.25 (d, J=7.42 Hz, 1H), 8.35 (br. s., 1H), 7.05 (d, J=6.64 Hz, 1H), 6.76 - 6.92 (m, 1H), 4.53 (d, J=3.90 Hz, 1H), 4.32 (br. s., 1H), 4.03 - 4.17 (m, 1H), 3.52 - 3.84 (m, 1H), 3.40 - 3.50 (m, 1H), 3.13 - 3.26 (m, 4H), 3.00 - 3.12 (m, 1H), 1.77 - 2.12 (m, 5H), 1.10 - 1.69 (m, 9H), 0.94 (d, J=6.64 Hz, 3H).
PEAK 4: 4-((1R,3S,4S)hydr0xymethylcyclohexylamin0) ((1r,4R)—4-methoxycyclohexylamin0)pyrimidine—5-carb0xamide. 0.4715 g, 1.249 mmol, 29.8 % yield. 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.46 - 9.75 (m, 1H), 8.35 (s, 1H), 4.57 (br. s., 1H), 4.35 (br. s., 1H), 3.60 - 3.79 (m, 1H), 3.23 (s, 4H), 3.00 - 3.15 (m, 1H), 2.87 - 2.99 (m, 1H), 1.75 - 2.06 (m, 5H), 1.06 - 1.73 (m, 11H), 0.94 (d, J=6.25 Hz, 3H).
Example 18: 4-((1R,3S)Hydr0xycyclohexylamino)—2-((1r,4R)—4- methoxycyclohexylamin0)pyrimidinecarboxamide /O”' N \ NH2 )L / A. 2-Cyclohexenyl—is0indole-1,3-di0ne. To a solution of 3-bromo- cyclohexene (500 g, 3.12 mmol) in DMF (2000 mL) was added potassium phtalimide (580 g, 3.12 mmol), and the reaction mixture was stirred at room temperature for 30 h.
The reaction mixture was diluted with water (1000 mL), ted with ethyl acetate (3 times), and the combined organic layers were concentrated to give 2-cyclohexenyl- ole-1,3-dione (520 g, 2.30 mol) as a gray solid.
B. rom0hydroxy-cyclohexyl)-is0indole-1,3-di0ne. 2-Cyclohex- 2-enyl-isoindole-1,3-dione (520 g, 2.30 mol) and N—bromosuccinimide (416 g, 2.35 mol) were mixed in a cosolvent of chloroform (3000 mL) and ethanol (120 mL) and stirred at room temperature for 16 h. When HPLC showed the starting material was ed, the on mixture was concentrated to give the residue, which was diluted with ATI—25 14175Vl hydrochloride solution (450 mL, 2.0 mol) and THF (2000 mL), and the reaction mixture was stirred at room temperature for 2 h. The reaction mixture was trated to give the crude product. The crude product was washed with ethanol (2 times) to give 2-(2-bromo- 3-hydroxy-cyclohexyl)-isoindole-1,3-dione (400 g, 1.23 mol) as a white solid. 1H NMR (CD301), 400 MHz): 5 ppm 7.87 (m, 4H), 4.70 (m, 1H), 4.33 (m, 1H),3.70 (m, 1H), 2.15 (m, 2H),1.88 (m, 2H), 1.53 (m, 2H).
C. 2-(3-Hydr0xy-cyclohexyl)-isoindole-1,3-dione. Tri-n-butyltin hydride (430 g, 1.48 mol) was added to a stirred solution of 2-(2-bromohydroxy-cyclohexyl)- isoindole-1,3-dione (400 g, 1.23 mol) and 2,2'-azobis(2-methylpropionitrile) (20 g) in toluene (4000 mL) and ol (400 mL) and the reaction e was stirred at 80 CC for 64 h. The reaction mixture was concentrated to give the crude product. The crude product was washed with eum ether and ethyl acetate (1 :1) to give 2-(3-hydroxy- cyclohexyl)-isoindole-1,3-dione (240 g, 0.98 mol) as a white solid. 1H NMR (CDgOD, 400 MHz): 5 ppm 7.82 (m, 4H), 4.16 (m, 1H), 3.63 (m, 1H), 2.13 (m, 2H), 1.98 (d, 2H), 1.89 (m, 1H),1.68 (t, 1H), 1.46 (m, 1H),1.31(m, 1H).
D. (1S,3R)(3-Hydroxy-cyclohexyl)-is0indole-1,3-di0ne and (1R,3S) acetic acid 3-(1,3-di0x0-1,3-dihydr0-isoind0]—2-yl)—cyclohexyl ester. A solution of 2-(3- hydroxy-cyclohexyl)-isoindole-1,3-dione (240 g, 0.98 mol) and Lipase B (87 g) in THF (3000 mL) was stirred at room temperature under nitrogen for 1~2 h. Then acetic acid vinyl ester (252 g, 2.88 mol) was added to the solution, and the reaction mixture was stirred at rt for 3~5 h. Then the reaction mixture was filtered, and the filtrate was concentrated to give crude product. The crude product was purified on silica gel (eluting with petroleum ether/ethyl acetate =10: 1) to give (1S,3R)(3-hydroxy-cyclohexyl)- isoindole-1,3-dione (Rf:0.6, petroleum ethyl e = 3/1, 90 g, 0.37 mol, yield: 39%) as a white solid. 1H NMR (CDgOD, 400 MHz): 5 ppm 7.82 (m, 4H), 4.16 (m, 1H), 3.63 (m, 1H), 2.13 (m, 2H), 1.98 (m, 2H), 1.89 (m, 1H), 1.68 (m, 1H), 1.46 (m, 1H), 1.31(m, 1H).
)Acetic acid 3-(l,3-dioxo-l,3-dihydro-isoindolyl)-cyclohexyl ester 2, petroleum ether/ethyl acetate = 3/ 1, 110 g, 0.38 mol, yield: 39%) as a white solid. 1H NMR (CD301), 400 MHz): 5 ppm 7.83 (m, 4H), 4.78 (m, 1H), 4.23 (m, 1H), 2.33 (m, 2H), 2.13 (m, 2H), 2.07 (m, 5H), 1.95 (m, 1H), 1.74 (m, 1H), 1.46 (m, 1H).
ATI—25 14175v1 E. (1S,3R)Amin0-cyclohexanol. To a solution of (1S,3R)(3- hydroxy-cyclohexyl)-isoindole-1,3-dione (90 g, 0.37 mol) in ethanol (1500 mL) was added hydrazine (90 g, 1.8 mol), and the reaction solution was stirred at 100 CC for 3h. The reaction solution was filtered, and the filtrate was concentrated to give (1S,3R)amino- cyclohexanol (25 g, 0.21 mmol) as a yellow solid. 1H NMR (CDgOD, 400 MHz) 5 ppm 3.54 (m, 1H), 2.65 (m, 1H), 2.12 (m, 1H), 1.92 (m, 1H), 1.89 (m, 2H), 1.32 (m, 1H), 1.06 (m, 2H).
F. Ethyl 4-((1R,3S)—3-hydr0xycyclohexylamin0)(methylthi0)- dine—S-carboxylate. To a stirring solution of ethyl 4-chloro(methylthio)- pyrimidinecarboxylate (4.38 g, 18.82 mmol) and (1S,3R)aminocyclohexanol (2.276 g, 19.76 mmol) in ethanol (75 ml) was added DIEA (4.93 ml, 28.2 mmol). The on mixture was heated to 60 CC for 2h, was removed from heat and concentrated, then d by silica gel chromatography (20-100% ethyl acetate/hexane). The desired product fractions were ed and trated to afford ethyl 4-((1R,3S) hydroxycyclohexylamino)(methylthio)pyrimidinecarboxylate (5 g, 16.06 mmol, 85 % yield) as a white foam. MS (ESI) m/z 312.1 [M+1]+.
G. 4-((1R,3S)—3-hydr0xycyclohexylamin0)(methylthi0)pyrimidine carboxylic acid. To a stirring solution of ethyl 4-((1R,3S)—3-hydroxycyclohexyl-amino)- hylthio)pyrimidinecarboxylate (5 g, 16.06 mmol) in ethanol (50 ml) was added 2M sodium hydroxide (20 ml, 40.0 mmol). Stirring was continued at room temperature for 1h. The reaction e was neutralized by addition of saturated citric acid. The resulting precipitate was filtered and dried to afford 4-((1R,3S)—3-hydroxycyclohexylamino )(methylthio)pyrimidinecarboxylic acid (4.5 g, 15.88 mmol, 99 % yield) as a white solid. 1H NMR (400 MHz, 6) 8 ppm 13.21 (br. s., 1 H), 8.44 - 8.54 (m, 2 H), 4.74 (d, J=3.90 Hz, 1 H), 3.97 - 4.12 (m, 1 H), 3.56 (d, J=3.12 Hz, 1 H), 2.46 (s, 3 H), 2.05 - 2.16 (m, 1 H), 1.84 (d, J=10.15 Hz, 1 H), 1.65 - 1.80 (m, 2 H), 1.11 - 1.36 (m, 4 H). MS (ESI) m/z 284.1 [M+1]+.
H. 4-((1R,3S)Hydr0xycyclohexylamino)—2-(methylthio)pyrimidine—5- carboxamide. 4-((1R,3 S)—3-hydroxycyclohexylamino)(methylthio)pyrimidine-5 - carboxylic acid (4.5 g, 15.88 mmol) and HATU (9.06 g, 23.82 mmol) were dissolved in DMF (75 ml) and allowed to stir for 5 min at room temperature before adding ammonium ATI—25 14175v1 de (4.25 g, 79 mmol) and DIEA (13.87 ml, 79 mmol). The reaction turned yellow upon addition of the base. The reaction was allowed to stir at room ature for 1 h then partitioned between water and ethyl acetate. The c layer was washed once with brine before drying over sodium sulfate, filtering, and concentrating to afford 4-((1R,3S)- 3-hydroxycyclohexylamino)(methylthio)pyrimidinecarboxamide (4.19 g, 14.84 mmol, 93 % yield) as an off-white solid. MS (ESI) m/z 283.2 .
I. 4-((1R,3S)Hydroxycyclohexylamin0)(methylsulfonyl)— pyrimidine—5-carb0xamide. To a stirring suspension of 4-((1R,3S)—3- hydroxycyclohexylamino)(methylthio)pyrimidinecarboxamide (302 mg, 1.070 mmol) in DCM (10 ml) and acetone (10 ml) was added 3-chloroperoxybenzoic acid (479 mg, 2.139 mmol). The reaction was stirred at room temperature for 1.5 h. The crude reaction mixture was quenched by addition of 10 mL of 10% sodium thiosulfate solution, stirred for 5 minutes, then partitioned between ethyl acetate and water. The organic layer was washed with saturated sodium bicarbonate and brine. The ed aqueous layers were extracted with ethyl acetate (3x50mL). The combined organic layers were dried over sodium sulfate, filtered, and trated to afford 4-((1R,3S)—3-hydroxycyclohexyl- amino)(methylsulfonyl)pyrimidinecarboxamide (259 mg, 0.824 mmol, 77 % yield) as a white solid. MS (ESI) m/z 315.2 [M+1]+.
] J. 4-((1R,3S)Hydr0xycyclohexylamin0)((1r,4R)—4-meth0xycyclo— hexylamin0)pyrimidine—5-carb0xamide. To a stirring solution of 4-((1R,3S)hydroxy- cyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (0.178 g, 0.566 mmol) and (1r,4r)methoxycyclohexanamine (0.146 g, 1.132 mmol) in DMSO (1 . 132 ml) was added DIEA (0.395 ml, 2.265 mmol). The reaction mixture was stirred at 100 CC for 17 h.
The crude reaction mixture was concentrated and purified by silica gel chromatography (0-15% methanol/ethyl acetate). The product fractions were combined and concentrated to afford 4-((1R,3 S)hydroxycyclohexylamino)((1r,4R)—4-methoxycyclohexylamino)- pyrimidinecarboxamide (0.08 g, 0.220 mmol, 38.9 % yield) as a light tan solid. 1H NMR (CDgOD, 400 MHz): 8 ppm 8.29 (s, 1H), 3.99-4.04 (m, 1H), 3.76 (m, 1H), 3.50 - 3.63 (m, 1H), 3.37 (s, 3H), 3.24 (m, 1H), 3.23 (m, 1H), 1.92-2.12 (m, 6H), .87 (m, 1H), 1.19-1.41(m, 8H). MS (ESI) m/z 364.6 [M+1]+.
ATI—25 14175V1 Example 19: 4-((1R,3S)—3-Hydr0xycycloheptylamino)—2-((1r,4R)—4- methoxycyclohexylamin0)pyrimidine—5-carb0nitrile /o,,, CN )L / N N NH H OOH A. 4-Chlor0((1R,4R)—4-methoxycyclohexylamin0)-pyrimidine—5- carbonitrile. (1r,4r)Methoxycyclohexanamine (6.37 g, 49.3 mmol), 2,4-dichloropyrimidinecarbonitrile (6 g, 34.5 mmol) and DIEA (9.03 mL, 51.7 mmol) were mixed in THF (180 mL) at -10 CC (salt ice bath) and stirred at the same temperature for 5 h. The reaction mixture was stirred further overnight while the temperature went up to room temperature slowly. Analysis of the reaction mixture by LC/MS showed formation of the ts with the ratio of about 3 :7. The reaction mixture was concentrated under reduced re, diluted with water (100 mL) and extracted with ethyl acetate (2x). The combined organic layers were washed with water and brine and dried over magnesium sulfate. The drying agent was removed by filtration and the solution was concentrated.
The ing crude mixture was purified by flash chromatography (0-20% ethyl acetate in hexanes) to give 4-chloro((1R,4R)methoxycyclohexylamino)pyrimidine carbonitrile (5.79 g, 21.71 mmol, 62.9 % yield; 1H NMR (DMSO-d6 ,400 MHz): 5 ppm 8.74 (d, J=8.2 Hz, 1 H), 8.60 - 8.72 (m, 1 H), 3.58 - 3.80 (m, 1 H), 3.18 (d, J=2.0 Hz, 3 H), 2.97 - 3.12 (m, 1 H), 1.96 (d, J=10.9 Hz, 2 H), 1.83 (d, J=10.5 Hz, 2 H), 1.21 - 1.39 (m, 2 H), 1.06 - 1.22 (m, 2 H); MS (ES) m/z 267.2 ) as a white solid and 2-chloro ((1R,4R)—4-methoxycyclohexylamino)pyrimidinecarbonitrile (2.5 g, 9.37 mmol, 27.2 % yield; 1H NMR (DMSO-d6 ,400 MHz): 5 ppm 8.50 (s, 1 H), 8.31 (d, J=7.8 Hz, 1 H), 3.90 (dtd, J=11.6, 7.6, 4.1 Hz, 1 H), 3.19 (s, 3 H), 2.98 - 3.10 (m, 1 H), 1.98 (d, J=10.5 Hz, 2 H), 1.77 (d, J=11.3 Hz, 2 H), 1.36 - 1.54 (m, 2 H), 1.05 - 1.22 (m, 2 H). MS (ES) m/z 267.1 [M+1]+) as a white solid.
The regiochemistry of the separated s was confirmed in the following experiments: ] 4-Chloro((1R,4R)methoxycyclohexylamino)pyrimidinecarbonitrile (50 mg, 0.187 mmol), diacetoxypalladium (42.1 mg, 0.187 mmol) and ammonium ATI—25 14175v1 formate (59.1 mg, 0.937 mmol) in methanol (1 mL) was stirred at 70 CC for 2 h. The on mixture was cooled down to room temperature and was filtered through a pad of celite. The resulting solution was trated and subjected to flash chromatography (0-15% methanol in DCM) to give 2-((1R,4R)—4-methoxycyclohexyl-amino)pyrimidine carbonitrile as a white solid. 1H NMR (DMSO-d6 ,400 MHz; measured at room ature): 8 ppm 8.66 (d, J=2.7 Hz, 1 H), 8.59 (d, J=3.1 Hz, 1 H), 8.22 (d, J=8.2 Hz, 1 H), 3.62 - 3.77 (m, 1 H), 3.18 (s, 3 H), 2.99 - 3.12 (m, 1 H), 1.90 - 2.02 (m, 2 H), 1.84 (d, J=10.5 Hz, 2 H), 1.21 - 1.37 (m, 2 H), 1.06 - 1.21 (m, 2 H). 1H NMR (DMSO-d6 ,400 MHz; measured at 80 oC): 8 ppm 8.57 (br. s., 1 H), 7.91 (d, J=7.0 Hz, 1 H), 3.68 - 3.85 (m, 1 H), 3.05 - 3.16 (m, 1 H), 3.02 (s, 3 H), 1.92 - 2.05 (m, 2 H), 1.87 (d, J=10.2 Hz, 2 H), 1.26 - 1.41 (m, 2 H), 1.11 - 1.26 (m, 2 H).
The same method described above was d to 2-chloro((1R,4R) methoxycyclohexylamino)pyrimidinecarbonitrile to give 4-((1R,4R)methoxy- exylamino)pyrimidinecarbonitrile as a white solid. 1H NMR (DMSO-dg, 400 MHz; measured at room temperature): 8 ppm 8.56 (d, J=15.2 Hz, 1 H), 7.74 - 7.91 (m, 1 H), 3.86 - 4.11 (m, 2 H), 3.19 (s, 3 H), 3.12 (d, J=5.1 Hz, 1 H), 1.92 - 2.04 (m, 2 H), 1.78 (d, J=10.5 Hz, 2 H), 1.35 - 1.52 (m, 2 H), 1.05 - 1.18 (m, 2 H). 1H NMR (DMSO-d6, 400 MHz; measured at 80 oC): 8 ppm 8.53 (d, J=28.1 Hz, 1 H), 7.47 (br. s., 1 H), 3.93 - 4.08 (m, 1 H), 3.16 (d, J=5.5 Hz, 1 H), 1.91 - 2.07 (m, 2 H), 1.84 (d, J=10.5 Hz, 2 H), 1.37 - 1.54 (m, 2 H), 1.10 - 1.21 (m, 2 H).
B. (1 S,3R)—3-Amin0cycloheptanol hydrochloride. To vigorously stirring methanol (60 mL) at 0 CC was added acetyl chloride (5.11 mL, 72.0 mmol) and the resulting mixture was allowed to stir for 30 min. tert-Butyl (1R,3S)—3-hydroxycycloheptylcarbamate (5500 mg, 23.98 mmol; synthesis described herein) was added and the resulting mixture was then stirred overnight at room temperature. The reaction mixture was trated under reduced pressure to an oil that was triturated with ethyl ether (100 mL) overnight. The solids were filtered, rinsed with diethyl ether, and dried under reduced pressure for a few hours to afford (1 S,3R)aminocycloheptanol hydrochloride (3780 mg, 22.82 mmol, 95 % yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.85 (br. s., 3 H), 4.69 (d, J=7.42 Hz, 1 H), 3.64 (t, J=8.79 Hz, 1 H), ATI—25 14175v1 3.07 - 3.21 (m, 1 H), 2.05 (d, J=12.89 Hz, 1 H), 1.69 - 1.94 (m, 2 H), 1.28 - 1.68 (m, 7 H).
MS (ESI) m/z 130.1 [M+1]+.
C. 4-((1R,3S)—3-Hydroxycycloheptylamino)—2-((11,4R) methoxycyclohexylamin0)pyrimidinecarb0nitrile. To a stirring solution of 4-chloro- 2-((1r,4r)methoxycyclohexylamino)pyrimidine-5 -carbonitrile (200 mg, 0.750 mmol) and (1S,3R)aminocycloheptanol hydrochloride (186 mg, 1.125 11111101) in DMF (4 mL) was added DIEA (0.458 mL, 2.62 11111101). The resulting mixture was stirred at 70 CC for 4 h and then allowed to cool to ambient ature overnight. DMF was removed under reduced re and the remaining residue was purified using silica gel chromatography (0-50% ethyl acetate + 10% 7 N ammonia in ol in hexanes) to afford 4-((1R,3S) hydroxycycloheptylamino)((1r,4R)methoxycyclohexylamino)pyrimidine carbonitrile (210 mg, 0.584 mmol, 78 % yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.07 - 8.23 (m, 1 H), 7.06 - 7.61 (m, 2 H), 4.49 - 4.63 (m, 1 H), 4.00 - 4.27 (m, 1 H), 3.55 - 3.82 (m, 2 H), 3.23 (s, 3 H), 3.00 - 3.16 (m, 1 H), 1.07 - 2.08 (m, 18 H). MS (ESI) m/z 360.5 [M+1]+.
Example 20: 2-((1r,4R)—4-Eth0xycyclohexylamin0)((1R,3S)—3-hydroxy methylcyclohexylamin0)pyrimidine—5-carb0nitrile ”(l “XAxN N N H H @011 A. (11, 4r)—4—(Tritylamin0)cyclohexanol. To a cooled (0 0C) solution of (11, 4r)aminocyclohexanol (10 g, 87 mmol) in DCM (250 mL) and TEA (11.2 g, 112 mmol) was added trityl chloride (24.2 g, 87 mmol). The resulting mixture was stirred at room temperature ght. Saturated aqueous sodium bicarbonate on was added to quench the reaction and the aqueous layer was extracted with ethyl acetate. The combined extracts were washed with brine, dried over anhydrous sodium sulfate. Filtration and concentration under vacuum gave crude product, which was purified by silica gel column chromatography (50 % ethyl e in DCM) to afford the title compound (24.8 g, 70 mmol, 80 % yield) as a white solid. MS (ESI) m/z 243.2 [M - ocyclohexanol]+. —124— ATI—25 14175v1 B. (1r,4r)—4-Ethoxy-N-tritylcyclohexanamine. To a cooled (0 0C) solution of )(tritylamino)cyclohexanol (15.0 g, 42 mmol) in DMF (100 mL) was added sodium hydride (3.4 g, 84 mmol, 60 % in mineral oil) and ethyl iodide (7.2 g, 46 mmol) at 0 0C. The resulting mixture was stirred at room temperature overnight under nitrogen atmosphere. Saturated aqueous ammonia chloride solution was added to quench the reaction and the aqueous layer was extracted with ethyl e. The combined ts were washed with brine and dried over anhydrous sodium sulfate. Filtration and concentration under vacuum gave crude product, which was purified by silica gel column chromatography (5 % ethyl acetate in petroleum ether) to afford the title compound (11.9 g, 31 mmol, 79 % yield) as a white solid. 1H NMR (400 MHz, CDClg) 5 ppm 7.56-7.54 (m, 6H), .20 (m, 6H), 7.18-7.14 (m, 3H), 3.42-3.38 (q, 2H), 3.09 - 3.04 (m, 1H), 2.25-2.21 (m, 1H), 1.80-1.78 (m, 2H), 1.38 (s, 1H), 1.27-1.25 (m, 2H), 1.10 (t, J: 7.2 Hz, 3H), 1.01-0.88 (m, 4H); MS (ESI) m/z 243.2 [M - 4-ethoxycyclohexanamine]+.
C. (1r,4r)—4-Eth0xycyclohexanamine. To a cooled (0 0C) solution of (1r, ethoxy-N-tritylcyclohexanamine (13.1 g, 34 mmol) in DCM (10 mL) was added trifluoroacetic acid (18 mL) at 0 0C. The resulting mixture was dark red. Triethylsilane (5 mL) was added until the resulting mixture was colourless. The reaction was stirred at 0 0C for additional 15 minutes. After removal of all volatiles in vacuo, the residue was further dried under high vacuum for 2 h to give the crude product as a white solid. The crude product was dissolved in ethyl acetate and aqueous hydrochloride solution (150 mL, 0.25 mol/L). The c layer was removed and the s layer was washed with ethyl e twice. tration of the aqueous layer under high vacuum gave the crude hydrochloride salt of the desired amine (6.1 g, 34 mmol, 100 % yield) as a white solid, which was used for the next step without further purification. MS (ESI) m/z 144.0 [M+H]+.
D. 4-Chlor0((1r,4r)—4-ethoxycyclohexylamino)pyrimidine—5- carbonitrile. To a cooled (-60 0C) solution of (1r,4r)ethoxycyclohexanamine (6.82 g, 37.9 mmol) and 2,4-dichloropyrimidinecarbonitrile (6 g, 34.5 mmol) in THF (115 mL) was added DIEA (15.02 mL, 86 mmol) dropwise. The resulting mixture was stirred at -60 0C for 1 h and then d to warm to room temperature overnight. The resulting mixture was concentrated and the residue was purified by silica gel column ATI—2514175v1 chromatography (14 % ethyl acetate in petroleum ether). The product fractions were combined and concentrated to afford 2-chloro((1r,4r)ethoxycyclohexylamino)- pyrimidinecarbonitrile (3.33 g, 11.86 mmol, 34.4 % yield) and 4-chloro((1r,4r) ethoxycyclohexylamino)pyrimidinecarbonitrile (5.5 g, 19.59 mmol, 56.8 % yield; 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.62 - 8.87 (m, 2 H), 3.61 - 3.86 (m, 1 H), 3.44 (dd, J=7.03, 2.34 Hz, 2 H), 3.12 - 3.26 (m, 1 H), 1.75 - 2.15 (m, 4 H), 1.13 - 1.44 (m, 4 H), 1.08 (td, J=6.93, 1.76 Hz, 3 H); MS (ESI) m/z 281.1 [M+1]+) as white solids. The regiochemistry of the major isomer was confirmed by comparison with the proton spectra of 4-chloro((1r,4r)methoxycyclohexylamino)pyrimidinecarbonitrile (synthesis described herein).
E. (1 S,3R)(Dibenzylamin0)cyclohexanol. To a suspension of (1S,3R)- 3-aminocyclohexanol (5 g, 43.4 mmol; prepared as described in Tetrahedron: try (2004) 056) and sodium bicarbonate (12.03 g,143 mmol) in ethanol (100 mL) was added (chloromethyl)benzene (15.01 mL, 130 mmol) at room temperature. The on mixture was heated at 75 CC overnight. Upon completion of the reaction as indicated by LCMS and TLC the reaction mixture was filtered and the e ammMmmd7mem$mwwwflmnmwdwdeCM(%0mLflmdwwmflwfihmmmm sodium hydroxide solution (1N, 2 x 100mL) and brine (2 x 100 mL). The combined organic layers were dried over anhydrous magnesium sulfate, concentrated, and purified by silica gel chromatography % ethyl e in hexanes) to yield (1S,3R)—3- (dibenzylamino)cyclohexanol (11.70 g, 91%) as a thick yellow oil . 1H NMR (400 MHz, DMSO-dg) 5 ppm 7.26 - 7.37 (m, 8H), 7.17 - 7.22 (m, 2H), 4.56 (d, J: 4.30 Hz, 1H), 3.57 (s, 4H), 3.16 - 3.26 (m, 1H), 2.41 (tt, J= 3.17, 11.86 Hz, 1H), 1.99 - 2.06 (m, 1H), 1.72 (d, J: 8.20 Hz, 2H), 1.63 - 1.69 (m, 1H), 1.18 - 1.28 (m, 2H), 0.98 (t, 2H); MS (ESI) m/z 296.4 [M+1]+.
] F. (R)—3-(Dibenzylamin0)cyclohexanone. Oxalyl chloride (3.81 mL, 43.6 mmol) was dissolved in dry DCM (150 mL) and cooled to -78 oC. DMSO (6.75 mL, 95.0 mmol) in dry DCM (20 mL) was added dropwise to the reaction mixture and the reaction was stirred for 15 minutes at -78 CC. Next, (1S,3R)(dibenzylamino)— cyclohexanol (11.7 g, 39.6 mmol) in dry DCM (100 mL) was added dropwise using an addition fiJnnel and the reaction was stirred at -78 CC for 15 minutes. TEA (27.6 mL, ATI—25 14175V1 2012/034349 198 mmol) was then added and the reaction was stirred at -78 CC for 1 h. The dry-ice bath was removed and the on mixture was allowed to warm to room temperature and stir overnight. The reaction mixture was washed with brine (5 x 500 mL), the organic layer was separated, dried over magnesium sulfate and concentrated to yield (R)(dibenzylamino)cyclohexanone (10.2 g, 88%) as a tan solid. 1H NMR (400 MHz, 6) 8 ppm 7.26 - 7.39 (m, 8H), 7.17 - 7.25 (m, 2H), 3.56 - 3.70 (m, 4H), 2.71 - 2.81 (m, 1H), 2.60 - 2.69 (m, 1H), 2.25 - 2.42 (m, 2H), 2.04 - 2.11 (m, 1H), 1.88 - 1.99 (m, 2H), 1.69 - 1.82 (m, 1H), 1.18 - 1.33 (m, 1H); MS (ESI) m/z 294.4 [M+1]+.
G. (3R,5R)—N,N-Dibenzyl—l-oxaspiro[2.5]octanamine and )— N,N-dibenzyl—1-0xaspir0[2.5]octan-S-amine. To a yellow on of (R)(dibenzylamino)cyclohexanone (10.2 g, 34.8mmol) and trimethylsulfonium iodide (14.90 g, 73.0 mmol) in dry DMSO (260 mL) was added sodium tert—butoxide (6.68 g, 69.5 mmol) n wise at room temperature under a nitrogen atmosphere . The reaction mixture was stirred at room temperature overnight under nitrogen. Upon completion of the reaction as indicated by LCMS and TLC the reaction was quenched with 1 L of water and extracted with ethyl acetate (2 x 800 mL). The organic phase was dried over sodium sulfate, concentrated and dried to yield a mixture of (3R,5R)-N,N—dibenzyl ro[2.5]octanamine and (3 S,5R)-N,N—dibenzyloxaspiro [2 . 5 ]octanamine (10.6g, 99%) as a yellow solid. The mixture was used as is in the next step without further purification. MS (ESI) m/z 308.4 [M+1]+.
H. (1R,3R)(Dibenzylamin0)—l-methylcyclohexanol and (1S,3R)—3- (dibenzylamino)—l-methylcyclohexanol. To a mixture of solid lithium aluminum hydride (95% LAH, 3.47 g, 87 mmol) in THF (230 mL) was added very slowly a clear pale yellow solution of the above described mixture of (3R,5R)—N,N—dibenzyloxaspiro[2.5]octan amine and (3S,5R)—N,N—dibenzyloxaspiro[2.5]octanamine (10.69 g, 34.8 mmol) in THF (230 mL) at 0 CC under nitrogen. The reaction mixture was brought to room temperature and then heated at 65 CC overnight under nitrogen. Upon completion of the reaction as indicated by LCMS and TLC the reaction mixture was transferred to a round- bottom flask and then diluted with THF (100 mL), cooled to 0 CC and while stirring a saturated on of aqueous sodium sulfate was added drop wise until the reaction stopped ng. The mixture was stirred ght at room temperature and then was ATI—25 14175v1 filtered through Celite in a coarse fritted funnel into a round-bottom flask. The filter cake was washed thoroughly with THF and the colorless filtrate was trated under reduced pressure. The ing crude mixture was purified by silica gel tography (0%-60% ethyl acetate in hexanes) to yield the faster eluting isomer (1R,3R) (dibenzylamino)methylcyclohexanol (5.5 g, 50%; 1H NMR (400 MHz, DMSO-dg) 8 ppm 7.25 - 7.36 (m, 8H), 7.15 - 7.21 (m, 2H), 3.85 (s, 1H), 3.55 (s, 4H), 2.86 (tt, J= 3.37, 12.06 Hz, 1H), 1.70 - 1.81 (m, 2H), 1.38 - 1.49 (m, 3H), 1.32 (t, J: 12.30 Hz, 1H), 1.15 - 1.27 (m, 2H), 1.12 (s, 3H); MS (ESI) m/z 310.4 [M+1]+) as a white solid, and then the slower eluting isomer (1 S,3R)(dibenzylamino)methylcyclohexanol (5.1 g, 47%; 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.26 - 7.36 (m, 8H), 7.16 - 7.23 (m, 2H), 4.38 (s, 1H), 3.49 - 3.63 (m, 4H), 2.46 (d, J: 8.98 Hz, 1H), 1.67 - 1.80 (m, 2H), 1.59 (d, J: 9.76 Hz, 1H), 1.33 - 1.47 (m, 2H), 1.17 - 1.30 (m, 2H), 0.98 - 1.13 (m, 1H), 0.89 (s, 3H); MS (ESI) m/z 310.4 [M+1]+) as a thick yellow oil. A COSY NMR experiment and the 1H NMR data reported above were used to verify the regiochemistry of the amino alcohols. For the slower eluting isomer an NOE (via NOESY ment) was observed between the protons on the amino group cyclohexyl-carbon and the methyl group carbon.
This verifies a cis relationship n these two onal groups (a trans relationship, lacking this spacial proximity, would not show such an effect), which in turn confirms this isomer to be (1 S,3R)(dibenzylamino)methylcyclohexanol.
I. (1 S,3R)—3-Amin0-l-methylcyclohexanol. A solution of (1S,3R) (dibenzylamino)methylcyclohexanol (5.0 g, 16.16 mmol) in ethanol (150 mL) was treated with palladium hydroxide on carbon and stirred under a balloon filled with hydrogen gas overnight. Upon completion of the on as indicated by LCMS the reaction mixture was filtered h a pad of celite and the filtrate concentrated to yield (1R,3R)aminomethylcyclohexanol (1.3g, 62%) as a thick yellow oil. 1H NMR (400 MHz, DMSO-dg) 5 ppm 2.87 (br. s., 1H), 1.64 - 1.74 (m, 1H), 1.47 - 1.59 (m, 2H), 1.21 - 1.40 (m, 4H), 1.11 - 1.20 (m, 1H), 1.02 (s, 3H); MS (ESI) m/z 130.2 [M+1]+.
J. 2-((1r,4R)—4-Eth0xycyclohexylamino)—4—((1R,3S)—3-hydr0xy methylcyclohexylamin0)pyrimidinecarb0nitrile. 4-Chloro((1r,4r) ethoxycyclohexylamino)pyrimidinecarbonitrile (350 mg, 1.247 mmol), (1S,3R) aminomethylcyclohexanol (242 mg, 1.870 mmol), and DIEA (0.327 mL, 1.870 mmol) ATI—25 14175v1 2012/034349 were dissolved in DMF (3.5 mL) in a screw-capped vial. The reaction mixture was stirred at 70 CC for 4 h. After completion of the reaction as indicated by LCMS and TLC the reaction mixture was condensed and the resulting residue was purified by silica gel tography using a gradient of 0-100% ethyl e in hexanes. The desired product fractions were combined and concentrated to give 2-((1r,4R)ethoxycyclohexylamino)- 4-((1R,3S)—3-hydroxymethylcyclohexylamino)pyrimidinecarbonitrile (0.356 g, 76 % yield, 98.0% pure) as a white solid. 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.09 - 8.24 (m, 1H), 7.28 - 7.63 (m, 2H), 4.71 - 4.90 (m, 1H), 4.21 (d, J: 3.51 Hz, 1H), 3.57 - 3.78 (m, 1H), 3.43 (q, J: 7.03 Hz, 2H), 3.11 - 3.23 (m, 1H), 1.78 - 2.02 (m, 4H), 1.48 - 1.74 (m, 5H), 1.16 - 1.47 (m, 6H), 1.11 - 1.16 (m, 4H), 1.08 (t, 3H); MS (ESI) m/z 374.5 [M+1]+.
Example 21: 2-((1r,4R)—4-Eth0xycyclohexylamin0)—4-((1R,3S) hydroxycyclohexylamin0)pyrimidine—5-carb0nitrile \/O/,'(1QCN N N NH A. 2-Chlor0((1R,3S)hydroxycyclohexylamino)pyrimidine—5- carbonitrile and 4-chlor0((1R,3S)hydr0xycyclohexylamin0)pyrimidine carbonitrile. 2,4-Dichloropyrimidinecarbonitrile (900 mg, 5.17 mmol) in anhydrous ethanol (5 mL) and (1R,3S)—3-amino-cyclohexanol (624 mg, 5.43 mmol; prepared as described in edron: Asymmetry 15 (2004) 2051—2056) in anhydrous ethanol (5 mL) were mixed at -60 OC and then DIEA (1.0 g, 7.75 mmol) was added dropwise. The mixture was stirred at -60 CC for 1 h and then at room temperature ght. Volatile fractions were removed and the residue was purified on silica gel ng with 9.1%-33% ethyl acetate in petroleum ether) to give 4-chloro((1R,3S)hydroxycyclohexylamino)- pyrimidinecarbonitrile (600 mg, 2.38 mmol, yield 46%) as a white solid and 2-chloro ((1R,3S)hydroxycyclohexylamino)pyrimidinecarbonitrile (270 mg, 1.07 mmol, yield %; 1H NMR (400 MHz, 6) 5 ppm 8.55 (s, 1 H), 8.46 (d, J=7.81 Hz, 1 H), 4.83 (d, J=3.90 Hz, 1 H), 3.95 - 4.11 (m, 1 H), 3.45 - 3.61 (m, 1 H), 1.93 (d, J=11.71 Hz, 1 H), ATI—25 14175v1 1.60 - 1.84 (m, 3 H), 1.03 - 1.50 (m, 4 H); MS (ESI) m/z 253.2 ) as a white solid. mmmydflmmmmmgmwmflwwmmmmwnmmpBMbw B. (1S,3R)(5-(Amin0methyl)pyrimidinylamin0)cyclohexanol. To a mixture of 2-chloro((1R,3 S)—3-hydroxycyclohexyl-amino)pyrimidine-5 -carbonitrile (70 mg, 0.277 mmol) and ammonium hydroxide (500 ul, 12.84 mmol) in ethanol (4 mL) was added Raney nickel (16.26 mg, 0.277 mmol). The resulting mixture was evacuated and then stirred under a n of hydrogen at atmospheric pressure and t temperature overnight. Ethanol was removed in vacuo and the remaining residue was d using reverse-phased preparative HPLC (5-80% acetonitrile + 0.1% TFA in water + 0.1% TFA, over 30 min). Fractions containing product were concentrated under reduced pressure. The resulting residue was redissolved in methanol (5 mL), passed over a Varian StratoSpheres HCO3 resin SPE tube for TFA l (0.9 mrnol bicarbonate equiv.), and then concentrated under reduced pressure to afford the title compound as an oil that was used for NMR purposes only. The regiochemistry was confirmed by the presence of two ct aromatic peaks in the 1H NMR spectrum and the observation of an NOE signal between the C5 methylene hydrogens and only one of these aromatic peaks in the NOESY spectrum. 1H NMR (400 MHz, DMSO-dg) 8 ppm 8.32 (s, 1 H), 7.92 (s, 1 H), 7.32 (d, J=7.48 Hz, 1 H), 4.67 (d, J=3.87 Hz, 1 H), 3.88 - 4.01 (m, 1 H), 3.59 (s, 2 H), 3.49 (d, J=3.61 Hz, 1 H), 2.06 - 2.16 (m, 1 H), 1.99 (br. s., 2 H), 1.81 (t, J=13.28 Hz, 2 H), 1.69 (dt, J=13.35, 3.51 Hz, 1 H), 1.01 - 1.35 (m, 4 H). MS (ESI) m/z 223.5 [M+1]+.
C. 2-((1r,4R)—4-Eth0xycyclohexylamin0)—4-((1R,3S) hydroxycyclohexylamin0)pyrimidine—5-carbonitrile. A mixture of 2-chloro((1R,3S)- 3-hydroxycyclohexylamino)pyrimidinecarbonitrile (270 mg, 1.07 mmol), (1r,4r) ethoxycyclohexanamine hydrochloride (289 mg, 1.60 mmol; synthesis described herein) and cesium carbonate (698 mg, 2.14 mmol) in anhydrous nol (10 mL) was stirred at 80 CC for 1 day and then at 120 CC for 5 h. The mixture was extracted between water and DCM. The c layer was combined, concentrated and purified on silica gel (eluting with 9.1%-33% ethyl acetate in petroleum ether) to give 2-((1r,4R)ethoxycyclohexyl- amino)((1R,3 S)—3-hydroxycyclohexylamino)pyrimidine-5 -carbonitrile (202 mg, 0.56 mmol, yield 52%) as a white . 1H NMR (400 MHz, DMSO-d6): 5 ppm 8.11 (s, 1H), 7.10 (s, 1H), 6.95 (s, 1H), 4.51 (d, J=3.6, 1H), 4.05 (s, 1H), 3.67 (s, 1H), 3.58-3.55 ATI—25 14175v1 (m,1H), 3.47 (q, J=6.8, 2H), 3.24—3.19 (m, 1H), 2.00—1.90 (m, 5H), 1.73 (s, 3H), 1.47—1.25 (m, 8H), 1.10 (t, J=7.2, 3H); MS(ESI): m/Z 359.9 [M+1]+. e 22: 2-(Cyclohexylamin0)—4-((1R,3S)—3-hydr0xycyclohexylamin0)— pyrimidinecarb0nitrile (1 “i”JL / N N NH A. 2-(Cyclohexylamin0)—4-((1R,3S)—3-hydr0xycyclohexyl- amin0)pyrimidinecarb0nitrile. A solution of cyclohexanamine (0.181 mL, 1.583 mmol) in DMSO (5 mL), 2-chloro((1R,3S)hydroxycyclohexylamino)- pyrimidinecarbonitrile (0.200 g, 0.791 mmol; synthesis described herein) and DIEA (0.276 mL, 1.583 mmol) was heated at 70 °C overnight. After completion of reaction as indicated by LCMS and TLC the reaction mixture was allowed to cool to room ature and trated. The resulting crude was purified by silica gel chromatography using a gradient of 0% - 70% ethyl acetate (containing 10% ammonia saturated methanol) in hexanes. The desired product fractions were combined and concentrated to yield lohexylamino)((1R,3 S)hydroxycyclohexylamino)- pyrimidinecarbonitrile (0.220 g, 88 % yield, 98.1% pure) as a pale yellow solid. 1H NMR (400 MHz, 6) 5 8.08 - 8.21 (m, 1H), 7.08 - 7.56 (m, 2H), 4.74 (d, J: 3.90 Hz, 1H), 3.93 - 4.13 (m, 1H), 3.44 - 3.77 (m, 2H), 1.83 - 2.02 (m, 2H), 1.65 - 1.82 (m, 6H), 1.58 (d, J: 14.84 Hz, 1H), 1.32 - 1.45 (m, 2H), 1.08 - 1.30 (m, 7H); MS (ESI) m/z 316.4 [M+1]+.
Example 23: 4-((1R,3S)—3-Hydr0xycycloheptylamino)—2—((1R,4R)—4-(2,2,2- trifluor0ethoxy)cyclohexylamin0)pyrimidinecarb0nitrile Fscvo/n CN N N NH H to.
ATI—2514l75vl A. 4-((1R,3S)Hydroxycycloheptylamin0)(methylthio)pyrimidine— -carb0nitrile. To a stirring solution of 4-chloro(methylthio)pyrimidinecarbonitrile (0.4 g, 2.155 mmol) and )—3-aminocycloheptanol hydrochloride (0.428 g, 2.59 mmol; synthesis described herein) in DMF (3 mL) was added DIEA (1.126 mL, 6.46 mmol) and stirred at 60 0C for 1h. LCMS shows the reaction to be completed. The reaction mixture was cooled to room temperature, and poured into water (50 mL). The suspension was stirred for five minutes, then d to afford 4-((1R,3S) hydroxycycloheptylamino)(methylthio)pyrimidinecarbonitrile (0.404 g, 1.451 mmol, 67.4 % yield) as an off-white solid; 1H NMR (DMSO-d6 ,400 MHz): 8 ppm 8.39 (s, 1 H), 8.00 (d, J=7.4 Hz, 1 H), 4.63 (d, J=3.9 Hz, 1 H), 4.21 (br. s., 1 H), 3.74 (td,J=8.5, 4.1 Hz, 1 H), 2.47 (s, 3 H), 1.66 - 1.97 (m, 5 H), 1.35 -1.66 ppm (m, 5 H); MS (ESI) m/z 138.1 [M+1]+.
B. 4-((1R,3S)Hydr0xycycloheptylamin0)((1r,4r)—4-(2,2,2- trifluoroethoxy)cyclohexylamin0)pyrimidine—5-carb0nitrile. 4-((1R,3S) Hydroxycycloheptylamino)(methylthio)pyrimidinecarbonitrile (381 mg, 1.369 mmol) was dissolved in NMP (7 mL). At 0 oC, mCPBA (675 mg, 3.01 mmol) was then added portionwise and the on was stirred at room temp for 90 min. To the reaction mixture was added )(2,2,2-trifiuoroethoxy)cyclohexanamine (0.405 g, 2.054 mmol) (prepared as described herein) and DIEA (0.956 mL, 5.48 mmol) and stirred for 5 min at room temp. The on mixture was further stirred at 80 CC for 30 min after which LCMS showed the tion of the reaction. The reaction mixture was cooled down to room temperature and diluted with water (30 mL). The e was let standing at room temperature for 4 h during which the desired product precipitated out. The product was filtered and the collected solid was washed with water then hexanes and dried under vacuum to give 4-((1R,3S)—3-hydroxycycloheptylamino)((1R,4R)—4-(2,2,2- trifiuoroethoxy)-cyclohexylamino)pyrimidinecarbonitrile (0.444 g, 1.039 mmol, 76 % yield) as a tan solidng NMR (DMSO-d6 ,400 MHZ): 5 ppm 8.08 - 8.22 (m, 1 H), 7.06 - 7.59 (m, 2 H), 4.51 - 4.60 (m, 1 H), 4.01 - 4.24 (m, 1 H), 3.54 - 3.78 (m, 2 H), 3.44 (q, J=701h,2H)311-3240n,1H)188-205(m,4HL112-1880n,P4HL108pmn (t, J=7.0 Hz, 3 H); MS (ESI) m/z 374.2 [M+1]+.
ATI—25 14175v1 Example 24: 4-((1R,3S)Hydr0xycycloheptylamin0)((1r,4R)—4- hydroxycyclohexylamin0)pyrimidinecarb0nitrile HO/h )LN/fiA/CN/ N N NH H 0..
A. ,3S)Hydr0xycycloheptylamin0)(methylsulfonyl)— pyrimidinecarb0nitrile. 4-((1R,3S)—3-Hydroxycycloheptylamino) (methylthio)pyrimidinecarbonitrile (457 mg, 1.642 mmol; synthesis described herein) was dissolved in NMP (10 mL). At 0 oC, mCPBA (736 mg, 3.28 mmol) was then added portionwise and the reaction was stirred for 2 h at room temperature. LCMS indicated consumption of the starting material and formation of sulfone ediate. This reaction was carried onto the next step without further purification. MS (ESI) m/z 311.1 [M+1]+.
B. 4-((1R,3S)Hydr0xycycloheptylamin0)((1r,4R)—4- hydroxycyclohexylamino)pyrimidine—5-carbonitrile. To the on mixture of the previous step was added (1r,4r)aminocyclohexanol hydrochloride (0.374 g, 2.465 mmol) and DIEA (1.435 mL, 8.22 mmol). The reaction mixture was stirred at 80 CC for 1 h and then the reaction mixture was cooled to room temperature. The solvent was evaporated under reduced re and the resulting residue was purified by flash tography (0-10% ammonia saturated methanol in DCM) to afford 4-((1R,3S) ycycloheptylamino)((1r,4R)hydroxycyclohexylamino)pyrimidine carbonitrile (0.250 g, 0.724 mmol, 44.0 % yield) as a tan solid. 1H NMR (400 MHz, DMSO-dg) 5 8.07 - 8.21 (m, 1H), 7.05 - 7.55 (m, 2H), 4.52 - 4.59 (m, 2H), 4.02 - 4.25 (m, 1H), 3.51 - 3.79 (m, 2H), 3.35 - 3.42 (m, 1H), 1.75 - 1.97 (m, 8H), 1.39 - 1.70 (m, 6H), 1.14 - 1.31 (m, 4H); MS (ESI) m/z 346.1 [M+1]+.
ATI—25 14175v1 Example 25: 2-(4-Hydr0xybicyclo[2.2.2]octanylamin0)—4-(is0pr0pylamin0)pyrimidine- 5- carboxamide HO\@\ )LN/YkNHZ/ A. Dimethyl cyclohexane—1,4-dicarb0xylate. To a d solution of cyclohexane-l,4-dicarboxylic acid (100 g, 0.58 mol) in anhydrous methanol (800 mL) was added sulfurous dichloride (208 g, 1.75 mol) at 0 oC. The reaction mixture was d at room temperature overnight. The solution was then concentrated and the residue was poured into water. The mixture was extracted with ethyl e (3 x 200 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. Concentration under reduced pressure afforded dimethyl cyclohexane-l,4- oxylate (105 g, 0.53 mol, 90.5 % yield), which was used in the next step without further purification.
B. Dimethyl 1-(2-chlor0ethyl)cyclohexane—1,4-dicarb0xylate. To a solution of diisopropylamine (88 mL, 0.62 mol) in anhydrous THF (500 mL) was added n-butyl lithium (240 mL, 0.6 mol, 2.5 M solution in hexane) over 20 minutes at -78 oC.
The e was stirred at 0 CC under nitrogen for 30 min. To a mixture of dimethyl exane-l,4-dicarboxylate (100 g, 0.5 mol) and thylphosphoramide (360 mL, 2 mol) in anhydrous THF (800 mL) was added lithium diisopropylamine (freshly prepared above) over 30 min at -40 CC. After stirring for l h at this temperature, l-bromo chloroethane (42 mL, 0.5 mol) was added over 1 h. The mixture was stirred for 3 h at -78 CC, then warmed to room temperature and stirred overnight. To the on mixture was added aqueous hydrochloric acid (3 N, 420 mL) and the mixture was stirred for min. The solvent was removed by evaporation under d pressure. The s layer was extracted with ethyl acetate (3 x 200 mL), and the combined extracts were washed with brine (2 x 300 mL) and dried over sodium sulfate. Concentration under reduced pressure afforded dimethyl l-(2-chloroethyl)cyclohexane-l,4-dicarboxylate (l 16 g, 88 % yield), which was used in the next step without further purificatioan NMR —134— ATI—2514l75vl (400 MHz, CDClg) 5 ppm 3.72 (s, 3H), 3.65 (s, 3H), 3.46-3.42 (m, 2H), 2.33-2.21 (m, 3H), 2.05-1.85 (m, 4H), 1.58-1.42 (m, 2H), 1.25-1.15 (m, 2H).
C. yl bicyclo[2.2.2]octane-1,4-dicarb0xylate. To a solution of diisopropylamine (77 mL, 0.54 mol) in anhydrous THF (500 mL) was added n-butyl lithium (210 mL, 0.53 mol, 2.5 M solution in hexane) over 20 min at -78 CC. Then the mixture was stirred at 0 CC under nitrogen for 30 min. To a mixture of dimethyl 1-(2- chloroethyl)cyclohexane-1,4-dicarboxylate (116 g, 0.44 mol) and hexamethylphosphoramide (317 ml, 1.7 mol) in anhydrous THF (800 mL) was added lithium diisopropylamine (freshly prepared above) over 30 min at -40 CC. The e was d for 2 h at -78 CC and then stirred overnight allowing warming to room ature. To the reaction mixture was added saturated aqueous ammonium chloride (200 mL) and the mixture was stirred for 10 min. The solvent was removed by evaporation under reduced pressure. The aqueous layer was extracted with ethyl acetate (3 x 200 mL).
The combined extracts were washed with brine (2 x 300 mL) and dried over sodium sulfate. Concentration under reduced pressure gave crude product, which was purified by silica gel column chromatography (10 % ethyl acetate in petroleum ether) to afford the title compound (58 g, 0.25 mol, 50 % yield in two steps). 1H NMR (400 MHz, CDClg) 8 ppm 3.65 (s, 6H), 1.81 (s, 12H).
D. 4-(Methoxycarbonyl)bicyclo[2.2.2]octane-l-carboxylic acid. A solution of dimethyl bicyclo[2.2.2]octane-1,4-dicarboxylate (58.0 g, 0.25 mol) in ol (600 mL) was heated under reflux. To this solution was added a solution of potassium hydroxide (9.8 g, 0.175 mol) in methanol (100 mL) and water (12 mL) over 30 minutes.
The reaction mixture was refluxed for 24 h. The solvent was then removed and the residue was diluted with water. The s solution was ted with ethyl acetate (2 x 200 mL) to recover starting material (22.0 g), and the s layer was acidified to pH 3 by on of hydrochloric acid. A precipitate was formed and extracted with ethyl acetate (3 x 300 mL). The combined extracted were washed with brine, dried over sodium sulfate and concentrated to give the titled product (30.0 g, 0.14 mol, 55 % yield).1H NMR (400 MHz, CDClg) 5 ppm 3.65 (s, 3H), 1.81 (s. 12H); MS (ESI) m/z 211.3 [M-H]'.
E. Methyl 4-br0m0bicyclo[2.2.2]octane-l-carboxylate. To a sion of 4-(methoxycarbonyl) bicyclo[2.2.2]octane-l-carboxylic acid (11.0 g, 51.8 mmol) in ATI—25 14175v1 acetone (80 mL) was added 1 M aqueous sodium hydroxide solution (51.8 mL, 51.8 mmol). Then a solution of silver nitrate (8.8 g, 51.9 mol) in water (10 mL) was added.
The formed precipitate was collected by filtration, washed with water, acetone and diethyl ether and dried in vacuo at 115 0C for 4 h. The obtained (4-(methoxycarbonyl)- bicyclo[2.2.2]octanecarbonyloxy)silver (15.3 g, 47.9 mmol) was suspended in hexane (125 mL), then e (7.7 g, 48.1 mmol) was added to the reaction mixture over s at room temperature. After the addition was complete, the reaction mixture was stirred at room ature for another 30 minutes. The reaction mixture was filtered to remove the solid, and the filter cake was washed with hexane (4 x 150 mL). The combined organic filtrates were washed with saturated sodium bicarbonate (2 x 150 mL) andbfine(200nfl),andthmidfiedovernwgnemunimflfifie Concennafionundervamunn gave crude product, which was purified by silica gel column chromatography (5 % ethyl e in petroleum ether) to afford the title compound (4.2 g, 0.17 mol, 33 % yield in two steps). 1H NMR (400 MHz, CDClg) 8 ppm 3.64 (s, 3H), 2.27-2.20 (m, 6H), .94 (m.
] F. 4-Hydr0xybicyclo[2.2.2]octane-l-carboxylic acid. Methyl 4- bromobicyclo [2.2.2]octane-l-carboxylate (17.0 g, 69.0 mol) was refluxed in aqueous sodium hydroxide on (1500 mL, 1 %) for 24 h. After cooling, the reaction solution was acidified with hydrochloric acid (6 N, 100 mL) and extracted with diethyl ether (6 x 500 mL). The combined ether layers were dried over magnesium e and concentrated to afford the title compound (10.4 g, 61.1 mmol, 89 % yield), which was used in the next step without further purification. MS (ESI) m/z 169.2 [M-H]'.
G. Benzyl 4-hydr0xybicyclo[2.2.2]octan-l-ylcarbamate. To a solution of 4-hydroxybicyclo[2.2.2]octanecarboxylic acid (10.4 g, 61.1 mmol) in dioxane (150 mL) was added DIEA (11.8 g, 91.5 mmol), diphenyl phosphoryl azide (25 g, 91.5 mmol) and benzyl alcohol (131 g, 1.22 mol). The mixture was stirred at 80 CC overnight. Then the reaction was concentrated under reduce pressure to remove e and benzyl alcohol (100 oC, 2 mm Hg). The residue was purified by silica gel column chromatography (5 % methanol in DCM) to give the title product (15.4 g, 54 mmol, yield 91%). 1H NMR (400 MHz, CDClg) 8 ppm 7.35-7.30 (m, 5H), 5.03 (s. 2H), 4.55 (br s, 1H), 2.01-1.95 (m, 6H), 1.77—1.72 (m, 6H); MS (ESI) m/z 276.3 [M+H]+.
ATI—25 14175v1 H. 4-Aminobicyclo[2.2.2]0ctanol hydrochloride. To a solution of benzyl 4-hydroxybicyclo[2.2.2]octanylcarbamate (14.8 g, 53 mmol) in methanol (200 mL) was added palladium on charcoal (0.5 g, 10%). The reaction mixture was stirred at 50 0C under hydrogen here (50 Psi) overnight and filtered through celite. The filtrate was concentrated and the residue was added to hloric acid in methanol (10%, 50 mL). The mixture was stirred for 2 h at room temperature. Then the mixture was concentrated again and THF (20 mL) was added. The mixture was stirred at room temperature for 1 h and the precipitate was collected and dried to give the title t (6.7 g, 36 mmol, 70 % yield).1H NMR dg) 8 8.00 (s, 3H), 4.48 (br s,lH) ,1.76- 1.80 (m, 6H), 1.58-1.61 (m, 6H). MS (ESI) m/z 142.1[M+1] +.
I. Ethyl 4-(isopropylamin0)(methylthio)pyrimidine—5-carboxylate. A mixture of ethyl 4-chloro(methylthio)pyrimidinecarboxylate (10 g, 43.0 mmol), propanamine (4.21 mL, 49.4 mmol) and DIEA (9.76 mL, 55.9 mmol) in ethanol (140 mL) was heated at 60 CC overnight. After g to room temperature, the solvent was concentrated under reduced pressure and water was added. The aqueous phase was extracted three times with ethyl acetate and the combined organic phases were dried over magnesium sulfate, d and evaporated to give ethyl 4-(isopropylamino) (methylthio)pyrimidinecarboxylate (10.10 g, 39.5 mmol, 92 % yield) as a colorless oil.
MS (ESI) m/z 256.1 [M+1]+.
J. 4-(Isopropylamin0)(methylthio)pyrimidine—5-carboxylic acid. In a round-bottomed flask ethyl 4-(isopropylamino)—2-(methylthio)pyrimidinecarboxylate (10.97 g, 43.0 mmol) was dissolved in ethanol (150 mL) ed by the addition of aqueous sodium hydroxide solution (1M, 129 ml, 129 mmol) . The resulting mixture was stirred overnight at room ature and then the solvent was evaporated under reduced pressure. Aqueous citric acid solution (2M, 129 ml, 258 mmol) was slowly added and the resulting mixture was stirred for 0.5 h at room temperature. The suspension was filtered and the solids were washed twice with water (2 x 50 mL) and then dried in the vacuum oven at 45 CC overnight to afford propylamino)(methylthio)pyrimidine ylic acid (8.38 g, 36.9 mmol, 86 % yield); MS (ESI) m/z 228.4 [M+1]+.
K. 4-(Isopropylamin0)(methylthio)pyrimidine—5-carboxamide. 4-(Isopropylamino)(methylthio)pyrimidinecarboxylic acid (8.38 g, 36.9 mmol) and ATI—25 14175v1 HATU (21.03 g, 55.3 mmol) were combined in a 250 mL bottomed flask followed by the addition ofDMF (92 mL). After that ammonia hydrochloride (9.86 g, 184 mmol) and DIEA (32 mL, 184 mmol) were added and the mixture was stirred at room temperature overnight. The solvent was concentrated under reduced re and water (150 mL) was added. The resulting suspension was filtered, washed with water and dried under high vacuum overnight to afford 4-(isopropylamino)(methylthio)pyrimidine carboxamide (8 g, 35.4 mmol, 96 % yield); MS (ESI) m/z 227.4 [M+1]+.
L. 4-(Isopr0pylamin0)—2-(methylsulfinyl)pyrimidine—5—carboxamide and 4-(Isopr0pylamin0)—2-(methylsulfonyl)pyrimidinecarboxamide. To a solution of 4-(isopropylamino)(methylthio)pyrimidinecarboxamide (641 mg, 2.8 mmol) in NMP (6 mL) was added mCPBA (852 mg, 4.2 mmol, 85% purity) at 0 CC. The reaction was stirred for 1 h at room temperature. Then water (25 mL) was added, which dissolved the suspension for a short time only to form another thick precipitate, which was filtered off The resulting filtrate was concentrated under reduced re to afford the title mixture in NMP, which was used in the next step without further purification. MS (ESI) m/z 2433,2592 [M+1]+.
M. 2-(4-Hydr0xybicyclo [2.2.2] octanylamin0)—4-(is0propylamin0)- pyrimidine— 5-carb0xamide. To the solution of 4-(isopropylamino)(methylsulfinyl)- pyrimidine carboxamide and 4-(isopropylamino)(methylsulfonyl)pyrimidine amide in NMP (from previous step, 2.8 mmol), 4-aminobicyclo[2.2.2]octanol hydrochloride (500 mg, 2.8 mmol), DIEA (1.1 g, 8.4 mmol) were added and heated at 100 0C for 3 d. The resulting e was concentrated under reduced pressure and the residue was purified by ative thin layer chromatography to afford the final product (74.1 mg, 8.2 % . 1H NMR (DMSO-dg) 8 ppm 8.88 (br s, 1H), 8.31 (s, 1H), 7.5-6.5 (br s, 2H), 5.76 (s, 1H), 4.25 (s, 1H), 4.11 (brs, 1H), 2.05-1.97 (m, 6H), 1.61-1.54 (m, 6H), 1.24-1.12(m, 6H); MS (ESI) m/z 320.2 [M+H]+; Purity = 96.5 % at 214 nm, Purity = 97.7 % at 254 nm.
ATI—2514175v1 Example 26: 4-(tert-Butylamin0)—2-((1r,4r)—4-hydr0xycyclohexylamin0)pyrimidine carboxamide HO/“Q [VI/\f'kNHZ A. Ethyl 4-(tert-butylamin0)(methylthi0)pyrimidine—5-carb0xylate.
A mixture of ethyl 4-chloro(methylthio)pyrimidinecarboxylate (3 g, 12.89 mmol), 2-methylpropanamine (1.565 mL, 14.83 mmol) and DIEA (2.93 mL, 16.76 mmol) in ethanol (20 mL) was heated at 60 CC overnight. After cooling to room temperature, the solvent was concentrated under reduced pressure and water was added. The aqueous phase was extracted three times with ethyl e and the combined organic phases were dried over anhydrous magnesium sulfate, filtered and evaporated to give ethyl 4-(tert- butylamino)(methylthio)pyrimidinecarboxylate (3.20 g, 11.86 mmol, 92% yield) as a ess oil. MS (ESI) m/z 269.5 [M+1]+.
B. 4-(tert—Butylamin0)(methylthi0)pyrimidine—5-carb0xylic acid. In a round-bottomed flask ethyl 4-(tert-butylamino)(methylthio)pyrimidinecarboxylate (3.2 g, 11.88 mmol) was dissolved in ethanol (40 mL) followed by the addition of 1M s sodium ide solution (35.6 mL, 35.6 mmol) . The resulting mixture was stirred overnight at room temperature and then the solvent was ated under reduced re. After that 2M s citric acid solution (35.6 mL, 71.3 mmol) was slowly added and the resulting mixture was stirred for 0.5 h at room temperature. The suspension was filtered and the solids were washed twice with water (2 x 50 mL) and then dried in a vacuum oven at 45 °C overnight to afford 4-(tert-butylamino)(methylthio)pyrimidine carboxylic acid (2.83 g, 11.73 mmol, 99% yield). MS (ESI) m/z 242.3 [M+1]+.
C. 4-(tert-Butylamin0)(methylthi0)pyrimidinecarboxamide. 4-(tert-Butylamino)—2-(methylthio)pyrimidinecarboxylic acid (2.83 g, 11.73 mmol) and [1 ,2,3 ]triazolo [4,5 -b]pyridin-3 -yl)- 1 ,1 ,3 ,3 -tetramethylisouronium hexafluoro- phosphate(V) (6.69 g, 17.59 mmol) were combined in a 250 mL round-bottomed flask followed by the addition ofDMF (29 mL). After that ammonia hydrochloride (3.14 g, 58.6 mmol) and DIEA (10.21 mL, 58.6 mmol) were added and the mixture was stirred at ATI—25 14175v1 room temperature overnight. The solvent was concentrated under reduced re and water (150 mL) was added. The resulting suspension was filtered, washed with water and dried under high vacuum overnight to afford 4-(tert-butylamino)(methylthio)- pyrimidinecarboxamide (2.54 g, 10.57 mmol, 90% yield). MS (ESI) m/z 241.2 [M+1]+.
] D. 4-(tert-Butylamin0)—2-(methylsulfonyl)pyrimidine—5-carb0xamide.
To a ng solution of 4-(tert—butylamino)(methylthio)pyrimidinecarboxamide (0.6 g, 2.497 mmol) in NMP (5 mL) was added mCPBA (0.839 g, 3.74 mmol) at 0 oC and stirring was continued at room temperature for 1h. LCMS showed te conversion to desired product. The reaction mixture was carried on to next reaction without further purification. MS (ESI) m/z 273.2 [M+1]+.
E. t-Butylamin0)—2-((1r,4r)—4-hydroxycyclohexylamino)— pyrimidine—5-carb0xamide. To the solution of 4-(tert-butylamino)(methylsulfonyl)- pyrimidinecarboxamide from the previous step was added DIEA (2.181 mL, 12.49 mmol) and (1r,4r)aminocyclohexanol hydrochloride (0.568 g, 3.75 mmol), and the reaction was stirred at 90 0C for 16 h. The solvent was evaporated under reduced pressure and the resulting residue was purified using e-phased preparative HPLC (0-50% itrile + 0.1% TFA in water + 0.1% TFA, over 30 min). Fractions containing product were concentrated under reduced pressure. The resulting residue was redissolved in a methanol (5 mL), passed over an Varian StratoSpheres HCO3 resin SPE tube for TFA removal (0.9 mmol bicarbonate equiv.), and then concentrated under reduced pressure to afford 4-(tert-butylamino)((1r,4r)hydroxycyclohexylamino)pyrimidine carboxamide (0.350 g, 1.139 mmol, 45.6% yield). 1H NMR (DMSO-d6 ,400 MHz) 5 ppm 0.82 - 0.88 (m, 1 H) 1.14 - 1.30 (m, 5 H) 1.42 (s, 9 H) 1.85 (d, J=4.30 Hz, 4 H) 3.38 (br. s, 1 H) 3.59 (d, J=7.42 Hz, 1 H) 4.54 (s, 1 H) 7.02 (d, J=7.81 Hz, 1 H) 8.32 (s, 1 H) 9.18 (s, 1 H); MS(ESI): m/Z 308.1 [M+1]+. —140— ATI—25 14175v1 Example 27 : 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0) (isopr0pylamin0)pyrimidine—5-carboxamide A )L /N \ NH2 N N NH _ OH A. 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0) (methylthi0)pyrimidine—5-carb0nitrile. To a stirring solution of 4-chloro(methylthio)- pyrimidinecarbonitrile (310 mg, 1.670 mmol) and (1R,2R,5R)aminomethylcyclo- hexanol hydrochloride (304 mg, 1.837 mmol; synthesis described herein) in DMF (2.4 mL) was added DIEA (0.873 mL, 5.01 mmol). The resulting mixture was d at 60 0C for 2 h. LCMS showed the reaction to be complete. The solvent was evaporated under reduced pressure and the residue diluted with 150 mL ethyl e and 50 mL water. The layers were separated and the aqueous phase was extracted with 75 mL ethyl acetate. The combined ethyl acetate layers were washed with brine (2 x 50 mL), dried over anhydrous magnesium sulfate, filtered and concentrated to an oil that solidified upon standing to afford 4-((1R,3R,4R)hydroxymethylcyclohexylamino) (methylthio)pyrimidinecarbonitrile (418 mg, 1.503 mmol, 90% yield). MS (ESI) m/z 279.3 [M+1]+.
B. 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0) (methylsulfonyl)pyrimidine—5-carb0nitrile. To a solution of 4-((1R,3R,4R)hydroxy- 4-methylcyclohexylamino)(methylthio)pyrimidinecarbonitrile (400 mg, 1.437 mmol) in NMP (5 mL) at 0 0C was added mCPBA (805 mg, 3.59 mmol) and the on was stirred for 3 h at room temperature. The resulting solution was used ly in the next step without further purification. MS (ESI) m/z 311.4 [M+1]+ ] C. 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0) (isopropylamin0)pyrimidine—5-carb0nitrile. To the on of 4-((1R,3R,4R)—3- hydroxymethylcyclohexylamino)(methylsulfonyl)pyrimidinecarbonitrile (446 mg, 1.437 mmol) from the preVious step was added propanamine (0.367 mL, 4.31 mmol) —141— ATI—25 14175V1 WO 45569 and DIEA (1.004 mL, 5.75 mmol). The reaction was stirred at 80 CC for 4 h and then allowed to cool to ambient temperature. The solvent was ated under reduced pressure and the residue was purified by silica gel chromatography (0-90% ethyl acetate in hexanes) to afford 4-((1R,3R,4R)hydroxymethylcyclohexylamino) (isopropylamino)pyrimidinecarbonitrile (381 mg, 1.317 mmol, 92% yield). MS (ESI) m/z 290.2 [M+1]+ D. 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0) (isopr0pylamin0)pyrimidine—5-carb0xamide. 4-((1R,3R,4R)—3-Hydroxymethylcyclohexylamino )(isopropylamino)pyrimidinecarbonitrile (0.38 g, 1.313 mmol) was dissolved in DMSO (5 mL). To this solution was added a 50% aqueous sodium hydroxide solution (0.103 ml, 1.970 mmol) and a 30% s hydrogen peroxide solution (0.101 ml, 0.985 mol) at room temperature. Then the reaction mixture was d at 50 CC for 1 h. After that, the reaction mixture was poured into ice water (15 mL) and the resulting e was stirred for 1 h. The ing precipitate was filtered and washed with water to afford 4-((1R,3R,4R)—3-hydroxymethylcyclohexylamino) (isopropylamino)pyrimidinecarboxamide (0.210 g, 0.683 mmol, 52% yield). 1H NMR (DMSO-d6 ,400 MHz) 8 ppm 0.94 (d, J=6.40 Hz, 4 H) 1.12 (d, J=5.91 Hz, 9 H) 1.65 (dd, J=13.29, 2.95 Hz, 1 H) 1.92 (br. s., 1 H) 2.15 (br. s., 1 H) 2.97 (br. s., 1 H) 3.81 - 4.14 (m, 2 H) 4.55 (d, J=5.41 Hz, 1 H) 6.70 - 7.02 (m, 1 H) 8.34 (br. s., 1 H) 8.92 (br. s., 1 H); MS (ESI) m/z 308.0 [M+1]+.
Example 28: 2-(Bicyclo[1.1.1]pentan-l-ylamino)—4-((1R,3R,4R)—3-hydr0xy methylcyclohexylamin0)pyrimidine—5-carboxamide & JL /N \ NH2 N N NH . OH A. 2-(Bicyclo[1.1.1]pentan-l-ylamino)—4-((1R,3R,4R)—3-hydr0xy methylcyclohexylamin0)pyrimidinecarb0nitrile. To a solution of 4-((1R,3R,4R) hydroxymethylcyclohexylamino)(methylsulfonyl)pyrimidinecarbonitrile (446 mg, ATI—2514175v1 1.437 mmol; synthesis described herein) in NMP (5 mL) was added bicyclo[1.1.1]pentan- 1-amine hydrochloride (258 mg, 2.155 mmol; prepared according to Org. Lett., 13(17): 4746-4748 (2011)) and DIEA (1.255 mL, 7.18 mmol). The reaction was stirred at 80 °C for 4 h and then allowed to cool to ambient ature. The solvent was ated under reduced pressure and the residue was d by flash chromatography (0-90% ethyl acetate in hexanes) to afford 2-(bicyclo[1.1.1]pentanylamino)((1R,3R,4R) hydroxymethylcyclohexylamino)pyrimidinecarbonitrile (381 mg, 1.216 mmol, 85% yield). MS(ESI): m/Z 314.0 .
B. 2-(Bicyclo[1.1.1]pentanylamin0)—4-((1R,3R,4R)—3-hydr0xy methylcyclohexylamin0)pyrimidine—5-carb0xamide. 2-(Bicyclo[1.1.1]pentan ylamino)((1R,3R,4R)hydroxymethylcyclohexylamino)pyrimidinecarbonitrile (0.38 g, 1.213 mmol) was dissolved in DMSO (26.9 mL) and to this solution was added a 50% aqueous sodium hydroxide solution (0.097 ml, 1.213 mmol) and a 30% aqueous hydrogen peroxide solution 0.137 mL, 1.213 mmol) at room temperature. Then the reaction e was stirred at 50 0C for 1 h. The reaction was cooled to room ature and it was poured into 50 mL of ice water. The white precipitate was collected on filter paper and was washed twice with water. The precipitate was dried to give 2- (bicyclo[1 . 1 . 1]pentanylamino)((1R,3R,4R)hydroxymethylcyclohexylamino)- pyrimidinecarboxamide (0.269 g, 0.812 mmol, 67% yield). 1H NMR (DMSO-d6, 400 MHz): 8 ppm 0.94 (d, J=6.25 Hz, 3 H) 1.10 - 1.26 (m, 3 H) 1.68 (d, J=10.54 Hz, 1 H) 1.92 (d, J=10.93 Hz, 1 H) 2.06 (s, 6 H) 2.17 (d, J=9.76 Hz, 1 H) 2.46 (s, 1 H) 2.98 (br. s., 1 H) 3.82 - 3.95 (m, 1 H) 4.63 (br. s., 1 H) 7.16 (br. s., 1 H) 7.77 (br. s., 1 H) 8.11 (br. s., 1 H) 8.33 (s, 1 H) 9.24 (br. s., 1 H); MS(ESI): m/z 332.3 [M+1]+. —143— ATI—25 14175v1 Example 29: 4-((2-Cyclopropylpr0panyl)amin0)—2-(((1r,4r)—4- hydroxycyclohexyl)amin0)pyrimidinecarb0xamide “00 1*)L / N N NH H 2% A. 4-((2-Cyclopropylpropanyl)amin0)(methylthi0)pyrimidine carbonitrile. To a solution of 4-chloro(methylthio)pyrimidinecarbonitrile (750 mg, 4.0 mmol) in DMF (8 mL) was added 2-cyclopropylpropanamine (400 mg, 4.0 mmol) and DIEA (1560 mg, 12.0 mmol). The resulting mixture was d at 60 0C for 2 h. Water was added and the mixture was ted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated to give the crude product, which was purified by silica gel column chromatography (0-30% ethyl acetate in eum ether) to afford the title compound (665 mg, 2.68 mmol, 66% yield) as a white solid. MS (ESI) m/z = 249.2 [M+H] +.
B. 4-((2-Cyclopr0pylpr0panyl)amin0)(methylthi0)pyrimidine carboxamide. To a stirring solution of 4-((2-cyclopropylpropanyl)amino) (methylthio)pyrimidinecarbonitrile (665 mg, 2.68 mmol) in DMSO (4 mL) was added aqueous hydrogen peroxide solution (0.162 mL, 30%) and aqueous sodium ide (2.2 mL, 6 mol/L) solution. The resulting mixture was stirred at 50 0C for 12 min, and then water was added. The d product was filtered and dried in vacuo to give the crude product (392 mg, 1.47 mmol, 55% yield) as a white solid. MS (ESI) m/z 267.3 [M+H] +. [003 85] C. 4-((2-Cyclopropylpr0panyl)amin0)—2-(methylsulfinyl)pyrimidine—5- carboxamide and 4-((2-cyclopropylpropan-Z-yl)amin0)(methylsulfonyl)pyrimidine carboxamide. To a cooled (0 0C) solution of 4-((2-cyclopropylpropanyl)amino) (methylthio)pyrimidine amide (390 mg, 1.46 mmol) in THF (6 mL) was added mCPBA (447 mg, 2.2 mmol) portion-wise. The reaction mixture was stirred for 0.5 h at 0 0C.
The mixture was concentrated under vacuum and purified by silica gel column chromatography (2.5%-10% DCM in methanol) to give a mixture of 4-((2-cyclopropylpropanyl)amino) (methylsulfinyl)pyrimidinecarboxamide and 4-((2-cyclopropylpropanyl)amino) ATI—25 14175v1 (methylsulfonyl)pyrimidinecarboxamide (382 mg, 1.32 mmol, 90% yield). MS (ESI) m/z 299.1 [M+H] [and 283.1 [M+H]+.
D. 4-((2-Cyclopr0pylpr0panyl)amin0)(((1r,4r)—4-hydr0xycyclohexyl)— amin0)pyrimidine—5-carb0xamide. A mixture of cyclopropylpropanyl)amino)—2- (methylsulfinyl)pyrimidinecarboxamide, 4-((2-cyclopropylpropanyl)amino) (methylsulfonyl)pyrimidinecarboxamide (191 mg, 0.66 mmol), (1r,4r)aminocyclohexanol (114 mg, 0.99 mmol), DIEA (256 g, 1.98 mmol) and NMP (5 mL) were combined and heated at 100 0C overnight. The resulting mixture was concentrated and the residue was purified by HPLC (5-95% acetonitrile in water) to afford the title compound (129.5 mg, 0.39 mmol, 59% . 1H NMR (400 MHz, CDClg): 8 ppm 8.96 (s, 1H), 8.09 (s, 1H ), 5.38 (s, 2H), 4.98-4.96 (m, 1H), 3.79-3.77 (m, 1H), 3.69-3.67 (m, 1H), 2.14-2.11 (d, J=11.6Hz, 2H), 2.04-2.01 (m, 2H), 1.48-1.41 (m, 4H), 1.38 (s, 6H), 1.32-1.22 (m, 2H), 0.48-0.39 (m, 4H); MS (ESI) m/z 334.3 [M+H] +. e 30: 4-Cyclobutylamin0(((1r,4r)—4—methoxycyclohexyl)amin0)pyrimidine- -carb0xamide /O, ' N \ NH2 )L / N N NH H <> A. 4-(Cyclobutylamin0)(methylthi0)pyrimidine—5-carbonitrile. To a ng solution of 4-chloro(methylthio)pyrimidinecarbonitrile (2 g, 10.8 mmol) in DMF (10 mL) was added DIEA (4.2 g, 32.4 mmol) and cyclobutanamine (2.3 g, 32.4 mol) at 0 CC.
The resulting mixture was stirred at 60 CC for 2 h. The on mixture was poured into saturated sodium chloride, and extracted with ethyl acetate three times. The ed organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to afford the crude product. The crude product was purified Via silica gel column chromatography (10% ethyl acetate in petroleum ether) to get the desired t as a white solid (1.7 g, 7.7 mmol, 71% yield). MS (ESI) m/Z = 221 .2[M+H] +.
B. 4-(Cyclobutylamino)—2-(methylsulfinyl)pyrimidine—5-carb0nitrile and 4- (cyclobutylamin0)(methylsulfonyl)pyrimidine—5-carb0nitrile. 4-(Cyclobutylamino) (methylthio)pyrimidinecarbonitrile (1.7 g, 7.7 mmol) was dissolved in DCM (30 mL) and —145— ATI—25 14175V1 cooled to 0 0C. To this mixture, mCPBA (4.6 g, 23 mmol) was added portionwise and the reaction was stirred for 1 h. The resulting mixture was poured into a ted aqueous sodium bicarbonate solution. The aqueous layer was extracted three times with DCM. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to afford a mixture of 4-(cyclobutyl-amino)(methylsulf1nyl)pyrimidinecarbonitrile and 4-(cyclobutylamino) (methyl-sulfonyl)pyrimidinecarbonitrile as a yellow thick oil (1.6 g crude), which was used in the next step without fithher purification. MS (ESI) m/z =237.2/253.2 [M+H] +.
C. lobutylamin0)(((1r,4r)—4-meth0xycyclohexyl)amin0)pyrimidine— -carb0nitrile. To the mixture of 4-(cyclobutylamino)(methylsulf1nyl)pyrimidine carbonitrile and 4-(cyclobutylamino)(methylsulfonyl)pyrimidinecarbonitrile (1.6 g, crude) from the previous step was added (1r,4r)methoxycyclohexanamine (0.96 g, 7.4 mmol), 1,4-dioxane (100 mL) and DIEA (4.3 g, 33.4 mmol). The resulting mixture was d at 100 CC overnight. After removal of all volatile solvents under reduced pressure, the residue was purified by silica gel column chromatography (33.3% ethyl acetate in petroleum ether) to provide the desired product as a white solid (1.3 g, 57% . MS (ESI) m/z =302.1 [M+H] +.
D. 4-(Cyclobutylamin0)(((1r,4r)—4-meth0xycyclohexyl)amin0)pyrimidine— -carb0xamide. To the mixture of lobutylamino)(((1r,4r)methoxycyclohexyl)- amino)pyrimidinecarbonitrile (700 mg, 2.3 mmol) in DMSO (8 mL) was added aqueous en peroxide solution (1.3 g, 30%, 11.5 mmol), and then aqueous sodium hydroxide solution (2 mL, 6 mol/L, 11.5 mmol). The mixture was d at 50 CC for 0.5 h, then water (30 mL) was added to the reaction mixture, the product was collected and dried to give the desired compound (400 mg, 1.25 mmol, 54% yield). 1H NMR (400 MHz, CDClg) 8 ppm 8.76 (s, 1H), 8.11 (s, 1H), 5.46 (s, 2H), 5.01 (s, 1H), 4.51-4.45 (m, 1H), 3.83-3.81 (m, 1H), 3.36 (s, 3H), 3.21-3.16 (m, 1H), 2.40-2.33 (m, 2H), 2.15-1.96 (m, 6H), 1.78 (s, 2H), 1.43-1.20 (m, 4H); MS (ESI) m/Z = 319.9 [M+H] +).
ATI—25 14175v1 Example 31: 4-(Cyclobutylamin0)—2-(((1r,4r)—4-hydroxycyclohexyl)amin0)- pyrimidine-S-carboxamide A. 4-(Cyclobutylamin0)(methylsulfinyl)pyrimidinecarb0nitrile and 4- (cyclobutylamin0)(methylsulfonyl)pyrimidinecarb0nitrile. 4-(Cyclobutylamino) (methylthio)pyrimidinecarbonitrile (889 mg, 4.04 mmol; synthesis described herein) was ved in DCM (20 mL) and cooled to 0 0C. To this mixture, mCPBA (2.1 g, 12.1 mmol) was added portion-wise and the reaction was stirred for 1 h. The resulting mixture was poured into a saturated aqueous sodium bicarbonate solution. The aqueous layer was extracted three times with DCM. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to afford a e of 4-(cyclobutylamino)(methylsulf1nyl)pyrimidine carbonitrile and 4-(cyclobutylamino)(methylsulfonyl)pyrimidinecarbonitrile as a yellow thick oil (893 mg crude), which was used in the next step without r purification. MS (ESI) m/Z =237.2/253.2 [M+H] +.
B. 4-(Cyclobutylamin0)(((1r,4r)—4-hydr0xycyclohexyl)amin0)pyrimidine— -carb0nitrile. To the mixture of 4-(cyclobutylamino)(methylsulf1nyl)pyrimidine itrile and 4-(cyclobutylamino)(methylsulfonyl)pyrimidinecarbonitrile (424 mg, about 1.93 mmol) from the previous step was added (1r,4r)aminocyclohexanol (244 mg, 2.12 mmol), 1,4-dioxane (50 mL) and DIEA (1.2 g, 9.65 mmol). The resulting mixture was stirred at 100 OC overnight. After removal of all volatile solvents under reduced pressure, the residue was purified by silica gel column chromatography (50% ethyl acetate in petroleum ether) to get the desired product as a white solid (305 mg, 1.06 mmol, 55.4% . MS (ESI) m/z =288.2 [M+H] f ] C. lobutylamin0)—2-(((1r,4r)—4-hydr0xycyclohexyl)amin0)pyrimidine— -carb0xamide. To a solution of lobutylamino)(((1r,4r)hydroxycyclohexyl)- amino)pyrimidinecarbonitrile (305 mg, 1.06 mmol) in DMSO (6 mL) was added aqueous hydrogen peroxide solution (600.1 mg, 30%, 5.3 mmol), and then aqueous sodium hydroxide —147— ATI—25 14175v1 solution (1 mL, 6 mol/L, 5.3 mmol). The mixture was stirred at 50 CC for 0.5 h, and then water was added and the mixture was extracted with ethyl e. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. Concentration under reduced pressure gave the crude product, which was purified by silica gel column chromatography (0-10% methanol in DCM) to afford the title compound (248.2 mg, 0.81 mmol, 76.6% yield). 1H NMR (400 MHz, CDClg) 8 ppm 8.74 (s, 1H), 8.12 (s, 1H), 5.46 (brs, 2H), 5.07- 4.96 (m, 1H), 4.53-4.43 (m, 1H), 3.80-3.79 (m, 1H), 3.70-3.65 (m, 1H), 2.40-2.34 (m, 2H), 2.15- 1.97 (m, 6H), 1.91-1.88 (s, 2H), 1.55-1.19 (m, 4H); MS (ESI) m/Z = 306.0 [M+H]+.
Example 32: 2-(tert-Butylamin0)—4-((1R,3R,4R)—3-hydr0xymethylcyclohexylamin0)- dine-S--carb0xamide 9N1?“ £1...
A. (R)—tert—Butyl 4-methylcyclohex—3-enylcarbamate and (S)-tert-butyl 4-methylcyclohexenylcarbamate. To a stirring solution of a mixture of (R) methylcyclohexenamine and (S)—4-methylcyclohexenamine (40.2973 g, 359 mmol; prepared as described in J. Org. Chem. 1992, 5 7, 3454 — 3462) in ethyl ether (498 mL) at 0 0C was added a solution of di-tert-butyl-dicarbonate (81 g, 362 mmol) in ethyl ether (100 mL) dropwise over 30 min. The reaction was stirred at 0 0C for 1 h, allowed to slowly reach room ature over 4 h, and then stirred at room temperature for 16 h.
The reaction mixture was concentrated under reduced re to afford the crude residue.
The crude residue was ated with s (500 mL), stirred for 5 min at 0 0C, then filtered and washed with hexanes (50 mL) to afford a first crop of product (~55 g). The filtrate was concentrated under d pressure, triturated with hexanes and the precipitate was filtered to afford a second crop of product (~15 g). This was repeated to afford a third crop of product (4.25 g). The three crops were combined to afford a mixture of (R)-tert-butyl 4-methylcyclohexenylcarbamate and (S)-tert—butyl 4-methylcyclohex- carbamate (74.25 g, 351 mmol, 98% yield) as a white solid. 1H NMR (400 MHz, ATI—25 14175V1 W0 45569 DMSO-d6) 8 ppm 6.72 (d, J=7.42 Hz, 1 H), 5.26 (br. s., 1 H), 3.35 - 3.47 (m, 1 H), 1.93 (br. s., 5 H), 1.60 (s, 3 H), 1.32 - 1.44 (m, 10 H).
] B. tert-Butyl (1R,3R,4R)—3-hydr0xymethylcyclohexylcarbamate, tert-butyl (1R,3S,4S)hydr0xymethylcyclohexylcarbamate, tert—butyl (1 S,3R,4R)—3-hydr0xymethylcyclohexylcarbamate, tert—butyl (1 S,3S,4S)—3- hydroxymethylcyclohexylcarbamate. To a ng solution of a mixture of rt- butyl 4-methylcyclohexenylcarbamate and (S)-tert-butyl 4-methylcyclohex enylcarbamate (13 g, 61.6 mmol) in THF (780 mL) at 0 CC was added 1 M borane THF complex (277 mL, 277 mmol). The solution was stirred at 0 oC, allowed to reach room temperature over 1 h, and then stirred at room temperature for 20 h. The reaction was quenched very slowly with water (330 mL), diluted with ethanol (326 mL) and basified with 5 N aqueous sodium hydroxide (308 mL, 1.54 mol). To the stirring biphasic mixture was slowly added 30% hydrogen peroxide (316 mL, 3.08 mol) and the resulting mixture was heated to 45 0C for 20 h. The crude reaction was quenched with saturated aqueous sodium sulfite (573 mL) and extracted with ethyl acetate (4 x 1 L). The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and trated under reduced pressure to give the crude product. The above reaction was repeated 5 times (using 3 x 13 g and 2 x 11 g starting material). The combined crude products from all 6 ons were purified by silica gel chromatography % ethyl acetate in hexanes).
The t containing fractions were combined and concentrated under reduced pressure to afford a mixture of tert-butyl (1R,3R,4R)hydroxymethylcyclohexylcarbamate, tert-butyl (1 R,3 S ,4S)-3 -hydroxymethylcyclohexyl-carbamate, tert-butyl (1 S,3R,4R)-3 - hydroxymethylcyclohexylcarbamate, and tert-butyl (1 S,3S,4S)hydroxy methylcyclohexylcarbamate (55 g, 242 mmol, 72% yield) as a solid. The 4 constituent stereoisomers were separated by ative chiral SFC utilizing multiple injections over a series of 3 separate columns. First column: ChiralPak AD-H, 250><50 mm I.D., isocratic % methanol in CO2. Second column: ChiralPak AD-H, 0 mm I.D., isocratic 25% methanol in CO2. Third column: Pak AD-H, 250><50 mm I.D., isocratic 15% ethanol in C02. The separated isomers were characterized on an analytical scale ChiralPak AD-3 column, 150><4.6 mm I.D., 5-40% methanol (with 0.05% diethylamine) in CO2 (15 min run time) and labeled as ediate 1 to Intermediate 4. —149— ATI—25 14175v1 2012/034349 ediate 1: 2.0 g (8.72 mmol, 3.6% yield fiom SFC purification).
Retention time: 4.77 min. MS (ESI) m/z 252.1 [M + 23]+.
Intermediate 2: 1.5 g (6.54 mmol, 2.7% yield from SFC purification).
Retention time: 5.08 min. MS (ESI) m/z 252.1 [M + 23]+. ediate 3: 16.0 g (69.78 mmol, 29.1% yield from SFC purification).
Retention time: 5.48 min. 1H NMR (400 MHz, DMSO-d6) 5 ppm 6.64 - 6.86 (m, 1 H), 4.43 - 4.60 (m, 1 H), 3.10 - 3.29 (m, 1 H), 2.81 - 2.96 (m, 1 H), 1.84 - 2.01 (m, 1 H), 1.49 - 1.72 (m, 2 H), 1.37 (s, 9 H), 0.98 - 1.14 (m, 3 H), 0.76 - 0.96 (m, 4 H). MS (ESI) m/z 252.1 [M + 23]+.
Intermediate 4: 18.5 g (80.68 mmol, 33.6% yield from SFC purification).
Retention time: 7.79 min. MS (ESI) m/z 252.1 [M + 23]+.
A small le X-ray crystal structure was solved for Intermediate 3 and the structure was demonstrated to be tert-butyl (1R,3R,4R)—3-hydr0xy methylcyclohexylcarbamate. The X-ray structure was solved as follows. Single crystal X-ray diffraction studies were carried out on a Bruker Kappa APEX CCD diffractometer equipped with Cu Ka radiation (1 = 1.5478). Crystals of the subject compound were grown by vapor diffilsion of hexanes into a hexanes/THF solution. A 0.21 x 0.05 x 0.03 mm colorless needle was mounted on a Cryoloop with ne oil. Data were ted in a en gas stream at 120(2) K using (I) and as scans. Crystal-to-detector distance was 60 mm using variable exposure time (2s-20s) depending on 0 with a scan width of 1.00. Data tion was 97.0% complete to 68.000 in 6’. A total of 9000 reflections were collected covering the indices, -33<=h<=32, -31<=k<=30, -6<=l<=5. 2554 reflections were found to be symmetry independent, with a Rint of 0.0602. Indexing and unit cell refinement indicated a rhombohedral, hexagonal lattice. The space group was found to be R3. The data were integrated using the Bruker SAINT software m and scaled using the SADABS software program. Solution by direct methods (SHELXS) produced a complete phasing model consistent with the structure of tert-butyl (1R,3R,4R)—3-hydr0xymethylcyclohexylcarbamate.
All non-hydrogen atoms were refined anisotropically by atrix least- squares (SHELXL-97). All hydrogen atoms were placed using a riding model. Their ATI—2514175v1 positions were constrained relative to their parent atom using the appropriate HFIX command in SHELXL-97.
C. (1R,2R,5R)—5-Amino-Z-methylcyclohexanol hydrochloride. To vigorously ng ol (149 mL) at 0 0C was added acetyl chloride (15.87 mL, 223 mmol) and the resulting mixture stirred for 10 min. To this solution was added tert- butyl (1R,3R,4R)hydroxymethylcyclohexylcarbamate (17.08 g, 74.5 mmol) and the mixture was stirred for 22 h at room temperature. The crude reaction was concentrated and then triturated with ethyl ether (2 x 300 mL) to afford (1R,2R,5R)—5-amino methylcyclohexanol hydrochloride (12.2 g, 73.6 mmol, 99% yield) as a white solid. 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.02 (br. s., 3 H), 4.77 (d, J=5.86 Hz, 1 H), 2.85 - 3.10 (m, 2 H), 2.03 - 2.21 (m, 1 H), 1.83 (d, J=11.71 Hz, 1 H), 1.65 (dd, J=13.47, 3.32 Hz, 1 H), 1.24 (q, J=11.58 Hz, 2 H), 1.12 (dd, J=6.05, 3.32 Hz, 1 H), 0.82 - 1.03 (m, 4 H).
D. 5-Br0m0-N-tert-butyl(methylthi0)pyrimidinamine. To -bromochloro(methylthio)pyrimidine (3 g, 12.53 mmol) in dioxane (12.53 mL) was added 2-methylpropanamine (7.93 mL, 75 mmol). The mixture was stirred at 100 CC overnight in a sealed vessel. The solvent was removed under reduced pressure and the residue was dissolved in 100 mL ethyl acetate and washed with 50 mL of a 1M aqueous solution of sodium en ate. The aqueous layer was back-extracted with 50 mL ethyl acetate. The combined organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated to give 5-bromo-N-tert-butyl(methylthio)pyrimidin amine (3.4 g, 12.31 mmol, 98% yield) as a solid. 1H NMR (400 MHz, 6) 5 ppm 8.61 - 8.72 (m, 1 H), 8.08 (s, 1 H), 6.95 - 7.17 (m, 1 H), 2.48 (s, 2 H), 1.38 (s, 9 H).
MS (ESI) m/z 276.0 [M+1]+ and 278.2 [M+1]+.
] E. 2-(tert—Butylamin0)(methylthi0)pyrimidine—5-carb0nitrile.
-Bromo-N-tert-butyl(methylthio)pyrimidinamine (3400 mg, 12.31 mmol), zinc dust (201 mg, 3.08 mmol), zinc cyanide (940 mg, 8.00 mmol), 1,1'-bis-(diphenylphosphino)- ferrocene (552 mg, 0.985 mmol), tris(dibenzylideneacetone)dipalladium(0) (564 mg, 0.616 mmol), and DMF (20.5 mL) were ed and heated at 90 oC overnight under nitrogen. The reaction mixture was d with 125 mL ethyl acetate and 50 mL of water and then filtered through a pad of Celite. The layers of filtrate were separated and the aqueous layer was ted with 75 mL ethyl acetate. The combined ethyl acetate layers -l5l- ATI—25 14175v1 waewwkflwfih2x50mLbmm¢Mmhwmaflwmmmnmgmmmnwflfimjflmmdmm concentrated to an oil under reduced re. The crude oil was purified by silica gel chromatography (0-30% ethyl acetate/hexane) to afford 2-(tert-butylamino) (methylthio)pyrimidinecarbonitrile (2.26 g, 10.17 mmol, 83% yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.37 - 8.51 (m, 1 H), 7.85 - 8.10 (m, 1 H), 2.53 - 2.62 (m, 3 H), 1.41 (s, 9 H). MS (ESI) m/z 223.1 [M+1]+.
F. 2-(tert-Butylamin0)(methylthi0)pyrimidinecarboxamide. To a stirring solution of 2-(tert-butylamino)(methylthio)pyrimidinecarbonitrile (0.6 g, 2.70 mmol) in DMSO (7 mL) was added 6M aqueous sodium hydroxide solution (2.249 mL, 13.49 mmol) and 30% aqueous hydrogen peroxide (1.530 mL, 13.49 mmol) solution at 0 CC. Then the mixture was stirred at 50 CC for 15 min. The on mixture was poured into water (40 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and ed. Concentration of the filtrate under reduced pressure gave the crude product, which was purified by silica gel column tography (5% methanol in DCM) to afford 2-(tert-butylamino)(methylthio)pyrimidinecarboxamide (0.462 g, 1.922 mmol, 71.2% yield) as a white solid. MS (ESI) m/z 241.0 [M+1]+.
G. 2-(tert-Butylamin0)—4-(methylsulfinyl)pyrimidine—5—carboxamide.
To a stirring solution of 2-(tert—butylamino)(methylthio)pyrimidinecarboxamide (0.1 g, 0.416 mmol) in chloroform (12 mL) was added portion-wise yl lsulfonyl)-1,2-oxaziridine (0.130 g, 0.499 mmol). The resulting pale yellow solution was stirred at ambient temperature overnight. The reaction solution was concentrated under reduced re to give the crude product as a white solid. Ethyl acetate (1 mL) was added and the slurry was stirred at room temp for 1 h. The solids were filtered, washed with ethyl acetate, and dried under vacuum to afford 2-(tert-butylamino)- 4-(methylsulfinyl)pyrimidinecarboxamide (0.092 g, 0.359 mmol, 86% yield).
MS (ESI) m/z 257.3 [M+1]+.
H. 2-(tert-Butylamin0)—4-((1R,3R,4R)—3-hydr0xymethylcyclohexyl- amino)pyrimidine—5-carboxamide. To a stirring suspension of 2-(tert-butylamino) (methylsulf1nyl)pyrimidinecarboxamide (0.092 g, 0.359 mmol) and (1R,2R,5R) aminomethylcyclohexanol hydrochloride (0.065 g, 0.395 mmol) in DMF (2 mL) was ATI—2514175v1 added N—ethyl-N-isopropylpropanamine (0.157 mL, 0.897 mmol) and the on was heated to 90 oC overnight. The crude reaction mixture was concentrated under reduced pressure and then ice-cold water (20 mL) was added to the residue. The resulting mixture was vigorously stirred for 1 h and then the product was filtered, washed with water and dried under vacuum to afford 2-(tert-butylamino)((1R,3R,4R)—3-hydroxy methylcyclohexylamino)pyrimidinecarboxamide (0.074 g, 0.230 mmol, 64.1% . 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.94 (br. s., 1 H), 8.34 (s, 1 H), 6.69 (br. s., 1 H), 4.59 (d, J=5.47 Hz, 1 H), 3.87 (br. s., 1 H), 2.92 - 3.01 (m, 1 H), 2.14 (d, J=10.15 Hz, 1 H), 1.91 (d, J=11.71 Hz, 1 H), 1.67 (dd, J=13.28, 3.12 Hz, 1H), 1.07 - 1.24 (m, 3 H), 0.91 — 0.99 (m, 4 H). MS (ESI) m/z 322.3 [M+1]+.
Example 33: 2-(Cyclopropylamino)—4-((1R,3R,4R)—3-hydr0xy methylcyclohexylamin0)pyrimidinecarboxamide N \ NH2 A JL / N N NH . OH A. 2-(Cyclopropylamino)—4-((1R,3R,4R)—3-hydr0xymethylcyclo- hexylamin0)pyrimidinecarb0nitrile. To a solution of 4-((1R,3R,4R)hydroxy methylcyclohexylamino)(methylsulfonyl)pyrimidinecarbonitrile (362 mg, 1.166 mmol; synthesis described herein) in NMP (5.832 mL), was added ropanamine (0.485 mL, 7.00 mmol). The reaction was stirred at 80 0C for 5 h in a sealed vessel and then allowed to cool to ambient temperature overnight. The reaction mixture was concentrated under reduced pressure to an oil that was purified by silica gel chromatography (0-60% ethyl acetate/hexane) to afford 2-(cyclopropylamino) ((1R,3R,4R)—3-hydroxymethylcyclohexylamino)pyrimidinecarbonitrile (265 mg, 0.922 mmol, 79% yield) as a solid. 1H NMR (400 MHz, 6) 8 ppm 6.99 - 8.32 (m, 3 H), 4.54 (d, J=5.08 Hz, 1 H), 3.85 - 4.13 (m, 1 H), 2.80 - 3.03 (m, 1 H), 2.58 - 2.75 (m, 1 H), 1.50 - 2.08 (m, 3 H), 1.03 - 1.42 (m, 3 H), 0.88 (d, J=6.25 Hz, 4 H), 0.29 - 0.68 (m, 4 H). MS (ESI) m/z 288.1 [M + 1]+.
ATI—2514175v1 B. 2-(Cyclopropylamino)—4-((1R,3R,4R)—3-hydr0xymethylcyclo- hexylamin0)pyrimidinecarb0xamide. 2-(Cyclopropylamino)((lR,3R,4R) hydroxymethylcyclohexylamino)pyrimidinecarbonitrile (265 mg, 0.922 mmol) was ved in DMSO (9.222 mL). To the solution was added 10 drops of 50% aqueous sodium hydroxide followed by 10 drops of 30% aqueous hydrogen peroxide at room temperature. The resulting on mixture was stirred at 50 CC for 2 h and was then added slowly to 60 mL of ice water. The resulting precipitate was stirred for 30 min, filtered, and washed with water. The solids were dried in a vacuum oven overnight at 45 CC to afford 2-(cyclopropylamino)((1R,3R,4R)hydroxymethylcyclohexyl- amino)pyrimidinecarboxamide (229 mg, 0.750 mmol, 81% yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.73 - 8.95 (m, l H), 8.18 - 8.42 (m, l H), 7.17 - 7.37 (m, l H), 4.39 - 4.62 (m, l H), 3.75 - 3.99 (m, l H), 2.81 - 3.06 (m, l H), 2.53 - 2.74 (m, l H), 2.09 - 2.34 (m, l H), 1.82 - 2.05 (m, l H), 1.50 - 1.69 (m, l H), 0.99 - 1.24 (m, 3 H), 0.81 - 0.98 (m, 4 H), 0.50 - 0.67 (m, 2 H), 0.26 - 0.47 (m, 2 H). MS (ESI) m/z 306.3 [M + l]+.
Example 34: t-Butylamin0)—4-((1R,3R)—3-hydr0xy-4,4- dimethylcyclohexylamin0)pyrimidinecarb0xamide >L )L /N \ NH2 N N NH A. 4,4-Dimethylcyclohex-Z-enol. Sodium borohydride (5.03 g, 133 mmol) was added portion-wise to a stirred solution of 4,4-dimethylcyclohexenone (15.0 g, 121 mmol) in ol (403 mL) in a water bath. Once addition was complete, the solution was allowed to stir under a nitrogen atmosphere at room temperature for 3 h. The reaction mixture was diluted with water (150 mL) and the majority of the methanol was d under reduced re. The aqueous layer was extracted with ethyl acetate (2 x 200 mL). The combine ethyl acetate extracts were dried over anhydrous magnesium sulfate, filtered, and concentrated to an oil under reduced pressure to afford methylcyclohexenol (13.4 g, 106 mmol, 88% yield). 1H NMR (400 MHz, —154— ATI—2514l75v1 6) 8 ppm 5.41 - 5.57 (m, 1 H), 5.24 - 5.42 (m, 1 H), 4.61 (d, J=5.47 Hz, 1 H), 3.82 - 3.99 (m, 1 H), 1.65 - 1.83 (m, 1 H), 1.21 - 1.59 (m, 3 H), 0.93 (d, J=15.23 Hz, 6 H).
B. 2-(4,4-Dimethylcyclohexenyl)isoindoline—1,3-di0ne. Resin supported nylphosphine (32.7 g, 106 mmol), oline-1,3-dione (17.19 g, 117 mmol), and 4,4-dimethylcyclohexenol (13.4 g, 106 mmol) were combined and THF (197 mL) was added. The mixture was stirred and cooled to 0 0C, then diisopropyl arboxylate (21.09 mL, 107 mmol) was added drop-wise over 2 min. The reaction was stirred and allowed to warm slowly to ambient temperature overnight. The reaction mixture was filtered and then concentrated to a yellow solid under reduced pressure. Ethyl acetate (200 mL) was added and the resulting solids were filtered off again. The ethyl acetate filtrate was washed with water (100 mL), the aqueous layer extracted with ethyl e (100 mL), the combined organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated to an oil that was purified by silica gel chromatography (0-20% ethyl acetate/hexane) to afford 2-(4,4-dimethylcyclohexenyl)isoindoline-1,3- dione (11.59 g, 45.4 mmol, 42.8% yield) as an oil. 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.84 (s, 4 H), 5.51 - 5.62 (m, 1 H), 5.43 (d, J=10.15 Hz, 1 H), 4.59 - 4.74 (m, 1H), 2.14 - 2.30 (m, 1 H), 1.67 - 1.81 (m, 1 H), 1.44 - 1.68 (m, 2 H), 1.09 (s, 3 H), 1.01 (s, 3 H); MS (ESI) m/z 256.2 [M + 1]+.
C. Mixture of 2-((1R,ZS,3S)br0m0hydr0xy-4,4- dimethylcyclohexyl)is0indoline-1,3-di0ne, 2-((1S,2R,3R)—2-br0m0hydr0xy-4,4- dimethylcyclohexyl)is0indoline-1,3-di0ne, 2-((1R,2R,3R)—3-br0m0hydr0xy-4,4- dimethylcyclohexyl)is0indoline-1,3-di0ne, 2-((1S,ZS,3S)br0m0hydr0xy-4,4- dimethylcyclohexyl)is0indoline-1,3-di0ne. To a solution of 2-(4,4-dimethylcyclohex enyl)isoindoline-l,3-dione (11.59 g, 45.4 mmol) in chloroform (110 mL) and ethanol (3.83 mL) was added N—bromosuccinimide (10.34 g, 58.1 mmol) as a solid over a few min at ambient temperature. After addition was complete, the reaction e was stirred at ambient temperature ght under nitrogen. The reaction e was washed with 1 M aqueous solution of sodium thiosulfate (100 mL). The aqueous layer was extracted with chloroform (100 mL) and the ed organic layers were dried over anhydrous magnesium sulfate, filtered and concentrated to an oil. To this oil was added 150 mL THF (150 mL) and a 1N aqueous solution of hydrochloric acid (30 mL). The resulting mixture ATI—25 14175V1 2012/034349 was d at ambient temperature for 2 h. The THF was removed under reduced pressure and the remaining aqueous layer was d with ethyl e (150 mL) and 75 mL of a 1:1 water:saturated aqueous sodium bicarbonate on e. The layers were separated and the aqueous was back-extracted aqueous with ethyl acetate (75 mL). The ed ethyl acetate layers were washed with brine (50 mL), dried over anhydrous magnesium sulfate, filtered and concentrated to an oil that was purified by silica gel chromatography (0-25% ethyl acetate/hexane) to afford a mixture of 2-((1R,2S,3S) bromohydroxy-4,4-dimethylcyclohexyl)isoindoline- 1 ,3-dione, 2-((1 S,2R,3R)—2-bromohydroxy-4,4-dimethylcyclohexyl)isoindoline- 1 ,3 -dione, 2-((1R,2R,3R)bromo hydroxy-4,4-dimethylcyclohexyl)isoindoline-1,3-dione, and 2-((1S,2S,3S)bromo hydroxy-4,4-dimethylcyclohexyl)isoindoline-1,3-dione (11.4 g, 32.4 mmol, 71.3% yield).
MS (ESI) m/z 374.5 [M + 11+ and 376.5 [M + 11+.
D. Mixture 2-((1R,3R)—3-hydroxy-4,4-dimethylcyclohexyl)is0indoline- 1,3-di0ne and ,3S)hydroxy-4,4-dimethylcyclohexyl)is0indoline-1,3-di0ne. A mixture of 2-((1R,2S ,3 S)bromohydroxy-4,4-dimethylcyclohexyl)isoindoline-1 ,3 - dione, 2-((1 S,2R,3R)—2-bromohydroxy-4,4-dimethylcyclohexyl)isoindoline-1 ,3 -dione, 2-((1R,2R,3R)bromohydroxy-4,4-dimethylcyclohexyl)isoindoline-1 ,3 -dione, and 2-((1S,2S,3S)—3-bromohydroxy-4,4-dimethylcyclohexyl)isoindoline-1,3-dione (5 .7 g, 16.18 mmol) was dissolved in e (90 mL) and methanol (9 mL). To the solution was added tributyltin hydride (5.66 mL, 21.04 mmol) over 10 min via syringe under nitrogen, followed by azobisisobutyronitrile (0.266 g, 1.618 mmol) in one portion. The mixture was allowed to stir at reflux under a nitrogen atmosphere overnight. The reaction mixture was concentrated under reduced pressure to afford a residue that was purified by silica gel chromatography (0-50% ethyl acetate/hexane) yielding a mixture of 2-((1R,3R) hydroxy-4,4-dimethylcyclohexyl)isoindoline-1,3-dione and 2-((1S,3S)hydroxy-4,4- dimethylcyclohexyl)isoindoline-1,3-dione (0.9 g, 3.29 mmol, 20.35% . 1H NMR (400 MHz, DMSO-d6) 5 ppm 7.82 (s, 4 H), 4.51 - 4.80 (m, 1 H), 3.80 - 4.03 (m, 2 H), 2.84 - 3.11 (m, 1 H), 1.49 - 1.67 (m, 3 H), 1.37 - 1.47 (m, 1 H), 1.22 - 1.35 (m, 1 H), 1.13 (s, 3 H), 0.80 - 0.93 (m, 3 H). MS (ESI) m/z 274.0 [M + 1]+. The regiochemistry ofthe product mixture was verified by small molecule X-ray crystal diffraction. The X-ray structure was solved as follows. The single crystal X-ray diffraction studies were carried ATI—25 14175v1 out on a Bruker Kappa APEX-II CCD diffiactometer equipped with Mo Kg radiation (7t = 3 A). Crystals were grown by vapor ion of pentane into a DCM solution. A 0.25 x 0.20 x 0.05 mm ess plate was mounted on a Cryoloop with ne oil. Data were ted in a nitrogen gas stream at 90(2) K using (I) and as scans. Crystal-to- or distance was 60 mm and exposure time was 20 seconds per frame using a scan width of 0.50. Data collection was 100% complete to 25.000 in 6’. A total of 10827 reflections were collected covering the indices, -21<=h<=12, -7<=k<=8, -28<=l<=29. 2892 reflections were found to be symmetry ndent, with a Rint of 0.0821. Indexing and unit cell refinement indicated a ive, orthorhombic lattice. The space group was found to be Pbcn. The data were integrated using the Bruker SAINT software program and scaled using the SADABS re m. Solution by direct methods (SHELXS) produced a complete phasing model consistent with the proposed structure of a mixture of the enantiomeric pair 2—((1R,3R)—3-hydroxy-4,4-dimethylcyclohexyl)isoindoline-1,3- dione and 2-((1S,3S)—3-hydr0xy-4,4-dimethylcyclohexyl)is0indoline-1,3-di0ne.
All non-hydrogen atoms were refined anisotropically by full-matrix least- squares (SHELXL-97). All hydrogen atoms were placed using a riding model. Their positions were constrained relative to their parent atom using the appropriate HFIX command in SHELXL-97.
E. Mixture of (1R,5R)—5-amin0-2,2-dimethylcyclohexanol hydrochloride and (1 S,5S)—5-amin0-2,2-dimethylcyclohexanol hydrochloride. To a mixture of 2-((1R,3R)—3-hydroxy-4,4-dimethylcyclohexyl)isoindoline-1 ,3-dione and ,3S)hydroxy-4,4-dimethylcyclohexyl)isoindoline-1,3-dione (1.1 g, 4.02 mmol) in ethanol (50 mL) was added hydrazine hydrate (0.195 mL, 4.02 mmol). The resulting solution was allowed to stir at reflux overnight under a nitrogen atmosphere. The reaction was cooled to ambient temperature and then the pH was adjusted to < 2 via addition of concentrated aqueous hydrochloric acid. The precipitate was filtered off, rinsed with ethanol, and then the resulting filtrate was concentrated to 20 mL under reduced pressure.
An equal volume of water was added and this mixture was stirred at ambient temperature for 15 min. The solids were filtered off, rinsed with water, and the filtrate was concentrated under reduced pressure to give a solid that was dried in a vacuum oven for a ATI—2514175v1 few hours to afford a mixture of (1R,5R)amino-2,2-dimethylcyclohexanol hydrochloride and (1S,5S)amino-2,2-dimethylcyclohexanol hydrochloride (835 mg, 4.65 mmol, 115% yield) that was used without further purification. 1H NMR (400 MHz, DMSO-dg) 5 ppm 7.87 - 8.21 (m, 3 H), 2.98 - 3.23 (m, 1 H), 2.75 - 3.02 (m, 1 H), 1.49 - 1.92 (m, 2H), 1.26 - 1.47 (m, 3 H), 0.96 - 1.16 (m, 1 H), 0.48 - 0.93 (m, 6 H) F. 2-(tert-Butylamin0)—4-((1R,3R)—3-hydr0xy-4,4-dimethylcyclohexyl- amin0)pyrimidinecarboxamide and 2-(tert-butylamin0)—4-((1S,3S)hydr0xy-4,4- dimethylcyclohexylamin0)pyrimidine—5-carb0xamide. To a stirring suspension of 2-(tert-butylamino)(methylsulfinyl)pyrimidinecarboxamide (375 mg, 1.46 mmol) and a mixture of )amino-2,2-dimethylcyclohexanol hydrochloride and (1S,5S)- -amino-2,2-dimethylcyclohexanol hydrochloride (342 mg, 1.902 mmol) in DMF (4.877 mL) was added DIEA (0.767 mL, 4.39 mmol) and the reaction was heated to 90 oC overnight. The crude reaction mixture was concentrated under reduced pressure and then purified using reverse-phased semi-preparative HPLC (5-80% acetonitrile + 0.1% TFA in water + 0.1% TFA, over 30 min). Fractions containing product were concentrated under reduced pressure and redissolved in a mixture of ethyl acetate (125 mL) and saturated aqueous sodium bicarbonate solution (50 mL). The layers were separated and the aqueous layer was extracted with ethyl e (75 mL). The combined ethyl acetate layers were dried over anhydrous magnesium sulfate, filtered and concentrated under reduced pressure. The resulting e was redissolved in a methanol (5 mL), passed over an Varian StratoSpheres HCO3 resin SPE tube for TFA l (0.9 mmol onate equivalent), and then concentrated under d pressure to afford an oil that was triturated with diethyl ether and reconcentrated to give solids. The solids were dried for a few hours in a vacuum oven at 45 CC to afford a mixture of 2-(tert-butylamino) ((1R,3R)—3-hydroxy-4,4-dimethylcyclohexylamino)pyrimidinecarboxamide and 2-(tert- butylamino)((1 S ,3 ydroxy-4,4-dimethylcyclohexylamino)pyrimidine-5 - carboxamide (373 mg, 1.112 mmol, 76% yield).
] G. SFC Separation of a mixture of 2-(tert-butylamin0)—4-((1R,3R) hydroxy-4,4-dimethylcyclohexylamin0)pyrimidine—5-carb0xamide and 2-(tert- butylamino)—4-((1S,3S)hydroxy-4,4-dimethylcyclohexylamino)pyrimidine—5- carboxamide. A mixture of the enantiomers 2-(tert-butylamino)((1R,3R)hydroxy- ATI—25 14175v1 4,4-dimethylcyclohexylamino)pyrimidinecarboxamide and t-butylamino) ((1S,3S)hydroxy-4,4-dimethylcyclohexylamino)pyrimidine-5 -carboxamide (373 mg) was separated by preparative chiral SFC utilizing a ChiralPak AD-H, 250><30mm ID. column with an isocratic 32% ethanol + 0.1% ammonium hydroxide in C02 gradient at 50 mL/min flow rate. The faster eluting isomer was denoted as peak 1 and 155 mg (0.462 mmol) was obtained. The slower eluting isomer was denoted as peak 2 and 170.0 mg (0.502 mmol) was obatined. Peak 1: 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.82 - 9.11 (m, 1 H), 8.34 (s, 1 H), 6.50 - 6.82 (m, 1 H), 4.40 - 4.72 (m, 1 H), 3.72 - 4.01 (m, 1 H), 3.01 - 3.25 (m, 1 H), 1.83 - 1.99 (m, 1 H), 1.65 - 1.81 (m, 1 H), 1.37 (s, 13 H), 0.89 - 0.95 (m, 3 H), 0.72 - 0.88 (m, 3 H). MS (ESI) m/z 336.2 [M+1]+. Peak 2: 1H NMR (400 MHz, DMSO-dg) 5 8.95 (s, 1H), 8.34 (s, 1H), 6.68 (s, 1H), 4.52 (d, J = 4.9 Hz, 1H), 3.86 (s, 1H), 3.15 (dt, J: 11.3, 4.6 Hz, 1H), 1.90 (d,.]= 11.9 Hz, 1H), 1.72 (s, 1H), 1.40 (s, 1H), 1.36 (s, 9H), 1.34 - 1.05 (m, 3H), 0.91 (s, 3H), 0.82 (s, 3H). MS (ESI) m/z 336.2 [M+1]+. By SAR y comparison with similar compounds ofknown absolute stereochemistry provided herein, Peak 1 was assigned as 2-(tert-butylamin0)—4-((1S,3S)- oxy-4,4-dimethylcyclohexylamin0)pyrimidinecarboxamide. Peak 2 was assigned as 2-(tert-butylamin0)—4-((1R,3R)—3-hydroxy-4,4-dimethylcyclohexylamin0)- dine-S-carboxamide.
Example 35: 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(isopr0pylamin0)pyrimidine carboxamide HO/“O‘ I‘ll/\kaHZ A. Ethyl 4-(is0propylamin0)(methylthi0)pyrimidinecarboxylate.
Ethyl 4-chloro(methylthio)pyrimidinecarboxylate (900 g, 3.87 mol) ethanol (12 L), DIEA (876 mL, 5.05 mol), and isopropylamine (379 mL, 4.45 mol) were combined and mixed at t temperature for 4 h. An onal amount of isopropylamine (50 mL, 0.59 mol) was added and the mixture was stirred overnight at ambient ature. The reaction was concentrated under reduced pressure and the crude product was diluted with water (2 L). The aqueous layer was extracted with chloroform (2 x 3 L). The combined ATI—25 14175v1 organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced re to afford ethyl propylamino)(methylthio)pyrimidine carboxylate (1042 g, 4.08 mol, >100% yield, inated with trace DIEA) as a pale brown oil that was used without fiarther purification. MS (ESI) m/z 256.4 [M+1]+.
B. 4-(Isopropylamino)—2-(methylthi0)pyrimidinecarboxylic acid.
Ethyl 4-(isopropylamino)(methylthio)pyrimidinecarboxylate (1042 g, 4.08 mol, >100%, contaminated with trace DIEA) was dissolved in ethanol (10 L) and to the resulting solution was added a 2M aqueous sodium hydroxide solution (3.87 L, 7.74 mol).
The resulting mixture was stirred overnight at ambient temperature and then concentrated under d pressure. The resulting residue was diluted with 2 L of water and then washed with methyl t-butyl ether (2 x 1.2 L). The pH of the aqueous layer was adjusted to pH 4.2-4.5 with a 2N aqueous hydrochloric acid solution. The resulting solids were collected by filtration, washed with water (2 L) and s (2 L) and then dried overnight in a vacuum oven at 45 ° C to afford 4-(isopropylamino)(methylthio)pyrimidine carboxylic acid (848 g, 96.5% yield over 2 steps) as an off-white solid. MS (ESI) m/z 228.1 [M+1]+.
C. 4-(Isopr0pylamin0)—2-(methylthi0)pyrimidine—5-carboxamide. To a solution of 4-(isopropylamino)(methylthio)pyrimidinecarboxylic acid (180 g, 0.792 mol) and HOBt (123 g, 0.911 mol) in anhydrous THF (6.9 L) was added drop-wise a mixture of EDC (174.6 g, 0.911 mol) in acetonitrile (3.7 L) at ambient temperature. The mixture was stirred for 1 h at ambient temperature and then an aqueous ammonium hydroxide solution (989 mL, 28-30% concentrated, 10 eq) was added drop-wise over min. The resulting mixture was heated to gentle reflux for 3 h and then concentrated under reduced pressure. The remaining e was diluted with a saturated aqueous sodium bicarbonate on (5 L) and then extracted twice with ethyl acetate (9 L and 2 L tively). The combined organic layers were washed with a ted aqueous sodium bicarbonate solution (4 L), water (4 L), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford 4-(isopropylamino)(methylthio)- pyrimidinecarboxamide (171.2 g, 95.5% yield) as a solid. MS (ESI) m/z 227.4 [M+1]+.
D. 4-(Is0pr0pylamin0)—2-(methylsulfinyl)pyrimidine—5-carb0xamide.
To a stirring solution of 4-(isopropylamino)(methylthio)pyrimidinecarboxamide ATI—25 14175v1 (170 g, 0.751 mol) in chloroform (22 L) was added portion-wise 3-phenyl (phenylsulfonyl)-1,2-oxaziridine (235.6 g, 0.902 mol) and the resulting solution was stirred at ambient ature overnight. An additional amount of 3-phenyl (phenylsulfonyl)-1,2-oxaziridine (19.6 g, 0.075 mol) was added and the mixture was then stirred ght at ambient temperature. The reaction on was concentrated under reduced pressure to give the crude product as a white solid. The solids were triturated with ethyl acetate (1.5 L) at ambient temperature for 1 h, filtered, washed with ethyl e (250 mL), and dried in a vacuum oven overnight at 45 CC to afford 4-(isopropylamino) (methylsulf1nyl)pyrimidinecarboxamide (167.7 g, 92% yield) as white solid. MS (ESI) m/z 243.2 [M+1]+.
E. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(is0pr0pylamin0)— pyrimidine—5-carb0xamide. A mixture of (1r,4r)aminocyclohexanol (262.4 g, 2.278 mol) and anhydrous DMF (79 mL) was heated to 100 0C. To this mixture was added portion-wise 4-(isopropylamino)(methylsulf1nyl)pyrimidinecarboxamide (157.7 g, 0.651 mol). The resulting mixture was allowed to stir a under nitrogen atmosphere at 110 OC ght. The reaction mixture was concentrated under reduced pressure to remove DMF. To the remaining residue was added water (2 L) and the mixture was extracted with ethyl acetate (3 x 2 L). The combined organic extracts were washed with water (2 x 2 L), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure until the remaining volume reached to 500 mL. The ing material was ed and washed with ethyl acetate (800 mL). The product was dried at 45 CC in a vacuum oven overnight. The product was triturated with water (2.1 L) for 4.5 h at 50 OC, filtered and then dried at 45 CC in a vacuum oven overnight to afford 2-((1r,4r) ycyclohexylamino)(isopropylamino)pyrimidinecarboxamide (134 g, 70% yield); 1H NMR (499 MHz, DMSO-d6) 8 ppm 8.71 - 9.01 (m, 1 H), 8.33 (br. s., 1 H), 7.42 - 7.70 (m, 1 H), 6.67 - 7.12 (m, 2 H), 4.51 (d, J=3.94 Hz, 1 H), 3.98 - 4.25 (m, 1 H), 3.51 - 3.77 (m, 1 H), 3.34 - 3.41 (m, 1 H), 1.82 (br. s., 4 H), 1.06 - 1.33 (m, 10 H). MS (ESI) m/z 294.1 [M+1]+.
ATI—25 14175v1 Example 36: (1R,4r)—4-(5-Cyan0((1R,3S)hydr0xymethylcyclohexylamin0)— pyrimidin-Z-ylamino)—N,N-dimethylcyclohexanecarboxamide \TJLIIQ )NL/jCN N N NH A. tert-Butyl )—4-(dimethylcarbamoyl)cyclohexylcarbamate. A mixture of )(tert-butoxycarbonylamino)cyclohexanecarboxylic acid (7.5 g, 30.8 mmol), EDC (20.09 g, 105 mmol), HOBt (16.05 g, 105 mmol) in NMP (60 mL) was stirred at room temperature for 2 h. A solution of dimethylamine (46.2 mL, 92 mmol) (2.0 M in THF) was added and the reaction mixture was stirred for 2 days. Water and ethyl acetate were added and the phases separated. The organic phase was washed twice with saturated aqueous potassium carbonate on, followed by 1M aqueous hydrogen chloride solution and brine. The organic phase was dried over ous magnesium sulfate, filtered, and the solvent was evaporated to give the desired product (5.8 g, 19.3 mmol, 96% yield). MS (ESI) m/z 271.4 .
B. (1r,4r)—4-Amin0-N,N-dimethylcyclohexanecarboxamide hydrochloride. tert-Butyl (1r,4r)(dimethylcarbamoyl)cyclohexylcarbamate (8.27 g, 30.6 mmol) was dissolved in e (110 mL) and hydrochloric acid (38.5 mL, 1267 mmol) was added. The reaction mixture was stirred at room temperature for 4 h, toluene was added and the solvent evaporated to give the product (6.0 g, 29.0 mmol, 95% yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.22 (br. S., 3 H), 3.01 (s, 3 H), 2.86 — 2.98 (m, 1 H), 2.79 (s, 3 H), 1.93 — 2.04 (m, 2 H), 1.72 (d, J=7.81 Hz, 2 H), 1.31 — 1.48 (m, 4 H). MS (ESI) m/z 171.4 [M+1]+ C. )—4-(5-Br0m0(methylthi0)pyrimidinylamin0)-N,N- dimethylcyclohexanecarboxamide. To a stirring suspension of 5-bromochloro (methylthio)pyrimidine (1.5 g, 6.26 mmol) in ethanol (7.5 mL) was added (1r,4r)amino-N,N— dimethylcyclohexanecarboxamide hloride (1.618 g, 7.83 mmol) and DIEA (3.28 mL, 18.79 mmol). The resulting mixture was stirred at 80 OC overnight. The ethanol was removed under reduced pressure and the remaining residue was purified using silica gel chromatography (0-90% ethyl acetate in hexanes) to afford (1r,4r)(5-bromo(methylthio)pyrimidin ATI—25 14175v1 ylamino)-N,N-dimethylcyclohexanecarboxamide (1085 mg, 2.91 mmol, 46.4% yield). MS (ESI) m/z 373.0 [M+1]+, 375.2 [M+1]+.
D. (1r,4r)—4-(5-Cyan0-4—(methylthi0)pyrimidinylamin0)-N,N- dimethylcyclohexanecarboxamide. (1r,4r)(5-Bromo(methylthio)pyrimidinylamino)— N,N-dimethylcyclohexanecarboxamide (500 mg, 1.339 mmol), zinc dust (21.90 mg, 0.335 mmol), zinc cyanide (102 mg, 0.871 mmol), 1,1'-bis-(diphenylphosphino)-ferrocene (60.0 mg, 0.107 mmol), tris(dibenzylideneacetone)dipalladium(0) (61.3 mg, 0.067 mmol), and N,N—dimethylacetamide (2.178 mL) were combined and the resulting mixture was mixed and heated at 90 CC ght. The reaction mixture was diluted with ethyl acetate (125 mL) and water (50 mL) and filtered through a pad of Celite. The filtrate layers were separated and the aqueous layer was extracted with ethyl acetate (75 mL). The combined ethyl acetate layers were washed with brine (50 mL), dried over anhydrous ium e, filtered, and concentrated to an oil under reduced pressure. The oil was purified using silica gel chromatography (0-10% methanol in DCM) to afford (1r,4r)(5-cyano(methylthio)pyrimidinylamino)-N,N— dimethylcyclohexanecarboxamide (400 mg, 1.252 mmol, 93% yield) as a solid. MS (ESI) m/z 320.2 [M+1]+.
E. (1r,4r)—4-(5-Cyan0(methylsulfonyl)pyrimidinylamin0)-N,N- dimethylcyclohexanecarboxamide. To a solution of (1r,4r)(5-cyano(methylthio)- pyrimidinylamino)-N,N-dimethylcyclohexanecarboxamide (400 mg, 1.252 mmol) in NMP (6.261 mL) at 0 °C was added mCPBA (702 mg, 3.13 mmol). The reaction was stirred for 1 h at 0 CC and then at ambient temperature overnight. The solution was then used ly in the next step without further purification assuming a theoretical yield of )(5 -cyano (methylsulfonyl)pyrimidinylamino)-N,N-dimethylcyclohexanecarboxamide (440 mg, 1.25 mmol, 100% yield). MS (ESI) m/z 352.3 [M+1]+.
F. (1R,4r)—4-(5-Cyan0((1R,3S)—3-hydr0xymethylcyclohexylamin0)- pyrimidin-Z-ylamino)—N,N-dimethylcyclohexanecarboxamide. To a solution of (1r,4r)—4-(5- cyano(methylsulfonyl)pyrimidinylamino)-N,N-dimethylcyclohexanecarboxamide (440 mg, 1.252 mmol) in NMP (6.261 mL) was added (1S,3R)aminomethylcyclohexanol hloride (259 mg, 1.565 mmol) and DIEA (1.312 mL, 7.51 mmol). The reaction was d at 80 CC for 4 h and then allowed to cool to ambient ature overnight The crude reaction mixture was concentrated under reduced pressure and purified using reverse-phased ATI—25 14175v1 semi-preparative HPLC (5-60% itrile + 0.1% TFA in water + 0.1% TFA, over 30 min).
Fractions ning product were concentrated under reduced pressure. The resulting residue was redissolved in a methanol (5 mL), passed over Varian StratoSpheres HCO3 resin SPE tubes for TFA removal (0.9 mmol bicarbonate equivalent per tube), concentrated under reduced pressure, and dried overnight in a vacuum oven at 45 0C to afford (1R,4r)(5-cyano ((1R,3S)hydroxymethylcyclohexylamino)pyrimidinylamino)-N,N-dimethyl- cyclohexanecarboxamide (138 mg, 0.345 mmol, 27.5% yield). 1H NMR (400 MHz, DMSO-d6) ppm 8.11 (s, 1 H), 7.58 (d, J=7.81 Hz, 2 H), 4.70 - 5.01 (m, 1 H), 4.12 - 4.42 (m, 2 H), 3.52 - 3.79 (m, 1 H), 3.00 (s, 3 H), 2.79 (s, 3 H), 1.21 - 2.07 (m, 16 H), 1.03 - 1.20 (m, 3 H). MS (ESI) m/z 401.5 [M+1]+.
Example 37: ,4r)—4-Hydr0xycyclohexylamin0)(tert—pentylamin0)pyrimidine carboxamide A. 2-(Methylthi0)—4-(tert-pentylamino)pyrimidine—5-carbonitrile. 4-Chloro (methylthio)pyrimidinecarbonitrile (500 mg, 2.69 mmol) was ved in DMF (5 mL), ylbutanamine (0.378 mL, 3.24 mmol) and DIEA (1.411 mL, 8.08 mmol) were added and the reaction was heated at 70 oC overnight. LCMS showed the desired product mass as the dominant peak and no starting material remaining. The reaction was removed from heat and partitioned between ethyl acetate and water. The organic layer was washed once with brine before drying over magnesium e, filtering, and condensing. The crude material was purified by silica gel chromatography (0-80% ethyl acetate in hexane over 1650 mL; 40 mL/min). t fractions were combined, evaporated, and dried under high vacuum to afford 2- (methylthio)(tert-pentylamino)pyrimidinecarbonitrile (359 mg, 1.519 mmol, 56.4% yield) as a white solid; MS (ESI) m/z 236.9 [M+1]+ B. 2-(Methylsulfonyl)(tert-pentylamin0)pyrimidinecarb0nitrile. 2-(Methylthio)(tert-pentylamino)pyrimidinecarbonitrile (350 mg, 1.481 mmol) was dissolved in NMP (5 mL), and cooled to 0 0C before adding mCPBA (664 mg, 2.96 mmol) ATI—2514175v1 portion-wise. The reaction was kept at 0 CC and allowed to slowly warm to room temperature.
After 2 h, LCMS showed the desired product as the dominant peak and no starting material present. The crude reaction mixture was used directly in the next step assuming theoretical yield ofthe e; MS (ESI) m/z 269.2 [M+1]+ C. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(tert-pentylamino)pyrimidine—5- itrile. To the crude reaction mixture from the previous step was added ) aminocyclohexanol hloride (269 mg, 1.775 mmol) and DIEA (1.034 mL, 5.92 mmol).
The reaction was then heated at 70 oC overnight. LCMS showed the desired t as the dominant peak and no starting material remaining. The reaction was removed from heat, and then partitioned between ethyl e and water. The organic layer was washed once with brine before drying over magnesium sulfate, filtering, and concentrating under reduced pressure. The material was purified by semi-preparative HPLC (5-80% acetonitrile + 0.1% TFA in water + 0.1% TFA). The product fractions were combined and concentrated under reduced pressure to a volume <5 mL. The material was then neutralized with saturated sodium bicarbonate and ted with ethyl acetate (2x). The combined organic layers were dried over magnesium sulfate, filtered, and concentrated under reduced pressure. The resulting solid was dissolved in methanol and washed through a StratoSpheres SPE PL-HCO3 MP-Resin column, eluting with methanol. ation of the solvent under reduced pressure gave 2-((1r,4r) hydroxycyclohexylamino)(tert—pentylamino)pyrimidinecarbonitrile (200 mg, 0.659 mmol, 45% yield) as a white solid; 1H NMR (400 MHz, DMSO-dg) 5 ppm 8.07 - 8.24 (m, 1 H), 7.24 - 7.63 (m, 1 H), 5.67 - 5.95 (m, 1 H), 4.56 (d, J=4.69 Hz, 1 H), 3.35 - 3.60 (m, 2 H), 1.74 - 1.96 (m, 6 H), 1.09 - 1.43 (m, 10 H), 0.72 - 0.86 (m, 3 H); MS (ESI) m/z 304.1 [M+1]+ D. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(tert-pentylamino)pyrimidine—5- carboxamide. 2-((1r,4r)Hydroxycyclohexylamino)(tert-pentylamino)pyrimidine carbonitrile (141 mg, 0.465 mmol) was dissolved in DMSO (3 mL) and 10 drops of 50% aqueous sodium hydroxide and 10 drops of 30% aqueous hydrogen peroxide were added at room ature. The reaction was then heated at 50 0C. After 30 min, LCMS showed the desired product as the dominant peak and no ng material remaining. The reaction was removed from heat and slowly added to ~50 mL of ice water. The resulting material was allowed to mix for 3 h before filtering and drying under high vacuum at 60 CC overnight. 2-((1r,4r) hydroxycyclohexylamino)(tert—pentylamino)pyrimidinecarboxamide (125 mg, 0.389 mmol, ATI—25 14175v1 84% yield) was obtained; 1H NMR (400 MHz, DMSO-d6) 5 ppm 9.14 (s, 1 H), 8.31 (s, 1 H), 7.00 (d, J=7.81 Hz, 1 H), 4.53 (d, J=4.29 Hz, 1 H), 3.38 (d, J=3.51 Hz, 2 H), 1.74 - 1.92 (m, 6 H), 1.36 (s, 7 H), 1.13 - 1.31 (m, 4 H), 0.80 (t, J=7.42 Hz, 3 H); MS (ESI) m/z 322.0 [M+1]+.
Example 38: 4-(Bicyclo[1.1.1]pentanylamin0)—2-((1R,4R)—4- hydroxycyclohexylamino)pyrimidine—5-carboxamide A. Ethyl 4-(bicyclo[1.1.1]pentanylamin0)—2-(methylthi0)pyrimidine- 0xylate. A mixture of ethyl ro(methylthio)pyrimidinecarboxylate (200 g, 0.86 mol), bicyclo[l . l.1]pentanamine hydrochloride (171 g, 1.11 mol; prepared according to Org. Lett., 13(17): 4746-4748 (2011)) and DIEA (278 g, 2.15 mol) in ethanol (2.4 L) was stirred overnight at room temperature. After completion of the reaction, the solvents were concentrated under reduced pressure to give the crude product, which was diluted with water (1 L). The aqueous layer was extracted with chloroform (2 x 1 L) and the combined organic layers were washed with brine and dried over anhydrous sodium sulfate. Evaporation of the solvents afforded the d compound (231 g, 0.83 mol, 96% yield) as a pale brown oil which was contaminated with trace amounts of DIEA. This product was used in the next reaction without further purification; 1H NMR (400 MHz, CDClg) 5 ppm 8.61 (s, 2H), 4.30 (q, J: 7.2 Hz, 2H), 2.54 (s, 3H), 2.51 (s, 1H), 2.20 (s, 6H), 1.35 (t, J: 7.2 Hz, 3H).
B. 4-(Bicyclo[1.1.1]pentan-l-ylamin0)—2-(methylthi0)pyrimidine carboxylic acid. Ethyl 4-(bicyclo[1.1.1]pentanylamino)(methylthio)pyrimidine carboxylate (120 g, 0.43 mol) was dissolved in ethanol (1.5 L) followed by the addition of an aqueous sodium hydroxide on (530 mL, 1.06 mol, 2M) and the resulting mixture was stirred ght at room temperature. After that the solvent was evaporated under reduced pressure (<42 oC bath ature). The mixture was d with 500 mL of water and washed with tert-butyl methyl ether (2 x 500 mL). The s layer was treated with aqueous hydrochloric acid solution (2N) to pH 4.2-4.5. The resulting solids ATI—2514l75vl were ted by filtration followed by washing with water (500 mL) and hexanes (500 mL). The wet cake was dried under reduced pressure overnight at 45 0C to afford the desired compound (108 g, 0.43 mol, 100% yield) as an off-white solid (116 mg, 0.365 mmol, 54.7% yield); 1H NMR (400 MHz, CDgOD) 8 ppm 8.52 (s, 1H), 2.55 (s, 3H), 2.52 (s, 1H), 2.24 (s, 6H) C. 4-(Bicyclo[1.1.1]pentanylamin0)(methylthi0)pyrimidine carboxamide. To a stirring solution of yclo[1.1.1]pentanylamino) (methylthio)pyrimidinecarboxylic acid (108 g, 0.43 mol) and HOBt (67 g, 0.49 mol) in anhydrous THF (4 L) was added drop-wise a mixture of EDC (94 g, 0.49 mol) in acetonitrile (2 L) at room temperature. After the reaction mixture was stirred for 1 hr at room temperature, aqueous ammonium hydroxide (600 mL, 28-30% concentrated, 10 eq) was added drop-wise over 30 min and then the mixture was heated to gentle reflux for 3 h (60::5 0C). After that all solvents were removed under reduced pressure, the residual paste/solid was diluted with saturated aqueous sodium bicarbonate on (2 L) and the resulting suspension was filtered to afford the desired compound (100 g, 0.4 mol, 93% yield) as an off-white fluffy solid; MS(ESI): m/z 251.1 [M+1]+.
D. 4-(Bicyclo[1.1.1]pentanylamin0)(methylsulfinyl)pyrimidine—5- carboxamide. To a stirring solution of yclo[1.1.1]pentanylamino) (methylthio)pyrimidinecarboxamide (76 g, 0.304 mol) in form (7 L) was added 3-phenyl(phenylsulfonyl)-1,2-oxaziridine (143 g, 0.547 mol, 1.8 equiv.) and the resulting pale yellow solution was stirred at ambient temperature overnight. The reaction solution was concentrated under d re to give the crude product as a white solid, which was d with ethyl acetate (1.5 L) and the slurry was stirred at room temperature for 1 h. The resulting suspension was ed, washed with ethyl acetate, and dried under vacuum to afford the desired compound (73 g, 0.274 mol, 90% yield) as a white solid; MS(ESI): m/Z 267.0 [M+1]+.
E. 4-(Bicyclo[1.1.1]pentan-l-ylamino)—2-((1r,4r)—4-hydr0xycyclohexyl— amin0)pyrimidine—5-carb0xamide. A mixture of (1r,4r)aminocyclohexanol (151.4 g, 1.314 mol) and anhydrous DMF (500 mL) was heated at ~100 c’C. To this mixture was added portion-wise 4-(bicyclo[1.1.1]pentanylamino)(methylsulf1nyl)pyrimidine carboxamide (100 g, 0.376 mole) under nitrogen atmosphere and the reaction mixture was ATI—25 14175v1 stirred at 110 CC overnight. The reaction mixture was cooled to ~45 CC and DMF was removed under reduced pressure followed by addition of water (500 mL) and ethyl acetate (500 mL). The resulting white slurry was filtered and washed with ethyl acetate (50 mL).
The solid was dried at 55 0C under vacuum for 48 hrs to afford 100 g of the crude product.
In order to get rid of traces of organic solvent the crude product was suspended in water (1 L) and d for 4.5 h g the internal temperature at 50::1 c’C. The slurry was filtered at 45—50 0C and rinsed with water (50 mL). The wet cake was dried under vacuum at 45—50 0C for 48 h to afford the d compound (92 g, 0.289 mol, 77% yield); 1H NMR (CDgOD, 400 MHz): 8 ppm 8.26 (s, 1H), 3.79 (m, 1H), 3.56-3.58 (m, 1H), 2.47 (s, 1H), 2.18 (s, 6H), 1.98-2.06 (m, 4H), 1.32-1.39 (m, 4H); MS(ESI): m/z 318.1 [M+1]+.
Example 39: 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(1- cyclopropylamin0)pyrimidine—5-carb0xamide A. (1r,4r)—4-(5-Br0m0(methylthi0)pyrimidinylamin0)- cyclohexanol. A 3-L, 3-neck, round-bottom flask was equipped with a J-KEM temperature controller, a mechanical stirrer, and a nitrogen inlet. The flask was charged with 5-bromochloro(methylthio)pyrimidine (100 g, 417.5 mmol), (1r,4r) yclohexanol (76.4 g, 663.4 mmol), and ethanol (1 L). DIEA (109 mL, 626.3 mmol) was added, and the mixture was heated to reflux overnight. TLC (1 :1 hexanes/ethyl acetate) analysis after 20 h indicated complete reaction. The reaction was allowed to cool to room temperature. Water (300 mL) was added, and a precipitate gradually formed. The solid was filtered and washed with water to give 111.7 g of a white solid. The filtrate was extracted with ethyl acetate (3 X 300 mL). The combined organic layers were dried over sodium e, filtered, and concentrated to a semi-solid. The semi-solid was slurried in 1:1 hexanes/ethyl acetate and d to give an additional 12.1 g of a white solid. The two batches were combined to give 123.8 g (93%) of (1r,4r)- 4-(5 (methylthio)pyrimidinylamino)cyclohexanol as a white solid. MS (ESI) m/z 318, 320 [M+1]+.
ATI—25 14175v1 WO 45569 B. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(methylthi0)pyrimidine itrile. A 2-L, 3-neck, round-bottom flask was equipped with a J-KEM temperature controller, a mechanical stirrer, and a nitrogen inlet. The flask was charged under nitrogen with (1r,4r)(5-bromo(methylthio)pyrimidinylamino)cyclohexanol (123 .8 g, 389 mmol), zinc cyanide (29.2 g, 249 mmol), zinc dust (6.36 g, 97 mmol), and N,N—dimethylacetamide (478 mL). Tris(dibenzylideneacetone)dipalladium(0) (17.8 g, 0.05 equiv) and 1,1'-bis-(diphenylphosphino)-ferrocene (17.25 g, 0.08 equiv) were added, and the e was purged with nitrogen. The reaction was heated to 100 °C overnight.
TLC (2:1 ethyl acetate/hexanes) analysis after 17 h indicated complete reaction. The reaction was allowed to cool to room temperature and diluted with ethyl acetate (2 L). The mixture was filtered through a short Celite-pad, and the pad was washed with ethyl acetate (3 X 400 mL). The combined organic layers were washed with water (1 L) and brine (400 mL), dried over sodium sulfate, and filtered. The filtrate was concentrated to remove most of the solvent, and a beige precipitate formed. The solids were filtered and washed with ethyl acetate (2 X 50 mL) to give 55 g (54%) of ,4r)hydroxycyclohexyl- amino)(methylthio)pyrimidinecarbonitrile as a beige solid. MS (ESI) m/z 265 [M + 11+.
C. ,4r)—4-Hydr0xycyclohexylamin0)(methylsulfinyl)— pyrimidine—5-carb0nitrile and 2-((1r,4r)—4-hydr0xycyclohexylamin0)—4- (methylsulfonyl)pyrimidine—5-carb0nitrile. To a stirred colorless solution of 2-((1r,4r)- 4-hydroxycyclohexylamino)(methylthio)pyrimidinecarbonitrile (0.700g, 2.65 mmol) in NMP (10 mL) was added mCPBA (1.306 g, 5.83 mmol) at 0 CC. The reaction mixture was then stirred at room ature for 2 h until completion of the reaction as indicated by LCMS. The reaction mixture was carried on to the next step without further ation. MS (ESI) m/z 281.4 [M+1] + and 297.2 [M+1] 3 D. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(1-methylcyclopr0pyl- amin0)pyrimidine—5-carb0nitrile. To the reaction mixture of 2-((1r,4r) hydroxycyclohexylamino)(methylsulfinyl)pyrimidinecarbonitrile and ,4r) hydroxycyclohexylamino)(methylsulfonyl)pyrimidinecarbonitrile from the previous step was added DIEA (2.78 mL, 15.89 mmol) and 1-methylcyclopropanamine hympchbnde(0627g,583nnnoD.Therawfionwmssfinedat90oCibr16h. Upon ATI—25 14175v1 completion of reaction as indicated by LCMS and TLC the reaction e was concentrated and purified by silica gel chromatography using a gradient of 0% - 80% ethyl e in hexanes. The product fractions were combined and concentrated to afford 2-(( 1 s,4s)hydroxycyclohexylamino)(1 -methylcyclopropylamino)pyrimidine-5 - carbonitrile (0.147g, 19% yield) as a pale yellow solid. MS (ESI) m/z 288.2 [M+1]+.
E. 2-((1r,4r)Hydr0xycyclohexylamin0)(l-methylcyclopropyl- pyrimidine—5-carboxamide. ,4r)Hydroxycyclohexylamino)(1- methylcyclopropylamino)pyrimidinecarbonitrile (0.225 g, 0.783 mmol) was dissolved in DMSO (15 ml). Sodium hydroxide (50% wt, 175 uL, 0.78 3 mmol) and 30% hydrogen peroxide (175 uL, 1.543 mmol) were added to the reaction mixture at room temperature.
The reaction mixture was stirred at 50 CC for 1 h. Upon completion of the reaction, as indicated by LCMS and TLC, the reaction mixture was cooled to room temperature and was poured into 300 mL of ice water. The aqueous layer was extracted with 20% isopropanol in form (x3) and the combined organic layers were dried over anhydrous magnesium sulfate and concentrated. The crude e was purified by reverse phase silica gel chromatography using a gradient of 0% - 90% methanol in water.
The product fractions were combined and concentrated to afford 2-((1r,4r) hydroxycyclohexylamino)( 1 lcyclopropylamino)pyrimidinecarboxamide (0.181 g, 76% yield); 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.10 (s, 1H), 8.33 (s, 1H), 7.06 (d, .1: 7.42 Hz, 1H), 4.53 (d, .1: 4.30 Hz, 1H), 3.59 - 3.70 (m, 1H), 3.35 - 3.46 (m, 1H), 1.96 (d, .1: 9.76 Hz, 1H), 1.85 (d, .1: 9.76 Hz, 2H), 1.40 (s, 3H), 1.20 - 1.34 (m, 4H), 0.81 - 0.89 (m, 1H), 0.58 - 0.71 (m, 4H); MS (ESI) m/z 306.4 [M+1]+ Example 40: 4-((R)Cyclopr0pylethylamin0)((1r,4R)—4- hydroxycyclohexylamino)pyrimidine—5-carboxamide A. 2-((1r,4r)—4-Hydr0xycyclohexylamino)(methylthi0)pyrimidine carboxamide. A 2-L, 3-neck, round-bottom flask was equipped with a J-KEM temperature ATI—25 14175V1 2012/034349 controller, a mechanical r, and a nitrogen inlet. The flask was charged with 2-((1r,4r) hydroxycyclohexylamino)(methylthio)pyrimidinecarbonitrile (54 g, 204.3 mmol; synthesis described herein) and DMSO (270 mL), and the solution was cooled to 0 oC. Sodium hydroxide solution (170 mL, 1021 mmol, 6 M in water) and en peroxide on (99 mL, 1021 mmol, 35% in water) were slowly added to the reaction mixture. An rm to 30 °C was observed. The reaction was heated to 50 0C for 15 min. TLC (10% methanol/ethyl acetate) analysis indicated complete reaction. The mixture was cooled to 10 CC, diluted with water (800 mL), and stirred for 10 min. The mixture was filtered to give 18 g of a crude solid. To recover more product, the e was extracted with ethyl acetate (18 X 250 mL). The combined organic layers were washed with brine (75 mL), dried over sodium sulfate, filtered, and concentrated to give 33 g of a crude solid. The crude solids were combined and purified on silica gel g with 0-15% ol/ethyl acetate to give 26 g of an off-white solid. The solid was slurried in itrile and filtered to give 19.5 g (33%) of 2-((1r,4r) hydroxycyclohexylamino)(methylthio)pyrimidinecarboxamide as an off-white solid; MS (ESI) m/z 283.3 [M+1] B. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(methylsulfinyl)pyrimidine carboxamide and 2-((1r,4r)—4-hydroxycyclohexylamino)—4-(methylsulfonyl)pyrimidine—5- carboxamide. To a stirred yellow solution of 2-((1r,4r)hydroxycyclohexylamino) (methylthio)pyrimidinecarboxamide (0.400 g, 1.42 mmol) in NMP (5 mL) was added mCPBA (0.635 g, 2.83 mmol) at 0 CC. The reaction mixture was then stirred at room temperature for 1.5 h until completion of the reaction as indicated by LCMS. The reaction mixture was carried on to next step without filrther purification. MS (ESI) m/z 298.0 [M+1] + and 315.1[M+1]+ C. 4-((R)Cyclopr0pylethylamin0)—2-((1r,4R)—4-hydroxycyclohexylamino)— pyrimidine—5-carb0xamide. To the reaction mixture of 2-((1r,4r)hydroxycyclohexylamino)- 4-(methylsulfinyl)pyrimidinecarboxamide and 2-((1r,4r)hydroxycyclohexylamino) (methylsulfonyl)pyrimidinecarboxamide from the previous reaction was added DIEA (1.56 mL, 8.95 mmol) and (R)cyclopropylethanamine (0.254 g, 2.98 mmol). The reaction was stirred at 90 °C for 16 h. Upon completion of reaction, as ted by LCMS and TLC, the reaction mixture was concentrated and purified by revere phase silica gel chromatography using a gradient of 0% - 90% methanol in water. The desired fractions were purified again by silica ATI—25 14175v1 gel tography using a gradient of 0% - 15% ol saturated with ammonia in DCM, ed by reverse-phased semi-preparative HPLC (0-50% acetonitrile + 0.1% TFA in water + 0.1% TFA, over 30 min). Fractions containing product were concentrated under reduced pressure. The ing residue was redissolved in methanol (5 mL), passed over Varian StratoSpheres HCO3 resin SPE tubes for TFA l (0.9 mmol bicarbonate equivalent per tube), and then concentrated under reduced pressure to afford 4-((R)cyclopropylethylamino)- 2-((1r,4R)—4-hydroxycyclohexylamino)pyrimidinecarboxamide (0.104 g, 22% ; 1H NMR (400 MHz, DMSO-dg) 5 ppm 8.87 - 9.10 (m, 1H), 8.32 (br. s., 1H), 6.68 - 7.02 (m, 1H), 4.51 (d, J: 3.94 Hz, 1H), 3.52 - 3.72 (m, 2H), 3.36 (br. s., 1H), 1.82 (br. s., 4H), 1.12 - 1.32 (m, 8H), 0.95 (d, J: 6.89 Hz, 1H), 0.25 - 0.50 (m, 3H), 0.10 - 0.23 (m, 1H); MS (ESI) m/z 320.1 [M+1]+.
Example 41: 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0)(1- methylcyclopropylamin0)pyrimidine—5-carb0xamide A. 5-Br0m0-N-(1-methylcyclopr0pyl)—4-(methylthi0)pyrimidinamine. To a stirring suspension of 5-bromochloro(methylthio)pyrimidine (1.0 g, 4.18 mmol) in ethanol (6.0 mL) was added 1-methylcyclopropanamine hydrochloride (0.674 g, 6.26 mmol) and DIEA (2.188 mL, 12.53 mmol). The mixture was stirred at 90 CC for 16 h. Upon completion of the reaction as indicated by LCMS and TLC the reaction mixture was concentrated and purified by silica gel chromatography using a gradient of 0% - 20% ethyl acetate in hexanes. The product fractions were combined and concentrated to afford 5-bromo-N-(1-methylcyclopropyl) (methylthio)pyrimidinamine (0.665 g, 58% yield) as a white solid. MS (ESI) m/z 276.2 [M+1]+.
B. 2-(1-Methylcyclopr0pylamin0)—4-(methylthi0)pyrimidine—5-carbonitrile.
A black suspension of 5-bromo-N-(1-methylcyclopropyl)(methylthio)pyrimidinamine (0.665 g, 2.425 mmol), zinc (0.040 g, 0.606 mmol), zinc e (0.185 g, 1.577 mmol), 1,1'-bis- ATI—25 14175v1 2012/034349 (diphenylphosphino)-ferrocene (0.109 g, 0.194 mmol), tris(dibenzylideneacetone)dipalladium(0) (0.111 g, 0.121 mmol), and N, N’ -dimethylacetamide (6 mL) was flushed with nitrogen and heated at 90 °C for 16 h. Upon completion of reaction, as indicated by LCMS and TLC, the reaction mixture was diluted with 125 mL ethyl acetate and 50 mL of water and filtered through a pad of Celite. The layers of the filtrate were separated and the aqueous layer was extracted with 75 mL of ethyl acetate. The combined ethyl e layers was washed with 2 x 50 mL brine, dried over anhydrous magnesium sulfate, filtered and concentrated to an oil. The crude oil was purified by silica gel chromatography using a gradient of 0% - 30% ethyl acetate in hexanes.
The product fractions were combined and concentrated to afford 2-(1-methylcyclopropylamino)- 4-(methylthio)pyrimidinecarbonitrile (0.449 g, 84% yield) as a pale yellow solid. MS (ESI) m/z 221.2 [M+1]+.
C. 2-(1-Methylcyclopropylamin0)(methylsulf0nyl)pyrimidine—5- carbonitrile. To a stirred colorless solution of 2-(1-methylcyclopropylamino)(methylthio)- pyrimidinecarbonitrile (0.449 g, 2.04 mmol) in NMP (6 mL) was added mCPBA (1.142 g, .10 mmol) at 0 CC. The reaction mixture was then stirred at room temperature for 1 h until completion of the reaction as indicated by LCMS. The reaction mixture was d on to next step without further ation. MS (ESI) m/z 253.3 [M+l] +.
D. 4-((3R,4R)—3-Hydr0xymethylcyclohexylamino)—2-(l-methylcyclopropyl- amin0)pyrimidine—5-carb0nitrile. To the reaction mixture of 2-(1-methylcyclopropylamino) (methylsulfonyl)pyrimidinecarbonitrile from the preVious step was added DIEA (2.14 mL, 12.22 mmol) and (1R,2R,5R)—5-aminomethylcyclohexanol hydrochloride (0.371 g, 2.241 mmol; synthesis described herein). The reaction was stirred at 90 0C for 16 h. Upon completion of reaction, as ted by LCMS and TLC, the on mixture was concentrated and purified by silica gel tography using a gradient of 0% - 60% ethyl e in hexanes.
The product fractions were combined and concentrated to afford 4-((3R,4R)hydroxy cyclohexylamino)(1-methylcyclopropylamino)pyrimidinecarbonitrile (0.185g, 30% yield) as a pale yellow solid. MS (ESI) m/z 302.1 [M+1]+ E. 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0)(1- methylcyclopropylamin0)pyrimidine—5-carb0xamide. 4-((3R,4R)Hydroxy methylcyclohexylamino)(1-methylcyclopropylamino)pyrimidinecarbonitrile (0. 1 8 1 g, 0.601 mmol) was ved in DMSO (6 mL). Ten drops of 50% aqueous sodium hydroxide ATI—25 14175V1 2012/034349 (0.047 mL, 0.901 mmol) and ten drops of 30% aqueous hydrogen peroxide were added at room temperature. The reaction mixture was stirred at 50 CC for 1 h. Upon completion of reaction, as indicated by LCMS and TLC, the reaction mixture was cooled to room temperature and was poured into 300 mL of the ice water. The aqueous layer was extracted with 20% iPrOH/chloroform (x3) and the combined c layers were dried over anhydrous magnesium sulfate and trated. The crude mixture was purified by reverse-phased semi-preparative HPLC (0-50% acetonitrile + 0.1% TFA in water + 0.1% TFA, over 30 min). Fractions containing product were concentrated under reduced pressure. The resulting residue was redissolved in methanol (5 mL), passed over Varian StratoSpheres HCO3 resin SPE tubes for TFA removal (0.9 mmol bicarbonate equiv. per tube), and then trated under reduced pressure to afford 4-((1R,3R,4R)—3-hydroxymethylcyclohexylamino)(1- methylcyclopropylamino)pyrimidinecarboxamide (0.069 g, 36% yield); 1H NMR (400 MHZ, DMSO-dg) 5 ppm 8.79 - 8.92 (m, 1H), 8.28 - 8.39 (m, 1H), 7.32 - 7.48 (m, 1H), 4.56 (s, 1H), 3.83 - 3.97 (m, 1H), 2.89 - 3.08 (m, 1H), 2.18 - 2.31 (m, 1H), 1.97 - 2.08 (m, 1H), 1.61 - 1.74 (m, 1H), 1.35 (s, 4H), 1.05 - 1.26 (m, 1H), 0.94 (d, J: 6.64 Hz, 5H), 0.66 (s, 3H), 0.54 (br. s., 3H); MS (ESI) m/z 320.2 [M+1]+.
Example 42: 4-(tert-Butylamin0)—2-((1r,4r)—4—cyclopropoxycyclohexylamin0)- pyrimidine-S-carboxamide V/O’“ N \ NH2 )L / N N NH H /|\ A. 2-((1r,4r)—4-Hydr0xycyclohexyl)is0indoline-1,3-di0ne. To a stirring solution of isobenzofuran-l, 3-dione (90 g, 0.6 mol) and (1r,4r)aminocyclohexanol (70 g, 0.6 mol) in toluene (250 mL) and DMF (250 mL) was stirred at 130 oC ght. The reaction was cooled to room temperature, water was added and the product was filtered, washed with water and dried under vacuum to give the desired product as a white solid, which was used for the next step without r purification (119 g, 0.48 mol, yield: 81%). 1H NMR (400 MHz, CDClg) 5 ppm 7.84-7.79 (m, 2H), 7.72-7.68 (m, 2H), 4.18-4.10 (m, 1H), 3.80-3.73 (m, 1H), .29 (qd, —174— ATI—25 14175v1 Jl=3.4Hz, J2=l30Hz, 2H), 2.12-2.09 (dd, J=6.2Hz, 2H), 1.78-1.74 (dd, J=6.6Hz, 2H), 1.49-1.39 (qd, Jl=3.3Hz, J2=l25Hz, 2H).
B. 2-((1r,4r)—4-(Vinyloxy)cyclohexyl)is0indoline-1,3-di0ne. Palladium acetate (ll g, 0.048 mol) in ethoxyethene (500 mL) was stirred at 0 0C for 15 min, followed by the addition of a on of 2-((lr,4r)hydroxycyclohexyl)isoindoline-l,3-dione (l 18 g, 0.48 mol) in ethoxyethene (1000 mL). The mixture was stirred at 60 0C for 3 days. After that the mixture was cooled to room temperature, filtered and washed with ethyl acetate. The resulting filtrate was concentrated to give the crude product, which was purified by silica gel column chromatography (l6.7% - 25% ethyl acetate in petroleum ether) to afford the title compound as a white solid (80 g 0.29 mol, yield: 61%). 1H NMR (400 MHz, CDClg) 8 ppm 7.84-7.80 (m, 2H), 7.72-7.68 (m, 1H), .32 (dd,J=6.4Hz, 1H), 4.34-4.30 (dd,J=l.2Hz, 1H), 4.20-4.12 (m, lH),4.04-4.02 (dd, z, 1H), 3.89-3.81 (m, 1H), 2.40-2.29 (qd, Hz, J2=l30.6Hz, 2H), 2.2 l -2. 17 (dd, J=64Hz, 2H), 182-1 .78 (dd, J=64Hz, 2H), 153-1 .43 =33Hz, J2=l26.6Hz, 2H).
C. 2-((1r,4r)—4-Cyclopr0p0xycyclohexyl)isoindoline-1,3-dione. To a stirred mixture of diethyl ether (100 mL) and potassium hydroxide (2.5 N, 50 mL) was added l-nitrosourea (38 g, 0.37 mol) at 0 CC. After 15 min, the ether phase was collected and carefully added to the mixture of 2-((lr,4r)(vinyloxy)cyclohexyl)isoindoline-l,3-dione (10 g, 36.9 mmol) and palladium acetate (826 mg, 3.69 mmol) in diethyl ether (100 mL) and DCM (100 mL). The resulting solution was stirred at 0 CC for 0.5 h. The procedure of generation and addition of the ethereal solution of diazomethane was then repeated three times. After the reaction was completed, the solid in the on mixture was filtered off, the filtrate was collected and concentrated to give the crude product which was purified by silica gel column chromatography (25% ethyl acetate in petroleum ether, then 33% ethyl e in petroleum ether) to give 2-((lr,4r)cyclopropoxycyclohexyl)isoindoline-l,3-dione (9.3 g, 32.6 mmol, yield: 88%). 1H NMR (400 MHz, CDClg) 5 ppm 7.84-7.79 (m, 2H), 7.72-7.67 (m, 1H), 4.18-4.09 (m, 1H), .48 (m, lH),3.37-3.33 (m, lH),2.38-2.27 (qd, Jl=3.6Hz, .6Hz, 2H),2.2l- 2.18 (dd, J=6.4Hz, lH), l.79-l .76 (dd, J=6.4Hz, 2H), l.50-l .33(qd, Jl=35Hz, J2=l26Hz, 2H), 0.60-0.56 (m, 2H), 0.50-0.46 (m, 2H).
D. (1r,4r)—4-Cyclopropoxycyclohexanamine. A mixture of 2-((lr,4r) ropoxycyclohexyl)isoindoline-l,3-dione (30 g, 0.10 mol) and hydrazine hydrate (l 8.9 g, -l75- ATI—2514l75vl 0.31 mol) in ol (600 mL) was heated at 80 0C for 2 h. The reaction was cooled to room temperature and filtered. The filtrate was collected and concentrated to give the crude t which was purified by silica gel column chromatography (3.33% methanol in DCM) to afford (h4)¢wdwmmquwammmmwamemflflOgwfimdeMd6W0lHNMR (400 MHz, CDClg) 8 ppm 3.40-3.28 (m, 2H), 2.72-2.65 (m, 1H), 2.06-2.03 (dd, J=3.8Hz, 2H), 1.88-1.85 (dd, J=6.2Hz, 2H), .24 (qd, J1=28Hz, J2=123.3Hz, 2H), 1.19-1.09 (qd, J1=28Hz, J2=123.3Hz, 2H), 0.56-0.51(m, 2H), 0.49-0.44(m, 2H); MS(ESI) m/z = 156.1 [M+1]+.
E. 4-(tert-Butylamin0)—2-(methylsulfonyl)pyrimidine—5-carb0xamide. To a stirred on of t-butylamino)(methylthio)pyrimidinecarboxamide (0.500 g, 2.081 mmol; synthesis described herein) in NMP (6 mL) was added mCPBA (0.933 g, 4.16 mmol) at 0 CC. The reaction mixture was stirred at room temperature for 1 h until completion of the reaction as indicated by LCMS. The reaction mixture was carried on to the next step without further purification. MS (ESI) m/z 273.2 [M+1] +.
F. 4-(tert-Butylamin0)—2-((1r,4r)—4-cyclopr0p0xycyclohexylamin0)— pyrimidine—5-carb0xamide. To the reaction mixture of 4-(tert-butylamino)(methylsulfonyl)- pyrimidinecarboxamide from the preVious step was added DIEA (1.5 mL, 8.33 mmol) and (1r,4r)cyclopropoxycyclohexanamine (0.485 g, 3.12 mmol). The reaction was d at 90 °C for 16 h. Upon completion of the reaction, as indicated by LCMS and TLC, the reaction mixture was slowly added to ~ 70 mL of ice water. This product was filtered, washed with water, followed by a minimum amount of ethanol and diethyl ether and then dried to give the title nd 4-(tert-butylamino)((1r,4r)cyclopropoxycyclohexylamino)pyrimidine carboxamide (0.214g, 30% yield); 1H NMR (400 MHz, DMSO-dg) 5 9.18 (s, 1H), 8.32 (s, 1H), 702-708(nn1HL357-3670n,HD,3300“]=293Ph,HHL199-2090nfiHD,L85- 1.94 (m, 2H), 1.41 (s, 11H), 1.28 (br. s., 5H), 0.37 - 0.46 (m, 4H); MS (ESI) m/z 348.3 .
ATI—25 14175Vl Example 43: 4-((1R,3S)—3-Hydr0xycyclohexylamino)—2-((1r,4R)—4- hydroxycyclohexylamin0)pyrimidinecarb0nitrile HOII'Q )LN/jCN/ N N NH A. 4-((1R,3S)—3-Hydr0xycyclohexylamino)—2-((1r,4R)—4— hydroxycyclohexylamino)pyrimidine—5-carbonitrile. 2-Chloro((1R,3S) hydroxycyclohexylamino)pyrimidinecarbonitrile (240 mg, 0.950 mmol; synthesis described ), (1R,4R)—4-aminocyclohexanol hydrochloride (216 mg, 1.425 mmol), and cesium carbonate (619 mg, 1.899 mmol) were suspended in 1,4-dioxane (10 mL) and heated at 80 0C for 3 h. LCMS showed the d product mass as the dominant peak and no starting material was remaining. The reaction mixture was concentrated and purified by silica gel column chromatography (10-100% ethyl acetate in hexanes, then 0-15% methanol in DCM) to give the title compound (159 mg, 0.480 mmol, 50.5% ; 1H NMR (DMSO-d6 ,400 MHz) 5 ppm 8.06 - 8.24 (m, 1 H), 7.09 - 7.57 (m, 2 H), 4.74 (d, J=4.3 Hz, 1 H), 4.48 - 4.63 (m, 1 H), 3.89 - 4.16 (m, 1 H), 3.34 - 3.74 (m, 3 H), 1.53 - 2.09 (m, 8 H), 0.99 — 1.51 ppm (m, 8 H); MS (ESI) m/Z 332.1 [M+1]+.
Example 44: 4-((1R,3S)—3-Hydr0xycyclohexylamin0)((1r,4R)—4-(2,2,2- trifluor0ethoxy)cyclohexylamin0)pyrimidinecarb0nitrile FFflvoh, QNACCCNN \N NH ” ii [0045 8] A. 4-((1R,3S)—3-Hydr0xycyclohexylamin0)((1r,4R)—4—(2,2,2- roethoxy)cyclohexylamin0)pyrimidinecarb0nitrile. To a stirring solution of 2-chloro((1R,3S)—3-hydroxycyclohexylamino)pyrimidinecarbonitrile (113 mg, 0.447 mmol; synthesis described herein) and (1r,4r)(2,2,2-trifluoroethoxy)cyclo- hexanamine (150 mg, 0.761 mmol; synthesis bed herein) in DMSO (6.0 mL) was ATI—25 14175V1 added DIEA (0.156 mL, 0.895 mmol). The resulting mixture was stirred at ambient temperature for 3 d. DMSO was removed under reduced pressure and the remaining residue was purified using silica gel chromatography (0-40% ethyl acetate + 10% 7 N ammonia in methanol in s) to afford the title compound (156 mg, 0.378 mmol, 84.6% yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.03 - 8.29 (m, 1 H), 7.10 - 7.62 (m, 2 H), 4.74 (d, J=3.90 Hz, 1 H), 4.05 (q, J=9.37 Hz, 3 H), 3.36 - 3.82 (m, 3 H), 1.55 - 2.13 (m, 8 H), 1.00 — 1.50 (m, 8 H). ”P NMR (376 MHz, DMSO-d6) 5 ppm -73.64 (t, J=9.19 Hz, 3 F). MS (ESI) m/z 414.2 [M+1]+.
Example 45: ,4R)—4-(Diflu0r0meth0xy)cyclohexylamin0)((1R,3S)—3- hydroxycyclohexylamin0)pyrimidine—5-carb0nitrile FYO/l' F l/jCN '1‘ N/ NH H i1...
A. 2-((1r,4R)—4-(Difluorometh0xy)cyclohexylamino)—4—((1R,3S)—3- hydroxycyclo-hexylamin0)pyrimidinecarb0nitrile. To a stirring solution of 2- ((1R,3 S)hydroxycyclohexylamino)pyrimidine-5 nitrile (100 mg, 0.396 mmol; synthesis described herein) and (1r,4r)(difluoromethoxy)cyclohexanamine (131 mg, 0.791 mmol; synthesis described herein) in DMSO (4.0 mL) was added DIEA (0.138 mL, 0.791 mmol). The resulting mixture was stirred at ambient temperature overnight. DMSO was removed under reduced pressure and the remaining residue was purified using silica gel chromatography (5-40% ethyl acetate + 10% ammonia saturated methanol in hexanes) to afford the title nd (130 mg, 0.340 mmol, 86.0% yield). 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.08 - 8.25 (m, 1 H), 7.12 - 7.62 (m, 2 H), 6.47 - 6.94 (m, 1 H), 4.74 (d, J=4.29 Hz, 1 H), 3.88 - 4.18 (m, 2 H), 3.41 - 3.85 (m, 2 H), 1.01 - 2.11 (m, 16 H). ”P NMR (376 MHz, DMSO-d6) 5 ppm —79.21 — -78.76 (m, 2 F). MS (ESI) m/z 382.3 [M+1]+.
ATI—25 14175v1 Example 46: 4-(tert—Butylamin0)(4-hydr0xybicyclo[2.2.1]heptan-l-ylamin0)pyrimidine— -carb0xamide HO\®\ I‘ll/EELNHZ A. Cyclopentane-l,3-dicarb0xylic acid. A 22-L, 3-neck, round bottom flask was equipped with a mechanical stirrer, a J-KEM temperature controller, and a nitrogen inlet. The flask was d with norbornene (200 g, 2.123 mol), ethyl acetate (1.95 L), and acetonitrile (1.95 L). The reaction mixture was cooled to 5 0C using an acetone/dry ice bath. Ruthenium trichloride (9.69 g, 46.72 mmol) was added in one portion followed by the slow addition of a suspension of sodium periodate (1.816 kg, 8.707 mol) in water (2.925 L) over 30 min. The reaction slowly began to exotherm and was monitored to keep the temperature between 10 OC and 15 0C. After 90 min the reaction mixture suddenly thickened to the point where stirring was difficult and exothermed rapidly up to 39 0C (a large amount of dry ice was added to the cooling bath to control the exotherm). The reaction mixture was allowed to cool to 20 0C, the dry ice/acetone bath was removed, and the mixture was d at room temperature overnight.
The solids were d by filtration through a pad of celite and the filtrate was concentrated to a solid which was triturated with hexane (2 L), filtered, and rinsed with hexane (2 X 500 mL) to yield 195 g (58%) of cyclopentane-l,3-dicarboxylic acid. 1H NMR (500 Hz, DMSO-dg) 8 ppm 12.07 (s, 2H), 2.66-2.73 (m, 2H), 2.06-2.12 (m, 1H), 1.85-1.89 (m, 1H), .85 (m, 4H).
B. Dimethyl cyclopentane—1,3-dicarb0xylate. A 5-L, 3-neck, round bottom flask was equipped with a mechanical r, a J-KEM temperature controller, and a reflux condenser. The flask was charged with cyclopentane-l,3-dicarboxylic acid (357 g, 2.262 mol) and ol (1.75 L). The solution was cooled to 7 0C using an ice/water bath. Concentrated sulfuric acid (70 mL) was added se over 30 min resulting in an rm up to 12 CC. The reaction mixture was heated to reflux and stirred for 16 h when TLC is (10 % methanol/ethyl acetate) indicated that the reaction was complete. The reaction e was concentrated, redissolved in methyl ATI—2514175v1 WO 45569 tert—butyl ether, and washed with saturated aqueous sodium bicarbonate (2 X 150 mL) and brine (2 X 150 mL). The organic layer was dried over sodium sulfate, filtered, and concentrated. The ing clear oil was dissolved in hexane (2 L) and treated with a 2 N aqueous sodium hydroxide solution (950 mL) until the pH ~ 10. The layers were separated and the aqueous layer was extracted with hexane (4 X 1 L). The organic layers were combined, dried over sodium sulfate, filtered, and concentrated to provide 360 g (100%) of dimethyl cyclopentane-l,3-dicarboxylate as a clear oil. 1H NMR (500 Hz, CDClg) 8 ppm 3.67 (s, 6H), 2.75-2.83 (m, 2H), 2.20-2.26 (m, 1H), 2.05-2.12 (m, 1H), 1.90-2.0 (m, 4H).
C. yl bicyclo[2.2.1]heptane—l,4-dicarb0xylate. A 22-L, 3-neck, round bottom flask was equipped with a J-KEM temperature controller, a mechanical stirrer, a nitrogen inlet, and an addition fiJnnel. The flask was flushed with nitrogen then charged with anhydrous THF (5 L) and ropylamine (731 mL, 5.219 mol). The on was cooled to —20 0C using a dry ice/acetone bath. The stirring mixture was slowly treated with 1.6 M llithium in s (3.02 L, 4.833 mol) via cannula over 1 h maintaining the temperature between —20 CC and —27 CC. The reaction mixture was cooled to — 40 OC and hexamethylphosphoramide (2.7 L, 16.317 mol) was slowly added via on funnel over 40 min. The on mixture was cooled to —73 °C and yl cyclopentane-l,3-dicarboxylate (360 g, 1.933 mol) dissolved in anhydrous THF (2 L) was slowly added via addition funnel over 2 h. The reaction mixture was warmed to —10 CC and stirred at that temperature for 30 min, then cooled to —70 CC and treated with 1-bromochloroethane (267 mL, 3.209 mol) via addition funnel over 4 h. The reaction mixture was allowed to slowly warm to room temperature over 12 h and then was quenched with saturated aqueous ammonium chloride (2 L) over 90 min. The reaction mixture was diluted with hexane (2 L), the layers were separated, and the aqueous layer was further extracted with hexane (3 X 2 L). The combined organic layers were washed with brine (2 X 1 L), dried over sodium sulfate, filtered, and concentrated to a brown oil.
The crude product was purified by silica gel purification (0—10 % ethyl acetate/hexane).
The product ning fractions were concentrated to near dryness and diluted with hexanes. The resulting crystalline solids were filtered and washed with hexane (200 mL) providing pure product as a clear crystalline solid. Additional batches of product were ATI—25 14175v1 obtained from the e in a similar manner. All product batches were combined to provide 208 g (51%) of dimethyl bicyclo[2.2. 1]heptane-1,4-dicarboxylate as a clear crystalline solid. 1H NMR (500 Hz, CDClg) 5 ppm 3.68 (s, 6H), 2.03 (d, J: 6.4 Hz, 4H), 1.90 (s, 2H), 1.67 (d, J: 7.0, 4H).
D. 4-(Methoxycarbonyl)bicyclo[2.2.1]heptanecarb0xylic acid. A 12-L, 3-neck, round bottom flask was equipped with a ical stirrer, a J-KEM temperature controller, and a 250 mL addition flannel. The flask was charged with dimethyl bicyclo[2.2.1]heptane-1,4-dicarboxylate (208 g, 980 mmol) and THF (6.7 L).
The resulting solution was cooled to 15 0C using an ice/water bath. Sodium hydroxide pellets (39.2 g, 980 mmol) were dissolved in methanol (400 mL) and slowly added to the stirring solution over 30 min. After the addition was te, white solids began to precipitate. The reaction mixture was stirred at room temp for 16 h when TLC analysis (100% ethyl acetate) indicated about 90% conversion. The reaction e was concentrated to dryness, ed in hexane (2 L), filtered, and washed with hexane (2 X 400 mL). The resulting sodium carboxylate salt was transferred to a 3-L, 3-neck, round bottom flask equipped with a mechanical stirrer, dissolved in water (1 L), and slowly treated with 2 N aqueous hydrochloric acid (430 mL) at 10 CC until pH ~ 4. The thick suspension was diluted with ethyl acetate (1 L) and transferred to a tory funnel. The layers were separated and the aqueous layer was fiarther extracted with ethyl acetate (2 X 500 mL), washed with brine (200 mL), dried over sodium sulfate, and concentrated to provide 170 g (87%) of 4-(methoxycarbonyl)bicyclo[2.2. 1]heptanecarboxylic acid. 1H NMR (500 Hz, DMSO-dg) 5 ppm 12.21 (s, 1H), 3.60 (s, 3H), 1.91 (d, J = 6.7 Hz, 4H), 1.75 (s, 2H), .62 (m, 4H).
E. 4-(Benzyloxycarbonylamin0)bicyclo[2.2.1]heptane—l-carboxylic acid.
A 2-L, 3-neck, round bottom flask was equipped with a mechanical stirrer, a J-KEM temperature controller, a reflux condenser, and a nitrogen inlet. The flask was charged with 4-(methoxycarbonyl)bicyclo[2.2.1]heptanecarboxylic acid (100 g, 0.504 mol) and anhydrous e (500 mL). The flask was cooled to 10 OC and DIEA (175 mL, 1.008 mol) was slowly added over 5 min resulting in a mild exotherm up to 14 OC.
Diphenylphosphonic azide (130 mL, 0.605 mol) was slowly added to the reaction mixture.
The mixture was heated to about 60 CC when ssing began. The reaction quickly ATI—25 14175v1 ted a large amount of gas and exothermed to reflux. The reaction was stirred at reflux (110 CC) for 2 h, then cooled to 50 CC and slowly treated with benzyl alcohol (104 mL, 1.008 mol) over 5 min. The reaction was heated again to 110 CC and stirred for 40 h. The reaction mixture was concentrated to an oil in vacuo, dissolved in ethyl acetate (2 L), washed with brine (500 mL), and the brine layer was extracted with ethyl acetate (2 X 400 mL). The ed organics were dried over sodium sulfate, filtered, and amwmmmdmamweMMmmhwwfiwfiflmdwaih3mmkmmmbmwmflwk equipped with a mechanical stirrer, a J-KEM temperature controller, and a nitrogen inlet.
The flask was charged with methanol (2 L). The mixture was cooled to 10 °C and treated with 2N sodium ide (500 mL) over 5 min resulting in an exotherm to 16 CC. The reaction was stirred at 60 CC for 20 h when TLC analysis (20% methanol/ethyl e) indicated a complete reaction. The reaction was cooled to room temperature and methanol was removed in vacuo. With cooling, the reaction mixture was acidified using 2 N hydrochloric acid (480 mL) to pH 2-3. The aqueous mixture was extracted using ethyl e (2 L, then 500 mL). The combined c layers were washed with brine (400 mL), dried over sodium sulfate, and concentrated to provide the crude product as a brown oil. The crude product was purified by silica gel chromatography (20—100% ethyl acetate/hexane) providing 132 g (91%) of 4-(benzyloxycarbonylamino)- o[2.2.1]heptanecarboxylic acid. 1H NMR (500 Hz, DMSO- d6) 5 ppm 12.12 (s, 1H), 7.56 (s, 1H), 7.34-7.38 (m, 4H), 7.28-7.32 (m, 1H), 4.98 (s, 2H), 1.80-1.93 (m, 4H), 1.79 (s, 2H), 1.64 (t, J: 9.1 Hz, 2H), 1.57 (t, J: 8.8 Hz, 2H).
F. 4-Amin0bicyclo[2.2.1]heptane—l-carboxylic acid. A 2-L Parr bottle was charged with 4-(benzyloxycarbonylamino)bicyclo[2.2.1]heptanecarboxylic acid (105 g, 363mmmDnndeoH5mhnLL08Nhymodmnmadd600mLme10%pdbmmnon carbon (38.6 g, 18.15 mmol, 50% wet with water). The reaction mixture was reacted on a Parr shaker under 30 gpsi hydrogen for 10 h when TLC is (10% methanol/ethyl e) indicated a te reaction. The catalyst was removed by filtration through a short plug of celite and the plug was washed thoroughly with water (4 X 100 mL). The filtrate was concentrated to provide 4-aminobicyclo[2.2.1]heptanecarboxylic acid (56.3 g, 100%) as a wet white solid which was used in the next step without any further purification; MS (ESI) m/z 156.2 [M+1]+ ATI—25 14175v1 G. 0xybicyclo[2.2.1]heptane—l-carboxylic acid. A 2-L, 3-neck, round bottom flask was equipped with a mechanical stirrer, a reflux ser, a J-KEM temperature controller, and a nitrogen inlet. The flask was charged with 4-aminobicyclo[2.2.1]heptane carboxylic acid (56.3 g, 363 mmol) and 10% aqueous acetic acid (340 mL). The reaction mixture was cooled to 10 oC and was slowly treated over 45 min with sodium nitrite (75.0 g, 1.088 mol) in water (125 mL) via addition filnnel. A significant amount of gas evolution was observed and the on mixture was stirred at 65 oC overnight. The reaction was cooled to oC and slowly treated over 30 min with potassium hydroxide (183 g, 3.265 mol) in ol (400 mL) via addition filnnel. The reaction was stirred at 65 CC for 5 h when TLC analysis (10% methanol/ethyl acetate) indicated a te reaction. The reaction mixture was concentrated to remove ol and the remaining aqueous mixture was extracted with ethyl acetate (2 X 400 mL). The aqueous layer was acidified to pH 3 using concentrated hydrochloric acid (210 mL) while cooling in ice/water bath. The resulting solids (impurities) were filtered and washed with water (2 X 30 mL). The filtrate was extracted with ethyl acetate (10 X 400 mL), dried over sodium sulfate, and concentrated to s to provide 53 g (94%) of 4-hydroxybicyclo[2.2.1]heptanecarboxylic acid; MS (ESI) m/z 155.2 [M-1]' H. Benzyl 4-hydr0xybicyclo[2.2.1]heptanylcarbamate. A 3-L, 3-neck, round bottom flask was equipped with a mechanical stirrer, a J-KEM temperature controller, a nitrogen inlet, and a 125 mL addition funnel. The flask was flushed with nitrogen and charged with 4-hydroxybicyclo[2.2.1]heptanecarboxylic acid (53 g, 339 mmol) and ous toluene (350 mL). The reaction mixture was cooled to 10 CC and treated with benzyl alcohol (175 mL, 1.695 mol). The reaction mixture was then slowly treated with DIEA (118 mL, 678 mmol) and diphenylphosphonic azide (87.7 mL, 407 mmol). The reaction mixture was slowly heated up to 50 °C when off-gassing began. The heating mantle was d and the reaction mixture slowly exothermed up to 75 0C while off-gassing considerably. The reaction was heated to 110 CC and stirred for 20 h when TLC is (10% methanol/ethyl acetate) indicated a complete reaction. The on mixture was concentrated to remove solvent and partitioned between ethyl acetate (1.2 L) and brine (700 mL). The organic layer was dried over sodium sulfate, concentrated, and purified by silica gel chromatography (20—60% ethyl e/hexane) providing 61 g (69%) ofbenzyl 4-hydroxybicyclo[2.2.1]heptanylcarbamate; MS (ESI) m/z 260.1 [M-1]' ATI—25 14175v1 WO 45569 ] I. 4-Amin0bicyclo[2.2.1]heptan-l-ol hloride. A 2-L Parr bottle was charged with benzyl 4-hydroxybicyclo[2.2.1]heptanylcarbamate (61 g, 233 mmol), methanol (400 mL), and water (200 mL). The Parr bottle was flushed with nitrogen and charged with % palladium on carbon (25 g, 11.65 mmol, 50% wet with water). The reaction was run under gpsi hydrogen on a parr shaker for 3 h when TLC analysis (15% methanol/DCM with 2% ammonium hydroxide) ted a complete reaction. The reaction was filtered through a celite plug and concentrated to remove ol. The resulting aqueous mixture was treated with 2 N hydrochloric acid (115 mL) until it reached pH 1-2. The aqueous mixture was extracted using ethyl acetate (5 X 400 mL) to remove an ty. The aqueous mixture was concentrated to dryness and dried in a vacuum oven at 40 oC overnight to provide 36.91 g (97%) of 4-aminobicyclo[2.2.1]heptanol hydrochloride; MS (ESI) m/z 128.1 [M+1]+ J. 4-(tert-Butylamin0)—2-(4-hydroxybicyclo[2.2.1]heptan-l-ylamin0)- pyrimidine—5-carb0xamide. 4-(tert-Butylamino)(methylsulfonyl)pyrimidinecarboxamide (0.165 g, 0.606 mmol; synthesis described herein), 4-aminobicyclo[2.2.1]heptanol hydrochloride (0.105 g, 0.644 mmol) and DIEA (0.337 mL, 1.931 mmol) were mixed in DMF (8 mL) and heated at 90°C overnight. The t was evaporated and the residue was d by silica gel column chromatography (0-10% methanol in ethyl acetate) to give the desired product. (41 mg, 0.127 mmol, 21 %). 1H NMR (400 MHz, DMSO-d6) 5 ppm 9.43 (br. s., 0 H) 8.32 (s, 1 H) 7.07 (br. s., 1 H) 4.88 (br. s., 1 H) 1.94 - 2.14 (m, 2 H) 1.61 - 1.87 (m, 6 H) 1.49 - 1.60 (m, 2 H) 1.42 (s, 9 H). MS (ESI) m/z 320.0 [M+1]+ Example 47: 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(3-methylbicyclo[1.1.1]pentan ylamin0)pyrimidine—5-carb0xamide A. 3-Methylbicyclo[1.1.1]pentane—1-carb0xylic acid. To a solution of 1-iodomethylbicyclo[1.1.1]pentane (1.62 g, 7.79 mmol; prepared according to Eur. J.
Org. Chem. 1137-1155 (2000)) in diethyl ether (26 mL), a solution of tert-butyllithium ATI—25 14175v1 (9.16 mL, 15.57 mmol, 1.7 M in pentane) was added over a period of 40 min at -78 0C.
After stirring the reaction mixture for 1 h at this temperature, carbon e gas was bubbled through the reaction mixture for 5 min and then the mixture was allowed to warm to room ature. The reaction mixture was extracted twice with a 5% aqueous sodium bicarbonate solution. The ed aqueous phases were acidified to pH 2- 3 with concentrated hydrochloric acid at 0 CC, ted with sodium chloride, and extracted with diethyl ether. The combined organic phases were dried and, after evaporation of the solvent under reduced pressure, purified by column chromatography (0-20% ethyl acetate in hexanes) to afford 3-methylbicyclo[1.1.1]pentanecarboxylic acid (0.5 g, 3.96 mmol, 50.9 % yield); 1H NMR (400 MHz, DMSO-d6) 8 ppm 12.21 (s, 1 H), 1.82 (s, 6 H), 1.14 (s, 3 H).
B. tert-Butyl 3-methylbicyclo[1.1.1]pentanylcarbamate.
Diphenylphosphonic azide (0.340 mL, 1.577 mmol) was added dropwise to a solution of 3-methylbicyclo[1.1.1]pentanecarboxylic acid (0.199 g, 1.577 mmol) and TEA (0.220 mL, 1.577 mmol) in dry tert-butanol (6 mL). The solution was stirred at room ature for 4 h and then heated to reflux for 24 h. The solvent was evaporated under reduced pressure and the residue was extracted three times with tert-butyl methyl ether.
The combined organic phase was washed with saturated aqueous sodium bicarconate solution and dried over anhydrous magnesium e. After filtration and evaporation of the solvent under reduced pressure the residue was purified by column chromatography (0-5% ethyl e in hexanes) to afford tert—butyl 3-methylbicyclo[1.1.1]pentan ylcarbamate (0.111 g, 0.563 mmol, 35.7 % yield); 1H NMR (400 MHz, 6) 5 ppm 1.74 (s, 6 H), 1.36 (s, 9 H), 1.17 (s, 3 H).
] C. 3-Methylbicyclo[1.1.1]pentanamine hydrochloride. To a solution of tert-butyl 3-methylbicyclo[1.1.1]pentanylcarbamate (0.1 g, 0.507 mmol) in ethyl acetate (10 mL) was added hydrochloric acid (0.760 mL, 3.04 mmol, 4M in e) at room temperature. The reaction was stirred at room temperature for 16 h. The reaction mixture was concentrated, the resulting solids were washed with diethyl ether and the ing suspension filtered to give 3-methylbicyclo[1.1.1]pentanamine hydrochloride (0.04 g, 0.299 mmol, 59.1 % yield). 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.59 (br. s., 3 H), 1.84 (s, 6 H), 1.22 (s, 3 H).
ATI—25 14175v1 D. ,4r)—4-Hydr0xycyclohexylamin0)(3- methylbicyclo[1.1.1]pentan-l-ylamin0)pyrimidinecarb0xamide. To a stirring suspension of 2-((1r,4r)hydroxycyclohexylamino)(methylsulf1nyl)pyrimidine carboxamide (0.25 g, 0.838 mmol; synthesis bed herein) and 3-methylbicyclo- [1.1.1]pentanamine hydrochloride (0.123 g, 0.922 mmol) in DMF (3 mL) was added DIEA (0.366 mL, 2.095 mmol) and the reaction was heated to 80 oC overnight. The crude reaction mixture was concentrated under reduced pressure and then ice-cold water (20 mL) was added to the residue. The resulting mixture was usly stirred for 1 h and then the formed precipitate was d, washed with water and dried under vacuum to afford 2-((1r,4r)hydroxycyclohexylamino)(3-methylbicyclo[1 . 1 . 1]pentan ylamino)pyrimidinecarboxamide (0.222 g, 0.670 mmol, 80 % . 1H NMR (400 MHz, DMSO-d6) 5 ppm 1.15 - 1.31 (m, 7 H), 1.81 - 1.93 (m, 4 H), 1.98 (s, 6 H), 3.35 - 3.44 (m, 1 H), 3.54 - 3.63 (m, 1 H), 4.56 (d, J=4.29 Hz, 1 H), 7.12 (d, J=7.81 Hz, 1 H), 8.35 (s, 1 H), 9.35 (s, 1 H); MS (ESI) m/z 332.4 [M+1]+.
Example 48: 4-((1R,3S)Hydr0xycyclohexylamin0)(4-methyltetrahydr0-2H-pyran ylamin0)pyrimidine—5-carb0xamide )L / N N NH A. 4-((1R,3S)—3-Hydr0xycyclohexylamin0)(4-methyltetrahydr0-2H— pyranylamin0)pyrimidine—5-carboxamide. 4-((1R,3S)—3-Hydroxycyclohexylamino)- 2-(methylsulfonyl)pyrimidinecarboxamide (0.740 g, 2.354 mmol; synthesis described herein), 4-methyltetrahydro-2H-pyranamine hydrochloride (0.892 g, 5.88 mmol), DIEA (1.645 mL, 9.42 mmol) and NMP (20 mL) were combined and heated for 1 h at 180 CC in a microwave . The solvent was concentrated under reduced pressure and the residue was purified by column chromatography (0-10% methanol in DCM) to afford 4-((1R,3S) hydroxycyclohexylamino)(4-methyltetrahydro-2H-pyranylamino)pyrimidine amide (88 mg, 0.252 mmol, 10.70 % yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm ATI—25 14175v1 8.96 (br. s., 1H), 8.35 (s, 1H), 6.76 (br. s., 1H), 4.65 (d, .1: 4.69 Hz, 1H), 3.56 (dd, .1: 3.32, 6.83 Hz, 4H), 3.40 — 3.47 (m, 1H), 2.20 — 2.29 (m, 2H), 2.10 (d, .1: 10.93 Hz, 1H), 1.76 — 1.89 (m, 2H), 1.66 — 1.73 (m, 1H), 1.50 — 1.59 (m, 2H), 1.39 (s, 3H), 1.16 — 1.26 (m, 1H), 1.03 _ 1.12 (m, 3H). MS (ESI)1n/z 350.4 [M+1]+.
Example 49: 4-((1R,3S)—3-Hydroxycyclohexylamino)—2—((1r,4r)—4—[(2H3)methyloxy]- exylamino) dine-S-carboxamide /O// D30 ‘ N \ NH2 N N NH A. (1r,4r)—4-[(2H3)methyloxy]-N-tritylcyclohexanamine. To a solution of (1r,4r)(tritylamino)cyclohexanol (2.34 g, 6.55 mmol) in dry THF (15 mL) was added sodium hydride (524 mg, 13.1 mmol, 60 % in mineral oil) at 0 0C. After the resulting mixture was stirred for 30 minutes at 0 0C under nitrogen atmosphere, a solution of iodo(2H3)methane (1.0 g, 6.89 mmol) in THF (5 mL) was added dropwise at 0 0C. The resulting e was stirred at room temperature ght under nitrogen atmosphere, then poured into ice water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was evaporated under vacuum to give crude product, which was purified by silica gel column chromatography (5 % ethyl acetate in petroleum ether) to afford the title nd (2.0 g, .35 mmol, 83 % yield) as a white solid. MS (ESI) m/z 375.0 [M+1] +.
B. (lr,4r)[(2H3)Methyloxy]cyclohexanamine. To a cooled (0 0C) solution of (1r,4r)[(2H3)methyloxy]-N-tritylcyclohexanamine (2.0 g, 5.35 mmol) in DCM (10 mL) was added TFA (3 mL) at 0 0C. The resulting mixture was dark red.
Triethylsilane (0.4 mL) was added until the resulting mixture was d to colourless.
The reaction was stirred at 0 0C for additional 15 min. After removal of all volatile solvents in vacuo, the residue was further dried under high vacuum for 2 h to give the crude product as a white solid. The crude t was dissolved in ethyl acetate and aqueous hydrochloride solution (20 mL, 0.25 mol/L) was added. The organic layer was removed and the inorganic layer was washed with ethyl acetate twice. Aqueous ammonia ATI—2514l75vl WO 45569 solution (2 mL) was added to the inorganic layer and the mixture was stirred for 15 min.
Concentration in vacuo gave the crude product as a white solid (706 mg, 5.35 mmol, 100% yield). MS (ESI) m/z 133.0 [M+1] C. 4-((1R,3S)—3-Hydr0xycyclohexylamino)—2-((1r,4r)—4- [(2H3)methyloxy]cyclohexanamin0)pyrimidine—5-carb0xamide. A mixture of 4- ((1 R,3 S)-3 -hydroxycyclohexylamino)(methylsulfonyl)pyrimidine carboxamide (160 mg, 0.5 mmol; synthesis described herein), )[(2H3)methyloxy]- cyclohexanamine (132 mg, 1.0 mmol), DIEA (194 mg, 1.5 mmol) and NMP (1 mL) were combined and heated at 80 0C overnight. The solvent was evaporated under reduced pressure and the residue was purified by column chromatography (0-15% methanol in DCM) to give the desired product as a white powder (120 mg, 0.33 mmol, 63 % yield). 1H NMR (400 MHz, CDgOD) 5 ppm 8.17 (s, 1H), 3.93-3.87 (m, 1H), 3.64 (s, 1H), 3.50 (d, J: 6.0 Hz, 1H), 3.13 (s, 1H), 2.22 (s, 1H), .72 (m, 7H), 1.29-0.99 (m, 8H); MS (ESI) m/z 367.3 [M+1] i e 50: 4-(tert-Butylamin0)—2-((1s,4s)—4—hydr0xy methylcyclohexylamin0)pyrimidine—5-carboxamide >22V; A. 4--(tert-Butylamin0)—2-((1s,4s)hydr0xymethylcyclohexyl- amino)pyrimidine—5-carboxamide. 4-(tert-Butylamino)(methylsulfonyl)pyrimidine carboxamide (0.110 g, 0.404 mmol; synthesis bed herein), )amino methylcyclohexanol (0.055 g, 0.429 mmol; prepared according to PCT Int. Appl. publication No. WO 2010027500) and DIEA (0.225 mL, 1.287 mmol) were mixed in DMF (5 mL) and heated at 90 OC overnight. The solvent was evaporated and the residue was purified by silica gel column chromatography (0-10% methanol in ethyl acetate) to give the desired product. (37 mg, 0.115 mmol, 27%). 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.08 — 9.23 (m, 1 H) 8.25 — 8.37 (m, 1 H) 6.89 — 7.06 (m, 1 H) 3.97 — 4.04 (m, 1 H) ATI—25 14175v1 3.46 — 3.62 (m, 1 H) 1.48 — 1.71 (m, 6 H) 1.40 (s, 9 H) 1.19 — 1.36 (m, 2 H) 1.10 (s, 4 H).
MS (ESI)m/z 322.4 [M+1]+ Example 51: 4-((1R,3R)Hydr0xy-4,4-dimethylcyclohexylamin0)(1- methylcyclopropylamin0)pyrimidine—5-carb0xamide N \ NH2 % JL /N N NH A. 2-(1-Methylcyclopr0pylamin0)—4-(methylthi0)pyrimidine carboxamide. To a ng solution of 2-(1-methylcyclopropylamino) (methylthio)pyrimidinecarbonitrile (0.653 g, 2.96 mmol, synthesis bed herein) in DMSO (8 mL) was added 6N aqueous sodium hydroxide (2.470 mL, 14.82 mmol) solution and 30% aqueous hydrogen peroxide solution (1.873 mL, 16.52 mmol) at 0 oC.
Then the e was stirred at 50 CC and for 20 minutes and was then diluted with 100 mL ethyl e and 30 mL water. The layers were separated and the aqueous layer was back extracted with 50 mL ethyl e. The combined ethyl acetate layers were dried over anhydrous magnesium sulfate, filtered and trated to solids that were dried in a vacuum oven at 45 CC to afford 2-(1-methylcyclopropylamino)—4- (methylthio)pyrimidinecarboxamide (305 mg, 1.280 mmol, 43.2 % yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm 8.31 - 8.54 (m, 1 H), 7.80 - 8.07 (m, 1 H), 6.97 - 7.77 (m, 1 H), 2.39 (br. s., 3 H), 1.37 (s, 3 H), 0.52 - 0.79 (m, 4 H). MS (ESI) m/z 239.0 [M+1]+.
B. 2-(1-Methylcyclopr0pylamin0)—4-(methylsulfinyl)pyrimidine—5— carboxamide. To a stirring solution of 2-(1-methylcyclopropylamino)(methylthio)- pyrimidinecarboxamide (305 mg, 1.280 mmol) in form (25.6 mL) was added portionwise 3-phenyl(phenylsulfonyl)-1,2-oxaziridine (502 mg, 1.920 mmol). The resulting pale yellow solution was stirred at ambient temperature overnight. The reaction solution was concentrated under reduced pressure to give crude product as a white solid.
Ethyl acetate (10 mL) was added and the slurry was stirred at room temperature for 1 h, filtered, washed with ethyl acetate, and dried in a vacuum oven for a few hours to afford ATI—25 14175v1 WO 45569 2-(1-methylcyclopropylamino)(methylsulf1nyl)pyrimidinecarboxamide (242 mg, 0.952 mmol, 74.4 % yield). 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.35 - 8.90 (m, 2 H), 788-8140n,1HL728-7590n,1HL274(&31D,L32-1460ng3Hx068-084 (m, 2 H), 0.50 — 0.70 (m, 2 H). MS (ESI) m/z 255.0 [M+1]+.
C. 4-((1R,3R)Hydr0xy-4,4-dimethylcyclohexylamin0)(1- methylcyclopropylamin0)pyrimidine—5-carb0xamide. To a stirring suspension of 2-(1-methylcyclopropylamino)(methylsulf1nyl)pyrimidinecarboxamide (242 mg, 0.952 mmol) and a mixture of (1R,5R)—5-amino-2,2-dimethylcyclohexanol hydrochloride and (1S,5S)—5-amino-2,2-dimethylcyclohexanol hydrochloride (222 mg, 1.237 mmol, synthesis described herein) in DMF (4.543 mL) was added DIEA (0.499 mL, 2.85 mmol) and the reaction was heated to 90 OC ght. The crude reaction mixture was poured into 50 mL of ice water. The resulting solids were slurried for ~ 1 h, filtered, rinsed with water and dried for a few hours in a vacuum oven at 45 CC to afford a mixture of ,3R)hydroxy-4,4-dimethylcyclohexylamino)(1 -methylcyclopropylamino)- pyrimidinecarboxamide and 4-((1R,3R)—3-hydroxy-4,4-dimethylcyclohexylamino) (1-methylcyclopropylamino)pyrimidinecarboxamide (169 mg, 0.507 mmol, 53.3 % yield). This material was separated by preparative chiral SFC utilizing a Pak AD-H, 250><30 mm ID. column with an isocratic 40% ethanol + 0.1% ammonium hydroxide in C02 gradient at 60 mL/min flow rate and at 38 CC. The faster eluting isomer was denoted as peak 1 and 49 mg (0.147 mmol) was obtained. The slower g isomer was denoted as peak 2 and 51 mg (0.153 mmol) was obatined. Peak 1: 1H NMR (400 MHz, DMSO-dg) 5 ppm 8.71 - 9.02 (m, 1 H), 8.21 - 8.43 (m, 1 H), 7.03 - 7.54 (m, 1HL444-4580n,1HL381-3990n,1HL307-3250n,1HL174-2080n,1HL 1.35 (s, 8 H), 0.87 - 0.97 (m, 3 H), 0.77 - 0.86 (m, 3 H), 0.62 - 0.70 (m, 2 H), 0.47 - 0.59 (m, 2 H). MS (ESI) m/z 334.2 [M+1]+. Peak 2: 1H NMR (400 MHz, DMSO-dg) 5 ppm 872-9000n,1HL818-8460n,1HL712-7600n,1HL438-4610n,1HL376- 3.98 (m, 1 H), 3.04 - 3.26 (m, 1 H), 1.75 - 2.10 (m, 1 H), 1.35 (s, 8 H), 0.92 (s, 3 H), 0.82 (s, 3 H), 0.61 - 0.72 (m, 2 H), 0.47 - 0.60 (m, 2 H). MS (ESI) m/z 334.2 [M+1]+. By SAR potency comparison with similar compounds ofknown absolute chemistry, Peak 1 was assigned as 4-((1S,3S)Hydr0xy-4,4-dimethylcyclohexylamin0)(l-methyl- cwhmmwhmhMmfimmmdfiwflmmmMePwk2wmwmmme4flkflfifi- ATI—2514l75vl 2012/034349 Hydroxy-4,4-dimethylcyclohexylamino)—2-(1-methylcyclopr0pylamin0)pyrimidine—5- carboxamide.
Example 52: yclo[1.1.1]pentanylamin0)—2-((1r,4r)—4- ycyclohexylamino)pyrimidine—5-carboxamide ] A. 2-Phenylbicyclo[1.1.1]pentanol. A stirring solution of utyl(phenyl)methanone (3.0 g, 18.8 mmol) in 800 ml of benzene was irradiated with a 1000 W mercury arc lamp for 48 h at room temperature in a nitrogen atmosphere. The solution was concentrated and the residue (3.1 g) was purified by column chromatography (10%-25% ethyl acetate in petrolium ether) to give the desired product as a white solid (0.9 g, 5.6 mmol, yield: 30%). 1H NMR (300 MHz, CDClg) 8 ppm 7.41-7.28 (m, 5 H), 3.04 (s, 2 H), 2.82 (dd, J1= 10.2 Hz, J2= 2.4 Hz, 1 H), 2.09 (s, 1 H), 1.78 (d, J = 2.4 Hz, 1 H), 1.50 (d, J = 3.3 Hz, 1 H), 1.33 (dd, J1= 9.9, J2= 3.3 Hz, 1 H).
B. 2-Phenylbicyclo[1.1.1]pentanyl acetate. To a stirring solution of 2.7 g (16.8 mmol) of ylbicyclo[1.1.1]pentanol in pyridine (15 mL) at 0 CC was added dropwise acetyl chloride (1.5 mL, 21.0 mmol) in a nitrogen atmosphere. The reaction mixture was stirred for 0.5 h at 0 CC and for 1 h at room temperature. The solution was poured onto ice and extracted with 50 mL of diethyl ether twice. The combined diethyl ether on was washed with saturated aqueous sodium bicarbonate solution and with saturated aqueous copper sulfate solution. After drying over anhydrous magnesium sulfate, the solution was concentrated and the crude product was purified by column chromatography (0-5% ethyl acetate in petrolium ether) to give the desired product (2.7 g, 13.4 mmol, yield: 80%). 1H NMR (400 MHz, CDClg) 5 ppm 7.47-7.26 (m, 5 H), 3.31 (s, 2 H), 2.43 (dd, J1= 10.4 Hz, J2= 2.8 Hz, 1 H), 1.95 (s, 3 H), 1.80 (d, J = 2.8 Hz, 1 H), 1.66 (d, J = 3.2 Hz, 1 H), 1.49 (dd, J1= 10.4, J2= 3.2 Hz, 1 H).
C. 2-Phenylbicyclo[1.1.1]pentane. A solution of 2-phenyl- bicyclo[1.1.1]pentanyl acetate (2.7 g, 13.4 mol) in 50 mL of anhydrous ether was ATI—25 14175V1 2012/034349 placed in a three-necked flask. The flask was cooled to -78 oC, and 150 mL of liquid ammonia was added. To the stirring mixture was added sodium (0.62 g, 27.0 mmol) in small pieces over a period of 10 minutes. ng was ued until the blue color was discharged. The reaction was quenched by the addition of saturated aqueous ammonia chloride solution (5 mL), and the solvent was allowed to evaporate overnight. Pentane (40 mL) was added to the residue, and the solution was washed with 30 mL saturated s sodium chloride on, dried over anhydrous magnesium sulfate, and then concentrated. The crude product was purified by column chromatography (100% pentane) to give the desired compound as a colorless liquid (1.8 g, 12.5 mmol, yield: 93% ). 1H NMR (300 MHz, CDC13)5 ppm 7.40-7.21 (m, 5 H), 3.48 (d, J = 6.9 Hz, 1 H), 2.81 (s, 2 H), 2.20 (dd, J1= 9.9Hz, J2= 2.7 Hz, 1 H), 2.00 (d, J = 2.1 Hz, 1 H), 1.94-1.85 (m, 2 H).
D. Bicyclo[1.1.1]pentane—2-carb0xylic acid. A mixture of 2-phenylbicyclo[l.l.l]pentane (2.8 g, 19.4 mmol), ruthenium(IV) oxide (0.1 g, 0.75 mmol), sodium periodate (41.5 g, 194 mmol), water (155 mL), perchloromethane (110 mL), and acetonitrile (110 mL) was stirred for 4 d. DCM (200 mL) was added to the mixture, and the solids were removed by filtration. The mother liquid was alkalized with aqueous sodium hydroxide solution (1 M), and the organic solvent was removed by reduced pressure. The residue nic layer was extracted with diethyl ether (100 mL x 2) and the water phase was acidified with aqueous hydrochloric acid (2 M) to pH < 3, then extracted with diethyl ether (100 mL x 5 ). The organic layers were combined and concentrated to give the crude acid as a yellow liquid. Then the crude product was purified by column chromatography (10% ethyl acetate in petrolium ether) to give 400 mg (3.57 mmol, yield: 18 %) of the desired t. 1H NMR (300 MHz, CDClg) 8 ppm 2.97 (d, J = 7.5 Hz, 1 H), 2.82 (s, 2 H), 2.49 (dd, J1= 7.2 Hz, J2= 3.3 Hz, 1 H), 1.92 (dd, J1= 7.2Hz, J2= 3.3 Hz, 1 H), 1.79-1.76 (m, 2 H).
E. Bicyclo[1.1.1]pentanamine hydrochloride. To a stirring on of bicyclo[l.1.1]pentanecarboxylic acid (1.0 g, 8.9 mmol) in anhydrous toluene (25 mL) and 2-methylpropanol (5 mL) was added DIEA (2.3 g, 17.8 mmol) and diphenyl phosphorazidate (2.9 g, 10.7 mmol). The e was heated at 90 CC for 16 h under nitrogen atmosphere. Then the reaction mixture was concentrated and the residue was purified by column tography (10% ethyl acetate in petrolium ether) to give crude ATI—2514l75vl tert-butyl bicyclo[1.1.1]pentanylcarbamate. The crude product was dissolved in 15 mL hydrochloric acid solution (1 M in methanol) and the solution was stirred at room temperature for 16 h. The solution was concentrated and the residue was suspended in 50 mL of diethyl ether. The mixture was stirred for 15 min and the itates were collected and dried to give the title compound (350 mg, yield: 33%). 1H NMR (DMSO-dg) 8 ppm 8.58 (brs, 3H), 3.35 (s, 1H), 2.62 (s, 2H), .57 (m, 1H), 1.90 (t, J: 5.0 Hz, 1H), 1.79 (d, J = 2.4 Hz, 1H), 1.50 (dd, J1= 10.0 Hz, J1= 2.8 Hz, 1H).
F. 4-(Bicyclo[1.1.1]pentan-Z-ylamino)—2-((1r,4r)—4- hydroxycyclohexylamino)pyrimidine—5-carboxamide. A mixture of 2-((1r,4r) hydroxycyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (300 mg, 0.95 mmol; synthesis described herein), bicyclo[1.1.1]pentanamine hydrochloride (220 mg, 1.84 mmol) and DIEA (300 mg, 2.33 mmol) in NMP (3 mL) was heated at 100 0C for 16 h. The resulting mixture was purified by preparatory high performance liquid chromatography twice (5-80 % acetonitrile in water) to give the crude product (105 mg), which was recrystallized from form to give the desired compound (85.2 mg, 0.27 mmol, yield: 28%). 1H NMR (400 MHz, CDgOD) 8 ppm: 8.33 (s, 1H), 4.19 (d, J = 6.8 Hz, 1H), 3.77-3.74 (m, 1H), 3.60-3.57 (m, 1H), 2.67 (s, 2H), 2.54 (dd, J1 = 9.6Hz, J2 = 3.2 Hz, 1H), 2.07-1.88 (m, 6H), 1.66 (d, J = 9.6 Hz, 1H), 1.39-1.34 (m, 4H); MS (ESI) m/z 318.1 .
Example 53: 2-((S)-sec-Butylamin0)—4-(((1R,3R,4R)—3-hydr0xy methylcyclohexyl)amin0)pyrimidine—5-carb0xamide A. 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0) lthi0)pyrimidine—5-carb0xamide. 4-((1R,3R,4R)Hydroxy methylcyclohexylamino)(methylthio)pyrimidinecarbonitrile (0.335 g, 1.203 mmol; sis described herein) was dissolved in DMSO (3 mL). Then aqueous sodium ATI—2514175v1 hydroxide solution (1.003 mL, 6 M, 6.02 mmol) and aqueous en peroxide solution (0.682 mL, 6.02 mmol, 30 %) were added at room ature. Then the reaction mixture was stirred at 50 °C for 2 h. The reaction was cooled to room temperature and it was poured into 100 mL of ice water. The white precipitate was collected and was washed twice with water. The solid was dried to give 4-((1R,3R,4R)—3-hydroxymethyl- cyclohexylamino)(methylthio)pyrimidinecarboxamide (0.260 g, 0.877 mmol, 72.9 % yield); MS (ESI) m/z 297.7 [M+1]+.
] B. 4-((1R,3R,4R)—3-Hydr0xymethylcyclohexylamin0) (methylsulfonyl)pyrimidine—5-carb0xamide. 4-((1R,3R,4R)Hydroxy methylcyclohexylamino)(methylthio)pyrimidinecarboxamide (315 mg, 1.063 mmol) was dissolved in NMP (4 mL) and cooled to 0 0C before adding mCPBA (476 mg, 2.126 mmol) portionwise. After 1 h, 75 mL of ld water was added directly to the reaction mixture and the slurry was allowed to stir for 2 h before filtering off the white precipitate. The filtrate was then condensed under reduced pressure to remove water. The NMP solution was used directly in the next step assuming quantitative conversion to the desired t; MS (ESI) m/z 329.4 [M+1]+.
C. 2-((S)-sec-Butylamin0)—4-(((1R,3R,4R)—3-hydr0xy methylcyclohexyl)amin0)pyrimidine—5-carb0xamide. To a stirring suspension of 4-((1R,3R,4R)hydroxymethylcyclohexylamino)(methylsulfonyl)pyrimidine carboxamide (0.2 g, 0.609 mmol) in DMF (3 mL) was added (S)—(+)—sec-butylamine (0.305 mL, 3.05 mmol) and the on was heated to 90 oC overnight. The crude reaction mixture was concentrated under reduced pressure and then purified using reverse-phased semi-preparative HPLC (5-75% methanol + 0.1% formic acid in water + 0.1% formic acid, over 26 min). Fractions containing product were concentrated under d pressure and the ing residue was redissolved in methanol (5 mL), passed over a Varian StratoSpheres HCO3 resin SPE tube for formic acid removal (0.9 mmol bicarbonate equiv.), and then concentrated under reduced pressure to afford 2-((S)-sec-butylamino) (((1R,3R,4R)hydroxymethylcyclohexyl)amino)pyrimidine-5 -carboxamide (0. 129 g, 0.402 mmol, 66 % yield). 1H NMR (400 MHz, DMSO-d6) 5 ppm 0.80 - 1.24 (m, 15 H) 1.35 - 1.45 (m, 1 H) 1.54 (br. s., 1 H) 1.66 (d, J=12.89 Hz, 1 H) 1.92 (br. s., 1 H) 2.14 (br. —194— ATI—25 14175v1 s., 1 H) 2.97 (br. s., 1 H) 3.71 — 3.99 (m, 1 H) 4.55 (d, J=5.86 Hz, 1 H) 6.96 (br. s., 1 H) 8.34 (s, 1 H) 8.90 (br. s., 1 H); MS (ESI) m/z 322.5 [M+1]+.
Example 54: ,4r)—4-Hydr0xycyclohexylamin0)(1- methylcyclobutylamin0)pyrimidine—5-carboxamide )L / N N NH H 6 A. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)—4—(1-methylcyclobutyl- amino)pyrimidine—5-carboxamide. To a mixture of 2-((1r,4r)hydroxycyclohexyl- amino)(methylsulf1nyl)pyrimidinecarboxamide and 2-((1r,4r)hydroxycyclohexyl- amino)(methylsulfonyl)pyrimidinecarboxamide (1.414 mmol total; synthesis described herein) in NMP (18 mL) was added 1-methylcyclobutanamine hydrochloride (0.189 g, 1.556 mmol) and DIEA (0.988 mL, 5.66 mmol). The ing solution was heated at 100 CC overnight. Ethyl acetate and water were added to the reaction mixture and the resulting layers were separated. The ethyl acetate layer was dried over anhydrous magnesium sulfate, filtered, and concentrated under reduced pressure to an oil that was d by silica gel chromatography (0-20% methanol in DCM) to afford 2-((1r,4r) hydroxycyclohexylamino)( 1 -methylcyclobutylamino)pyrimidinecarboxamide (0.193 g, 0.604 mmol, 42.7% yield). 1H NMR (400 MHz, DMSO-d6) 8 ppm 9.13 (s, 1 H) 8.33 (s, 1 H) 7.57 (br. s., 1 H) 7.01 (d, J=7.81 Hz, 2 H) 4.53 (d, J=4.69 Hz, 1 H) 3.58 (br. s., 1 H) 2.32 (d, 4 Hz, 2 H) 1.94 - 2.11 (m, 2 H) 1.84 (d, J=5.08 Hz, 6 H) 1.52 (s, 3 H) 1.05 — 1.36 (m, 4 H). MS (ESI) m/z 320.1 [M+1]+.
ATI—2514l75vl 2012/034349 Example 55: 4-(Bicyclo[2.1.1]hexanylamin0)—2-(((1r,4r)—4- ycyclohexyl)amin0)pyrimidine—5-carb0xamide N N NH H @ A. 2-Methylbicyclo[2.2.1]heptan-Z-ol. To a solution of bicyclo[2.2. l]heptanone (25 g, 0.23 mol) in diethyl ether (250 mL) was added a solution of methylmagnesium bromide in THF (3 mol/L, 90 mL, 0.27 mol) over 1 h at 0 CC. The reaction mixture was stirred for an onal 30 min as it was warmed to room temperature. Then the reaction was quenched with 40 mL saturated aqueous ammonium chloride solution. The resulting mixture was extracted with l ether (3 x 80 mL).
The c layers were combined and concentrated to give the desired product (26 g, 0.205 mol, yield: 90%). 1H NMR (400 MHz, CDClg) 5 ppm 2.20 (s, l H), 2.01 (s, 1H), 1.94 (s, l H), 1.61-1.49 (m, 3 H), 1.34-1.20 (m, 7H).
] B. 1-Methylbicyclo[2.2.1]heptan-Z-ol. To a solution of 2-methyl- bicyclo[2.2.l]heptanol (26 g, 0.20 mol) in acetic acid (50 g, 0.83 mol) was added 1 mL sulfuric acid. The mixture was refluxed for 3 h and then concentrated under reduced presssure. The e was poured into 100 mL water. The resulting mixture was extracted with ethyl acetate (3 x 50 mL) and the combined extracts were washed with saturated aqueous sodium bicarbonate on (100 mL), brine (100 mL) and then dried over anhydrous sodium sulfate. Removal of the solvent gave l-methyl- bicyclo[2.2.l]heptanyl acetate (31 g, 0.173 mol, yield: 89%) as a colorless oil, which was used without fithher purification.
A mixture of l-methylbicyclo[2.2. l]heptanyl acetate (31 g, 0. 17 mol) and aqueous sodium hydroxide solution (5%, 250 mL) was refluxed for 2 h. After being cooled to room temperature, the reaction mixture was extracted with diethyl ether (3 x 100 mL) and the combined organic layers were dried over magnesium e and concentrated to afford the desired compound (21 g, 0.167 mol, yield: 82%), which was used in the next step without further purificatioan NMR (400 MHz, CDClg) 5 ppm 3.47- -l96- ATI—2514l75vl 3.46 (m, 1H), 2.16-2.14 (m, 1H), .75(m, 1H), 1.57-1.52(m, 1H), 1.43-1.30(m, 3H), 1.15(brs, 3H), 1.01-0.97(m, 2H).
C. Methylenebicyclo[2.2.1]heptanyl romethanesulfonate. To a solution of oxalyl dichloride (31.75 g,0.25 mol) in DCM (300 mL) was added at -78 0C in consecutive order DMSO (19.5 g, 0.25 mol), a on of 1-methylbicyclo[2.2.1]heptan- 2-ol (21g, 0.167 mol) in DCM (50 mL) and then triethylamine (50 g, 0.5 mol). The mixture was stirred at room temperature ght and then poured into 200 mL water.
The resulting mixture was extracted with DCM (2 x 100 mL). The combined extracts were washed with saturated aqueous sodium bicarbonate solution (50 mL), brine (50 mL) and dried over anhydrous sodium sulfate. Concentration under reduced pressure gave crude methylbicyclo[2.2.1]heptanone, which was purified by silica gel column chromatography (10 % ethyl acetate in eum ether) to afford 12.4 g (0.10 mol, yield: 60%) of this compound.
To a solution of methylbicyclo[2.2. 1]heptanone (26 g, 0.209 mol) and 2,6-di-tert-butylmethyl-pyridine (64 g, 0.315 mol) in DCM (300 mL) was added se trifluoromethanesulfonic anhydride (88 g, 0.315 mol) in DCM (50 mL) at 0 CC.
The on mixture was stirred at room temperature for 24 h. The mixture was poured into water, and extracted with DCM (2 x 100 mL). The combined extracts were washed with a 10% hydrochloric acid solution (3 x 100 mL), saturated aqueous sodium bicarbonate solution (50 mL), brine (50 mL) and dried over sodium sulfate. Concentration under reduced pressure gave the crude t, which was purified by silica gel column chromatography (100% n-pentane) to give the desired product (7.6 g, 0.03 mol, 14%). 1H NMR (400 MHz, : 5 ppm 5.15 (brs, 1H), 4.89 (brs, 1H), 2.51-2.39 (m, 2H), 2.26-2.17 (m, 2H), 2.12-2.05 (m, 2H), 1.99-1.85 (m, 2H), 1.53-1.50 (m, 1H).
D. 2-Ox0bicyclo[2.2.1]heptan-l-yl trifluoromethanesulfonate. A solution of methylenebicyclo[2.2. 1]heptanyl trifluoromethanesulfonate (7.6 g, 0.03 mol) in methanol (100 mL) was cooled to -40°C and an ozone stream was passed through the reaction mixture. When the color of the mixture turned to blue, the ozone was eliminated by passing a stream of argon through the solution for 10 min. Dimethyl sulfide (5 mL) was added to the mixture and the on mixture was allowed to warm to room temperature. The resulting mixture was poured into water and extracted with DCM (2 x ATI—2514l75vl 2012/034349 40 mL). The combined extracts were washed with brine ) and dried over anhydrous sodium sulfate. The solution was concentrated under reduced pressure to give the crude product, which was purified by silica gel column chromatography (10 % ethyl acetate in eum ether) to give the desired product (4.6 g, 18.1 mmol, yield: 60%) as a yellowish oil. 1H NMR (400 MHz, CDClg) 5 ppm rs, 1H), 2.44-2.34 (m, 2H), 2.26- 2.13 (m, 4H), 2.06-2.02(m, 1H), 1.70-1.66 (m, 1H).
E. Bicyclo[2.1.l]hexane-l-carboxylic acid. A mixture of 2-oxobicyclo[2.2. l]heptan-l-yl trifiuoromethanesulfonate (4.6 g,18.1 mmol) in 600 mL ethanol/water (60% w/w) and TEA (3.7 g 36.2 mmol) was heated at 130 0C for 100 h.
The e was concentrated under reduced pressure and the residue was poured into aqueous hydrochloric acid solution (1 mol/L, 150 mL) and extracted with diethyl ether (3 x 100 mL). The combined extracts were washed with brine (100 mL) and dried over anhydrous sodium sulfate. The solution was concentrated under reduced pressure to give the crude product which was purified by silica gel column chromatography (10 % ethyl acetate in petroleum ether) to give the desired product (1.00 g, 8.09 mmol, yield: 45 %). 1H NMR (400 MHz, CDClg) 8 ppm 2.47-2.45(m, 1H), 1.94-1.90 (m, 4H),1.79-1.74 (m, 2H), .35 (m, 2H).
F. Benzyl o[2.l.1]hexan-l-ylcarbamate. To a on of bicyclo[2.1.1]hexanecarboxylic acid (1.0 g, 8.09 mmol) in dioxane (20 mL) was added DIEA (2.5g, 12.4 mmol), diphenylphosphorylazide (3.1 g, 11.2 mmol) and phenylmethanol (1.5 g, 14.2 mmol). The mixture was stirred at 80 °C overnight under nitrogen atmosphere and concentrated under reduce pressure. The residue was purified by silica gel column chromatography (10 % ethyl acetate in petroleum ether) to give the desired product (1.2 g, 5.2 mmol, yield: 65 %). MS (ESI) m/z = 231.2[M+H]+.
G. Bicyclo[2.1.1]hexan-l-amine hydrochloride. To a solution of benzyl bicyclo[2.1.1]hexanylcarbamate (1.2 g, 5.2 mmol) in methanol (30 mL) was added palladium on charcoal (10%, 0.1 g). The reaction mixture was stirred at 50 °C under hydrogen atmosphere (50 Psi) overnight and filtered through . To the filtrate was added a hydrochloric acid solution (10% in methanol, 20 mL). The mixture was concentrated and the residue was suspended in THF (20 mL). The mixture was stirred at room temperature for 1 h. The precipitate was ted and dried to give the desired ATI—25 14175v1 product (550 mg, 4.13 mmol, yield: 79 %). 1H-NMR (400MHz, DMSO-dg) 8 ppm 8.73 (br. s, 3H), 2.4 (br. s, 1H), 1.71-1.69 (m, 6H), 1.32-1.31 (m, 2H). MS (ESI) m/z = 98.2 [M+H]+.
H. yclo [2. 1.1] hexanylamin0)(methylthio)pyrimidine—5- carbonitrile. To a stirred solution of 4-chloro(methylthio)pyrimidinecarbonitrile (427 mg, 2.3 mmol) in 1,4-dioxane (10 mL) was added DIEA (890 mg, 6.9 mmol) and bicyclo[2.1.1]hexanamine hloride (320 mg, 2.4 mmol). The resulting mixture was stirred at 60 CC for 2 h. The reaction mixture was poured into a saturated brine solution and then extracted with ethyl acetate (20 mL) three times. The combined organic layers were dried over anhydrous sodium e, filtered and concentrated to afford the crude product, which was purified by silica gel column tography (15 % ethyl e in petroleum ether) to give the desired product (502 mg, 2.0 mmol, 89% .
MS (ESI) m/Z =247.2 [M+1] +.
] I. yclo[2.1.1] hexanylamin0)—2-(methylthi0)pyrimidine carboxamide. To a solution of 4-(bicyclo[2.1.1]hexanylamino) (methylthio)pyrimidinecarbonitrile (502.0 mg, 2.0 mmol) in DMSO (5 mL) was added aqueous hydrogen peroxide solution (1.2 g, 30%, 10.2 mmol) and aqueous sodium hydroxide solution (1.7 mL, 6 mol/L, 10.2 mol) at 0 CC. The mixture was stirred at 50 0C for 15 min and then quenched by the addition of water (30 mL). The solid formed was collected and dried under vacuum to give the desired product (500 mg, 1.9 mmol, 93% yield). MS (ESI) m/Z =265.3 [M+1] +.
J. 4-(Bicyclo[2.1.1] hexan-l-ylamin0)—2-(methylsulfinyl)pyrimidine—5- carboxamide and 4-(bicyclo[2.1.1]hexanylamin0)—2-(methylsulfonyl)pyrimidine carboxamide. To a solution of 4-(bicyclo[2.1.1]hexanylamino)(methylthio)- pyrimidinecarboxamide (500 mg, 1.9 mmol) in THF (10 mL) at 0 °C was added mCPBA (490 mg, 2.8 mmol) portion-wise. After 1 h, the reaction mixture was concentrated. The resulting crude material was purified by silica gel column chromatography (5-10% methanol in DCM) to give 510 mg of the desired mixture.
MS (ESI) m/Z =281.3, 297.3 [M+1] +.
K. yclo[2.1.1]hexanylamin0)—2-(((1r,4r)—4-hydr0xycyclo- hexyl)amin0)pyrimidine—5-carb0xamide. To the mixture of 4-(bicyclo[2.1.1]hexan ATI—2514175v1 ylamino)(methylsulfinyl)pyrimidinecarboxamide and 4-(bicyclo[2.1.1]hexan ylamino)(methylsulfonyl)pyrimidinecarboxamide (200 mg, about 0.71 mmol ) and (1r,4r)aminocyclohexanol (123.3 mg, 1.02 mmol) in NMP (10 mL) was added DIEA (183.0 mg, 1.42 mmol). The resulting mixture was stirred at 100 CC overnight. The reaction mixture was d by preparative HPLC to give the desired product (187.3 mg, 0.56 mmol, 72% yield). 1H NMR (400 MHZ, CDgOD) 5 ppm 8.29(s, 1H), 3.77(m, 1H), , 1H), 2.43(s, 1H), 2.07-1.93(m, 8H), .77(m, 2H), 1.60(s, 2H), l.4l-l.35(m, 4H). MS (ESI) m/Z =332.2 [M+H] +.
Example 56: 4-(1-Ethylcyclopentylamino)—2-((1r,4r)—4- ycyclohexylamino)pyrimidine—5-carboxamide A. Ethyl cyclopentenecarb0xylate. To a stirred solution of cyclopent- 3-enecarboxylic acid (10 g, 89.3 mmol) in anhydrous ethanol (30 mL) was added sulfurous dichloride (15.9 g, 134 mmol) at 0 0C. The on mixture was stirred at room temperature overnight and then concentrated. The residue was poured into water. The resulting mixture was extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated and the residue was d by silica gel column chromatography (2.5 % ethyl acetate in petroleum ether) to give the desired product (6.7 g, 47.9 mmol, yield = 54%) as a yellow oil which was used in the next step without further purification. 1H NMR (400 MHz, CDClg) 5 ppm 5.66 (s, 2H), 4.18-4.10 (m, 2H), 3.16- 3.05(m, 1H), 2.66 (s, 2H), 2.63 (s, 2H), 1.29-1.24 (t, J=9.4, 3H).
B. Ethyl 1-ethylcyclopentenecarboxylate. To a on of diisopropylamine (10 mL, 72 mmol) in anhydrous THF (50 mL) was added l lithium (29 mL, 72 mmol, 2.5 M solution in hexane) over 20 min at -78 0C. The mixture was stirred at 0 0C under nitrogen for 1 h. The freshly prepared lithium diisopropylamide was added to a mixture of ethyl cyclopentenecarboxylate (6.7 g, 47.9 mmol) in ATI—2514175v1 anhydrous THF (50 mL) over 20 minutes at -78 0C. After another 1 hour at this temperature, iodoethane (11.2 g, 72 mmol) was added over 20 min. Then the mixture was stirred at room temperature for 2 h and quenched with saturated aqueous ammonium chloride solution. The aqueous layer was extracted with ether (3 x 100 mL). The combined organic layers were washed with diluted hydrochloric acid (1 N), then washed with brine, dried over anhydrous sodium e and filtered. Concentration gave the titled compound (7 g crude) which was used in the next step without fithher purification. 1H NMR (400 MHz, CDClg) 8 ppm 5.60 (s, 2H), 4.21-4.12 (m, 2H), 2.92-2.84 (m, 2H), 2.31-2.25 (m, 2H), 1.75-1.67 (m, 2H), 1.29-1.24 (m, 3H), 0.88-0.82 (m, 3H).
] C. 1-Ethylcyclopent—3-enecarboxylic acid. A solution of ethyl l-ethylcyclopentenecarboxylate (3.5 g, 20.8 mmol) and aqueous sodium hydroxide solution (20.8 mL, 2 mol/L) in methanol (30 mL) was stirred at 80 CC for 2 h and then concentrated. The e was poured into water, and extracted with ether (3 x 100 mL).
The water layer was acidified with 4 mol/L s hydrochloric acid solution to pH < 3, and extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. Concentration under reduced pressure gave the crude product, which was purified by silica gel column chromatography (5 % ethyl e in eum ether) to afford the title compound (1.9 g, 13.6 mmol, 65.5 % yield). 1H NMR (400 MHz, CDC13)8 ppm 5.61 (s, 2H), 2.91 (d, J=l4.8, 2H), 2.32 (d, J=l4.4, 2H), 1.78-1.72 (m, 2H), 0.92-0.88 (t, J=7.4, 3H).
D. Benzyl (1-ethylcyclopentenyl)carbamate. To a solution of l-ethylcyclopentenecarboxylic acid (1.9 g, 13.6 mmol) in dioxane (30 mL) was added DIEA ( 5.3 g, 41 mmol), diphenyl phosphoryl azide (4.5 g, 16.3 mmol) and benzylalcohol (2.2 g, 20.4 mmol). The mixture was stirred at 80 CC overnight and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (2.5 % ethyl e in petroleum ether) to give the title product. (1.3 g, 5.3 mmol, yield: 39 %). MS (ESI) m/Z = 246.2 [M+1] +.
] E. l-Ethylcyclopentanamine hydrochloride. To a on of benzyl (1-ethylcyclopentenyl)carbamate (1.3 g, 5.3 mmol) in methanol (20 mL) was added palladium on charcoal (130 mg, 10 %). The reaction mixture was stirred at 50 °C under hydrogen atmosphere overnight. A solution of hydrochloric acid (20 mL, 1 mol/L in ATI—2514l75vl methanol) was added to the reaction and the reaction mixture was filtered through celite.
The filtrate was concentrated under reduced pressure to give the crude product (900 mg), which was used for the next step without filrther ation. MS (ESI) m/z = 114.2 [M+H] F. 4-(1-Ethylcyclopentylamin0)(methylthio)pyrimidine—5- awbmfinfla Toasfiflngsobfionof4£hbny2{nwflwhhmfiwfinnmne5fimbomfifle (984 mg, 5.3 mmol) in DMSO (10 mL) was added DIEA (2.1 g, 15.9 mmol) and 1-ethylcyclopentanamine hydrochloride (900 mg crude). The resulting mixture was stirred at 60 CC for 2 h. The reaction mixture was poured into water and extracted with ethyl e. The combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to afford the crude product. The crude product was purified by silica gel column tography (5 % ethyl acetate in petroleum ether) to get the desired product as a yellow oil (830 mg, 3.2 mmol, 53.5 % yield). MS (ESI) m/z = 263.2 [M+H] +. [0051 1] G. 4-(1-Ethylcyclopentylamin0)(methylthi0)pyrimidine carboxamide. To a stirred on of 4-(1-ethylcyclopentylamino)(methylthio)- dinecarbonitrile (830 mg, 3.2 mmol) in DMSO (10 mL) was added aqueous hydrogen peroxide (1.8 g, 30%, 16 mmol), and aqueous sodium hydroxide solution (2.7 mL, 6 mol/L, 16 mmol) at 0 CC. The mixture was stirred at 50 CC for 15 min and diluted with water. The mixture was extracted with ethyl acetate (3 x 100 mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. Concentration under reduced re gave the crude product, which was purified by silica gel column chromatography (5 % methanol in DCM) to afford the d product as a white solid (800 mg, 2.9 mmol, 90% yield). MS (ESI) m/z = 281.2 [M+1] H. 4-(1-Ethylcyclopentylamin0)(methylsulfonyl)pyrimidine—5- carboxamide. At 0 0C, mCPBA (873 mg, 4.3 mmol) was added portion-wise to a on of 4-(1-ethylcyclopentylamino)(methylthio)pyrimidinecarboxamide (800 mg, 2.9 mmol) in THF (30 mL). After 1 h, the reaction mixture was concentrated and the resulting crude was d by silica gel column chromatography (5 % methanol in DCM) to afford the desired product (800 mg, 2.56 mmol, 90% yield). MS (ESI) m/z = 313.2 [M+1] +.
ATI—25 14175V1 I. 4-(1-Ethylcyclopentylamin0)((1r,4r)—4-hydr0xycyclohexylamin0)- pyrimidine—5-carb0xamide. To a mixture of 4-(1-ethylcyclopentylamino)(methyl- sulfonyl)pyrimidinecarboxamide (200 mg, 0.64 mmol) and (1r,4r)aminocyclohexanol (115 mg, 1 mmol) in NMP (10 mL) was added DIEA (255 mg, 1.98 mmol).
The resulting mixture was stirred at 100 CC ght. The reaction mixture was purified by preparative HPLC to give the desired product (117.0 mg, 0.34 mmol, yield 51%). 1H NMR (400 MHz, CDgOD) 5 ppm 8.17 (s, 1H), 3.66 (s, 1H), 3.46 (s, 1H), 2.06-2.04 (m, 2H), 1.98-1.89 (m, 6H), 1.66-1.58 (m, 6H), 1.26-1.24 (m, 4H), .73 (t, J=7.2 Hz, 3H); MS (ESI) m/z = 348.2 [M+1]+.
Example 57: 4-(((1R,3S)Hydr0xycyclohexyl)amin0)((4-meth0xybicyclo[2.2.2]octan-l- yl)amin0)pyrimidinecarboxamide A. yl 1-(2-chlor0ethyl)cyclohexane-1,4-dicarb0xylate. To a stirring solution of cyclohexane-1,4-dicarboxylic acid (100 g, 0.58 mol) in anhydrous methanol (800 mL) was added sulfiarous dichloride (208g, 1.75 mol) at 0 0C. The reaction mixture was stirred at room temperature overnight and then concentrated under d pressure. The residue was poured into water. The ing mixture was extracted with ethyl acetate (200 mL x 3). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate and filtered. Concentration gave crude dimethyl cyclohexane-1,4-dicarboxylate (105 g, 0.53 mol, yield 90.5%) which was used without further purification.
To a solution of diisopropylamine (88 mL, 0.62 mol) in ous THF (500 mL) was added n-butyl lithium (240 mL, 0.6 mol, 2.5 M solution in hexane) over min at -78 0C. The mixture was stirred at 0 0C under nitrogen for 30 min.
] To the above described mixture of crude yl cyclohexane-1,4- dicarboxylate (100 g, 0.5 mol) and hexamethylphosphoramide (360 mL, 2 mol) in anhydrous THF (800 mL) was added the freshly prepared lithium diisopropylamide ATI—25 14175v1 solution (preparation described above) over 30 min at -40 0C. After stirring for l h at this temperature, l-bromochloroethane (42 mL, 0.5 mol) was added over 1. The mixture was stirred for 3 h at -78 0C and then stirred ght at room temperature. The reaction was quenched by the addition of aqueous hydrochloric acid on (3 N, 420 mL). The solvent was removed under reduced pressure. The aqueous layer was extracted with ethyl acetate (200 mL x 3). The combined extracts were washed with brine (2x300 mL) and dried over sodium e. Concentration gave the title nd (1 16 g, yield 88%) which was used in the next step without further purification; 1H NMR (400 MHz, CDClg) ppm 3.72 (s, 3H), 3.65 (s. 3H), 3.46-3.42 (m, 2H), 2.33-2.21 (m, 3H), 2.05-l.85(m, 4H), 158-1 .42 (m, 2H), l.25-l.l5(m, 2H).
B. Dimethyl bicyclo[2.2.2]octane-1,4-dicarb0xylate. To a solution of ropylamine (77 mL, 0.54 mol) in anhydrous THF (500 mL) was added n-butyl lithium (210 mL, 0.53 mol, 2.5 M solution in hexane) over 20 min at -78 0C. The mixture was stirred at 0 0C under nitrogen for 30 min. [005 l 8] To a mixture of dimethyl l-(2-chloroethyl)cyclohexane-l ,4-dicarboxylate (1 16 g, 0.44 mol) and hexamethylphosphoramide (3 17 mL, 1.7 mol) in anhydrous THF (800 mL) was added y prepared lithium diisopropylamide (preparation described above) over 30 min at -40 0C. The e was stirred for 2 h at -78 0C and then stirred overnight allowing warming to room temperature. To the reaction mixture was added saturated aqueous ammonium chloride solution (200 mL) and the mixture was stirred for min. The volatile ts were removed by evaporation under reduced pressure. The aqueous layer was extracted with ethyl acetate (3 x 200 mL) and the combined extracts were washed with brine (300 mL x 2) and dried over sodium e. Concentration under reduced pressure gave crude product, which was purified by silica gel column chromatography (10 % ethyl acetate in petroleum ether) to afford the title compound (58.0 g, 0.25 mol, yield 50% over two steps). 1H NMR (400 MHz, CDC13)C 8 ppm 3.65 (s, 6H), 1.81 (s. 12H).
C. 4-(Methoxycarbonyl)bicyclo[2.2.2]octane-l-carboxylic acid. A solution of dimethyl bicyclo[2.2.2]octane-l,4-dicarboxylate (58.0 g, 0.25 mol) in methanol (600 mL) was heated to reflux and then a solution of potassium hydroxide (9.8 g, 0. 175 mol) in methanol (100 mL) and water (12 mL) was added over 30 min. The on —204— ATI—2514l75vl mixture was refluxed for 24 h and concentrated. The residue was diluted with water and ted with ethyl e (200 mL x 2) to recover some starting material (22.0 g). The rammgammmmwawwadmfimhomfl=3bymMMmufihmeMMmaddmfl extracted with ethyl acetate (300 mL x 3). The combined organic layers were washed with brine, dried over sodium sulfate and concentrated to give the titled product (30.0 g, 0.14 mol, yield 55%). 1H NMR (400 MHz, CDClg) 8 ppm 3.65 (s, 3H), 1.81 (s. 12H); MS (ESI) m/z = 211.3 [M-H]'.
D. Methyl 4-br0m0bicyclo[2.2.2]octane-l-carboxylate. To a sion of 4-(methoxycarbonyl)bicyclo[2.2.2]octanecarboxylic acid (11.0 g, 51.8 mmol) in acetone (80 mL) was added aqueous sodium hydroxide solution (1 M, 51.8 mL, 51.8 mmol) and a solution of silver nitrate (8.8 g, 51.9 mol) in water (10 mL). The precipitate formed was ted by filtration, washed with water, acetone and diethyl ether and dried in vacuo at 115 0C for 4 h. The obtained ((4-(methoxycarbonyl)- bicyclo[2.2.2]octanecarbonyl)oxy)silver (15.3 g, 47.9 mmol) was suspended in hexane (125 mL), followed by addition of bromine (7.7 g, 48.1 mmol) over 30 min at room temperature. After the addition was completed, the reaction mixture was stirred at room temperature for another 30 min. The on mixture was filtered to remove the solid, and the filter cake was washed with hexane (150 mL x 4). The combined organic filtrates were washed with saturated aqueous sodium bicarbonate solution (150 mL x 2) and brine (200 mL), then dried over magnesium sulfate. Concentration under reduced pressure gave the crude product, which was purified by silica gel column chromatography (5 % ethyl acetate in petroleum ether) to afford the title compound (4.2 g, 0.17 mol, 33 % yield over two steps). 1H NMR (400 MHz, CDClg): 8 ppm 3.64 (s, 3H), 2.27-2.20 (m, 6H), 1.98-1.94 0n.6H) E. 4-Hydr0xybicyclo[2.2.2]octane-l-carboxylic acid. Methyl 4- bromobicyclo [2.2.2]octane-l-carboxylate (17.0 g, 69.0 mmol) was refluxed in 1.5 L of 1% aqeous sodium hydroxide solution for 24 h. After cooling down, the reaction solution was acidified with hloric acid (6N, 100 mL) and extracted with l ether (6 x 500 mL). The combined organic layers were dried over magnesium sulfate and trated to afford the titled compound (10.4 g, 61.1 mmol, yield: 89 %), which was used in the next step without further purification; MS (ESI) m/z = 169.2 [M-H]'.
ATI—25 14175v1 F. Methyl 0xybicyclo[2.2.2]octane-l-carboxylate. To a solution of 4-hydroxybicyclo[2.2.2]octane-l-carboxylic acid (14 g, 82.4 mmol) in methanol (300 mL) was added concentrated sulfuric acid (1 mL). The mixture was refluxed for 10 h and then concentrated. The reside was dissolved in water (100 mL) and the mixture was extracted with ethyl acetate (200 mL x 2). The combined organic phases were washed with saturated aqueous sodium en carbonate solution (100 mL), brine (100 mL) and dried over sodium sulfate. Concentration gave the title compound (14.5 g, yield 96%). 1H NMR (400 MHz, CDClg) 8 ppm 3.64 (s, 3H), 1.96-1.89 (m, 6H), 1.69-1.64 (m. 6H).
G. Methyl 4-meth0xybicyclo[2.2.2]octane-l-carboxylate. To a solution of methyl 4-hydroxybicyclo[2.2.2]octane-l-carboxylate (8.9 g, 48 mmol) in anhydrous THF (150 mL) at -78 CC was added n-butyl lithium (23 mL, 57.5 mmol, 2.5 M solution in hexane) over 30 min, ed by the slow addition of iodomethane (14 g, 98 mmol).
The mixture was stirred at 60 0C for 3 h and ed by addition of saturated aqueous ammonium chloride solution (50 mL). The resulting solution was concentrated and the residue was extracted with ethyl acetate (150 mL x 2). The organic layers were ed and washed with brine, dried over magnesium sulfate and then concentrated. The residue was purified by silica gel column chromatography (10% ethyl acetate in petroleum ether) to give the title product (6.5 g, 67%). 1H NMR (400 MHz, CDClg) 8 ppm3.64 (s, 3H), 3.18 (s, 3H), 1.95-1.89 (m, 6H), 1.70-1.65 (m. 6H).
] H. 0xybicyclo[2.2.2]octane-l-carboxylic acid. Methyl 4- methoxybicyclo[2.2.2]octane-l-carboxylate (6.5 g, 33 mmol) was refluxed in aqueous sodium hydroxide solution (5%, 150 mL) for 2 h. After cooling down, the reaction solution was acidified with hydrochloric acid solution (6 N, 50 mL) and extracted with ethyl acetate (100 mL x 2). The combined organic layers were dried over magnesium sulfate and concentrated to afford the title compound (5.9 g, 32 mmol, yield 97%) which was used in the next step without further purification. MS (ESI) m/z = 183.2 [M-H]'.
I. Benzyl h0xybicyclo[2.2.2]0ctanyl)carbamate. To a solution of 4-methoxybicyclo[2.2.2]octane-l-carboxylic acid (5.9 g, 32 mmol) in e (80 mL) was added DIEA (8.3 g, 64.2 mmol), diphenyl oryl azide (13.2 g, 48 mmol) and methanol (17.3 g, 160 mmol). The mixture was stirred at 80 CC overnight and then concentrated under reduce pressure. The residue was purified by silica gel column ATI—2514l75vl chromatography (5 % methanol in DCM) to give the desired product. (9 g, 31 mmol, yield 96%). MS (ESI) m/Z = 290.2 [M+H]+.
J. 4-Meth0xybicyclo[2.2.2]octan-l-amine hloride. To a on of benzyl (4-methoxybicyclo[2.2.2]octanyl)carbamate (9 g, 31 mmol) in methanol (150 mL) was added palladium on charcoal (0.5 g, 10%). The reaction mixture was stirred at 50 0C under hydrogen atmosphere (50 psi) overnight and then filtered h celite.
The filtrate was concentrated under reduced pressure. The residue was dissolved in hydrochloric acid solution (10% in methanol, 50 mL) and stirred for 2 h at room temperature. The mixture was concentrated and the resulting residue was suspended in THF (20 mL). The mixture was stirred at room temperature for 1 h and the itate was collected and dried to give the desired product (3.8 g, 20 mmol, yield: 68%). 1H-NMR (400MHz, ) 8 ppm 8.14 (brs, 3H), 3.04 (s, 3H), 1.83-1.80 (m, 6H), 1.68-1.64 (m, 6H); MS (ESI) m/Z = 156.1[M+1]+.
K. 4-((1R,3S)Hydr0xycyclohexylamin0)(4-meth0xy— o[2.2.2]octanylamin0)pyrimidine—5-carboxamide. To a solution of 4-((1R,3S)- 3-hydroxycyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (1.2 g, 3.8 mmol; sis described herein) and 4-methoxybicyclo[2.2.2]octanamine hydrochloride (600 mg, 3.1 mmol) in NMP (12 mL) was added DIEA (1.03 g, 8 mmol).
Theremflfingnnxunewmssfinedat130°Clnubrnnmowmveinadmfionfor3h.The resulting mixture was purified by reverse phase column tography to give the desired product (230 mg, 0.6 mmol, 15.5 % yield). 1H-NMR (400MHz, CDClg) 5 ppm 8.70 (brs, 1H), 8.08 (s, 1H), 5.40 (brs, 2H), 5.01 (br. s, 1H), 3.99-3.96 (m, 1H), 3.75-3.70 (m, 1H), 3.19 (s, 3H), 2.37-2.34 (m, 1H), 2.15-2.11 (m, 6H), 1.99-1.97 (m, 2H), 1.90-1.86 (m, 1H), 1.80-1.76 (m, 6H), 1.38-1.22 (m, 4H). MS (ESI) m/z = 390.2[M+1]+.
ATI—25 14175v1 Example 58: ,4r)—4-Hydr0xycyclohexylamino)—4-(3- (trifluoromethyl)bicyclo[1.1. 1]pentan-l-ylamin0)pyrimidinecarboxamide A. 2-((1r,4r)—4-Hydr0xycyclohexylamin0)(3-(triflu0r0methyl)- bicyclo[1.1.1]pentanylamin0)pyrimidine—5-carb0xamide. To a 5 mL NMP solution of (2-((1r,4r)hydroxycyclohexylamino)(methylsulfonyl)pyrimidinecarboxamide (445 mg, 1.416 mmol)) was added 3-(trifluoromethyl)bicyclo[1.1.1]pentanamine hydrochloride (266 mg, 1.416 mmol) and DIEA (0.494 mL, 2.83 mmol). The reaction was then heated at 90 oC overnight. LCMS showed the desired product mass as the dominant peak. The crude reaction was purified directly on a semi-prep HPLC (method: 5-80% acetonitrile + 0.1% TFA in water + 0.1% TFA). Product fractions were combined and evaporated under reduced pressure to a volume <5 mL. The material was then neutralized with saturated sodium bicarbonate and extracted with ethyl acetate (2X). The combined organic layers were dried over ium sulfate, filtered, and concentrated. The resulting solid was dissolved in methanol and washed through a StratoSpheres SPE PL-HCO3 in column, g with methanol. ation of solvent under reduced pressure gave 2-((1r,4r)hydroxycyclohexylamino)(3-(trifiuoromethyl)bicyclo [1 . 1 . 1]pentan ylamino)pyrimidinecarboxamide. The material was purified by silica gel chromatography (0-10% methanol in DCM over 1650 mL; 40 mL/min). Product fractions were ed and evaporated to dryness to afford 2-((1r,4r)hydroxycyclohexyl- amino)(3-(trifluoromethyl)bicyclo[1 . 1 . anylamino)pyrimidinecarboxamide (135 mg, 0.35 mmol, 24.8% yield); 1H NMR (400 MHz, DMSO-d6) 5 ppm 9.54 (s, 1H), 8.41 (s, 1H), 7.57 - 7.86 (m, 1H), 7.26 - 7.32 (m, 1H), 6.91 - 7.17 (m, 1H), 4.60 (d, J=4.29 Hz, 1H), 3.49 - 3.73 (m, 1H), 3.35 - 3.46 (m, 1H), 2.38 (s, 6H), 1.86 (d, J=11.32 Hz, 4H), 1.26 (br. s., 4H); MS (ESI) m/z 386.0 [M+1]+.
ATI—25 14175v1 Example 59: 4-(tert—Butylamino)—2-(((1R,3S)hydr0xycyclohexyl)amin0)pyrimidine carboxamide HO11 1*”*N/ )2 A. 4-(Tert-butylamino)—2-(((1R,3S)hydr0xycyclohexyl)— amin0)pyrimidinecarb0xamide. To a mixture of 4-(tert-butylamino) (methylsulfonyl)pyrimidinecarboxamide (250 mg, 0.918 mmol; synthesis bed herein) and (1S,3R)aminocyclohexanol (138 mg, 1.2 mmol; prepared as described in Tetrahedron.‘ Asymmetry 15:2051—2056 (2004)) in NMP (10 mL) was added DIEA (374 mg, 2.9 mmol). The resulting mixture was stirred at 100 CC overnight. The reaction mixture was purified by preparative HPLC to give the desired product (212.2 mg, 0.69 mmol, yield 75%). 1H NMR (400 MHz, CDClg) 5 ppm 8.86 (brs, 1H), 8.12 (s, 1H), .53 (brs, 3H), 3.93-3.91 (m, 1H), 3.88-3.82 (m, 1H), 2.27-2.25 (m, 1H), 1.92-1.85 (m, 3H), 1.47 (s, 9H), 1.42-1.25 (m, 4H); MS (ESI) m/z = 308.2 [M+1]+.
Example 60: 4-(Bicyclo[1.1.1]pentanylamin0)((tetrahydro-ZH-pyran yl)amin0)pyrimidinecarboxamide o N \ NH2 )L / N N NH ” <> A. yclo[1.1.1]pentanylamin0)(methylthi0)pyrimidine carbonitrile. To a d solution of 4-chloro(methylthio)pyrimidinecarbonitrile (3.0 g, 16 mmol) in DMSO (20 mL) was added DIEA (6.2 g, 48 mmol) and bicyclo[1.1.1]pentanamine hydrochloride (1.9 g, 16 mmol; prepared according to Org.
Lett., 13(17): 4746-4748 ). The resulting e was stirred at 60 CC for 2 h and then poured into water. The mixture was extracted with ethyl acetate and the combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated to afford the crude t, which was purified by silica gel column chromatography (10 % ATI—25 14175v1 emflawmmmpambmnaMfimgWemewmmdmmmmG5gJ49mmdfl3% yield). MS (ESI) m/Z = 233.2 [M+1]+.
B. yclo[1.1.1]pentan-l-ylamin0)—2-(methylthi0)pyrimidine amide. To a solution of yclo[1.1.1]pentan-l-ylamino)—2-(methylthio)- pyrimidinecarbonitrile (3.45 g, 14.9 mmol) in DMSO (15 mL) was added aqueous hydrogen peroxide solution (8.4 g, 30%, 74.5 mmol) and aqueous sodium hydroxide yflufion(124¢nL,6rnoUL,7451nnufl)at0°C.Thennxuuewmssfinedat50OClbr min and then quenched by the addition of water (30 mL). The solid formed was collected and dried under vacuum to give the desired product (3.5 g, 14.0 mmol, 94% yield). MS (ESI) m/Z =251.2 [M+1]+.
C. 4-(Bicyclo[1.1.1]pentanylamin0)—2-(methylsulfinyl)pyrimidine—5- carboxamide and 4-(bicyclo[1.1.1]pentanylamin0)—2-(methylsulfonyl)pyrimidine—5- carboxamide. mCPBA (4.3 g, 21 mmol) was added portion-wise to a solution of 4-(bicyclo[1.1.1]pentanylamino)(methylthio)pyrimidinecarboxamide (3 .5 g, 14 mmol) in THF (40 mL) at 0 0C. The reaction was monitored by thin layer chromatography. After 1 h, the reaction e was concentrated. The resulting crude mmmdwwpmfiwbywkaghmmmmMmegmm45%mmMmflmDCMflo afford 3.1 g of the titled mixture. MS (ESI) m/Z =267.2 [M+1] +/283.2 [M+1]+.
D. 4-(Bicyclo[1.1.1]pentanylamin0)((tetrahydro-ZH-pyran yl)amin0)pyrimidinecarboxamide. To a e of yclo[1 . 1.1]pentan ylamino)—2-(methylsulf1nyl)pyrimidinecarboxamide, 4-(bicyclo[1 . 1 . 1]pentan ylamino)(methylsulfonyl)pyrimidinecarboxamide (300 mg) and tetrahydro-2H- pyranamine hydrochloride (181.5 mg, 1.32 mmol) in NMP (10 mL) was added DIEA (426rng,331nnnfl) Theremflfingnnxuuewmssfinedat100°C(nwnnght Thenthe reaction mixture was purified by ative HPLC to give the desired product (221.3 mg, 0.73 mmol, yield 67 %) as a yellow powder. 1H NMR (400 MHz, CDgOD) 8 ppm 8.19 (s, 1H), 3.95-3.88 (m, 3H), 3.44-3.38 (m, 2H), 2.40 (s, 1H), 2.09 (s, 6H), 1.88-1.85 (m, 2H), 1.55—1.45 (m, 2H); MS (ESI) m/z = 304.2 [M+1]+.
ATI—25 14175v1 Example 61: 4-(Bicyclo[1.1.1]pentanylamin0)(((1R,3S) hydroxycyclohexyl)amin0)pyrimidinecarb0xamide A. 4-(bicyclo[1.1.1]pentanylamin0)(methylsulfinyl)pyrimidine carboxamide and 4-(bicyclo[1.1.1]pentanylamin0)(methylsulfonyl)pyrimidine carboxamide. 3-Chlorobenzoperoxoic acid (4.27 g, 21 mmol) was added portion-wise to a solution of 4-(bicyclo[l . l . l]pentan-l -ylamino)(methylthio)pyrimidinecarboxamide (3.50 mg, 14 mmol, synthesis described herein) in THF (40 mL) at 0 CC. The resulting solution was mixed at 0 0C for l h and then concentrated under reduced pressure to an oil that was purified by silica gel column chromatography (5 % methanol in DCM) to afford 310 mg of a mixture of 4-(bicyclo[l . l .l]pentan-l-ylamino)(methylsulfinyl)pyrimidine- -carboxamide and 4-(bicyclo[l . l . l]pentan- l -ylamino)(methylsulfonyl)pyrimidine carboxamide as a yellow solid. MS (ESI) m/Z =267.2 [M+l] +/283.2 [M+l]+.
B. 4-(Bicyclo[1.1.1]pentanylamin0)(((1R,3S)hydr0xycyclo- hexyl)amin0)pyrimidinecarb0xamide. To a mixture of 4-(bicyclo[l.l.l]pentan-l- ylamino)(methylsulfinyl)pyrimidinecarboxamide and 4-(bicyclo[l - . l . l n- l ylamino)(methylsulfonyl)pyrimidinecarboxamide (300 mg) and (l 3- aminocyclohexanol (152 mg, 1.32 mmol; prepared as described in Tetrahedron: Asymmetry 152051—2056 (2004)) in NMP (10 mL) was added DIEA (426 mg, 3.3 mmol).
The ing e was stirred at 100 CC overnight. The reaction mixture was purified by preparative HPLC to give the desired t (230.2 mg, 0.73 mmol, yield 66 %). 1H NMR (400 MHz, CDgOD) 5 ppm 8.29 (s, 1H), 3.91-3.89 (m, 1H), .62 (m, 1H), 2.50 (s, 1H), 2.29-2.25 (m, 1H), 2.20 (s, 6H), 1.97-1.83 (m, 3H), 1.41-1.21 (m, 4H); MS (ESI) m/z = 318.2 [M+1]+. -2ll- ATI—2514l75vl Example 62: 2-(Bicyclo[1.1.1]pentan-l-ylamino)—4-((1R,3R)—3-hydr0xy-4,4- dimethylcyclohexylamin0)pyrimidinecarb0xamide 3 JL / NH2 N N NH A. N-(Bicyclo[1.1.1]pentanyl)br0m0(methylthio)pyrimidin-Z- amine. To a stirring solution of 5-bromochloro(methylthio)pyrimidine (1.0 g, 4.18 mmol) in NMP (10.0 mL) was added bicyclo[1.1.1]pentanamine hydrochloride (749 mg, 6.26 mmol, prepared according to Org. Lett., 13(17): 4746-4748 (2011)) and DIEA (2.19 mL, 12.53 mmol). The mixture was stirred at 100 CC for 16 h and then the solvent was d under reduced pressure. The residue was d in 100 mL ethyl acetate and 50 mL 1M aquoues solution of sodium hydrogen phosphate. The layers were separated and the aqueous layer was extracted with 50 mL ethyl acetate. The combined organic layers were dried over anyhydrous ium sulfate, d and concentrated under reduced pressure to afford N-(bicyclo[1.1.1]pentan-l-yl)bromo (methylthio)pyrimidinamine (1250 mg, 4.37 mmol, 105 % yield) as a solid that was used without further purification. MS (ESI) m/z 286.0 [M+1]+ and 288.0 .
B. 2-(Bicyclo[1.1.1]pentan-l-ylamin0)—4-(methylthi0)pyrimidine carbonitrile. N-(Bicyclo[1 . 1 . 1]pentanyl)-5 -bromo(methylthio)pyrimidinamine (1250 mg, 4.37 mmol), zinc dust (71.4 mg, 1.092 mmol), zinc e (333 mg, 2.84 mmol), 1,1'-bis-(diphenylphosphino)-ferrocene (196 mg, 0.349 mmol), tris(dibenzylideneacetone)dipalladium(0) (200 mg, 0.218 mmol), and DMA (8.35 mL) were combined and heated overnight at 90 0C under a nitrogen atmosphere. The reaction mixture was diluted with 125 mL ethyl acetate and 50 mL water and filtered through a pad of . The layers of the filtrate were separated and the aqueous layer was extracted with 75 mL ethyl acetate. The combined ethyl acetate layers were washed with 2 x 50 mL brine, dried over anhydrous magnesium sulfate, filtered and concentrated to an oil. The crude oil was d by silica gel chromatography (0% - 30% ethyl e in hexanes).
The product containing fractions were combined and concentrated to afford 2- ATI—2514175v1 lo[1.1.1]pentan-l-ylamino)(methylthio)pyrimidinecarbonitrile (566 mg, 2.436 mmol, 55.8 % yield) as a solid. 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.81 - 9.08 (m, 1 H), 8.31 - 8.57 (m, 1 H), 2.55 - 2.63 (m, 3 H), 2.11 (s, 7 H). MS (ESI) m/z 233.3 [M+1]+.
C. 2-(Bicyclo[1.1.1]pentanylamin0)(methylthio)pyrimidine—5- carboxamide. To a stirring solution of 2-(bicyclo[1 . 1.1]pentanylamino)—4- (methylthio)pyrimidinecarbonitrile (566 mg, 2.436 mmol) in DMSO (7 mL) was added 6 M aqueous sodium hydroxide solution (2.030 mL, 12.18 mmol) and 30% aqueous hydrogen peroxide solution (1.381 mL, 12.18 mol) at 0 CC. The mixture was then stirred at 50 CC for 30 min and then was poured into ice water (50 mL). The solids that formed were stirred in an ice bath for 30 min, d, washed with water, and then dried in a vacuum oven overnight at 45 0C to afford 2-(bicyclo[1.1.1]pentanylamino) (methylthio)pyrimidinecarboxamide (410 mg, 1.638 mmol, 67.2 % yield). 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.41 (s, 1 H), 7.03 - 8.34 (m, 2 H), 2.43 - 2.48 (m, 1 H), 2.35 (br. s., 3 H), 2.09 (s, 6 H). MS (ESI) m/z 251.3 [M+1]+.
D. 2-(Bicyclo[1.1.1]pentanylamin0)—4-(methylsulfinyl)pyrimidine—5- carboxamide. To a stirring solution of 2-(bicyclo[1 . 1.1]pentanylamino)—4- (methylthio)pyrimidinecarboxamide (410 mg, 1.638 mmol) in chloroform (39.0 mL) was added portionwise, 3-phenyl(phenylsulfonyl)-1,2-oxaziridine (514 mg, 1.965 mmol). The ing pale yellow solution was stirred at ambient temperature for two nights under nitrogen. The reaction mixture was trated under reduced pressure to give the crude product as a white solid. Ethyl acetate (15 mL) was added to these solids and the slurry was stirred at room temp for 1 h, filtered, washed with ethyl acetate, and dried in a vacuum oven for a few hours to afford 2-(bicyclo[1.1.1]pentanylamino)—4- (methylsulf1nyl)pyrimidinecarboxamide (397 mg, 1.491 mmol, 91 % yield) as white solid. 1H NMR (400 MHz, DMSO-dg) 8 ppm 8.67 - 9.14 (m, 2 H), 7.93 - 8.11 (m, 1 H), 7.41 - 7.54 (m, 1 H), 2.68 - 2.79 (m, 3 H), 2.43 - 2.48 (m, 1 H), 2.02 - 2.21 (m, 6 H). MS (ESI) m/z 267.0 [M+1]+.
] E. 2-(Bicyclo[1.1.1]pentanylamin0)—4-((1R,3R)—3-hydr0xy-4,4- dimethylcyclohexylamin0)pyrimidine—5-carb0xamide. To a stirring suspension of 2-(bicyclo[1 . 1 .1]pentanylamino)(methylsulf1nyl)pyrimidinecarboxamide (397 mg, ATI—2514175v1 1.491 mmol) and and a mixture of (1R,5R)amino-2,2-dimethylcyclohexanol hydrochloride and (1S,5S)—5-amino-2,2-dimethylcyclohexanol hydrochloride (348 mg, 1.938 mmol, sis bed herein) in DMF (4.88 mL) was added DIEA (0.78 mL, 4.47 mmol) and the reaction was heated to 90 CC overnight. The crude reaction mixture was poured into 60 mL of ice water. The resulting solids were slurried for ~ 1 h, filtered, rinsed with water and dried for a few hours in a vacuum oven at 45 CC to afford a mixture of 2-(bicyclo[1 . 1 . anylamino)((1R,3R)—3-hydroxy-4,4-dimethyl- cyclohexylamino)pyrimidinecarboxamide and 2-(bicyclo[1.1.1]pentanylamino) ((1S,3S)—3-hydroxy-4,4-dimethylcyclohexylamino)pyrimidinecarboxamide (481 mg, 1.392 mmol, 93 % yield). This material was separated by preparative chiral SFC utilizing a ChiralPak AD-H, 250><30mm ID. column with an isocratic 40% ethanol + 0.1% um hydroxide in C02 gradient at 120 mL/min flow rate and at 38 CC. The faster eluting isomer was denoted as peak 1 and 132 mg (0.382 mmol) was obtained. The slower g isomer was denoted as peak 2 and 120 mg (0.349 mmol) was obtained.
Peak 1: 1H NMR (400 MHz, DMSO-d6) 8 ppm 8.85 - 9.12 (m, 1H), 8.14 - 8.44 (m, 1H), 7.63 - 7.90 (m, 1H), 4.47 - 4.63 (m, 1H), 3.76 - 3.96 (m, 1H), 3.08 - 3.23 (m, 1H), 2.43 (s, 1H), 2.04 (s, 6H), 1.87 - 1.99 (m, 1H), 1.66 - 1.83 (m, 1H), 1.04 - 1.47 (m, 4H), 0.92 (s, 3H), 0.82 (s, 3H) MS (ESI) m/z 346.3 [M+1]+.
Peak 2: 1H NMR (400 MHz, DMSO-dg) 8 ppm 8.88 - 9.07 (m, 1H), 8.25 - 8.45 (m, 1H), 7.57 - 7.90 (m, 1H), 4.46 - 4.59 (m, 1H), 3.78 - 3.96 (m, 1H), 3.09 - 3.22 (m, 1H), 2.35 - 2.47 (m, 1H), 2.04 (s, 6H), 1.87 - 1.99 (m, 1H), 1.66 - 1.80 (m, 1H), 1.37 - 1.48 (m, 1H), 1.12 - 1.34 (m, 3H), 0.88 - 0.94 (m, 3H), 0.71 - 0.86 (m, 3H). MS (ESI) m/z 346.3 [M+1]+. By SAR potency comparison with similar compounds ofknown absolute stereochemistry described herein, Peak 1 was assigned as 2—(bicyclo[1.1.1]pentan ylamino)—4-((1S,3S)hydroxy-4,4-dimethylcyclohexylamino)pyrimidine—5- carboxamide. Peak 2 was assigned as 2-(bicyclo[1.1.1]pentanylamin0)—4-((1R,3R)— 3-hydroxy-4,4-dimethylcyclohexylamin0)pyrimidinecarboxamide. —214— ATI—25 14175v1 ASSAYS BIOCHEMICAL ASSAYS A. Time resolved fluorescence assays JNKl Assay. A 384-well time resolved fluorescence assay was used to monitor JNKl activity. The JNKl assay was run in the following assay buffer: 50 mM HEPES, 10 mM MgClz, 1 mM EGTA, 2 mM DTT, and 0.01% Tween 20. To initiate the reaction 100 nM of ULightTM-labeled 4EBPl peptide (Perkin-Elmer) and 5 [LM ofATP were mixed with 500 pM of JNKl (Carna Biosciences), for a total assay volume of 20 [LL in each well. The assay was incubated at room temperature for l h and terminated using a mixture of 30 mM EDTA and 4 nM i-4EBPl, by adding 20 [LL of stop solution to each well. Plates were read on a -Elmer Envision Reader.
] JNK2 Assay. A 384-well time ed fluorescence assay was used to monitor JNK2 activity. The JNK2 assay was run in the following assay buffer: 50 mM HEPES, 10 mM MgClz, 1 mM EGTA, 2 mM DTT, and 0.01% Tween 20. To initiate the reaction 100 nM of ULightTM-labeled 4EBPl e (Perkin-Elmer) and 5 [LM ofATP were mixed with 500 pM 0f JNK2 (Carna Biosciences), for a total assay volume of 20 [LL in each well. The assay was incubated at room temperature for l h and terminated using a e of 30 mM EDTA and 4 nM Eu-anti-4EBPl, by adding 20 [LL of stop solution to each well. Plates were read on a Perkin-Elmer Envision Reader.
B. Z’-LYTE® Cascade Assays JNKl Assay. The JNKl Z’-LYTE® Cascade kinase assay was run in the following buffer: 50 mM HEPES at pH 7.5, 0.01% BRlJ-35, 10 mM MgClz, 1 mM EGTA, and 1 mM DTT. A 10 [LL kinase reaction mixture was prepared containing 1.81 - 7.25 ng JNKl, 25 ng inactive MAPKAPK2, 100 [LM ATP, and 2 [LM Ser/Thr 04 peptide. The assay was incubated at room temperature for l h. Next, 5 [LL of a 1:512 dilution of Development Reagent A (lnvitrogen, PV3295) was added to the reaction e and incubated at room temperature for an additional hour. The data was then read on a fluorescence plate reader and analyzed.
JNK2 assay. The JNK2 E® Cascade kinase assay was run in the following : 50 mM HEPES pH 7.5, 0.01% BRlJ-35, 10 mM MgClz, 1 mM EGTA, 2 mM DTT. A 10 [LL kinase reaction mixture was prepared containing 0.38 - 1.5 ng ATl—2514l75vl JNK2, 100 ng inactive MAPKAPK2, 100 uM ATP, and 2 uM Ser/Thr 04 peptide. The assay was incubated at room temperature for 1 h. Next, 5 uL of a 1:512 dilution of Development Reagent A (Invitrogen, PV3295) was added to the reaction mixture and incubated at room temperature for an additional hour. The data was then read on a fluorescence plate reader and analyzed.
C. Radioactive Assays JNK1 assay. The radioactive JNK kinase assay was carried out in a 96-well plate format at a final volume of 100 uL. The final assay concentration was 6.6 uM ATP (3-fold ATP Km), 2.64 to 5 ug/mL JNKl, and 100 ug/mL cJUN. JNKl was diluted in the following dilution buffer (20 mM HEPES pH 7.6, 0.1 mM EDTA, 2.5 mM MgC12, 0.004%(w/V) Triton X100, 2 ug/ml Leupeptin, 20 mM B-glycerol phosphate, 0.1 mM N33VO4 dithiothreitol) and then pre-mixed with cJun diluted in the substrate on buffer (20 mM HEPES pH 7.6, 50 mM NaCl, 0.1 mM EDTA, 2.5 mM MgClz, 0.05%(w/V) Triton X100). The JNKl/cJun mix (85 ul) was added to the inhibitor (5 ul) diluted in 100% DMSO to give a final DMSO assay concentration of 5%(V/V). The enzyme, substrate and inhibitor mixture was allowed to equilibrate at room temperature for minutes. The reaction was started by the addition of 10[LL of 10X ATP in kinase buffer (130 mM MgCl; 6 mM dithiothreitol, 150 mM itrophenyl phosphate, 100 uCi/ml y-[33P]-ATP). Reactions were allowed to proceed for 60 minutes before precipitation of protein Via trichloroacetic acid (7.2% TCA final). After a 30 minute incubation with TCA, reaction ts were collected onto glass microfilter 96-well plates pore MAHF CIH60) using a Packard Filtermate. The precipitate was washed with ate Buffered Saline and the amount of phosphate incorporated into cJun was quantified by scintillation counting using a d nt-NXT. All assays were conducted under conditions where phosphate incorporation was linear with respect to time and enzyme tration.
The IC50 values were calculated as the concentration of the inhibitor at which the c-Jun phosphorylation was reduced to 50% of the control value.
JNK2 assay. The assay was carried out in a l plate format at a final volume of 100 uL. The final assay concentrations were 6.6 uM ATP (3-fold ATP Km), 0.2 to 0.53 ug/mL JNK2, and 100 ug/mL cJUN. JNK2 was d in the following dilution buffer (20 mM HEPES pH 7.6, 0.1 mM EDTA, 2.5 mM MgClz, 0.004%(w/V) ATI—25 14175v1 2012/034349 Triton X100, 2 ug/ml Leupeptin, 20 mM B-glycerol phosphate, 0.1 mM N33VO4 dithiothreitol) and then pre-mixed with cJun diluted in the substrate on buffer (20 mM HEPES pH 7.6, 50 mM NaCl, 0.1 mM EDTA, 2.5 mM MgC12, 0.05% (w/v) Triton X100). The JNK2/cJun mix (85 ul) was added to the inhibitor (5 ul) diluted in 100% DMSO to give a final DMSO assay tration of 5%(V/V). The enzyme, ate and inhibitor mixture was allowed to equilibrate at room temperature for minutes. The reaction was started by the addition of 10uL of 10X ATP in kinase buffer (130 mM MgCl; 6 mM dithiothreitol, 150 mM itrophenyl phosphate, 100 uCi/ml y-[33P]-ATP). Reactions were allowed to proceed for 60 minutes before precipitation of protein Via trichloroacetic acid (7.2% TCA final). After a 30 minute incubation with TCA, reaction ts are collected onto glass microfilter 96-well plates (Millipore MAHF CIH60) using a Packard mate. The precipitate was washed with Phosphate Buffered Saline and the amount of phosphate incorporated into cJun was quantified by scintillation counting using a Packard nt-NXT. All assays were conducted under conditions where phosphate incorporation was linear with respect to time and enzyme concentration.
The IC50 values were calculated as the tration of the inhibitor at which the c-Jun phosphorylation was reduced to 50% of the control value.
CELL ASSAYS RAW264.7 Phosp_ho-cJun Whole Cell Assay. RAW264.7 cells were purchased from the American Tissue Culture Collection and maintained in growth media ting of 90% high glucose Dulbecco's Modified Eagle Medium rogen), 10% fetal bovine serum (Hyclone), and 2 mM L-glutamine (Invitrogen). All cells were cultured at 37 0C in 95% air and 5% C02. Cells were plated at a density of 1.0 x 105 cells per well in a 96-well plate in 120 uL of growth media. Diaminopyrimidine Compound stock (30 mM) was diluted serially in DMSO, fithher diluted in growth media, and was added to each well as a 10x concentrated solution in a volume of 15 uL, mixed, and allowed to incubate with cells. The compound vehicle (DMSO) was maintained at a final concentration of 0.2% in all wells. After 30 minutes, the cells were activated with lipopolysaccharide (ALEXIS Biochemicals) at a final concentration of 25 ng/mL.
Lipopolysaccharide was added as a 10x concentrated solution in growth media and added in a volume of 15 uL per well. Cell plates were cultured for 1 h, after which the cell media ATI—25 14175v1 was d. The level of c-Jun protein which was phosphorylated at serine 63 was measured according to the manufacturer’s instructions for the Whole Cell Lysate Kit- Phospho-c-Jun (Ser 63) Assay (Meso Scale Discovery) with the exception that the concentration ofNaCl in the lysis buffer was increased to a final concentration of 350 mM. The IC50 values were calculated as the concentration of Diaminopyrimidine Compound at which the level of phosphorylated c-Jun protein was reduced to 50% of the signal window. Certain compounds of Table l, 2 and 3 have an IC50 value ranging from 0.01 - 30 uM in this assay.
Jurkat T-cell IL-2 Production Assay. Jurkat T cells (clone E6-1) are purchased from the an Tissue Culture Collection and maintained in growth media consisting of RPMI 1640 medium containing 2 mM L-glutamine (Mediatech), with 10% fetal bovine serum (Hyclone) and penicillin/streptomycin. All cells are cultured at 37 °C in 95% air and 5% C02. Cells are plated at a density of 1 x 105 cells per well in 120 uL of media in a 96-well plate. opyrimidine Compound stock (20 mM) is diluted in growth media and added to each well as a 10 x concentrated solution in a volume of uL, mixed, and allowed to pre-incubate with cells for 30 min. The compound vehicle (dimethylsulfoxide) is maintained at a final concentration of 0.2% in all samples. After min the cells are activated with PMA (phorbol myristate acetate; final concentration 50 ng/mL) and PHA (phytohemagglutinin; final tration 1 ug/mL). PMA and PHA are added as a 10 x concentrated solution made up in growth media and added in a volume of 15 uL per well. Cell plates are ed for 6 h. Cells are pelleted by centrifilgation and the media removed and stored at -20 CC. Media aliquots are analyzed according the manufacturers instructions for the Human IL-2 Tissue Culture Kit (Meso Scale Discovery). The IC50 values are calculated as the concentration of the Diaminopyrimidine nd at which the IL-2 production was reduced to 50% of the signal window. n compounds from Table l, 2 and 3 have an IC50 value ranging from 0.01 - 10 uM in this assay.
ANIMAL MODELS Rat or mouse in viva LPS-induced TNF-u tion and chun Assay.
Male CD rats or 6 mice procured from Charles River Laboratories, at 7 weeks of age or 20 g weight respectively, were allowed to acclimate for one week prior to use. In ATI—2514l75vl the rat, the lateral tail vein was cannulated percutaneously with a 22-gage over-the-needle catheter under brief isoflurane anesthesia and 20 ug LPS (E. Coli 055:BS) and catheters were flushed with 2.5 mL/kg of normal injectable saline. In the mouse, 1 mg/kg LPS (E.
Coli ) in saline was administered intraperitoneally in a volume of 200 ul. The animals were administered the Diaminopyrimidine Compound by oral gavage 15 to 180 min prior to injection of LPS. Blood was collected via cardiac puncture 90 s or 2 h after LPS challenge for rats and mice respectively, and the liver and epididymal fat was ed for chun analysis using the ale Discovery Platform. The plasma was prepared using lithium heparin tion tubes and frozen at -80 CC until analyzed. The TNF-u levels were determined using a rat or mouse specif1c TNF-u ELISA kit (Mesoscale Discovery). The liver and epididymal fat were homogenized and sonicated in Mesoscale lysis buffer, the protein t was determined (BCA protein determination kit) and the chun levels were measured (Mesoscale ery). The ED50 values were calculated as the dose of the Diaminopyrimidine nd at which the TNF-u or chun levels were d to 50% of the control value. Certain compounds from Table l, 2 and 3 were shown, or will be shown, to have an ED50 value ranging from 30-100 mg/kg in this assay.
Choline def1cient amino acid supplement induced non-alcoholic steatohep_atitis g NASH) model in the rat. Male Wistar rats obtained from Charles River Laboratories at 7 weeks of age are allowed to acclimate for one week prior to use. Rats are fed Choline Def1cientL Amino Acid Supplemented (CDAA) diet (Dyets Inc) for up to 12 weeks. Rats are administered Diaminopyrimidine Compound by oral gavage once or twice daily starting at the time of introduction to the diet for between 2 and 12 weeks. At the termination of the study blood is collected via cardiac puncture and the liver and epididymal fat isolated. The liver tissue is processed for histology and the amount of steatosis or fibrosis is quantitated following H&E oxylin and eosin) or picro Sirius red staining. The liver fianction is ed by is of liver enzymes, for example, ALT and AST in plasma or serum. In addition, levels of phospho-cJun in the liver were quantitated by IHC, as described by Ma F. et. al. Lab Invest.; 89(4):470-84 (2009). The ED50 values are calculated as the dose of Diaminopyrimidine Compound at which the f1brosis, steatosis, liver enzyme, and/or p-cJun levels are reduced to 50% of the control -2l9- ATI—2514l75vl value. Certain compounds from Table l, 2 and 3 have, or will be shown to have, an ED50 value ranging from 10-100 mg/kg in this assay.
Bile duct ligation model of Fibrosis. Male BALB/c mice g) were obtained from Charles River Laboratories, and were d to acclimate for one week prior to surgery.
A ketamine (80-100 mg/kg)/xylazine(8mg/kg) anesthetic delivered IP was used for the bile duct ligation surgical procedure. An abdominal incision provided access to the bile duct which was ligated with two 6.0 silk sutures placed rostral to the bile duct connection with the intestines. 3-0 vicryl suture was used to close the abdominal wall, and skin. Following y, mice were administered 1 mL of lactated s solution sub cutaneous daily as well as wet food for the on of the study. Test compounds were administered by oral gavage starting the day after surgery. Studies were terminated 14 days post bile duct ligation. Blood was collected via cardiac puncture and serum was isolated for clinical chemistry analysis of liver function (ALT, AST, and bilirubin). Liver tissue was collected and processed histologically, stained with H&E (hematoxylin and eosin) or Picro Sirius red, and used to assess the amount of ation, rtal hyperplasia, necrosis and s induced by the bile duct ligation surgical procedure.
In addition, levels of phospho-cJun in the liver were quantitated by IHC, as described by Ma F. et. al. Lab Invest.; 89(4):470-84 (2009). Certain compounds from Table l, 2 and 3 show, or will show, statistically significant inhibition of inflammation, periportal hyperplasia, necrosis, fibrosis, and/or phosphor-cJun levels at dose levels of 10-300 mg/kg QD or BID.
Carbon Tetrachloride Model of Fibrosis. Male C57Bl/6 mice obtained from Harlan weighing 22-24g at arrival were allowed to acclimate for one week prior to use. Fibrosis was established via intraperitoneal injection of CCl4 (0.75 mL/kg) in mineral oil (15% v/v CCl4 in mineral oil) 3 times a week. The animals were administered the Diaminopyrimidine Compound by oral gavage l h prior to the initial CCl4 injection and then daily or twice daily through the end of the study, 28 days later. At the termination of the study blood was ted via cardiac puncture and the liver was isolated. The liver tissue was processed for histology and the amount of steatosis or fibrosis was quantitated following H&E (hematoxylin and eosin) or picro Sirius red staining. The liver function was ed by analysis of liver enzymes, for example, ALT and AST in plasma or serum. In addition, levels of o-cJun in the liver were tated by IHC, as described by Ma F. et. al. Lab Invest.; 470-84 (2009). Certain compounds from ATI—2514l75vl Table l, 2 and 3 show, or will show, tically significant inhibition of inflammation, periportal hyperplasia, necrosis, fibrosis, and/or phosphor-cJun levels at dose levels of -300 mg/kg QD or BID.
Skin Bleomycin model: Male or female DBA/2 mice are obtained from Harlan weighing 22-24g at arrival and are allowed to acclimate for one week prior to use.
Skin fibrosis is induced by intradermal injections of bleomycin (0.5 mg/mL in a total volume of 100 uL) every other day for 6 weeks in defined areas of the upper back. The animals are administered the Diaminopyrimidine Compound by oral gavage l h prior to the l bleomycin injection and then daily or twice daily through the end of the study, 3 or 6 weeks later. At the termination of the study blood is collected via cardiac puncture and the skin is isolated. The skin is processed for histology and the dermal thickness is quantitated following H&E (hematoxylin and eosin) staining. Certain compounds from Table l, 2 and 3 will show tically significant inhibition of skin thickness at dose levels of 10-300 mg/kg QD or BID.
Lung Bleomycin model: Male C57Bl/6 mice are obtained from Charles River tories and animals are allowed to acclimate for at least 5 days. Animals are kept on a l2-hours light/dark cycle and are approximately 8 weeks old at the beginning of the study. Mice are lightly anesthetized with isofiurane and the ae exposed. On Day 0, bleomycin (30 uL; 0.05U) is injected into the trachea. Following injection of bleomycin, the wound is closed using wound clips and the animal is allowed to recover.
Prednisolone ive control, dosed intraperitoneally), vehicle (dosed orally), or Diaminopyrimidine Compound (dosed orally) is administered 30 min to 2 hours prior to bleomycin administration (prophylactic model) or 4 days after bleomycin (therapeutic . Dosing with vehicle or Diaminopyrimidine Compound is carried out twice daily hout all studies, dosing with prednisolone is carried out once daily. Mice are red daily for adverse effects from test article administration, including effect on body . Mice are euthanized on Day 13. Bronchoalveolar lavage is med using 1 mL phosphate buffered saline (PBS) and total WBC counts are assessed. Results are expressed as total cells retrieved. Macrophages, eosinophils, neutrophils, and cytes are quantified in BALF samples. Lung lobes are processed for histology using H&E (hematoxylin and eosin), Trichrome or TUNEL ng. Trichrome sections are scored -22l- ATI—2514l75v1 for fibrosis ty as described previously (Ashcroft et al. J. Clin. Pat/ml. 41 :467-470 (1988)). Certain compounds from Table 1, 2 and 3 will show tically significant inhibition of lung fibrosis at dose levels of 10-300 mg/kg QD or BID.
Lupus Model: Female NZB/NZW Fl mice are obtained from Jackson tories between 4-12 weeks of age at l and are allowed to acclimate for one week prior to use and to develop spontaneous SLE-like disease. The animals are administered the Diaminopyrimidine Compound by oral gavage at the initiation of the study and then daily or twice daily through the end of the study up to 6 months later.
Blood is collected throughout the study and serum isolated to measure occurrence of dsDNA via a standard ELISA method and proteinurea is measured in the urine. Certain compounds from Table 1, 2 and 3 will show statistically significant inhibition of nurea or dsDNA at dose levels of 10-300 mg/kg QD or BID.
UUO Model of Kidney Fibrosis: Male CD-IGS rats are obtained from Charles River Laboratories at a body weight of approximately 150 — 160 g and are allowed to acclimate for one week prior to tion of the study. Rats are anesthetized using ketamine/xylazine and the surgical area is sterilized with 70% ethanol and ne. The ureter is exposed after a midline incision is made and ligated twice using 5.0 silk suture placed 1 cm apart and cut between the ligations. The on is closed using 3.0 silk suture and the animal is allowed to recover. The animals are administered the Diaminopyrimidine Compound by oral gavage 1 h prior to the ureter ligation and then daily or twice daily through the end of the study 7 days later. At the termination of the study blood is collected via cardiac puncture and the kidney isolated. The kidney is processed for histology and the morphology and amount of fibrosis is quantitated following H&E oxylin and eosin), picro sirius red or alpha-smooth muscle actin staining. Certain compounds from Table 1, 2 and 3 will show statistically significant inhibition of fibrosis or alpha-smooth muscle actin staining at dose levels of 10-300 mg/kg QD or BID.
ACTIVITY TABLES ] Each of the compounds in Tables 1, 2 and 3 was tested in one or more of the INK1 biochemical assays and was found to have activity therein, with all of the ATI—25 14175v1 WO 45569 compounds having an IC50 below 10 MM in the assay, with some compounds having an IC50 below 200 nM (activity level D), some an IC50 between 200 nM and 800 nM ity level C), some an IC50 between 800 nM and 1 MM (activity level B), and others having an IC50 between 1 MM and 10 MM (activity level A).
Table 1. 4-(isopropylan1ino)—2-((lr,4r)- ,2-trifluoroethoxy)— cyclohexylaniino)pyrin1idine-5 - carboxaniide 2-(tert-butylan1ino)((lR,3S)- 3-hydroxycyclohexylaniino)- pyrimidine-S-carboxaniide 2—(4,4— difluorocyclohexylaniino) (isopropylaniino)pyrin1idine-5 - carboxaniide 2-((lr,4R) D (difluoroniethoxy)cyclohexyla mino)((lR,2R) hydroxycyclopentylaniino)- pyrimidine-S-carboxaniide PM: 2-(4,4- 356.3 D “jug [\ ] difluorocyclohexylaniino) :l ]; ((1R,2R) M NH :3; hydroxycyclopentylaniino)- NE at pyrimidine-S-carboxaniide ATI—2514l75vl Cmpd Structure NAME MH+ Act.
A- Level 6 x49! 2-((1r,4r)(2-hydr0xypropan- 336.2 D wk] yclohexylamino) ‘T" [\i (isopropylamino)pyrirnidine carboxamide r f11:?” "if «Q. 4-((1R,2R) hydroxycyclopentylarnino) (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4R) methoxycyclohexylamino) ((R)-tetrahydr0-2H-pyran ylarnino)pyrirnidine-5 - carboxamide 4-(isopr0pylarnino)—2-((1r,4r)- 4-meth0xycyc10hexy1arnino)- dine-S-carboxamide 2-((1r,4r)—4- (dimethylcarbarnoyl)cyclohexyl amino)—4- (isopropylamino)pyrirnidine-5 - amide 2-(1,4-dioxaspiro[4.5]decan ylarnino)—4- (isopropylamino)pyrirnidine-5 - carboxamide —224— ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 12 :9” 2-((1r,4r)(hydr0xymethyl)- 308.3 D -"‘:‘\I cyclohexylamino) “T” L?" (isopropylamino)pyrirnidine arnide fl ‘1}: *1 Peak 4 Peak 3 Peak 2 Peak 1 One of: 4-(5-hydr0xytetrahydro-2H- pyrany1arnin0)—2-((1r,4r) methoxycyclohexylarnino)- pyrimidine-S-carboxarnide; 4-(5-hydr0xytetrahydro-2H- pyrany1arnin0)—2-((1r,4r) methoxycyclohexylarnino)- pyrimidine-S-carboxarnide; 4-(5-hydr0xytetrahydro-2H- pyrany1arnin0)—2-((1r,4r) methoxycyclohexylarnino)- pyrimidine-S-carboxamide; or 4-(5-hydr0xytetrahydro-2H- pyrany1arnin0)—2-((1r,4r) methoxycyclohexylarnino)- dine-S-carboxarnide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 17 4-((1R,3S,4S)—3-hydroxy 378 D [‘T‘m [JR] methylcyclohexylarnino) 3 i ((1r,4R)—4- methoxycyclohexylamino)- H “Hafijiky El pyrimidine-S-carboxamide 4-((1 S ,3R,4R)-3 xy methylcyclohexylarnino) (( 1r,4S) methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3R,4R)hydroxy methylcyclohexylarnino) (( 1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 4'((1S,3S,4S)hydroxy—4- cyclohexylamino)_2- ((1r,4S)-4_ methoxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1r,4R) (difluorornethoxy)cyclohexyla mino)((1R,3 S)-3 - hydroxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1r,4R)hydr0xy methylcyc10hexy1arnino) (( 1 R, 3 S)-3 - ycyclohexylamino)- pyrimidine-S-carboxamide ATI—ZS 14175V1 Structure NAME ,3R)hydroxy-3 - methylcyclohexylarnino) (( 1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3 ydroxy-3 - methylcyclohexylarnino) (( 1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1r,4S) methoxycyclohexylamino) ((S)0X0piperidin no)pyrirnidine-5 - carboxamide 2-((1r,4R) methoxycyclohexylamino) ((R)0X0piperidin ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4R) (dimethylcarbamoyl)cyclohexyl amino)((1R,3 S)—3 - hydroxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((1r,4R)—4-(2,2,2- trifluoroethoxy)cyclohexy1arnin o)pyrirnidinecarb0xarnide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ A°t° A- Level 29 E.T 4-((1R,3S) 432 D hydroxycyclohexylamino) 1:1” 2”;r ((ls,4S)(2,2,2- Efibffl'ffixgi trifluoroethoxy)cyclohexyl- "= ”gr ' w?“ amino)pyrimidine carboxamide 4—((1R,3S) w...“ i. 7 hydroxycycloheptylamlno)‘2_ (( 1 1‘,4R) ycyclohexylamino)- pyrimidine-S-carboxamide 31 4-((1R,3R) hydroxycycloheptylarnino) (( 1r,4R)—4- ycyclohexylamino)- pyrimidine-S-carboxamide 32 4-((IS,3R) ‘ ‘ hydroxycycloheptylarnino) ((1r,4S) ycyclohexylamino)- pyrimidine-S-carboxamide 33 4-((IS,3S) 378.3 D hydroxycycloheptylarnino) ((1r,4S) methoxycyclohexylamino)- pyrimidine-S-carboxamide ATI—ZS 14175V1 Structure NAME 4-((1r,4r)—4- hydroxycyclohexylamino) (( 1r,4r) methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-(4-hydr0xyrnethylbutan ylamino)((1r,4r) methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,ZS) (hydroxyrnethyl)cyclopentylarni n0)((1r,4R) methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1 S,2R) (hydroxyrnethyl)cyclopentylarni n0)((1r,4S) methoxycyclohexylamino)- dine-S-carboxamide 4-((1r,4r)—4- acetamidocyclohexylamino) (( methoxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1r,4R) acetamidocyclohexylamino) (( 1 R, 3 S)-3 - hydroxycyclohexylamino)- dine-S-carboxamide ATI—ZS 14175V1 Structure NAME 4-((1 S,3S) hydroxycyclohexylarnino) (( 1r,4S) methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3R)-3 - hydroxycyclohexylarnino) (( 1r,4R)—4- methoxycyclohexylamino)- dine-S-carboxamide 4-(3-hydroxy butylarnino)((1r,4r) methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,2R) hydroxycyclohexylamino) ((1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3 S)-3 - ycyclohexylamino) (( 1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 2-(4,4- difluorocyclohexylamino) (( 1 R, 3 S)-3 - hydroxycyclohexylamino)- pyrimidine-S-carboxamide ATI—ZS 14175V1 Structure NAME 4-((1R,3 S)-3 - hydroxycyclopentylarnino) (( 1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1r,4R) methoxycyclohexylamino) ((R)-piperidin ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4S) methoxycyclohexylamino) ((S)-piperidin no)pyrirnidine-5 - carboxamide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) (( 1r,4R)—4- (methylarnino)cyclohexylamino )pyrimidine-S-carb0xarnide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((R)-tetrahydr0-2H-pyran-3 - no)pyrirnidine-5 - carboxamide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) (isopropylamino)pyrirnidine-5 - amide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A' Level 52 3.93“. M mm 4-((1R,3S) 336.2 D LN] {ij hydroxycyclohexylamino) H EL». E. H ((S)-tetrahydro-2H-pyran-3 - 3f. mm W. ylarnino)pyrirnidine My “V1: fl amide 53 2-(cyclohexylarnino) 334.2 D .3. "h ‘ ((1R,3S) ycyclohexylamino)- pyrimidine-S-carboxamide 54 4-((1R,3S) 350.3 D ‘ hydroxycyclohexylamino) (( 1r,4R)—4- hydroxycyclohexylamino)- pyrimidine-S-carboxamide 55 3 2-((1r,4r)—4- 350.1 C L“: [H] methoxycyclohexylamino) “1'” ET“ (tetrahydro-ZH-pyran menujw ylarnino)pyrirnidine E NW. carboxamide (“I 3;. 4-((1R,3S) 391.2 D 3.. m 1.3 hydroxycyclohexylamino) L331“ 3LT} ((1r,4R)—4- (methylcarbarnoyl)cyclo- H m‘hnfiw3 .3.» “g” hexylamino)pyrimidine carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 57 4-((1R,3S) 336.2 D hydroxycyclohexylamino) (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4R) ethoxycyclohexylamino) (( 1 R, 3 S)-3 - hydroxycyclohexylamino)- pyrimidine-S-carboxamide ,4S) ethoxycyclohexylamino) (( 1 S ,3R)-3 - hydroxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1r,4S) ethoxycyclohexylamino) etrahydro-2H-pyran-3 - ylarnino)pyrirnidine-5 - carboxamide ,3R) hydroxycyclohexylarnino) (( 1r,4S) methoxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1 s,4R) ethoxycyclohexylamino) (( 1 S ,2 S) hydroxycyclopentylamino)- pyrimidine-S-carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 63 2-((1r,4S) 364.3 C ethoxycyclohexylamino) \a—5LI‘1,1. UP«.7.‘ ~ H§V€ ' N ((18,2R) 1: ‘r' hydroxycyclopentylamino)- pyrimidine-S-carboxamide 2-((1 S,3S) ethoxycyclopentylarnino) (( 1 S ,2 S) hydroxycyclopentylarnino)- pyrimidine-S-carboxamide ,2R) ycyclopentylarnino) (( 1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 2-((1r,4R) ethoxycyclohexylamino) (( 1 R,2R) hydroxycyclopentylarnino)- pyrimidine-S-carboxamide ,4R) ethoxycyclohexylamino) ((R)hydr0xypropyl- amino)pyrimidine-5 - carboxamide 4-((ls,3s) hydroxycyclobutylarnino)(1 - (3 -rnethy1butanoy1)piperidin ylarnino)pyrirnidine-5 - carboxamide —234— ATI—ZS 14175V1 2012/034349 Cmpd. Structure NAME MH+ Act.
A" Level 69 4-((ls,4s)hydroxy 378, 3 D K r‘juj methylcyclohexylarnino) «U ,9 N‘ \L «3:: ((1r,4r)rneth0xycyclohexyl- :92, \] pyrimidine carboxamide 2-((1 s,4R)hydroxy phenylcyclohexylarnino) ((1 S ,2S)hydroxycyclopentylamino )pyrimidine-5 - carboxamide 2-((1r,4r)—4- ethoxycyclohexylarnino)(2- hydroxyethylarnino)pyrimidine- oxarnide 2-((1r,4R) ethoxycyclohexylamino) ((R)-tetrahydr0-2H-pyran ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4S) ethoxycyclohexylamino) ((1 S ,2S)ethoxycyclo- pentylamino)pyrirnidine-5 - carboxamide 2-((1r,3S) ethoxycyclobutylarnino) ((1 S ,2S)hydroxycyclopentylamino )pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 75 2-((1r,4S)(2- 382.3 D A: ,LATgxi'n‘al fluoroethoxy)cyclohexy1amino) ‘ C “ ‘1; ((1S,2S)hydroxycyclo- ., if” it; pentylamino)pyrimidine carboxamide 2-((1R,3R)-3 - ethoxycyclopentylarnino) ((1 S ,2S)hydroxycyclopentylamino )pyrirnidine-5 - carboxamide 2-((1r,4S) ethoxycyclohexylamino) ((1 S -hydroxycyclohexylamino )pyrimidine-5 - carboxamide 4-( 1 - (hydroxyrnethyl)cyclopentylarni ((1r,4r)methoxycyclohexylamino )pyrimidine-5 - carboxamide 2-((1r,4r)—4- hydroxycyclohexylamino)(4- methyltetrahydro-2H-pyran ylarnino)pyrirnidine-5 - carboxamide 4-((1S,3R) hydroxycyclopentylarnino) ((1r,4S)rnethoxycyclohexylamino idine-5 - carboxamide ATI—ZS 14175V1 2012/034349 Cmpd. Structure NAME MH+ Act.
A- Level 81 i am 2—((1s,3S)—3— 350.3 D "I :11 :1 hydroxycyclopentylarnino) ((ls,4R)methoxycyclo- \ E: I: \1 :3 hexylamino)pyrimidine : My carboxamide 4-(1 -acety1piperidin ylamino)((1r,4r) (methoxymethyl)cyclohexylamino )pyrimidine-5 - carboxamide 2-(1 -acety1piperidin ylamino)((1 s,4s) (methoxymethyl)cyclohexy1- amino)pyrimidine-5 - carboxamide 2-((R)acety1pyrrolidin—3 - o)((1 s,4S) (methoxymethyl)cyclohexylamino )pyrimidine-5 - carboxamide 4-((S)acety1pyrrolidin-3 - ylamino)—2-((1r,4S) (methoxymethyl)cyclohexyl- amino)pyrimidine carboxamide 2-((1R,3R)-3 - hydroxycyclopentylarnino) (1 -(3 -rnethylbutanoyl)piperidin- 4-ylamino)pyrimidine-5 - amide ATI—ZS 14175V1 WO 45569 ure NAME 4-((1S,3R) hydroxycyclopentylarnino) (1 -(3 -rnethylbutanoyl)piperidin- 4-ylamino)pyrimidine-5 - carboxamide 2-((1r,4r)—4- hydroxycyclohexylamino) (isobutylarnino)pyrimidine-5 - carboxamide 4-((1 s,4s) hydroxycyc10hexy1arnino)—2-(1 - propionylpiperidin ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4r)—4- hydroxycyc10hexy1arnino)—4-(1 - propionylpiperidin ylarnino)pyrirnidine-5 - carboxamide 4-((1 S ,2S) hydroxycyclopentylarnino) ((1r,4S)rnethoxycyclo- hexylamino)pyrimidine-5 - carboxamide 4-((1 S - hydroxycyclopentylarnino) ((1r,4S)(rnethoxyrnethyl)— cyclohexylamino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Cmpd Structure NAME MH+ Act.
A- Level 93 ‘j ,3S) 364.3 D Ewing, m hydroxycyclohexylamino) “(fix ((1r,4R)—4-methoxycyclohexyl- t: 3R. , amino)pyrimidine : carboxarmde. 4-((1 S,3R) hydroxycyclopentylarnino) ((1r,4S)(rnethoxyrnethyl)— exylamino)pyrirnidine-5 - carboxamide 4-((1 s,4S)—4- ethoxycyclohexylamino) (( 1 R, 3R)-3 -hydroxycyclopentylamino )pyrirnidine-5 - carboxamide 2-((1r,4R) ethoxycyclohexylamino) ((1R,3 S)—3-hydroxycyclopentylamino )pyrimidine amide 2-((1R,3R)-3 - (hydroxyrnethyl)cyclopentylarni no)—4-((1s,4S)methoxy- cyclohexylamino)pyrirnidine-5 - carboxamide 4-((1 S,3S) (hydroxyrnethyl)cyclopentylarni no)—2-((1r,4S)rnethoxycyclohexylamino )pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 99 j W ,3S) 364.3 D m: :11 4;: hydroxycyclopentylarnino) :i N E: ((1s,4R)—4-(rnethoxyrnethy1)- km] a}; cyclohexylamino)pyrimidine 3‘9 carboxamide 2-((1r,4r)—4- hydroxycyclohexylarnino) ((1 s,4s)meth0xycyclohexylamino )pyrimidine-5 - carboxamide 4-((1 s,4s) hydroxycyclohexylarnino) ((1r,4r)methoxycyclohexylamino )pyrimidine-5 - carboxamide ,4S) ethoxycyclohexylamino) ((S)hydr0xypr0py1— amino)pyrimidine-5 - carboxamide 2-((1r,4S) ethoxycyclohexylamino) ((1 S ,2S)rnethoxycyclo- pentylamino)pyrirnidine-5 - carboxamide 2-((1r,4R) ethoxycyclohexylamino) (( 1 R,ZS)hydroxycyclopentylamino )pyrimidine carboxamide —240— ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 105 2-((1r,4S) 350.3 D ethoxycyclohexylamino) ( fng\[j :11; A x ”:33 ((S)-tetrahydr0fi1ran ylarnino)pyrirnidine-5 - carboxamide 4-((1 s,4s) ethoxycyclohexylamino)(1 - hydroxy-Z-methylpropan-Z- ylarnino)pyrirnidine-5 - carboxamide 4-(cyclobuty1arnino)(1-(3 - methylbutanoyl)piperidin ylarnino)pyrirnidine-5 - carboxamide 4-((ls,4s) (aminornethyl)cyclohexylamino etrahydro-2H-pyran—4- ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4r)—4- hydroxycyclohexylarnino) ((1 s,4s)(methoxymethyl)- cyclohexylamino)pyrirnidine-5 - carboxamide 2-( 1 -(3 - methylbutanoyl)piperidin no)(tetrahydro-2H- 4-y1arnino)pyrirnidine-5 - carboxamide —241— ATI—ZS 14175V1 Structure NAME 4-((1S,4S)(2-hydroxypropan- 2-y1)cyclohexylamino) (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - carboxamide 4-((1s,4s)(pyrrolidine carbonyl)cyclohexylamino) (tetrahydro-ZH-pyran no)pyrirnidine-5 - carboxamide 4-((1 s,4s) cyclohexylamino)(1 - propionylpiperidin ylarnino)pyrirnidine-5 - carboxamide 4-(Cyc10penty1arnino)-2—(1- propionylpiperidin_4_ ylarnino)pyrirnidine-5 - amide 4-((ls,4s) ethoxycyclohexylamino) (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - carboxamide 4-((ls,4s) (dimethylcarbarnoyl)cyclohexyl amino)(tetrahydr0-2H-pyran- 4-ylamino)pyrimidine-5 - carboxamide —242— ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 117 2-((11@4S) 364-3 D QM“ ethoxycyclohexylamino) fT‘I‘I ((IS,ZS) . . a \n, v \v ‘3’ hydroxycyc10penty1arn1no)pyr1 midine-S-carboxarnide 4-((1 4- (dimethylcarbamoyl)cyclohexyl amino)((1r,4r)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((ls,4s) xyrnethyl)cyclohexylami no)((1r,4r)methoxycyclohexylamino )pyrimidine-5 - carboxamide 4-((1 s,4s) aminocyclohexylamino) ((1r,4r)rneth0xycyclohexylamino )pyrimidine-5 - carboxamide 4-(1,4-dioxaspiro[4.5]decan ylamino)((1r,4r)_4_ methoxycyclohexylamino)- dine-S-carboxamide 2-((S) cyclohexylethylamino) ((1 s,4R)—4-(hydroxyrnethyl)— cyclohexylamino)pyrirnidine-5 - carboxamide —243— ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 123 A? methyl 4-(5-carbarnoyl 377.1 D [3’] (cyclohexylamino)pyrirnidin f“ x :5; ylarnino)piperidine “vl‘xf; carboxylate 2-((S) cyclohexylethylamino) (( 1 s ,4R)—4- propionamidocyclohexylarnino) pyrimidine-S-carboxamide 2-((S) cyclohexylethylamino) ((1 4-hydr0xycyclohexylamino )pyrimidine-5 - amide 4-((1 s,4R) aminocyclohexylamino)((S)cyclohexylethylamino)- pyrimidine-S-carboxamide (1-aC€ty1piperidin-4— ylamino)(1- cyclohexylethylarnino)- pyrimidine-S-carboxamide 2-(Cyclopentylamino)-4_ ((1 S,4s) hydroxycyclohexylamino)- pyrimidine-S-carboxamide —244— ATI—ZS 14175V1 2012/034349 Cmpd. Structure NAME MH+ Act.
A- Level 129 2-(cyclopentylamino) 375 .3 C 1 s> \ihfilr 31'9“ propionarnidocyclohexylarnino) mf\ pyrimidine-S-carboxamide 2-(cyclopentylarnino) (( 1 s ,4s) (methylcarbam0y1)cyclohexy1- amino)pyrimidine-5 - carboxamide 4-((1s,4s)(arnin0rnethyl)— cyclohexylamino) (cyclopentylarnino)pyrirnidine- -carboxarnide 2—(cyc10buty1arnino)((1 5,45)- 4-(cyclopropylcarbarnoyl)- cyclohexylamino)pyrirnidine-5 - carboxamide 2—(cyc10buty1arnino)((1 5,45)- 4-(piperidine carbonyl)cyclohexylamino)- pyrimidine-S-carboxamide 2—(cyc10buty1arnino)((1 5,45)- (dimethylcarbarnoyl)cyclohexyl amino)pyrimidine carboxamide —245— ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 135 ‘1 2-(cyclobutylamino)((1s,4s)- 334.3 C D[A] 4-ethoxycyclohexylamino)- “Rf; pyrimidine-S-carboxamide 2—(cyclopropy1arnino) (( 1 s ,4s) (hydroxyrnethyl)cyclohexylamino )pyrimidine-5 - carboxamide 2—(cyclopropy1arnino) (( 1 s ,4s) (ethylcarbarnoyl)cyclohexylamino )pyrimidine-5 - carboxamide 2—(cyclopropy1arnino) ((ls,4s)(3- methy1butanamido)cyclohexylamino )pyrimidine-5 - carboxamide ,4s) (hydroxyrnethyl)cyclohexylamino )—2- (isopropylamino)pyrirnidine-5 - amide 4-((1s,4s)(2-hydroxypropan- 2-y1)cyclohexylamino) opylarnino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Cmpd Structure NAME MH+ Act.
A- Level 141 4-((ls,4s) 349.3 A :1:U (ethylcarbarnoyl)cyclohexylarni 2?: :r Vt“? no)—2-(isopr0pylarnino)- w- :1 pyrimidine-S-carboxamide 4-((ls,4s) isobutyramidocyclohexyl- amino)—2- (isopropylamino)pyrirnidine-5 - carboxamide 2-(isopr0pylarnino)((15,45)- 4-(pyrrolidine carbonyl)cyclohexylamino)- pyrimidine-S-carboxamide 4-(cyclohexy1arnino)((1r,4r)- 0xycyc10hexy1arnino)— pyrimidine-S-carboxamide ,4r)—4- hydroxycyclohexylamino) (isopropylamino)pyrirnidine-5 - carboxamide 4-((ls,4s) carbamoylcyclohexylamino) (cyclohexylamino)pyrirnidine- -carboxarnide —247— ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 147 if 2-(cyc10hexylarnino)((1s,4s)- 334.2 D [ j [ \j 4-hydr0xycyc10hexy1arnino)— \f \T pyrimidine-S-carboxamide 4-(Cyclopentylamino)-2_ (( 1r,4r)-4— hydroxycyclohexylamino)- pyrimidine-S-carboxamide 2'(Cy010hexylarnino)—4_ Pentylamino)pyrimidine- -carboxarnide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((1r,4R)—4-((2H3)rnethyloxy)- cyclohexylamino)pyrirnidine-5 - carboxamide 4-((1R,3 S) hydroxycyclohexylamino)—2-(4- methyltetrahydro-2H-pyran ylarnino)pyrirnidine-5 - carboxamide 4-((R)-3 ,3 - difluorocyclohexylamino) R)—4-methoxycyclohexylamino )pyrimidine-5 - carboxamide ATI—ZS 14175V1 Cmpd. ure NAME MH+ Act.
A- Level 153 4-((1R,3 S)-3 - 403.3 D {it}. xx.
, “LN‘LM hydroxycyclohexylarnino) NAM; S“ ((1r,4R)(pyrr01idin fl’VwfiL --'[‘\_lr lohexylarnino)pyrirnidine carboxarnide 154 4-((S)—3 ,3 - 384.2 D difluorocyclohexylamino) ((1r,4S)rnethoxycyclohexylamino )pyrimidine-5 - carboxamide 155 4-((1R,2S) 364.2 D ycyclohexylarnino) ((1r,4R)—4-methoxycyclohexylamino )pyrimidine-5 - carboxamide 156 4-((1R,2R) 418.2 D hydroxycyclopentylarnino) ((1r,4R)—4-(2,2,2- trifluoroethoxy)cyclohexylamino )pyrimidine carboxamide 157 i 2-((1r,4R) 392.3 D ethoxycyclohexylamino) ((1R,3 S)—3-hydroxy-3 - methylcyclohexylamino)- pyrimidine-S-carboxamide —249— ATI—25 14175V1 Cmpd Structure NAME MH+ Act.
A- Level 158 2-((1r,4R) 392.5 D {’N‘ng ethoxycyclohexylamino) 2?“ * S) w?"“1:3“fi‘w’fi hydroxycycloheptylamino)- pyrimidine-S-carboxamide yc10[2.2.2]octan ylamino)((1R,3 S)—3- hydroxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3 S)-3 - hydroxycycloheptylarnino) ((1r,4R)—4-(2,2,2-trifluoroethoxy )cyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3 S)—3-ethy1—3- ycyclohexylamino) ((1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((1 S ,4R)—4-methoxy- cycloheptylamino)pyrirnidine- -carboxarnide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((1R,4S)—4-methoxycycloheptylamino )pyrirnidine- -carboxarnide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A' Level 164 l 319.9 D 7 .3“ 4-(cyclobutylamino)((1r,4r)- T 4-rneth0xycyc10hexy1arnino)- w, :JT pyrimidinecarboxarnide 2-((1r,4R) (difluorornethoxy)cyclohexyl- amino)((1R,3 S)hydr0xycycloheptylamino )pyrirnidine- -carboxarnide 4-((1R,3 S)-3 - hydroxycycloheptylarnino) (tetrahydro-2H-pyran ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4R) ethoxycyclohexylamino) ((R)-tetrahydr0furan-3 - ylarnino)pyrirnidine-5 - carboxamide 4-((1R,3 S)—3-hydroxy-3 - cyclohexylarnino) ((1r,4R)—4-(2,2,2-trifluoro- ethoxy)cyclohexylamino)- dine-S-carboxamide 4-((1R,3 S)—3-hydroxy-3 - methylcyclohexylarnino) (tetrahydro-2H-pyran ylarnino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 170 f; 2—((1r,4R) 413.7 D (difluorornethoxy)cyclohexyla mino)((1R,3 ydr0xy-3 - methylcyclohexylamino)- pyrimidine-S-carboxamide 171 4-((1R,3 S)-3 - 364 D hydroxycyc10hepty1arnino) ((1r,4R)—4-hydroxycyclohexylamino )pyrimidine-5 - carboxamide 172 2-((1r,4R)—4- 391.8 D cyclohexylamino) (( 1 Ra3R,4R)-3 -hydr0xy—4_ methylcyclohexylamino)- pyrimidine-S-carboxamide 173 2-((1r,4R) 413.7 D (difluorornethoxy)cyclohexyla mino)((1R,3R,4R)—3- hydroxyrnethy1- exy1amino)pyrirnidine-5 - carboxamide 174 4- -.»-’ 4-(cyclobuty1amino)((1r,4r)- 387.5 D ,a‘fiik‘w“ if? 1L1x 4-(2,2,2-trifluoroethoxy)— cyc10hexy1amino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Structure NAME 4-(cyclopropy1arnino) ((1r,4r)hydr0xycyclohexy1— pyrimidine-5 - carboxamide 4-(cyclobutylamino)((1r,4r)- 0xycyc10hexy1arnino)— pyrimidine-S-carboxamide 4-((1R,3 S)—3-hydroxy-3 - methylcyclohexylarnino) ((1r,4R)—4-hydroxycyclohexylamino )pyrimidine-5 - amide 4-((1R,3R,4R)hydroxy methylcyclohexylarnino) ((1r,4R)—4-(2,2,2-trifluoroethoxy )cyclohexylamino)- pyrimidine-S-carboxamide 4-((1R,3R,4R)hydroxy methylcyclohexylarnino) ((1r,4R)—4-hydroxycyclohexyl- amino)pyrimidine carboxamide 4-((1R,3R)-3 - hydroxycyclohexylarnino) ((1r,4R)—4-(2,2,2-trifluoroethoxy )cyclohexylamino)- pyrimidine-S-carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 181 3' ,3S) 364 D {wanna :13: hydroxycycloheptylarnino) w. \,**;’m (4-rnethy1tetrahydro-2H-pyran- memn" 4-ylamino)pyrirnidine REM: carboxarnide 2-(tert-butylamino)((1R,3S)hydroxycyc10hepty1arnino)— pyrimidine-S-carboxamide ,3 S)—3-hydroxy-3 - methylcycloheptylarnino) ((1r,4R)—4-methoxycyclohexylamino )pyrimidine-5 - carboxamide 4-((1 S ,3 S)-3 -hydr0xy-3 - methylcycloheptylarnino) ((1r,4S)methoxycyclohexylamino )pyrimidine-5 - amide 4-((1 S ,3R)—3 -hydroxy-3 - methylcycloheptylarnino) ((1r,4S)methoxycyclohexyl- amino)pyrimidine-5 - carboxamide 4-((1R,3R)hydroxy methylcycloheptylarnino) ((1r,4R)—4-methoxycyclohexylamino )pyrimidine-5 - carboxamide —254— ATI—ZS 14175V1 PCT/U82012/034349 Cmpd Structure NAME MH+ Act.
A- Level 187 384.6 D w =55? 2-(4,4- difluorocyclohexylamino) (( 1 R, 3 S)—3 -hydr0xy-3 -rnethy1— exylamino)pyrirnidine-5 - carboxamide 4-((R)-tetrahydr0-2H-pyran-3 - ylarnino)((1r,4R)(2,2,2- roethoxy)cyclohexylamino )pyrimidine-5 - carboxamide -butylamino)((1r,4R)- 4-hydr0xycyc10hexy1arnino)— pyrimidine-S-carboxamide 4-(Cyclopentylamino)-2_ ((1TAT)rnethoxycyclohexy1— amino)pyrirnidine_5 _ carboxamide 2-((1r,4r)—4- (difluorornethoxy)cyclohexylamino )—4- (isopropylamino)pyrirnidine-5 - carboxamide 2-(4,4- difluorocyclohexylamino) ((1R,3 S)—3-hydroxycycloheptylamino )pyrimidine-5 - carboxamide ATI—ZS 14175V1 WO 45569 Cmpd Structure NAME MH+ Act.
A- Level 193 2-((1r,4R) 336.5 D l I: [fix] hydroxycyclohexylamino) T“ \T ((R)-tetrahydr0-2H-pyran m \ T3»! f ylarnino)pyrirnidine-5 - carboxamide 2-((1r,4R) (dimethylcarbarnoyl)cyclohexyl amino)((R)-tetrahydro-2H- pyranylarnin0)pyrirnidine-5 - carboxamide 4-(sec-butylamino)((1r,4S)— 0xycyc10hexy1arnino)— pyrimidine-S-carboxamide 4-(tert-buty1arnino)((1r,4r)- 4-hydr0xycyc10hexy1arnino)— pyrimidine-S-carboxamide 4-(cyclobutylamino)((1r,4r)- 4-ethoxycyclohexylamino)- pyrimidine-S-carboxamide 2-(4,4- difluorocyclohexylamino) (( 1 Ra3R,4R)-3 -hydr0xy—4_ methylcyclohexylamino)- pyrimidine-S-carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 199 m _.o pr0pylarnino)—2—((1r,4r)- 335 .2 D ”Twnxfwat-T 4-(rnethylcarbarnoy1)- cyclohexylamino)pyrirnidine-5 - carboxamide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((1 S ,4S)rnethoxycycloheptylamino )pyrimidine-5 - carboxamide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((1R,4R)rnethoxycycloheptylamino idine-5 - carboxamide 2-(4- hydroxybicyclo[2.2.2]octan no)—4- (isopropylamino)pyrirnidine-5 - carboxamide 2-((1r,4r)—4- acetamidocyclohexylamino) (isopropylamino)pyrirnidine-5 - carboxamide 2—(tert-butylarnino)-4_ ((1R,3R,4R)hydr0xy—4_ methylcyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Structure NAME 2-((1r,4R) cyclopropoxycyclohexylarnino) ((1R,3 S)hydroxycyclohexylamino )pyrirnidine-5 - carboxamide 4-((1R,3 S)-3 - hydroxycyclohexylarnino) ((1r,4R)—4-methoxycyclohexylamino )-N-methylpyrirnidine-5 - carboxamide 4-((1R,3 S)—3-hydroxy-3 - methylcyclohexylarnino) R)—4-((2H3)rnethyloxy)- cyclohexylamino)pyrirnidine-5 - carboxamide ,3R,4R)hydroxy methylcyclohexylarnino) (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - amide 2-((1r,4R) ethoxycyclohexylamino) ((1R,2S)—2-(hydr0xyrnethyl)- cyclopentylamino)pyrimidine- -carboxarnide 4-(tert-buty1arnino)((1r,4r)- 4-methoxycyclohexy1- amino)pyrimidine-5 - carboxamide ATI—ZS 14175V1 Cmpd Structure NAME MH+ Act.
A- Level 2 1 1 "xi 4-(tert-buty1arnino)((1r,4r)- 336.4 D f; 4-ethoxycyclohexylamino)- xi [TR pyrimidine-S-carboxamide ~55"; ""N‘izf’m a 53$}ng 4-(tert-buty1arnino)((1r,4r)- ,2-trifluoroethoxy)— cyclohexylamino)pyrirnidine-5 - carboxamide t-butylamino)((1R,3S)- 3-hydr0xyrnethy1cyclohexyl- amino)pyrimidine-5 - carboxamide 4-(isopr0pylarnino)—2-((1r,4r)- 4-(rnethylarnino)cyclohexylamino )pyrimidine-5 - carboxamide 4-((1R,3 S)—3-hydr0xy- 1 _ methylcyclohexylarnino) ((1r,4R)—4-methoxy- cyclohexylamino)pyrirnidine-5 - carboxamide 4-((1R,3 S)-3 - ycyclohexylarnino) ((1r,4R)—4-rnethoxy-1 -methy1— cyclohexylamino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Cmpd Structure NAME MH+ A°t° A- Level 217 - 342 D <\F [fl] difluorocyclobutylarnino) \T‘ ((1r,4r)hydr0xycyclohexy1— m" {\KT‘M' amino)pyrimidine H 419%?” carboxamide 4-(2-cyclopropylethylarnino) ((1r,4r)hydr0xycyclohexy1— amino)pyrimidine-5 - carboxamide 4-(2-cyclobutylethylarnino) ((1r,4r)hydr0xycyclohexy1— amino)pyrimidine-5 - carboxamide 2-((1r,4r)—4- hydroxycyclohexylamino) (isopentylarnino)pyrimidine-5 - carboxamide lobutylmethylarnino) ((1r,4r)hydr0xycyclohexy1— amino)pyrimidine-5 - amide 4-(isopr0pylarnino) (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 2012/034349 Cmpd Structure NAME MH+ Act.
A- Level 223 WL 4-(bicyclo[1.1.1]pentan 318.6 D E: 3 ylamino)((1r,4r) is}: g» 3 hydroxycyclohexylamino)- “’11 5 ‘5: .-"N at“: pyrlmldlne-S-carboxarmde. . . .
H “‘3; 3“ ,4r)—4- ethoxycyclohexylamino) (isopropylamino)pyrirnidine-5 - carboxamide 2-((1r,4r)—4- hydroxycyclohexylamino) (tert-pentylarnino)pyrimidine-5 - carboxamide 4-(cyclopropylrnethylarnino) ((1r,4r)hydr0xycyclohexy1— amino)pyrimidine-5 - carboxamide 2-(tert-butylarnino) (isopropylamino)pyrirnidine-5 - carboxamide 2-((1r,4r)—4- hydroxycyclohexylamino) (neopentylarnino)pyrimidine-5 - carboxamide ATI—ZS 14175V1 Cmpd. ure NAME MH‘ Act.
A- Level 229 2-(4- 376.2 D iiifw hydroxybicyclo[2.2.2]octan 1 “‘1‘. “£19.! ylamino)((1R,3S)—3- hydroxycyclohexylamino)- dine-S-carboxamide 230 1-: ~. 2-((ls,4s) 294.1 C hydroxycyclohexylamino) (isopropylamino)pyrirnidine-5 - carboxamide 230 -. ,4s) 306.4 D ‘ hydroxycyclohexylamino)—4-(1- methylcyclopropylarnino)- pyrimidine-S-carboxamide 231 4-((1R,3S) 390.2 D :5.ij hydroxycyclohexylamino)—2-(4- WAN methoxybicyclo[2.2.2]octan #J\«”I\SET‘JR 33% ylarnino)pyrirnidine “= " “C carboxamide 232 N Am 4-(cyc10butylarnino)(4- 332.2 D mLEEI hydroxybicyclo [2.2 .2]octan M Mg.” ylarnino)pyrirnidine ‘-'-+“'“xi...4§\.i:;€-“ carboxamide +1.3 "55*: 233 Hi? 3"”“3 2-((1 S ,4R) 308 D {\W.-“‘“-‘§* hydroxycycloheptylarnino) 54.5%}. (isopropylamino)pyrirnidine-5 - -. ":1 . 3 2%"... carboxarmde ““? 3.
ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 234 H5 .~ fan.” 2-((1R,4S) 308 D hydroxycycloheptylarnino) (isopropylamino)pyrirnidine-5 - carboxamide 235 2-((1r,4r)—4- 308.5 D hydroxycyclohexylamino) (oxetany1amino)pyrimidine- -carboxarnide 236 [0001] (R)(tert- 294 C butylarnino)(tetrahydr0-2H- pyranylarnin0)pyrirnidine-5 - carboxamide 237 4-(tert-butylarnino)(4- 334.2 hydroxybicyclo[2.2.2]0ctan ylarnino)pyrirnidine-5 - amide 238 2,4-bis(tert- 266.4 butylarnino)pyrimidine-S - amide 239 2-((1r,4r)—4- 320.2 hydroxycyc10hexy1arnino)—4-(1 - methylcyclobutylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 240 | 4-((1R,3R,4R)hydroxy 308 D [1 x} methylcyc10hexy1arnino) ;: (isopropylamino)pyrirnidine fl 1 7| ‘3“ch carboxamide H \g V «,9- N55“: 241 :Kr 2-(4,4-difluorocyclohexy1- 370.6 D MW E; L. ] amino)((1R,2S)—2- w! I; (hydroxyrnethyl)cyclopentyl- amino)-pyrirnidine E1 1 carboxamide 242 7; 2-((1r,4r)rnethoxy- 334.2 exylamino)(2- methylcyclobutylamino)- pyrimidine-S-carboxamide 243 2-((1r,4r)rnethoxycyclo- 334.2 hexylamino)—4-(2- methylcyclobutylamino)- pyrimidine-S-carboxamide 244 »~ .. 2-((1r,4r)—4- 320.2 ‘ ‘ hydroxycyclohexylamino)—4-(2- methylcyclobutylamino)- pyrimidine-S-carboxamide 245 ‘ 320-2 : 2-((1r,4r)—4- hydroxycyclohexylamino)—4-(2- cyclobutylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 246 .L 2-((1r,4r)—4- 320.2 ycyclohexylamino)—4-(2- methylcyclobutylamino)- pyrimidine-S-carboxamide 247 2-((1r,4r)—4- 320.2 hydroxycyclohexylamino)—4-(2- methylcyclobutylamino)- pyrimidine-S-carboxamide 248 lobutylamino)—2-(4_ 320 hydTOXycycloheptylamino)_ pyrimidine-S-carboxamide 249 4-(Cyclobutylamino)—2-(4_ 320 hydroxycycloheptylamino)- pyrimidine-S-carboxamide 250 4-(tert-butylarnino)(4- 322 hydroxycycloheptylamino)- pyrimidine-S-carboxamide 4-(tert-butylarnino)(4- 322.5 hydroxycycloheptylamino)- pyrimidine-S-carboxamide .
ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level ’0‘ 4-((1R,ZS) 336 D m”“5““. l: \l (hydroxyrnethyl)cyclopentyl- E» ” x I amino)(tetrahydr0-2H-pyran- H V 4-ylamino)pyrirnidine-5 - .. 11:? 3T M carboxarnide 253 .5»qu e * 2-(4-fluorobicyclo[2.2.2]octan- 322.1 D w” (:33 1 -y1arnino) .1ng (isopropylarnino)pyrirnidine LVJLVL carboxamide {55, 254 2-((1r,4r)—4- 334.2 methoxycyclohexylamino) (2-rnethy1cyclobutylarnino)- pyrimidine-S-carboxamide 255 2-((1r,4r)—4- 334.2 methoxycyclohexylamino) (2-rnethy1cyclobutylarnino)- pyrimidine-S-carboxamide 256 nethylcyclobutylarnino)- 402.2 ,4r)(2,2,2- trifluoroethoxy)cyclohexyl- amino)pyrimidine-5 - carboxamide 257 nethylcyclobutylarnino)- 402.2 2-((1r,4r)(2,2,2- trifluoroethoxy)cyclohexylamino )pyrimidine carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 258 4-(2-rnethylcyclobutylarnino)- 402.2 D 2-((1r,4r)(2,2,2- trifluoroethoxy)cyclohexylamino )pyrimidine-5 - carboxamide 259 J 2-((1r,4R) 3782 D ethoxycyclohexylamino) (( 1 R,2R) (hydroxyrnethyl)cyclopentylamino )pyrimidine-5 - carboxamide 260 g“ ,2R) 364.2 D (hydroxyrnethyl)cyclopentylamino )((1r,4R)—4- methoxycyclohexylamino)- pyrimidine-S-carboxamide 261 3 4—<<R> 320.2 D cyclopropylethylamino) (( 1r,4R)—4- hydroxycyclohexylamino)- pyrimidine-S-carboxamide 262 2-((1r,4r)—4- 3362 D hydroxycyclohexylamino)—4-(3- methylpentan-3 - no)pyrirnidine-5 - carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 263 EN —l-cyclopropylethyl- 320.2 D ”‘ ' 3-44., 15x...»| am1n0)—2—(( 1 r,4S) T ycyclohexylam1no)-- T} 2k?“ pyrimidine-S-carboxamide Hfix _«- 6;?“N 264 ”If 2-(bicyclo[1.1.1]pentan 332.2 D g 3;” ylarnino)((1R,3R,4R) m \ hydroxy 4 i :5" RATE»: methylcyclohexylamino)- Swflkam‘; 7 pyrimidine-S-carboxamide 265 4-(2-rnethylcyclobutylarnino)- 402.2 2-((1r,4r)(2,2,2- trifluoroethoxy)cyclohexylamino )pyrimidine carboxamide 266 4-((1R,ZS)(hydroxyl- 432.2 methy1)cyc10penty1arnino)—2- ((1r,4R)—4-(2,2,2-trifluoroethoxy )cyclohexylamino)- pyrimidine-S-carboxamide 267 2-(4-flu0robicyclo [2 .2.2]0ctan- 378.2 1-y1arnino)((1R,3 S)-3 - hydroxycyclohexylarnino)pyrirn idine-S-carboxarnide 268 2-((1r,4R)(diflu0r0- 400.2 methoxy)cyclohexylamino) ((1R,2S)—2-(hydr0xyrnethyl)- entylamino)pyrirnidine- -carboxarnide ATI—25 14175V1 Cmpd. ure NAME MH+ Act.
A- Level 4-(ethylamino)((1r,4r) 280.1 D hydroxycyclohexylamino)- pyrimidine-S-carboxamide 270 2-((1r,4r)—4- 266.1 C hydroxycyclohexylamino) (methylamino)pyrimidine-5 - carboxamide 271 ;- 2-((1r,4r)—4- 338.3 hydroxycyclohexylamino)—4-(1- methoxyrnethylpr0pan ylarnino)pyrirnidine-5 - carboxamide 272 306 , 3 .V 2-(cyclopropy1arnino) . ..- ' ’— ((1R,3R,4R)hydroxy methylcyclohexylamino)- pyrimidine-S-carboxamide 273 »: 4-(2,3-dirnethylbutan 364.2 i ylamino)((1r,4r) ethoxycyclohexylamino)- pyrimidine-S-carboxamide 274 : . 4-(2,3-dirnethylbutan 350.2 3 i 3 ylamino)((1r,4r) methoxycyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 275 4-(2,3-dirnethylbutan 336.3 D MR 15..
Ix F ”I ylamino)((1r,4r) T it]; ycyclohexylamino)- *N pyrimidine-S-carboxamide [ a1 276 i“ =9 4-((S)—3,3-dirnethy1butan 364.2 D ‘i ”Wm-1 ylamino)—2-((1r,4S) \ffl ethoxycyclohexylamino)- “ [KL pyrimidinecarb0xarnide 277 4-((R)-3,3-dimethylbutan 364.2 no)((1r,4R)_4_ ethoxycyclohexylamino)- pyrimidine-S-carboxamide 278 4-((S)—3,3-dirnethy1butan 350.2 ylamino)—2-((1r,4S) methoxycyclohexylamino)- pyrimidine-S-carboxamide 279 4-((R)-3,3-dirnethylbutan 350.2 ylarnino)((1r,4R) methoxycyclohexylamino)- pyrimidine-S-carboxamide 280 4-((S)—3,3-dirnethy1butan 336.2 ylamino)—2-((1r,4S) hydroxycyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 WO 45569 2012/034349 Cmpd. Structure NAME MH+ Act.
A- Level m! 15:. 4-((R)-3,3-dirnethylbutan 336.2 D \L L. ylamino)((1r,4R)—4- '2 ital hydroxycyclohexylamino)- "‘1- pyrimidine-S-carboxamide [ a1 282 M .2. 2-((1r,4S) 350.2 D 'i‘iin’fifE’Q}' ethoxycyclohexylamino) ; -rnethylbutan m [Tel ylarnino)pyrirnidine-5 - [IA carboxamide 283 2-((1r,4S) 336.2 methoxycyclohexylamino) ((S)-3 -rnethy1butan ylarnino)pyrirnidine-5 - carboxamide 284 2-((1r,4S) 322.2 hydroxycyclohexylamino) ((S)-3 -rnethy1butan ylarnino)pyrirnidine-5 - carboxamide 285 2'(Cy010butylarnino)_4- 320.2 (( 1 Ra3R,4R)-3 -hydr0xy—4_ methylcyclohexylamino)- pyrimidine-S-carboxamide 286 4-((1R,3R,4R)hydroxy 336.2 methylcyclohexylarnino) (tert-pentylarnino)pyrimidine-5 - carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 287 1 4-((1R,3R,4R)hydroxy 334.1 D i: \f a“ methy1cyclohexy1arnino)(1- ;; methylcyclobutylamino)- m FL “I pyrimidine-S-carboxamide . x?» x 288 1 4-((1R,3R,4R)hydroxy 320.2 D L].. .m methy1cyclohexy1arnino)(1- ‘f‘w’ «:3 methylcyclopropylarnino)- fl» :1” pyrimidine-S-carboxamide M :1] :31 289 4-(tert-buty1arnino)(4- 336.2 fluorobicyclo[2.2.2]octan ylarnino)pyrirnidine-5 - carboxamide 290 2-((1r,4R) 350.2 ethoxycyclohexylamino) ((R)—3-methy1butan—2- ylarnino)pyrirnidine-5 - carboxamide 291 ,4R) 336.2 methoxycyclohexylamino) ((R)—3-methy1butan—2- ylarnino)pyrirnidine-5 - carboxamide 292 2-((1r,4R)—4- 322.2 hydroxycyclohexylamino)-4_ -rnethylbutan ylarnino)pyrirnidine-5 - carboxamide ATI—25 14175V1 Cmpd. ure NAME MH+ Act.
A- Level 293 H a, 4-((S)— 1 -cyclobutylethylarnino)- 362.2 D ‘S-‘*'l;"»- ”ax M‘wx 2 ((1r,4S) 4 Y - - - *1 1 ”f“ ethoxycyclohexylamino)- “I: l dine-S-carboxamide 294 4-((R) 362.2 A cyclobutylethylamino) ((1r,4R)—4- ethoxycyclohexylamino)- pyrimidine-S-carboxamide 295 4-((S)— 1 -cyc10butylethylarnino)- 348.2 2-((1r,4S) methoxycyclohexylamino)- pyrimidine-S-carboxamide 296 4-<<R> 348.2 cyclobutylethylamino) (( 1r,4R)—4- ycyclohexylamino)- pyrimidine-S-carboxamide 297 4-((S)— 1 -cyc10butylethylarnino)- 334.2 2-((1r,4S) hydroxycyclohexylamino)- pyrimidine-S-carboxamide 298 4-<<R> 334.2 cyclobutylethylamino) (( 1r,4R)—4- hydroxycyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 PCT/U52012/034349 Cmpd. Structure NAME MH+ Act.
A- Level 299 WT: 4-(tert-butylamino) 308.2 D N \TEIW‘X’ ((1 R,3R)_3 ' ””1”“ hydroxycyclohexyl- m [a] amino)pyrimidine-5 - XE: carboxarnide 300 H94 a «.39 (S)(tert-butylamino) 294. 1 D ' (tetrahydro-2H-pyran ylarnino)pyrirnidine-5 - amide 301 .V (R)(tert-buty1arnino) 294.1 (tetrahydro-2H-pyran ylarnino)pyrirnidine-5 - carboxamide 302 ' w: 4-(tert-buty1arnino)((1r,4r)- 322.2 4-hydr0xy methylcyclohexylamino)- pyrimidine-S-carboxamide 303 5-: '5 : 4-(tert-butylarnino)(oxetan- 266.1 3 -y1arnin0)pyrirnidine-5 - carboxamide 304 g: 4-(tert-buty1arnino)((1r,4r)- 348.3 4-cyclopropoxycyclohexylamino )pyrimidine carboxamide —274— ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 305 4-(tert-butylamino)((1S,3S)- 308.1 D KTRHRI 3-hydr0xycyc10hexy1arnino)— ””1”“ pyrimidine-S-carboxamide 306 .MVD 4-(tert-butylamino)((1S,3R)- 308.2 D ' 3-hydr0xycyc10hexy1arnino)— pyrimidine-S-carboxamide 307 9‘s 3 4-(tert-butylamino)((1R,3S)- 308.2 3-hydr0xycyc10hexy1arnino)— pyrimidine-S-carboxamide 308 4-(tert-buty1arnino)((1r,4r)- 363.2 nethylcarbamoyl)- cyclohexylamino)pyrirnidine-5 - carboxamide 309 2-((1r,4r)—4- |||||||||||!||||||||||||!||||||||||||!||||||||||||!|353.2 ‘ ethoxycyclohexylarnino)(1- hydroxymethylpropan ylarnino)pyrirnidine-5 - carboxamide 310 4-(1-hydr0xyrnethylpropan- 338.2 2-y1arnino)((1r,4r) ycyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. ure NAME MH+ Act.
A- Level 3 1 1 1:1: 4-(1-hydr0xyrnethylpropan- 324.2 D $112» 1* 2-y1arnino)((1r,4r) b 1L1?! hydroxycyclohexylamino)- 1- x pyrimidinecarb0xarnide [.1 L31 312 1 4-((1R,3R,4R)hydr0xy 322.3 D methylcyc10hexy1arnino) (oxetany1amino)pyrimidine- -carboxarnide 313 2-((1r,4r)—4- 348.3 ethoxycyclohexylamino) (( 1 r, 3r)-3 - methylcyclobutylamino)- pyrimidine-S-carboxamide 314 4-(2-cyclopr0pylpropan_2_ 362.2 ylamino)((1r,4r)_4_ ethoxycyclohexylamino)- pyrimidine-S-carboxamide 315 4-(2-cyclopr0pylpropan_2_ 348.2 ylamino)((1r,4r)_4_ ycyclohexylamino)- pyrimidine-S-carboxamide 316 4-(2-cyclopr0pylpropan_2_ 334.2 ylamino)((1r,4r)_4_ hydroxycyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 317 “a. 4-(tert-butylarnino) 294.1 D :1- ”#3 a 5.. -. « (tetrahydro-2H-pyran 1";558 ylarnino)pyrirnidine «11‘ fix carboxamide L l _ 318 m * 4-(tert-butylamino)(4,4- 328.1 D difluorocyclohexylamino)- dine-S-carboxamide 319 4-(tert-buty1arnino)((1r,4r)- 358.2 u0rornethoxy)cyclohexylamino )pyrimidine-5 - carboxamide 320 4-(bicyclo[1.1.1]pentan 332.1 ylamino)((1r,4r) methoxycyclohexylamino) pyrimidine-S-carboxamide 321 2-(4-fluorobicyclo[2.2.2]0ctan- 392.4 1-y1arnino)((3 S)hydroxymethylcyclohexylarnino) pyrimidine-S-carboxamide 322 2-((1r,4r)—4- 334.2 hydroxycyc10hexy1arnino)—4-(1 - methylcyclopentylamino) pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level V, x, w 2-(tert-butylamino)((1 S,3 S)- 336.2 C R} 3-hydr0xy-4,4- dimethylcyclohexylamino) :1 pyrimidine-S-carboxamide 324 2-(tert-butylamino)((1R,3R)- 336.2 D 3-hydr0xy-4,4- dimethylcyclohexylamino) dine-S-carboxamide 325 2-((1r,4r)—4- 320.2 hydroxycyclohexylamino) (( 1 r, 3r)-3 - methylcyclobutylamino) pyrimidine-S-carboxamide 326 2-((1r,4r)—4- 348.2 ethoxycyclohexylamino) (( 1 s ,3 s)-3 - methylcyclobutylamino) dine-S-carboxamide 327 ,4r)—4- 320.2 ‘ ( hydroxycyclohexylamino) ((1 s,3s) methylcyclobutylamino) pyrimidine-S-carboxamide 328 4-((1R,2R) 336.1 ‘ (hydroxyrnethyl)cyclopentyl amino)(tetrahydr0-2H-pyran- 4-ylamino)pyrirnidine carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 329 9* 9:949 2-((1r,4R)(difluoro- 400.2 D {“3 ,1: “fl" methoxy)cyclohexy1 ”1 amino)((1R,2R) If [SIX (hydroxyrnethyl)cyclopentyl- F 919.“ amino)pyrimidine-5 - carboxamide 2-(tert-butylamino)((1R,2R)- r0xyrnethy1)— cyclopentylamino)pyrirnidine- -carboxarnide 4-((1R,2R)(hydr0xymethyl)- cyclopentylamino)—2-((1r,4R)- 4-(2,2,2-trifluoroethoxy)— cyclohexylamino)pyrirnidine-5 - carboxamide 2-((1r,4r)—4- ethoxycyclohexylamino) (2 , 3 , 3 -trimethylbutan ylarnino)pyrirnidine-5 - carboxamide ,4r)—4- methoxycyclohexylamino) (2 , 3 , 3 -trimethylbutan ylarnino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 WO 45569 Cmpd. Structure NAME MH+ Act.
A- Level m15.. 2-((1r,4r)—4- 350.2 D \Lx R .m. hydroxycyclohexylamino) T L1 (2,3,3-trirnethylbutan ylarnino)pyrirnidine [Vim carboxamide 335 2-(4-fluorobicyclo[2.2.2]octan- 392.4 D 1-y1arnin0)((1R,3 S)-3 - ycycloheptylamino)- pyrimidine-S-carboxamide 336 2-((1r,4r)—4- 334.2 methoxycyclohexylamino) ((1r,3r)-3 -rnethy1cyc10buty1— amino)pyrimidine-5 - carboxamide 337 2-((1r,4r)—4- 334.2 methoxycyclohexylamino) ((1 5,3 s)-3 -rnethy1cyclobutylamino )pyrimidine-5 - carboxamide 338 4-((1R,3 S)—3-hydroxy 308.2 methylcyc10hexy1arnino) (isopropylamino)pyrirnidine-5 - carboxamide 339 2'(Cy010butylarnino)_4- 320.2 ((1R93 S)'3 'hYdI'OXycyclo_ heptylamin0)pyrirnidine—5 _ carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level w. ”.3 lobuty1arnino) 320.2 D 3.; g; T: ((1R,3S)—3-hydroxy I: W] EM]; methylcyclohexylamino)- w 51: pyrimidine-S-carboxamide 341 lobutylarnino) 306.2 D M V0 H3 («my iii \. :1: ((1R,3 S)—3-hydr0xycyclohexyl- Iw] E :1 amino)pyrimidine carboxamide H; “ 342 _';. - 2-(tert-butylarnino)(2- 336.2 hydroxy-4,4-dirnethy1— cyclohexylamino)pyrirnidine-5 - carboxamide 343 1-4 2-(tert-butylarnino)(2- 336.2 hydroxy-4,4-dirnethy1— cyclohexylamino)pyrirnidine-5 - carboxamide 344 -. 4-(bicyclo[1.1.1]pentan 346.4 ‘ ylamino)((1r,4r)—4-ethoxycyclohexylamino )pyrirnidine-5 - carboxamide 345 4-((1R,3R,4R)hydroxy 350.5 methy1cyclohexylarnino)(3- methylpentan-3 - ylarnino)pyrirnidine-5 - carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 346 2-(bicyclo[1.1.1]pentan 332.2 D ylamino)((1R,3 S)—3 - hydroxycycloheptylamino)- pyrimidine-S-carboxamide 347 332.2 D . 2-(bicyclo[1.1.1]pentan K F“. ”ask g. {is ylam1no)((1R,3S)—3-_ EM?“ “YE; hydroxymethy1— 513:: “gm: cyclohexylamino)pyrirnidine-5 - carboxamide 348 2-(bicyclo[1.1.1]pentan 318.1 ., _ ‘ ylamino)((1R,3S)—3- hydroxycyclohexylamino)- dine-S-carboxamide 349 . 4-((1R,3S) 336.2 ' hydroxycycloheptylarnino) (tert-pentylarnino)pyrimidine-5 - carboxamide 350 4-((1R,3S)—3-hydr0xy 336.2 I \‘ '7 cyc10hexy1arnino) (tert-pentylarnino)pyrimidine-5 - carboxamide 351 4-((1R,3S) 332.2 " hydroxycyclohexylamino) (tert-pentylarnino)pyrimidine-5 - carboxamide ATI—25 14175V1 2012/034349 Cmpd. Structure NAME MH+ Act.
A- Level 352 3* 4-((1R,3S) 334.2 D hydroxycycloheptylarnino) (1-methylcyclobutylamino )pyrimidine-5 - carboxamide 353 4-((1R,3S)—3-hydroxy 334.2 D methylcyclohexylamino)(1 - methylcyclobutylamino)- pyrimidine-S-carboxamide 354 4-((1R,3S) 320.2 .V ‘ hydroxycyc10hexy1arnino)—2-(1 - cyclobutylamino)- pyrimidine-S-carboxamide 355 ‘=. 4-(bi(cycloprop)ylarnino) 332,4 a“ 3. ‘ ((118410hydroxycyclohexylamino )pyrimidine carboxamide 356 _ 4-(1-ethylcyclopropylarnino) 320.3 ((1r,4r)hydr0xycyclohexy1— amino)pyrimidine-5 - carboxamide 357 is ;- 2-((1r,4r)hydroxycyclo- 348.2 hexylamino)—4-(1 - methylcyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 358 2-((1r,4r)—4- 362.2 D “34K” ”3%I methoxycyclohexylamino) L39] N \f“ (1 -rnethylcyclohexy1arnino)- WT “L pyrimidine-S-carboxamide Nuax‘u 359 2-((1r,4r)—4- 376.3 D ethoxycyclohexylamino)(1 - methylcyclohexylamino)- dine-S-carboxamide 360 2-((1r,4r)—4- 362.2 ethoxycyclohexylamino)(1 - ethylcyclobutylamino)- pyrimidine-S-carboxamide 361 4-(1-ethylcyclobutylarnino) 348.2 ((1r,4r)rneth0xycyclohexylamino )pyrimidine-5 - carboxamide 362 4-(1-ethylcyclobutylarnino) 334.2 ((1r,4r)hydr0xycyclohexy1— pyrimidine-5 - carboxamide 363 2-((1r,4r)—4- 322.4 hydroxycyclohexylamino) (pentany1arnino)pyrirnidine- -carboxarnide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 364 E 2-(cyc10pentylarnino) 334.2 D [: :] yJJJ {—3 R,4R)hydroxy J: methylcyclohexylamino)- JJ 9? pyrimidinecarboxarnide J TI“ :1: 365 2—(tert-butylamino)((1R,2S)- 308.3 D R‘W‘NJJ a} .4 r0xyrnethy1)cyclopentyl- :flxfi -J-QN‘S’ amino)pyrimidine “In, It? carboxarnide 366 2—(2,3-dimethylbutan 350.4 ylarnino)((1R,3R,4R) hydroxyrnethylcyclohexylamino )pyrimidine carboxamide 367 4-((1R,3R,4R)hydroxy 336.2 methylcyclohexylarnino) (neopentylarnino)pyrimidine-5 - carboxamide 368 4-(bicyc10[1.1.1]pentan 358.3 ylamino)((1r,4r) cyclopropoxycyclohexylarnino) pyrimidinecarboxarnide 369 4-(2,4-dimethylpentan 350.3 ylamino)((1r,4r) hydroxycyclohexylamino) dinecarboxarnide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 370 flew, v w \ 4-((3 S)—3 -hydr0xy-3 - 404.4 D m £31 methy1cyclohexylarnino)(4- N{R :\ methoxybicyclo[2.2.2]octan “wk“\\ ylamino)pyrirnidine-5 - 31““ H carboxarnide 371 W3N3 4-(tert-buty1arnino)((1 s,4s)- 3222 D 4-hydr0xyrnethy1— \_ [ i N x};‘ cyclohexylamino)pyrimidine ‘11 7: carboxamide H :“4 kg,“ x69“ 372 4-((S)—1- 348.2 ropylpropylamino) ((1r,4S)rnethoxycyclohexylamino )pyrirnidine-5 - amide 373 4-((S) 362.2 cyclopropylpropylamino) (( 1 r,4S)ethoxycyc10hexy1— amino)pyrimidine-5 - carboxamide 374 4-((R) 348.2 cyclopropylpropylamino) ((1r,4R)—4-rnethoxycyclohexyl- amino)pyrimidine-5 - carboxamide 375 4—<<R> 362.3 cyclopropylpropylamino) ((1r,4R)—4-eth0xycyclohexy1— pyrimidine carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level ”WAN t-butylamino)((1R,3S)- 3222 D S {jifim 3-hydr0xycyc10heptylarnino)— H)“; ”3““; :3 pyrimidine-S-carboxamide 377 f;- 2-(cyclopropylamino) 306.2 D :k} m ((1R,3S)—3-hydroxycyclo- iu-a _’ “L17 heptylamino)pyrimidine miga‘u} carboxarnide 378 2-(cyclopropylamino) 306.2 ((1R,3 S)—3-hydroxy-3 - methylcyclohexylamino)- pyrimidine-S-carboxamide 379 2-(cyclopropy1arnino) 292.1 ((1R,3 ydroxycyclohexylamino )pyrirnidine-5 - carboxamide 380 4-(tert-butylamino)((1R,3S)- 322.2 3-hydr0xyrnethy1cyclohexyl- amino)pyrimidine carboxamide 381 2-((1r,4r) 336.2 methoxycyclohexylamino) (tert-pentylarnino)pyrimidine-5 - carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 382 fig 2-((1r,4r) 362.3 D W; cyclopropoxycyclohexylarnino) LY: LY] (tert-pentylarnino)- W pyrimidinecarb0xarnide fl “m‘3le x“ «m 383 L 2-(sec-butylarnino) 322.3 D L L .1 ((1R,3R,4R)hydroxy 1;; g ‘1: methylcyclohexylamino)- ‘ ML U :L pyrimidine-S-carboxamide 384 4-((S) 334.2 ropylpropylamino) S)hydr0xycyclohexylamino )pyrimidine-5 - carboxamide 385 4-((R) 334.2 cyclopropylpropylamino) ((1r,4R)—4-hydroxycyclohexylamino )pyrimidine-5 - carboxamide 386 4-(tert-butylamino)((1R,3R)- 322.2 3-hydroxycycloheptylamino )pyrimidine-5 - carboxamide 387 4-(tert-butylamino)((1S,3S)- 322.2 3 -hydroxycyc10hepty1arnino)— pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level _:¢,_.m\ 4-(tert-butylarnino)((1 S,3R)- 322.2 D I'VE? 3-hydr0xycycloheptylarnino)- m: Rafi-wk };j__ pyrimidine-S-carboxamide “if": 389 4-(tert-butylarnino)(4- 308.2 D methyltetrahydro-2H-pyran ylarnino)pyrirnidine-5 - carboxamide 390 2-((1r,4r)—4- 336.2 hydroxycyclohexylamino)—4-(2- methylpentan ylarnino)pyrirnidine-5 - carboxamide 391 ,4r)—4- 364.3 ethoxycyclohexylarnino)(2- pentan ylarnino)pyrirnidine-5 - carboxamide 392 2-((1r,4r)—4- 350.2 methoxycyclohexylamino) (2-rnethylpentan ylarnino)pyrirnidine-5 - carboxamide 393 " 2-((1r,4r)—4- 332.3 ‘ hydroxycyclohexylamino)—4-(3- methylbicyclofl . 1 . 1]pentan ylarnino)pyrirnidine amide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 394 i 2-((R) 334.4 D “we“ a.\ “: L J cyclopropylethylamino) :5 N L: ((1R,3R,4R)hydroxy I! j methylcyclohexylamino)- U My pyrimidine-S-carboxamide 395 2-((S) 334.2 D cyclopropylethylamino)-4_ (( 1 Ra3R,4R)-3 -hydr0xy—4_ cyclohexylamino)- pyrimidine-S-carboxamide 396 4-((1R,3R,4R)hydroxy 346.3 methylcyclohexylamino)(3 - methylbicyclo [1 . 1 . 1]pentan ylarnino)pyrirnidine-5 - carboxamide 397 2-(2-cyclopr0pylpr0pan 348.3 ylarnino)((1R,3R,4R) hydroxyrnethylcyclohexy1— amino)pyrimidine-5 - carboxamide 398 2-(sec-butylarnino)-4 _ 322.3 (( 1 Ra3R,4R)-3 xy—4_ methylcyclohexylamino)- pyrimidine-S-carboxamide 399 4-(tert-butylarnino)(4- 320.2 ybicyclo [2.2. 1]heptan ylarnino)pyrirnidine-5 - carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 400 Hg.‘ V. {3a 2-((1r,4r)—4- 360.2 D cyclopropoxycyclohexylarnino) ( 1 -rnethy1cyclobutylarnino)- pyrimidine-S-carboxamide 401 2-((1r,4r)—4- 348.2 D ethoxycyclohexylamino)(1 - cyclobutylamino)- dine-S-carboxamide 402 2-((1r,4r)—4- 334.2 methoxycyclohexylamino) (1 -rnethylcyclobuty1arnino)- pyrimidine-S-carboxamide 403 (S)(1- 306.2 methylcyclobutylamino) (tetrahydro-2H-pyran-3 - ylarnino)pyrirnidine-5 - carboxamide 2-((1R,3 S)-3 - 320.2 hydroxycyc10hexy1arnino)—4-(1 - methylcyclobutylamino)- pyrimidine-S-carboxamide 405 (S)(bicyc10[1.1.1]pentan 304.2 ylarnino)(tetrahydro-2H- pyranylarnin0)pyrirnidine-5 - carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 406 4-(bicyc10[1.1.1]pentan 3182 D o)((1R,3 S)—3- hydroxycyclohexylamino)- ‘2‘ pyrimidinecarb0xarnide ”:3“? 407 4-(bicyc10[1.1.1]pentan 3042 D ylarnino)(tetrahydro-2H- pyrany1arnino)pyrirnidine-5 - carboxamide 408 (R)(1- 346.2 cyclopropylethylamino)(4- hydroxybicyc10[2.2.2]0ctan ylarnino)pyrirnidine-5 - carboxamide 4-((R) 402.2 cyclopropylethylamino) ((1r,4R)—4-(2,2,2-trifluoroethoxy )cyclohexylamino)- pyrimidine-S-carboxamide 410 2-((1r,4R) 360.2 cyclopropoxycyclohexylarnino) ((R)-1 -cyclopr0pylethyl- amino)pyrimidine-5 - carboxamide 411 348.2 ropylethylamino) ((1r,4R)—4-eth0xycyclohexy1— amino)pyrimidine carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 412 xx. «1 2-(4- 346.2 D m«EXT ybicyclo [2.2 .2]octan <> ”reg”! ylarnino)—4-(1 -methylcyclo- M“‘Vh”ux§: butylamino)pyrimidine carboxarnide fo‘ in... 413 H.» *- 3:9 4-(1 -rnethy1cyclobuty1arnino)- 306.2 D N 3.3; le 2-(tetrahydr0-2H-pyran <> 1L : w‘ ylarnino)pyrirnidine-5 - mm m“ carboxarnide L .1 414 2-((1r,4r) 375.2 (dimethylcarbarnoyl)cyclohexyl amino)—4-(1 -rnethy1cyclobuty1- amino)pyrimidine-5 - carboxamide 415 4-(1 y1cyclobuty1arnino)- 402.2 ,4r)(2,2,2-triflu0roethoxy )cyclohexylamino)- pyrimidine-S-carboxamide 416 2-((1r,4r)—4- 370.2 (difluorornethoxy)cyclohexyla mino)(1 -rnethy1cyclobuty1- amino)pyrimidine-5 - carboxamide 417 4-(bicyc10[1.1.1]pentan 344.2 ylamino)(4-hydroxybicyclo [2.2.2]octan ylarnino)pyrirnidine-5 - carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 418 4-(bicyclo[1.1.1]pentan 373.3 D ylamino)((1r,4r) hylcarbarnoyl)cyclohexyl amino)pyrimidine-5 - carboxamide 419 4-(bicyc10[1.1.1]pentan 400.2 D ylarnino)((1r,4r)(2,2,2- trifluoroethoxy)cyclohexylamino )pyrimidine-5 - carboxamide 420 4-(bicyc10[1.1.1]pentan 368.2 ylamino)((1r,4r) (difluorornethoxy)cyclohexylamino )pyrimidine-5 - carboxamide 421 4—<<R> 334.2 cyclopropylethylamino) ((1r,4R)—4-meth0xycyclohexylamino )pyrimidine-5 - carboxamide 422 4-((R) 306.2 cyclopropylethylamino)((S)- tetrahydro-2H-pyran-3 - ylarnino)pyrirnidine-5 - amide 423 4—<<R> 320.2 ropylethylamino) (( 1 R, 3 S)—3 -hydr0xycyclohexylamino )pyrimidine carboxamide —294— ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 424 w... Nc. (R)(1- 306.2 D 3:3. I .«1 cyclopropylethylamino) lmya hydro-ZH-pyran m A.
E .a x. ylarnino)pyrirnidine 1 r‘1 : carboxarmde- 425 4-((R) 375.2 D —%\.~5 .3; cyclopropylethylamino) ((1r,4R)—4- L.l~é (dimethylcarbarnoyl)cyclohexyl “ amino)pyrimidine carboxamide 426 m .5: 4-((R) 370.2 D 3“»me_ cYclopr0py1€thylamin0)'2' ((1r,4R)—4-(diflu0r0rnethoxy)- 21L” cyclohexylamino)pyrimidine carboxarnide 427 2-(bicyclo[1.1.1]pentan 346.3 C ylarnino)(3-hydr0xy_4,4_ y1cyclohexylarnino)- pyrimidine-S-carboxamide 428 "’7, ‘ 2-(cyclobutylamino)(3- 334.2 D hydroxy-4,4- dimethy1cyclohexylarnino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A' Level 429 fix an 2-(cyclobutylamino)(3- 334.2 D L J 33:} hydroxy-4,4- H1 ,s-‘k >34 dimethy1cyclohexylarnino)- M x“ fl pyrimidine-S-carboxamide 430 4-(3-hydroxy-4,4- 334.2 D dimethylcyclohexylamino) (1 -rnethylcyc10propy1arnino)- pyrimidine-S-carboxamide 431 :4. - is-_ . 4-(3 -hydr0xy-4,4- 334.2 dimethylcyclohexylamino) (1 -rnethylcyc10propy1arnino)- pyrimidine-S-carboxamide 432 2-(bicyclo[1.1.1]pentan 3463 { “ ylarnino)(3-hydr0xy-4,4- y1cyclohexylarnino)- pyrimidine-S-carboxamide 433 4-(cyclobuty1arnino) 306.2 (( 1 R, 3 S)-3 - ycyclohexylamino)- pyrimidine-S-carboxamide 434 -‘ I” 4-(cyclobutylamino)((1r,4r)- 346.2 N ' cyclopropoxycyclohexylarnino) pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 435 Why? 4-(cyclobutylarnino) 292.2 D PM}JEWL (tetrahydro-2H-pyran LJ “[4]“ ylarnino)pyrirnidine ”Ex fem carboxarnide L ~. 436 “-‘k V lobutylamino)((1r,4r)- 3562 D 4-(difluor0rnethoxy)— cyclohexylamino)pyrirnidine-5 - carboxamide 437 4-(cyclobutylamino)((1r,4r)- 361.2 4-(dirnethylcarbamoyl)- cyclohexylamino)pyrirnidine-5 - carboxamide 438 4-((1R,3 S) 306.2 hydroxycyc10hexy1arnino)—2-(1 - methylcyclopropylarnino)- pyrimidine-S-carboxamide 439 2-(Cyclopentylarnino)-4_ 320.2 (( 1R3 S)'3 'hydTOXYCYCIOhexyI- amino)pyrirnidine_5 _ carboxamide 2-((1r,4r)—4- 386 D ycyclohexylamino)—4-(3- (trifluorornethyl)bicyclo[1 .1 . 1]- pentanylamino)pyrimidine- -carboxarnide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 441 4-((1R,3R,4R)hydroxy 400 methylcyclohexylamino)(3 - (trifluorornethyl)bicyclo[1 .1 . 1]- pentanylamino)pyrimidine- -carboxarnide 442 2-((1r,4r)—4- 372.3 cyclopropoxycyclohexylarnino) -4 _(3 _ methylbicyclo [1 . 1 . 1]pentan ylarnino)pyrirnidine-5 - carboxamide 443 ; 2-((1r,4r)—4- 360.3 cyc10hexy1arnino)(3 - methylbicyclo [1 . 1 . 1]pentan ylarnino)pyrirnidine-5 - carboxamide 444 Hm 2-(3 ,3 - 356.1 difluorocyclobutylarnino) ((1R,3R,4R)hydr0xy methylcyclohexylamino)- pyrimidine-S-carboxamide 445 yclopr0pylpr0pan 346.3 o)(4- hydroxybicyclo [2.2. 1]heptan ylarnino)pyrirnidine-5 - carboxamide ATI—ZS 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 446 gum 2—(4— 346.3 D :“: hydroxybicyclo [2.2. 1 ]heptan ylarnino)(1 - a :‘mmk LE: 133;th “ methylcyclopentylarnino)- fix“ pyrimidine-S-carboxamide 447 4-(cyclobuty1arnino)-2—(4- 318.2 D hydroxybicyclo [2.2. 1]heptan ylarnino)pyrirnidine-5 - carboxamide 448 (cyclobutylamino) 292.1 (tetrahydro-2H-pyran-3 - ylarnino)pyrirnidine-5 - carboxamide lobutylamino)—2-(4_ 306.2 methyltetrahydro-2H-pyran_4_ ylarnino)pyrirnidine-5 - carboxamide 450 (R)(1 - 332.2 cyclopropylethylamino)(4- hydroxybicyclo [2.2. 1]heptan ylarnino)pyrirnidine-5 - carboxamide 451 2-(4- 332.2 hydroxybicyclo [2.2. 1]heptan no)—4-(1-rnethylcyclobutylamino )pyrimidine carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 452 2—(4- 334.2 D hydroxybicyclo [2.2. 1]heptan ylamino)(tertpentylamino )pyrimidine carboxamide MNL 4-(bicyclo[1.1.1]pentan-2— 318.1 D $.ngTIE ylamino)-2—(( 1r,4r) AM 9Ti“ hydroxycyclohexylamino)- »~"\ pyrimidine-S-carboxamide 454 .. 4-(1-ethylcyclopentylamin0)-2' 362-2 . ((1r,4r)—4-methoxycyclohexy1- amino)pyrimidine-5 - carboxamide 455 a. 4-(1 -ethylcyc10penty1arnino)-2— 348 .2 ((1r,4r)hydr0xycyclohexy1— pyrimidine-5 - carboxamide 456 ‘ 4-(2-cyclopropy1propan 334_2 ylamino)-2—((IS,3S) hydroxycyclohexylamino)- pyrimidine-S-carboxamide 457 -‘ . 4-(2-cyclopr0pylpr0pan 334.2 - ylamino)-2—((IS,3R) ycyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 W0 45569 Cmpd. Structure NAME MH+ Act.
A- Level 458 (1:39.- yclo[1.1.1]pentan 330 D sfi> ylamino)((1r,4r) ,5 ”a.“ hydroxybicyclo [2.2. 1]heptan 11“? w ylarnino)pyrirnidine 51-13;, P {A 1‘1 carboxamide 459 A“ ‘34 2-((R) 348.3 D l j i“; cyclobutylethylamino) : . '1 M .4 L ((1R,3R,4R)hydroxy \Tr 1:? methylcyclohexylamino)- ”Wink? xx? pyrimidine-S-carboxamide 2-((S)— 1 -cyc10butylethy1arnino)- 348.3 4-((1R,3R,4R)hydroxy methylcyclohexylamino)- pyrimidine-S-carboxamide 461 2-((R)-3,3-dimethylbutan 350.2 ylarnino)((1R,3R,4R) hydroxy methylcyclohexylamino)pyrimi dine-S-carboxarnide 462 4-(bicyclo[2.1.1]hexan 360.2 ylamino)((1r,4r) ethoxycyclohexylarnino)pyrirni dine-S-carboxarnide 463 ,4r)cyclopropoxy- 388.2 cyclohexylamino)(1- ethylcyclopentylarnino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 464 2-((1r,4r)ethoxy- 376.2 D L H “fir; K:"jgl‘“““~t] exylamino)(1 - 3“"?' i’ N “15.: “j” yclopentylarmno)-. 3 3 [xx]: pyrimidine-S-carboxamide \x‘i‘wfl 465 “NT” 4-(2-cyclopr0pylpr0pan 334.2 D RWY“?! ylamino)((1R,3R) N "’3’” hydroxycyclohexylarnino)pyrirn idine-S-carboxarnide 4-(2-cyclopr0pylpropan 334.2 ylamino)((1R,3 S)—3 - hydroxycyclohexylamino)- pyrimidine-S-carboxamide 467 4-(2-cyclopr0pylpr0pan 320.2 ylarnino)(tetrahydro-2H- pyrany1arnino)pyrirnidine-5 - carboxamide 468 (S)(2-cyclopr0py1propan 320.2 ylarnino)(tetrahydro-2H- pyranylarnin0)pyrirnidine-5 - carboxamide —3,3-dirnethy1butan 350.3 ylarnino)((1R,3R,4R) hydroxy methylcyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 470 1 ,3R,4R)hydroxy 336.2 D I“ ] methylcyc10hexy1arnino) \Rm" a... ff a“ m r L ((R)—3-rnethylbutan VT} i1: ylarnino)pyrirnidine-5 - K v?! m": carboxamide 471 «L‘- 4-((1R,3R,4R)hydr0xy 336.3 D [ :1 cyc10hexy1arnino) _, . ] ”fix-l ((S)rnethy1butan—2- l ylarnino)pyrirnidine-5 - . ‘ ‘1 carboxamide 472 2-(cyclopropylrnethylarnino) 320.1 ((1R,3R,4R)hydr0xy methylcyclohexylamino)- pyrimidine-S-carboxamide 473 2-((1r,4r)—4- 348.3 methoxycyclohexylamino) (1 -methylcyclopentylarnino)- pyrimidine-S-carboxamide 474 2'(‘DKCYC10101V0p)ylamino)-4— 346.2 (( 1 Ra3R,4R)-3 -hydr0xy—4_ methylcyclohexylamino)- pyrimidine-S-carboxamide 475 4-((1R,3R,4R)hydroxy 348.2 methylcyclohexylamino)(1 - methylcyclopentylarnino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. ure NAME MH+ Act.
A- Level 476 “a“ 1 #1 4-(bicyclo[1.1.1]pentan 346.2 D gmtl ylarnino)((1r,4r) N T“ ethoxycyclohexylamino)- Lil” pyrimidine-S-carboxamide 477 :n 4-(bicyclo[2.1.1]hexan 332.2 D o)((1r,4r)_4_ hydroxycyclohexylamino)- pyrimidine-S-carboxamide 478 ‘r"- 2-((1r,4r) 390.2 ‘ " ethoxycyclohexylarnino)(1- ethylcyclohexylarnino)- pyrimidine-S-carboxamide 479 4-(2-cyclopropylpropan 360.2 , 4 __L ylamino)(4-hydroxybicyclo [2.2.2]octan no)pyrirnidine carboxamide 480 4-(2-cyclopr0pylpr0pan 389_2 ,= ., ylamino)((1r,4r) (dimethylcarbarnoyl)cyclohexyl amino)pyrimidine-5 - carboxamide 481 i: - -6 4-(2-cyclopr0pylpr0pan 384.2 e" ' ‘ ylamino)((1r,4r) (difluorornethoxy)cyclohexyla mino)pyrirnidine-5 - carboxamide —304_ ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 482 H: R Tb 4-(2-cyclopr0pylpr0pan 416.2 D *fiwfi ylamino)((1r,4r)(2,2,2- :3; a.“ trifluoroethoxy)cyclohexy1arnin LVMLCE o)pyrirnidinecarb0xarnide 23?: 483 374.2 D \ FT“ 2-((1r,4r)cyclopr0poxy- 3 3T3 ”11“‘3é‘“‘~']. exylamino)(2- :33 ropylpropan I L. 1‘ ylarnino)pyrirnidine i: carboxamide 484 4-(bicyclo[1.1.1]pentan 318.1 ylarnino)((1S,3 S) hydroxycyclohexylamino)- pyrimidine-S-carboxamide 485 4-(bicyclo[1.1.1]pentan 318.2 ylamino)((1R,3R) hydroxycyclohexylamino)- pyrimidine-S-carboxamide 486 4-(bicyclo[1.1.1]pentan 358.2 o)((1r,4r) cyclopropoxycyclohexylarnino) pyrimidine-S-carboxamide 487 4-(bicyclo[2.1.1]hexan 372.2 ylamino)((1r,4r)_4_ cyclopropoxycyclohexylarnino) pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 488 J 2-((1r,4r)—4- 362.3 D ethoxycyclohexylamino)(1 - methylcyclopentylamino)- dine-S-carboxamide 489 f 2-(3-tert- 388.4 D . butylbicyclo [1 . 1 . 1]pentan ylarnino)((1R,3R,4R) hydroxyrnethylcyclohexy1— : pyrimidine carboxamide 490 it? 2-(4-hydr0xy- 344 D fi?§«:§ bicyclo [2 .2. 1]heptan $2“ «3;; ylamino)—4-(3-rnethy1— sifikk‘éwa bic clo[1.1.1] entan-l- N; y p SEN“: ylarnino)pyrirnidine carboxamide 491 2-(tert-butylarnino) 322.3 D (2+: f T .. ((1R,3S,4S)hydroxy :J ‘Itf methylcyclohexylamino)- dine-S-carboxamide H z I if1 KW 492 jg; 2-((1r,4R) 404.2 D cyclopropoxycyclohexylarnino) ((1R,3R,4R)hydr0xy methylcyclohexylamino)- pyrimidine-S-carboxamide ATI—25 14175V1 Cmpd. Structure NAME MH+ Act.
A- Level 493 I“ 4-(3 -tert- 374.4 D butylbicyclo[1 . 1 .1]pentan ylamino)((1r,4r) hydroxycyclohexylamino)- pyrimidinecarb0xarnide 494 2-(ethy1amino)—4-((1R,3R,4R)- 294 D 3-hydr0xy methylcyclohexylamino)- pyrimidine-S-carboxamide 495 4-((1R,3 S)-3 - 308.2 hydroxycyclohexylarnino) (isobutylarnino)pyrimidine-5 - carboxamide 2-(sec-buty1arnino)((1R,3S)- 308.2 3 -hydroxycyc10hexy1arnino)— pyrimidine-S-carboxamide 497 2-(sec-buty1arnino)((1R,3S)- 308.2 3 xycyc10hexy1arnino)— pyrimidine-S-carboxamide 498 4-((1R,3 S)-3 - 322.2 hydroxycyclohexylarnino) (pentany1arnin0)pyrirnidine- oxarnide ATI—25 14175V1 Table2 Structure nd Name Level 2-(4-ethoxyphenylarnino) 376.2 ((1 S ,2S)—2- hydroxycyclopentylamino)- pyrimidine-S-carboxamide 4-(quin01inylamino) 365.2 (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - arnide 4-(1H-indaz01ylamino) (tetrahydro-ZH-pyran ylarnino)pyrirnidine-5 - carboxarnide 2-((1r,4r)—4- hydroxycyclohexylamino) (phenylarnino)pyrimidine-5 - carboxarnide 2-((1r,4r)—4- (methylsulfonamido)cyclohexyla mino)—4- (phenylarnino)pyrimidine carboxarnide ATI—ZS 14175V1 Cmpd Structure Compound Name MH+ Act.
B- Level 6 2-((1r,4r)—4- acetamidocyclohexylamino) (phenylarnino)pyrimidine arnide 7 2-((1r,4r)—4- aminocyclohexylarnino) (phenylarnino)pyrimidine-5 - carboxarnide 8 f" 2-((1r,4r)—4- (methylcarbarnoyl)cyclohexy1arn ino) (phenylarnino)pyrimidine carboxarnide 9 - .sé. ’ nylarnino)(piperidin ylarnino)pyrirnidine carboxarnide "1. - -. 2-(1-acety1piperidinylarnino)- 4-(phenylarnino)pyrirnidine carboxarnide a 4-(phenylarnino)—2-(tetrahydro- 2H-pyrany1arnino)pyrirnidine- -carboxarnide ATI—ZS 14175V1 2012/034349 Cmpd Structure Compound Name MH+ Act.
B- iLevel 12 s; ., 2-(tert-butylarnino) (phenylarnino)pyrimidine carboxarnide 13 2-(4- (diethylamino)phenylamino) (phenylarnino)pyrimidine carboxarnide . 4-(phenylarnino)(5,6,7,8- ' tetrahydronaphthalen ylarnino)pyrirnidine-5 - carboxarnide 2-(2-methoxyphenylarnino) (phenylarnino)pyrimidine carboxarnide 16 2—(6-methoxypyridin ylarnino) (phenylarnino)pyrimidine arnide 17 2-(3,4-dimethoxyphenylamino)- 4-(phenylamino)pyrimidine-5 - carboxarnide ATI—ZS 14175V1 WO 45569 Cmpd Structure Compound Name MH+ Act.
B- Level 18 2-(4-rnethoxy methylphenylamino) (phenylarnino)pyrimidine-5 - arnide 19 4-(phenylarnino)-2—(3 ,4,5- trimethoxyphenylarnino)pyrimid ine-S-carboxarnide 2-(2,4-dimethoxyphenylamino)- 4-(phenylarnino)pyrirnidine carboxarnide 21 ' ‘ 1 _:, ”5"“ 2-(4-rnethylpyrirnidin ylarnino)—4- (phenylarnino)pyrimidine-5 - carboxarnide 22 2-(2-phen0xyphenylamino) (phenylarnino)pyrimidine carboxarnide 23 2-(3,5-dimethoxyphenylarnino)- 4-(phenylarnino)pyrirnidine carboxarnide ATI—25 14175V1 Cmpd Structure Compound Name MH+ Act. 24 x ._ if“. I 2-(3-fluorophenylarnino) ' (phenylarnino)pyrimidine carboxarnide .A 1 _ 2-(3-methoxypheny1arnino) (phenylarnino)pyrimidine-5 - carboxarnide 26 “" ‘ 2-(3-ethoxyphenylarnino) " (phenylarnino)pyrimidine carboxarnide 27 2-(3-fluor0 ‘ : methoxyphenylamino) (phenylarnino)pyrimidine-5 - carboxarnide 28 1 ,J - - 4-(pheny1arnino)—2-(rntolylamino )pyrirnidine arnide 29 , 1“: 2-(4-methoxybiphenyl no) (phenylarnino)pyrimidine carboxarnide ATI—25 14175V1 Cmpd Structure nd Name MH+ Act.
B- Level ~ = ,4 _ 4-(phenylamino)(0- i E ‘ tolylamino)pyrirnidine carboxarnide 31 2-(2,3-dihydro-lH-inden-l- ylarnino) (phenylarnino)pyrimidine-5 - arnide 32 V 2-(5-acetamidO > " methoxyphenylamino) (phenylarnino)pyrimidine-5 - carboxarnide 33 .. 2-(5-rnethy1—1H-pyraz01—3- g ' ylarnino) (phenylarnino)pyrimidine carboxarnide 34 2-(5-rnethoxy a «t h methylphenylamino) (phenylarnino)pyrimidine carboxarnide 2-(cyclopropylmethylamino) (phenylarnino)pyrimidine carboxarnide ATI—ZS 14175V1 WO 45569 Cmpd Structure Compound Name MH+ Act.
B- Level 36 1. 2-(cyclopr0py1arnino) ‘ ' (phenylarnino)pyrimidine carboxarnide 37 =fi-:‘= 2-(4-carbamoylpiperidiny1) ' (phenylarnino)pyrimidine carboxarnide 38 2-(4-methoxyphenethylarnino)- 4-(phenylamino)pyrimidine-5 - carboxarnide 39 2-(3-fluor0benzylarnino)—4- (phenylarnino)pyrimidine carboxarnide g 2-(4-morpholinopheny1amino)- 4-(phenylarnino)pyrirnidine arnide 41 ' 2-(2-acetamidoethylamino)—4- (phenylarnino)pyrimidine carboxarnide —314— ATI—ZS 14175V1 Cmpd Structure Compound Name MH+ Act. 42 ‘33“ >5 4-(phenylarnino)(2-(thi0phen- 2-y1)ethy1arnino)pyrirnidine carboxarnide 43 a -dimethoxybenzylamino)- " " '" nylarnino)pyrirnidine carboxarnide 44 , , 2-(cyc10pentylamino)—4- (phenylarnino)pyrimidine carboxarnide 45 - i; .. 2-(cyclohexylamino) ‘ ‘ (phenylarnino)pyrimidine carboxarnide 46 V": 2-(SeC-butylarnino) ' (phenylarnino)pyrimidine carboxarnide 47 a ‘ 2-(4-rnethylcyclohexylamino) (phenylarnino)pyrimidine carboxarnide ATI—ZS 14175V1 Cmpd Structure Compound Name MH+ Act. 48 ethyl 4-(5 -carbarn0y1—4- lamino)pyrimidin ylamino)piperidine carboxylate 49 I -‘ 2-(cyclohexylmethylarnino) (phenylarnino)pyrimidine carboxarnide 50 2-(1 -benzy1piperidin no) (phenylarnino)pyrimidine carboxarnide 51 4-(phenylarnino)(pyridin ' ylmethylamino)pyrirnidine carboxarnide 52 F i ' ‘ 7“ 2-(3,5-difluorobenzy1amino)—4- “ (phenylarnino)pyrimidine carboxarnide 53 4-(phenylamino)(1- ' “'17. phenylethylamino)pyrimidine carboxarnide ATI—ZS 14175V1 2012/034349 Cmpd Structure Compound Name MH+ Act.
B- Level 54 v 3‘ 2—(2-ethoxybenzylamino) (phenylarnino)pyrimidine carboxarnide 55 2-(2,3-dirneth0xybenzy1arnino)- 4-(phenylarnino)pyrirnidine carboxarnide 56 9"“; 4-(phenylamino)(1- ' _»_~_-- phenylpropylamino)pyrimidine- -carboxarnide 57 2-(2,4-diflu0r0benzylarnino)—4- (phenylarnino)pyrimidine carboxarnide 58 .- 2-((2,2-dirnethy1—1,3 -di0x01an "" y1)rnethylarnino) (phenylarnino)pyrimidine-5 - carboxarnide 59 ethylbutylamino)—4- (phenylarnino)pyrimidine carboxarnide ATI—ZS 14175V1 WO 45569 Cmpd Structure Compound Name MH+ Act. 60 2-(2-rnethoxybenzylamino) " (phenylarnino)pyrimidine-5_ carboxarnide h 2-(2- (dimethylamino)ethy1arnino) (phenylarnino)pyrimidine carboxarnide 62 ' 1-rnethy1pyrrolidin ' " yl)ethylarnino) (phenylarnino)pyrimidine-5 - carboxarnide 63 :j 4-(pheny1arnino)(2-(pyridin- 2-y1)ethy1arnino)pyrirnidine carboxarnide 64 a- 4-(phenylarnino)_2_ ((tetrahydrofuran—zyl )rnethy1amin0)pyrimidine carboxarnide 65 ' 2-(furany1rnethylarnino) (phenylarnino)pyrimidine carboxarnide ATI—ZS 14175V1 Cmpd Structure nd Name MH+ Act. 66 a 2-(3,4- dimethoxyphenethylamino) (phenylarnino)pyrimidine-5 - carboxarnide 67 f 2-(4-fluor0benzylarnino)—4- (phenylarnino)pyrimidine carboxarnide 68 " f 4-(phenylarnino) (propylamino)pyrirnidine carboxarnide 69 2-(2-methoxyethylarnino) (phenylarnino)pyrimidine carboxarnide 70 F 2-(3,4-diflu0r0benzylarnino)—4- (phenylarnino)pyrimidine carboxarnide 71 nethylarnino) (phenylarnino)pyrimidine carboxarnide ATI—ZS 14175V1 2012/034349 Cmpd Structure Compound Name MH+ Act.
B- Level 72 :j 4-(phenylamino)(2- ‘ phenylpropylamino)pyrimidine- oxarnide 73 2-(benzylarnino) :1: (phenylarnino)pyrimidine carboxarnide 74 i"? 4-((1R,3S) hydroxycyclohexylamino) ((R)-1,2,3,4- tetrahydronaphthalen ylarnino)pyrirnidine-5 - carboxarnide 75 4-((1R,3S)—3- hydroxycyclohexylamino) ((S)-1,2,3,4- tetrahydronaphthalen ylarnino)pyrirnidine-5 - carboxarnide 76 4-((1R,3S)—3- hydroxycyclohexylamino) ((R)—5 ,6 ,7 , 8-tetrahydr0quinolin- 6-ylamino)pyrirnidine-5 - carboxarnide ATI—ZS 14175V1 Cmpd Structure Compound Name MH+ Act. 77 4-((1R,3S)—3- ycyclohexylamino) ((S)-5 ,6 ,7 , 8 -tetrahydr0quinolin- 6-ylamino)pyrirnidine-5 - carboxarnide V: -chrornany1amino) ((1 R,3 S)-3 - hydroxycyclohexylarnino)pyrirni dine-S-carboxarnide 5_ 2-((R)-chr0rnanylamino) ‘ " ((1R,3S) hydroxycyclohexylarnino)pyrirni dine-S-carboxarnide 80 (S)(isopropy1amino) (5,6,7,8-tetrahydroquinolin ylarnino)pyrirnidine-5 - carboxarnide 81 (R)(isopropylarnino) (5,6,7,8-tetrahydroquinolin ylarnino)pyrirnidine-5 - carboxarnide 82 (S)(isopr0pylarnino) (1 ,2,3 ,4-tetrahydr0naphthalen ylarnino)pyrirnidine-5 - carboxarnide ATI—ZS 14175V1 2012/034349 Cmpd Structure Compound Name MH+ Act.
B- Level 83 (R)(isopropylarnino) (1 ,2,3 ,4-tetrahydr0naphthalen ylarnino)pyrirnidine carboxarnide N 4-((1R,3S)—3- hydroxycyclohexylamino) ((R)-1,2,3,4- tetrahydronaphthalen-Z- ylarnino)pyrirnidine carboxarnide _: 4-((1R,3S)—3- I 8 hydroxycyclohexylamino) ((S)-1,2,3,4- tetrahydronaphthalen-Z- ylarnino)pyrirnidine-5 - arnide 86 j : 2-(cyclohexylamino) (phenylarnino)pyrimidine-5 - carbonitrile 87 4-((1R,3S)—3- hydroxycyclohexylamino) ((S)-5 ,6 ,7 , 8 -tetrahydr0quinolin- 7-ylamino)pyrirnidine-5 - carboxarnide ATI—ZS 14175V1 2012/034349 Cmpd Structure Compound Name MH+ Act.
B- Level 88 ,3S)—3- hydroxycyclohexylamino) ((R)—5 ,6 ,7 , 8-tetrahydr0quinolin- 7-ylamino)pyrirnidine-5 - carboxarnide 89 2-((R)(4- . . . ‘ ‘ :3. fluoropheny1)ethylarnino) ((1 R,3 S)-3 - hydroxycyclohexylarnino)pyrirni dine-S-carboxarnide 90 —1-(4- _ ' . . 3‘ fluoropheny1)ethylarnino) ((1R,3S) hydroxycyclohexylarnino)pyrirni dine-S-carboxarnide 91 (S)(isopr0pylarnino) (5,6,7,8-tetrahydroisoquinolin ylarnino)pyrirnidine-5 - carboxarnide 92 (R)(isopropylarnino) (5,6,7,8-tetrahydroisoquinolin ylarnino)pyrirnidine-5 - carboxarnide 93 4-((1R,3R,4R)hydroxy methylcyclohexylarnino)((S)- ,6,7,8-tetrahydroquinolin ylarnino)pyrirnidine-5 - carboxarnide ATI—ZS 14175V1 Cmpd Structure Compound Name MH+ Act.
B- Level 94 4-((1R,3R,4R)hydroxy methylcyclohexylamino)((R)- ,6,7,8-tetrahydroquinolin ylarnino)pyrirnidine-5 - carboxarnide 95 . 4-((1R,3S)—3- ”h g“ hydroxycyclohexylamino) ,6,7,8- tetrahydroisoquinolin ylarnino)pyrirnidine carboxarnide ‘ 4-((1R,3S)—3- ' V ' ' hydroxycyclohexylamino) ((R)-5,6,7,8- tetrahydroisoquinolin ylarnino)pyrirnidine-5 - carboxarnide 97 4-((1R,3S)—3- hydroxycycloheptylarnino) ((S)-5 ,6 ,7 , 8 -tetrahydr0quinolin- 6-ylamino)pyrirnidine-5 - carboxarnide 98 4-((1R,3S)—3- hydroxycycloheptylarnino) ((R)—5 ,6 ,7 , ahydr0quinolin- ino)pyrirnidine-5 - carboxarnide —324— ATI—ZS 14175V1 Cmpd. Structure Compound Name MH+ Act.
Level lohexy1arnino)((1r,4r)- 344.2 \x La, “z” 4-ethoxycyclohexyl- LMLg -"L"\‘n K“: amino)pyrirnidine-5 -carb0nitrile

Claims (65)

What is claimed
1. A compound of formula (I): and ceutically acceptable salts, tautomers, isomers, enantiomers, and isotopologues f, wherein: R1 is substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl, substituted or unsubstituted alkylcycloalkyl, or substituted or unsubstituted alkylheterocyclyl, provided that R1 is not 1-aminocyclohexyl; R2 is substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted saturated cycloalkyl, substituted or unsubstituted alkylcycloalkyl, or substituted or unsubstituted non-aromatic heterocyclyl; provided the compound is not minoethylamino)(methylamino)pyrimidinecarboxamide, 2-(2-aminopropylamino)(cyclohexylamino)pyrimidinecarboxamide, 2-(2-aminooxoethylamino)(cyclohexylamino)pyrimidinecarboxamide, 2-(2-aminoethylamino)(cyclohexylamino)pyrimidinecarboxamide, (S)(2-aminopropylamino)(cyclobutylamino)pyrimidinecarboxamide, (R)(1-aminomethyloxobutanylamino)(cyclobutylamino)pyrimidine carboxamide, lopentylamino)(methylamino)pyrimidinecarboxamide, or 2-(1-acetylpiperidinylamino)(cyclopropylamino)pyrimidinecarboxamide, wherein when a C1-8 alky group is substituted, the C1-8 alkyl group is tuted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, amine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, ocyanate, e, thiocyanate, , or O(alkyl)aminocarbonyl; wherein when a group, other than a C1-8 alkyl group, is substituted, the group is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, her, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, de, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
2. The nd of claim 1, wherein R1 is a branched C1-8 alkyl.
3. The nd of claim 2, wherein R1 is isopropyl, sec-butyl, isobutyl, tert-butyl, 2,3-dimethylbutyl, isopentyl, 2-methylpentyl, neopentyl, tert-pentyl, or 3-methylpentyl.
4. The compound of claim 1, wherein R1 is a substituted or tituted cycloalkyl, wherein when the cycloalkyl is substituted, the cycloalkyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, yl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, lkyl, or a heterocyclyl.
5. The compound of claim 4, wherein the cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, o[1.1.1]pentyl, bicyclo[2.2.1]heptyl, or bicyclo[2.2.2]octyl.
6. The compound of claim 4, wherein the cycloalkyl is substituted with one or more halogen, -CF3, -(C1-4 , -(C1-6 cycloalkyl), -NR2, -(C0-3 alkyl)OR, -(C0-3 alkyl)OR”, -NRC(O)R’, -C(O)R’, -C(O)NR2, -C(O)OR’, or -NRS(O)2R’, n each R is independently H, or C1-4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated, and each R” is independently a C1-6 cycloalkyl, n the C1-6 cycloalkyl is optionally nated.
7. The compound of claim 4, wherein the cycloalkyl is substituted with one or more methyl, ethyl, t-butyl, cyclopropyl, -CF3, -F, -OH, -OCH3, -OCHF2, -OCF3, - OCH2CH3, -OCH2CH2F, F3, -O(cyclopropyl), -CH2OH, -CH2OCH3, -C(CH3)2OH, -NH2, -NH(CH3), -NHC(O)CH3, -C(O)NHCH3, -C(O)N(CH3)2, or -NHSO2CH3.
8. The compound of claim 1, wherein R1 is a substituted or unsubstituted nonaromatic heterocyclyl, wherein when the heterocyclyl is substituted, the heterocyclyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, mino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, e, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, e, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, e, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
9. The compound of claim 8, wherein the heterocyclyl is oxetanyl, pyrrolidinyl, tetrahydropyranyl, 1,4-dioxaspiro[4.5]decanyl, or piperidyl.
10. The compound of claim 9, n the piperidyl is substituted with -C(O)R’, or -C(O)OR’, wherein R’ is C1-4 alkyl, wherein the alkyl is optionally fluorinated.
11. The compound of claim 1, wherein R1 is a substituted or unsubstituted alkylcycloalkyl, wherein when the alkylcycloalkyl is substituted, the ycloalkyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, ino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, amide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
12. The compound of claim 11, wherein R1 is (C1-3 alkyl)cyclopropyl, (C1-3 alkyl)cyclobutyl, (C1-3 alkyl)cyclopentyl, or (C1-3 alkyl)cyclohexyl.
13. The nd of claim 11, wherein R1 is –(CH2)cyclopropyl, -CH(CH3)cyclopropyl, -CH(CH3)cyclobutyl,–CH(CH3)cyclohexyl, or -C(CH3)2cyclopropyl.
14. The compound of claim 1, wherein R1 is a substituted or unsubstituted alkylheterocyclyl, wherein when the alkylheterocyclyl is substituted, the eterocyclyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, rbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
15. The compound of claim 14, wherein R1 is -(C1-4 tetrahydrofuranyl, -(C1-4 alkyl)dioxolanyl, (C1-4 alkyl)furanyl, (C1-3 thiophenyl, or -(C1-3 alkyl)pyridyl.
16. The compound of claim 1, wherein R2 is a tuted or unsubstituted C1-8 alkyl, wherein when the C1-8 alky group, excluding cycloalkyl, heteroaryl and aryl groups, is tuted, the C1-8 alkyl group is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, B(OH)2, or O(alkyl)aminocarbonyl.
17. The compound of claim 16, wherein R2 is methyl, ethyl, n-propyl, isopropyl, n-butyl, tyl, isobutyl, tert-butyl, 3-methylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 2,3,3-trimethylbutyl, tert-pentyl, isopentyl, 3-pentyl, 3-methylpentyl, 2-methylpentyl, or 2,4-dimethylpentyl.
18. The compound of claim 16, wherein R2 is substituted with one or more -(C1-4 alkyl), -(C0-3 alkyl)OR, -C(O)NR2 or -NRCOR’, wherein each R is independently H, or C1-4 alkyl, wherein the alkyl is optionally nated, and each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated.
19. The compound of claim 16, wherein R2 is substituted with one or more -OH, -OCH3, or -CH3.
20. The compound of claim 1, wherein R2 is a substituted or unsubstituted cycloalkyl, wherein when the cycloalkyl is substituted, the cycloalkyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, y, nitro, cyano, thiol, thioether, imine, imide, amidine, ine, enamine, aminocarbonyl, acylamino, onato, phosphine, thiocarbonyl, yl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, one, azide, isocyanate, isothiocyanate, cyanate, anate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
21. The compound of claim 20, wherein R2 is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[1.1.1]pentyl, o[2.1.1]hexyl, or bicyclo[2.2.1]heptyl.
22. The compound of claim 20, wherein R2 is substituted with one or more -CF3, -(C1-4 alkyl), -(C1-6 cycloalkyl)-(C0-3 alkyl)OR, -(C0-3 alkyl)C(O)NR2, -NR2, or -NRCOR’, n each R is independently H, or C1-4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated, and wherein the cycloalkyl is optionally fluorinated.
23. The compound of claim 20, wherein R2 is substituted with one or more methyl, ethyl, isopropyl, -cyclopropyl, -CF3, -CH2OH, -OH, -OCH3, -OCH2CH3, -C(O)NH2, -NHC(O)CH3, or -NHC(O)CH2CH3.
24. The compound of claim 1, n R2 is a substituted or unsubstituted ycloalkyl, wherein when the alkylcycloalkyl is substituted, the alkylcycloalkyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, ne, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, ine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, lkyl, or a heterocyclyl.
25. The compound of claim 24, wherein R2 is a tuted or unsubstituted (C1-3 alkyl)cyclopropyl, (C1-3 cyclobutyl, (C1-3 alkyl)cyclopentyl, or (C1-3 alkyl)cyclohexyl, wherein when the R2 is substituted, the R2 is substituted with halogen, alkyl, hydroxyl, alkoxy, alkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, e, aminocarbonyl, acylamino, onato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, ine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
26. The compound of claim 24, wherein R2 is -(CH2)cyclopropyl, -(CH2)cyclobutyl, -CH(CH3)cyclopropyl, -CH(CH3)cyclobutyl, -CH(CH2CH3)cyclopropyl, -C(CH3)2cyclopropyl, or -CH2CH2cyclobutyl.
27. The compound of claim 1, wherein R2 is a substituted or unsubstituted non-aromatic heterocyclyl, wherein when the heterocyclyl is substituted, the heterocyclyl is substituted with halogen, alkyl, yl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, ino, phosphonato, phosphine, rbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, anate, oxo, B(OH)2, O(alkyl)aminocarbonyl, lkyl, or a heterocyclyl.
28. The compound of claim 27, wherein R2 is oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, piperidyl, piperidinonyl or oxaspiro[4.5]decanyl.
29. The compound of claim 27, wherein R2 is tuted with one or more -(C1-4 alkyl), -(C0-3 alkyl)OR, or -C(O)R’, wherein each R is independently H, or C1-4 alkyl, wherein the alkyl is optionally fluorinated, and each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated.
30. The compound of claim 20, wherein R1 is a cycloalkyl, optionally substituted with one or more halogen, -CF3, -(C1-4 alkyl), -NR2, -(C0-3 alkyl)OR, -(C0-3 OR”, -NRC(O)R’, -C(O)R’, -C(O)NR2, -C(O)OR’, or -NRS(O)2R’, wherein each R is independently H, or C1-4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated, and each R” is independently a C1-6 cycloalkyl, wherein the cycloalkyl is optionally fluorinated.
31. The nd of claim 1, wherein the compound at a concentration of 10 μM ts JNK1 by at least about 50%.
32. The compound of claim 1, wherein the compound is selected from or a pharmaceutically able salt, tautomer, stereoisomer, or enantiomer thereof.
33. A compound selected from or a ceutically able salt, tautomer, isomer, or enantiomer thereof.
34. A compound of formula (IB): (IB) and pharmaceutically acceptable salts, tautomers, stereoisomers, enantiomers, and isotopologues thereof, wherein: R3 is substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted non-aromatic heterocyclyl, substituted or unsubstituted alkylcycloalkyl, or substituted or unsubstituted alkyl-(non-aromatic heterocyclyl); R4 is substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted non-aromatic cyclyl; provided the compound is not 4-(isopentylamino)(3-(2-methylpiperidinyl)propylamino)pyrimidinecarbonitrile; (2S,2'S)-dimethyl 2,2'-(5-cyanopyrimidine-2,4-diyl)bis(azanediyl)bis(4- methylpentanoate); (2S,2'S)-diethyl 2,2'-(5-cyanopyrimidine-2,4-diyl)bis(azanediyl)bis(3-methylbutanoate); 4-(cycloheptylamino)(3-(2-methylpiperidinyl)propylamino)pyrimidine carbonitrile; 4-(4-methylcyclohexylamino)(3-(2-methylpiperidinyl)propylamino)pyrimidine carbonitrile; or 2-(3-(diethylamino)propylamino)(4-methylcyclohexylamino)pyrimidinecarbonitrile, wherein when a C1-8 alky group is substituted, the C1-8 alkyl group is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, y, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, rbonyl, sulfonyl, e, sulfonamide, ketone, aldehyde, ester, urea, ne, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, ocyanate, e, thiocyanate, B(OH)2, or O(alkyl)aminocarbonyl; wherein when a group, other than a C1-8 alky group, is substituted, the group is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, e, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, xyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a cyclyl.
35. The compound of claim 34, wherein R3 is a branched C1-8 alkyl.
36. The compound of claim 35, wherein R3 is isopropyl, sec-butyl, isobutyl, tert-butyl, 2,3-dimethylbutyl, isopentyl, 2-methylpentyl, neopentyl, tert-pentyl, or 3-methylpentyl.
37. The compound of claim 34, wherein R3 is a substituted or unsubstituted cycloalkyl, wherein when the cycloalkyl is substituted, the lkyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
38. The compound of claim 37, wherein the cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[1.1.1]pentyl, bicyclo[2.2.1]heptyl, or bicyclo[2.2.2]octyl.
39. The compound of claim 37, wherein the cycloalkyl is substituted with one or more halogen, -CF3, -(C1-4 alkyl), -(C1-6 cycloalkyl), -NR2, -(C0-3 alkyl)OR, -(C0-3 alkyl)OR”, -NRC(O)R’, -C(O)R’, -(C0-3 C(O)NR2, -C(O)OR’, or -NRS(O)2R’, n each R is independently H, or C1-4 alkyl, n the alkyl is optionally fluorinated, and each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated, and each R” is independently a C1-6 cycloalkyl, wherein the cycloalkyl is optionally fluorinated.
40. The compound of claim 37, wherein the lkyl is substituted with one or more methyl, ethyl, t-butyl, ropyl, -CF3, -F, -OH, -OCH3, -OCHF2, -OCF3, -OCH2CH3, -OCH2CH2F, -OCH2CF3, -O(cyclopropyl), -CH2OH, -CH2OCH3, )2OH, -NH2, -NH(CH3), -NHC(O)CH3, )2C(O)N(CH3)2, -C(O)NHCH3, -C(O)N(CH3)2, or -NHSO2CH3.
41. The compound of claim 34, wherein R3 is a substituted or unsubstituted non-aromatic heterocyclyl, wherein when the heterocyclyl is substituted, the heterocyclyl is tuted with n, alkyl, hydroxyl, alkoxy, alkyl, amino, mino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, amide, ketone, aldehyde, ester, urea, ne, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, e, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
42. The compound of claim 41, wherein the cyclyl is oxetanyl, tetrahydrofuranyl, ydropyranyl, piperidyl, piperidinonyl or 1,4-dioxaspiro[4.5]decanyl.
43. The compound of claim 34, wherein R4 is a substituted or unsubstituted C1-8 alkyl, wherein when the C1-8 alky group, excluding cycloalkyl, heteroaryl and aryl groups, is substituted, the C1-8 alkyl group is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, her, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, e, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, amine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, nate, isothiocyanate, cyanate, thiocyanate, B(OH)2, or O(alkyl)aminocarbonyl.
44. The compound of claim 43, wherein R4 is methyl, ethyl, yl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, 3-methylbutyl, 2,3-dimethylbutyl, 3,3-dimethylbutyl, 2,3,3-trimethylbutyl, tert-pentyl, isopentyl, 3-pentyl, 3-methylpentyl, 2-methylpentyl, or 2,4-dimethylpentyl.
45. The compound of claim 34, wherein R4 is a substituted or unsubstituted lkyl, wherein when the cycloalkyl is substituted, the cycloalkyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, ine, e, aminocarbonyl, acylamino, phosphonato, phosphine, thiocarbonyl, sulfonyl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
46. The compound of claim 45, wherein R4 is cyclopropyl, utyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, bicyclo[1.1.1]pentyl, bicyclo[2.1.1]hexyl, or bicyclo[2.2.1]heptyl.
47. The compound of claim 45, wherein R4 is substituted with one or more halogen, -CF3, -(C1-4 alkyl), -(C1-6 cycloalkyl), -(C0-3 alkyl)OR, -(C0-3 alkyl)C(O)NR2, -NR2, or -NRCOR’, wherein each R is independently H, or C1-4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated, and wherein the cycloalkyl is optionally fluorinated.
48. The compound of claim 45, wherein R4 is tuted with one or more methyl, ethyl, isopropyl, -cyclopropyl, -CF3, -CH2OH, -OH, -OCH3, -OCH2CH3, -C(O)NH2, -NHC(O)CH3, or -NHC(O)CH2CH3.
49. The compound of claim 34, wherein R4 is a substituted or unsubstituted non-aromatic heterocyclyl, wherein when the cyclyl is substituted, the heterocyclyl is substituted with halogen, alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxy, nitro, cyano, thiol, thioether, imine, imide, amidine, guanidine, enamine, aminocarbonyl, acylamino, onato, phosphine, rbonyl, yl, sulfone, sulfonamide, ketone, aldehyde, ester, urea, urethane, oxime, hydroxyl amine, alkoxyamine, xyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, e, thiocyanate, oxo, B(OH)2, O(alkyl)aminocarbonyl, cycloalkyl, or a heterocyclyl.
50. The compound of claim 49, wherein R4 is oxetanyl, tetrahydrofuranyl, ydropyranyl, piperidyl, piperidinonyl or 1,4-dioxaspiro[4.5]decanyl.
51. The compound of claim 45, wherein R3 is a cycloalkyl, optionally substituted with one or more halogen, -CF3, -(C1-4 alkyl), -NR2, -(C0-3 alkyl)OR, -(C0-3 alkyl)OR”, )R’, ’, -C(O)NR2, -C(O)OR’, or -NRS(O)2R’, wherein each R is independently H, or C1-4 alkyl, wherein the alkyl is optionally fluorinated, each R’ is independently C1-4 alkyl, wherein the alkyl is optionally fluorinated, and each R” is independently a C1-6 cycloalkyl, wherein the lkyl is optionally fluorinated.
52. The compound of claim 34, wherein the compound at a concentration of 10 μM inhibits JNK1 by at least about 50%.
53. The compound of claim 34, wherein the compound is selected from or a pharmaceutically acceptable salt, tautomer, stereoisomer, or enantiomer thereof.
54. A pharmaceutical composition comprising an ive amount of a compound of claim 1, 32, 33, 34, or 53, or a pharmaceutically acceptable salt, tautomer, isotopologue, or stereoisomer thereof, and a pharmaceutically acceptable carrier, excipient or vehicle.
55. An in vitro method of inhibiting a JNK1 or JNK2 kinase in a cell expressing said JNK1 or JNK2 kinase, comprising contacting said cell with an effective amount of a compound of claim 1, 32, 33, 34, or 53, or a pharmaceutically acceptable salt, tautomer, isotopologue, or stereoisomer thereof.
56. The use of a compound of any one of claims 1 to 53, in the manufacture of a medicament for inhibiting a JNK1 or JNK2 .
57. The use of a compound of any one of claims 1 to 53 in the manufacture of a medicament for the ent of a liver ic disorder, or diabetes or metabolic syndrome leading to a liver fibrotic disorder.
58. The use of claim 57, wherein the liver fibrotic disorder is non-alcoholic steatohepatitis, steatosis, cirrhosis, primary sclerosing gitis, primary biliary cirrhosis, tis, hepatocellular oma, or liver fibrosis coincident with chronic or repeated alcohol ingestion, with infection, with liver transplant, or with drug induced liver injury.
59. The use of claim 57, wherein the liver fibrotic disorder is non-alcoholic steatohepatitis, steatosis, hepatitis, or cirrhosis.
60. A compound according to claim 1, ntially as herein described or exemplified.
61. A compound according to claim 34, substantially as herein described or ified.
62. A pharmaceutical composition according to claim 54, substantially as herein described or exemplified.
63. A method according to claim 55, substantially as herein described or exemplified.
64. A use according to claim 56, substantially as herein bed or exemplified.
65. A use according to claim 57, substantially as herein described or exemplified.
NZ614776A 2011-04-22 2012-04-20 Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith NZ614776B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161478076P 2011-04-22 2011-04-22
US61/478,076 2011-04-22
US201161555339P 2011-11-03 2011-11-03
US61/555,339 2011-11-03
PCT/US2012/034349 WO2012145569A1 (en) 2011-04-22 2012-04-20 Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith

Publications (2)

Publication Number Publication Date
NZ614776A NZ614776A (en) 2015-06-26
NZ614776B2 true NZ614776B2 (en) 2015-09-29

Family

ID=

Similar Documents

Publication Publication Date Title
US11325890B2 (en) Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
AU2015201030B2 (en) Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
AU2016244228B2 (en) Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith
NZ614776B2 (en) Substituted diaminocarboxamide and diaminocarbonitrile pyrimidines, compositions thereof, and methods of treatment therewith