HK1191328A - 1,4-oxazepines as bace1 and/or bace2 inhibitors - Google Patents
1,4-oxazepines as bace1 and/or bace2 inhibitors Download PDFInfo
- Publication number
- HK1191328A HK1191328A HK14104402.8A HK14104402A HK1191328A HK 1191328 A HK1191328 A HK 1191328A HK 14104402 A HK14104402 A HK 14104402A HK 1191328 A HK1191328 A HK 1191328A
- Authority
- HK
- Hong Kong
- Prior art keywords
- compound
- formula
- disease
- methyl
- tetrahydro
- Prior art date
Links
Description
Background
Alzheimer's Disease (AD) is a neurodegenerative disease of the central nervous system and leads to the development of progressive dementia in the middle-aged and older population. The clinical symptoms are impairment of memory, cognition, temporal and local orientation, judgment and reasoning, and severe emotional disturbances. There is currently no effective treatment that can prevent the disease or its progression or stably reverse its clinical symptoms. AD is a major health problem in all societies with a high life expectancy and also becomes a significant economic burden for these societal health systems.
AD is characterized by 2 major pathological features in the Central Nervous System (CNS): the appearance of amyloid plaques and neurofibrillary tangles (Hardy et al, The amyloid hypothesised amyloid Alzheimer's disease: progress and protocols on The road to therapy (amyloid hypothesis of Alzheimer's disease: progress and problem to The route to therapy), science.2002Jul 19; 297 (5580): 353-6, Selkoe, Cell biology of The amyloid beta-protein precursor and The mechanismith of Alzheimer's disease, Annu Rev Cell I.1994; 10: 373 403). Both pathological features are also commonly found in patients with down syndrome (trisomy 21), which also exhibit AD-like symptoms at an early stage. Neurofibrillary tangles are intracellular aggregates of microtubule-associated protein tau (MAPT). Amyloid plaques occur in the extracellular space; their main component is the a β -peptide. The latter is a group of proteolytic fragments derived from the β -Amyloid Precursor Protein (APP) via a series of proteolytic cleavage steps. Several forms of APP have been identified, the largest of which are proteins of 695, 751 and 770 amino acids in length. They are all produced from one gene via different splicing. A β -peptides are derived from the same domain as APP but differ at their N-and C-termini, with the main species being 40 and 42 amino acids in length. There are several chains of evidence that strongly suggest that aggregated a β -peptide is an essential molecule in the pathogenesis of AD: 1) amyloid plaques formed by Α β -peptide are an invariant part of AD pathology; 2) a β -peptide is toxic to neurons; 3) in Familial Alzheimer's Disease (FAD), mutations in the causative genes APP, PSN1, PSN2 result in increased levels of a β -peptide and early cerebral amyloidosis; 4) transgenic mice expressing this FAD gene exhibit pathology with many similarities to human disease. The A β -peptide is produced by APP through the sequential action of 2 proteolytic enzymes named β -and γ -secretases. The β -secretase enzyme first cleaves the extracellular domain of APP of approximately 28 amino acids outside the transmembrane domain (TM) to generate a C-terminal fragment of APP containing the TM-and cytoplasmic domains (CTF β). CTF β is a substrate for γ -secretase, which cleaves at several adjacent positions within the TM to produce a β peptide and cytoplasmic fragments. Gamma-secretase is a complex of at least 4 different proteins whose catalytic subunits closely resemble presenilin proteins (PSEN1, PSEN 2). Beta-secretase (BACE1, Asp 2; BACE stands for Beta-site APP lyase) is an aspartyl protease anchored in the membrane via a transmembrane domain (Vassar et al, Beta-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. Beta. -secretase cleavage of the Alzheimer's amyloid precursor protein), science.199Oct 22; 286 (5440): 735). It is expressed in many tissues of the human body, but its levels are particularly high in the CNS. The gene ablation of the BACE1 gene in Mice clearly shows that its activity is important for APP processing leading to the production of A β -peptide, without the production of BACE1 (Luo et al, Mice discovery in BACE1, the Alzheimer's beta-secretase, havingnormal phenotypy and aborolished beta-amyloid generation (Alzheimer's beta-secretase, BACE1 deficient Mice have normal phenotype and eliminated β -amyloid production), Nat neurosci.2001Mar, (4): 231-2, Roberss et al, knockmicoe allegial cleavage of the primary beta-secretase activity in brans: polypeptides for Alzheimer's disease, BACE expression in Mice, although the basal secretion of BACE in Mice is a healthy brain-knockout method of the enzyme activity of JP-10. A-peptide-10 (this is a basic therapeutic method for the brain-secretion of BACE 3512). Mice that express the human APP gene by genetic engineering and form extensive amyloid plaques and Alzheimer's disease-like pathology during senescence no longer exhibit this when β -secretase activity is reduced by gene ablation of one of the BACE1 alleles (McConlogue et al, Partial reduction of BACE1 hadramatic effects on Alzheimer's disease and synthetic pathology in APPtransgenic Mice (Partial reduction of BACE1has a profound effect on Alzheimer's plaques and synaptic pathology in APP transgenic Mice) J Biol Chem.2007 Sep7; 282 (36): 26326). It is therefore speculated that inhibitors of BACE1 activity may be agents useful in the therapeutic intervention of AD.
It is therefore an object of the present invention to provide selective BACE1 inhibitors with enhanced therapeutic and pharmacological properties compared to compounds known in the art. Such compounds are useful as therapeutically active substances, especially in the control or prevention of alzheimer's disease. Furthermore, the formation, or formation and deposition, of β -amyloid plaques in, on or around neural tissue (e.g., the brain) is inhibited by such compounds by blocking the production of a β from APP or an APP fragment.
Type II diabetes (T2D) is caused by insulin resistance and insufficient insulin secretion from pancreatic β -cells resulting in poor glycemic control and hyperglycemia (M Prentki and CJ Nolan, "Islet beta-cell failure in type2diabetes mellitus". j.clin.investig.2006, 116(7), 1802-. Patients with T2D are at increased risk for microvascular and macrovascular disease and a variety of associated complications, including diabetic nephropathy, retinopathy and cardiovascular disease. In 2000, it was estimated that one hundred million people had the disease, and it was expected that this number will double by 2030 (S Wild, G Roglic, A Green, R.Sicree and H King, "Global prediction of Diabetes", Diabetes Care2004, 27(5), 1047-. The rise in the incidence of T2D is associated with an increasing sedentary lifestyle and high energy food intake in the world population (P Zimmet, KGMM Alberti and J Shaw, "Global and social interventions of the diabetes epidemic" Nature2001, 414, 782- "787).
Beta cell failure and the resulting marked decrease in insulin secretion and hyperglycemia marked the onset of T2D. Most of the current treatments do not prevent the loss of the amount of beta cells characteristic of dominant T2D. However, recent studies with the GLP-1 analog gastrin and other drugs have shown that it is possible to achieve preservation and proliferation of beta cells, leading to improvement in glucose tolerance and slowing down progression to dominant T2D (LL Baggio and DJ Drucker, "Therapeutic approaches to preservesilet mass in type2diabetes (Therapeutic measures for preserving islet mass in type II diabetes)", annu.
Tmem27 has been identified as a protein that promotes beta Cell proliferation (P Akpinar, S Kuwajima, JKrultzfeldt, M Stoffel, "Tmem 27: A cleaned and plated membrane proteins across cultures promotion (Tmem 27: cleaved and shed plasma membrane proteins that stimulate pancreatic islet beta-Cell proliferation)," Cell Metab.2005, 2, 385-. Tmem27 is a 42kDa membrane glycoprotein constitutively shed from the surface of beta cells, degraded from full-length cells Tmem 27. Over-expression of Tmem27 in transgenic mice increased beta-cell mass and improved glucose tolerance in a diet-induced obesity DIO model of diabetes. Furthermore, siRNA knock-out of Tmem27 in rodent beta cell proliferation assays (e.g., using INS1e cells) reduced the proliferation rate, indicating a role for Tmem27 in controlling beta-cell mass.
In the same proliferation assay, BACE2 inhibitors also increased proliferation. However, BACE2 inhibition combined with Tmem27siRNA knockdown resulted in lower proliferation rates. Therefore, BACE2 was concluded to be a protease associated with degradation of Tmem 27. Furthermore, BACE2 cleaves peptides based on the Tmem27 sequence in vitro. The closely related protease BACE1 does not cleave this peptide and selective inhibition of BACE1 alone does not enhance beta cell proliferation.
The proximal homolog BACE2is a membrane-bound aspartyl protease and is co-localized with Tmem27 in beta cells of the human pancreas (G Finzi, F Franzi, C Placidi, F Acquati et al, "BACE 2is stored in secretory granules of mouse and rat pancreatic beta cells (BACE2 stored in secretory granules of mouse and rat pancreatic beta cells)", Ultrasucctpethol.2008, 32(6), 246-. It is also known to degrade APP (I Hussain, D Powell, D Howlett, G Chapman et al, "ASP 1(BACE2) cleaves the amyloid precursor protein at the beta-secretase site (ASP1(BACE2) cleaves amyloid precursor protein at the beta-secretase site)" Mol Cell neurosci.2000, 16, 609-619 "), IL-1R2(P Kuhn, EMarjaux, A Imhof, B De Strooper et al," Regulated transmembrane protein catalysis of the interleukin-1receptor by alpha-, beta-and gamma-secretase) "intramembrane proteolysis of interleukin-1receptor J.biol.Chem.2007, 11982), (11982). The ability to degrade ACE2 suggests a possible role for BACE2 in hypertension control.
Therefore, inhibition of BACE2 has been proposed as a treatment for T2D, taking advantage of the potential to preserve and restore β -cell mass and stimulate insulin secretion in pre-diabetic and diabetic patients. It is therefore an object of the present invention to provide selective BACE2 inhibitors with enhanced therapeutic and pharmacological properties compared to compounds known in the art. Such compounds are useful as therapeutically active substances, especially for the treatment and/or prevention of diseases which are associated with the inhibition of BACE2, such as type II diabetes.
In addition, the formation or formation and deposition of β -amyloid peptide in, on or around neural tissue (e.g., brain) is inhibited by the compounds of the present invention, i.e., inhibiting production of a β from APP or an APP fragment.
Furthermore BACE1 and/or BACE2 inhibitors may be useful in the treatment of the following diseases: IBM (Inclusion body myositis) (Vattemi G. et al, Lancet)2001Dec 8; 358(9297): 1962-4), Down's syndrome (Barbiero L. et al, Exp neurol.2003Aug; 182(2): 335-45), Wilson's disease (Sugimoto I. et al, J Biol chem.2007Nov30; 282(48): 34896-903), Whipple's disease (Desnues B. et al, Clin Vaccine Immunol.2006Feb; 13(2): 170-8), spinocerebellar ataxia type 1 and spinocerebellar ataxia type 7 (Gatchel j.r. et al, Proc Natl Acad Sci USA2008Jan 29; 105(4): 1291-6), dermatomyositis (Greenberg S.A. et al, Ann neurol.2005Can; 57(5): 664-78 and Greenberg S.A. et al, Neurol2005 Can; 57(5): 664-78), Kaposi's sarcoma (Lagos D. et al, Blood, 2007Feb 15; 109(4): 1550-8), glioblastoma multiforme (E-MEXP-2576,http://www.ebi.ac.uk/microarray-as/aer/resultqueryFor= PhysicalArrayDesign&aAccession=AMEXP-258), rheumatoid arthritis (ungenthem u. et al, GSE2053), amyotrophic lateral sclerosis (Koistinen h. et al, muscelenerve.2006 oct; 34(4): 444-50 and Li Q.X. et al, Aging cell.2006Apr; 5(2): 153-65), Huntington's disease (Kim Y.J. et al, Neurobiol Dis.2006Can; 22(2): 346-56. Epub2006Jan19 and Hodges a. et al, Hum Mol genet.2006mar15; 15(6): 965-77. Epub2006Feb8), Multiple myeloma (Multiple myeloma) (kiharay et al, Proc Natl Acad Sci U S a.2009dec22; 106(51): 21807-12), malignant melanoma (Talantov D. et al, Clin Cancer Res.2005Oct 15; 11(20): 7234-42), sjogren's syndrome (Basset C. et al, Scand J Immunol.2000Mar; 51(3): 307-11), lupus erythematosus (Grewal p.k. et al, Mol Cell biol.2006, Jul; 26(13): 4970-81), macrophage myofascitis (Macrophagic myofasciitis), juvenile idiopathic arthritis, granulomatous arthritis, breast Cancer (Hedlund M. et al, Cancer Res.2008Jan 15; 68(2): 388-94 and Kondoh k. et al, break Cancer Res treat.2003mar; 78(1): 37-44), gastrointestinal disease (Hoffmeister A. et al, JOP.2009Sep4; 10(5): 501-6), autoimmune/inflammatory diseases (Woodard-Grice a.v. et al, J Biol chem.2008sep26; 283(39): 26364-73.Epub2008Jul23), rheumatoid arthritis (to egel s. et al, Osteoarthritis cartilage.2010feb; 18(2): 240-8.Epub2009Sep22), inflammatory response (lichtenshaler S.F. Etc., J Biol chem.2003dec5; 278(49): 48713-9.Epub2003Sep24), arterial thrombosis (Merten m. et al, Z kardiol.2004nov; 93(11): 855-63), cardiovascular diseases such as myocardial infarction and stroke (Maugeri n. et al, Srp Arh cellok lek.2010jan; 138Suppl 1: 50-2) and Graves disease (J. Etc., thyoid.2005jul; 15(7): 645-52).
Technical Field
The present invention relates to 2, 5, 6, 7-tetrahydro- [1, 4 ] having BACE1 and/or BACE2 inhibitory properties]Oxazazem-3-amines, their preparation, pharmaceutical compositions containing them and their use as therapeutically active substances.
In particular, the present invention relates to compounds of formula I, or their pharmaceutically acceptable salts
Wherein R is1To R3As described below.
The compounds of the present invention have Asp2(β -secretase, BACE1 or Memapsin-2) inhibitory activity and may therefore be used in the therapeutic and/or prophylactic treatment of diseases and disorders characterized by elevated β -amyloid levels and/or β -amyloid oligomers and/or β -amyloid plaques and further deposits, in particular alzheimer's disease. Furthermore, the compounds of formula I have BACE2 inhibitory activity and may therefore be used in the therapeutic and/or prophylactic treatment of diseases and disorders such as type2diabetes and other metabolic diseases.
Detailed Description
Objects of the present invention are therefore novel compounds of formula I having BACE1 and/or BACE2 inhibitory properties, their manufacture, medicaments comprising a compound of the invention, the manufacture of such medicaments and the use of compounds of formula I in the treatment or prevention of diseases such as alzheimer's disease and type2 diabetes.
The following definitions are used to illustrate and define the meaning and scope of the various terms used to describe the invention, unless otherwise indicated.
The term "halogen", alone or in combination with other groups, refers to fluorine, chlorine, bromine and iodine, especially fluorine (F).
The term "amido", alone or in combination with other groups, means-C (═ O) -NH2。
The term "C1-7Alkyl ", alone or in combination with other groups, denotes a linear or branched alkyl group having from 1 to 7 carbon atoms, in particular a linear or branched alkyl group having from 1 to 6 carbon atoms, more particularly a linear or branched alkyl group having from 1 to 4 carbon atoms. Straight and branched chains C1-7Examples of alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, the isomeric pentyl, hexyl and heptyl groups, in particular methyl (Me) and ethyl (Et). More particularly methyl.
The term "C1-7-alkoxy ", alone or in combination with other groups, refers to the group R '-O-, wherein R' is C as defined hereinbefore1-7-an alkyl group. C1-7Examples of-alkoxy include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, sec-butoxy and tert-butoxy, preferably methoxy (MeO).
The term "C3-7-cycloalkyl ", alone or in combination with other groups, means a monovalent saturated monocyclic or bicyclic hydrocarbon radical of 3 to 7 ring carbon atoms, especially a monovalent saturated monocyclic hydrocarbon radical of 3 to 5 ring carbon atoms. Double rings are meant to be composed of twoTwo saturated carbocyclic rings of a common carbon atom are formed, i.e. the bridge separating the two rings is a single bond or a chain of one or two carbon atoms. In particular C3-7-cycloalkyl is monocyclic. Examples are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. An example of bicycloalkyl is bicyclo [2.2.1]Heptyl, bicyclo [2.2.2]Octyl or adamantyl. In particular "C3-7-cycloalkyl "is cyclopropyl.
The term "heteroaryl", alone or in combination with other groups, refers to an aromatic carbocyclic group having a single 4 to 8 membered ring or multiple fused rings containing 6 to 14, especially 6 to 10 ring atoms and containing 1, 2 or 3 heteroatoms each selected from N, O and S, especially N and O, in which group at least one heterocyclic ring is aromatic. Examples of "heteroaryl" include benzofuranyl, benzimidazolyl, 1H-benzimidazolyl, benzoxazinyl, benzoxazolyl, benzothiazinyl, benzothiazolyl, benzothienyl, benzotriazolyl, furanyl, imidazolyl, indazolyl, 1H-indazolyl, indolyl, isoquinolyl, isothiazolyl, isoxazolyl, oxazolyl, pyrazinyl, pyrazolyl, 1H-pyrazolyl, pyrazolo [1, 5-a ] pyridyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, quinolinyl, tetrazolyl, thiazolyl, thienyl, triazolyl, 6, 7-dihydro-5H- [1] azoindenyl, and the like. Particular "heteroaryl" groups are pyridyl, oxazolyl, pyrazinyl and thiazolyl. In particular pyridin-2-yl, oxazol-4-yl, pyrazin-2-yl and thiazol-2-yl.
The term "C2-7-alkynyl ", alone or in combination with other groups, means a monovalent straight-chain or branched saturated hydrocarbon radical of 2 to 7 carbon atoms, in particular of 2 to 4 carbon atoms, and comprising one, two or three triple bonds. C2-7Examples of the-alkynyl group include ethynyl, propynyl, prop-2-ynyl, isopropynyl and n-butynyl groups. In particular ethynyl and propynyl.
The term "C1-7-alkoxy-C2-7-alkynyl ", alone or in combination with other groups, means via" C "as defined herein2-7-alkynyl "linked as defined herein“C1-7-alkoxy groups ". In particular 3-methoxy-prop-1-ynyl.
The term "C3-7-cycloalkyl-C2-7-alkynyl ", alone or in combination with other groups, means via" C "as defined herein2-7-alkynyl "linked" C as defined herein3-7-cycloalkyl ". In particular cyclopropylethynyl.
The term "C3-7-cycloalkyl-C1-7-alkoxy ", alone or in combination with other groups, means via" C "as defined herein1-7-alkoxy "linked to" C "as defined herein3-7-cycloalkyl ". In particular cyclopropylmethoxy.
The term "C3-7-cycloalkyl-C1-7-alkyl ", alone or in combination with other groups, means via" C "as defined herein1-7-alkyl "linked" C as defined herein3-7-cycloalkyl ". In particular cyclopropylmethyl.
The term "C1-7-alkyl-S- ", alone or in combination with other groups, means C as defined herein linked via-S-"1-7-an alkyl group.
The term "coupling agent" refers to an agent selected from the group consisting of: carbodiimides or ureas, for example N, N ' -Carbonyldiimidazole (CDI), N, N ' -Dicyclohexylcarbodiimide (DCC), N- (3-dimethylaminopropyl) -N ' -ethyl-carbodiimide-hydrochloride (EDCI), O- (benzotriazol-1-yl) -N, N, N ', N ' -tetramethylurea tetrafluoroborate (TBTU) and 1- [ bis (dimethylamino) methylene ] -1H-1, 2, 3-triazolo [4, 5-b ] pyridine-3-oxide (HATU) hexafluorophosphate.
The term "under basic conditions" means in the presence of a base, especially an alkylamine such as Diisopropylethylamine (DIEA) or Triethylamine (TEA), or a tertiary amine such as N-methylmorpholine or 4- (dimethylamino) -pyridine.
The term "triazine derivative" for example means 4- (4, 6-dimethoxy-1, 3, 5-triazin-2-yl) -4-methyl-morpholine chloride.
The term "protic solvent" refers to a solvent having a hydrogen bonded to oxygen, as in a hydroxyl group, or to nitrogen, as in an amine group, which hydrogen is dissociable. Examples are alcohols, especially ethanol or methanol.
The term "polar solvent" refers to a molecule whose charge is distributed unequally within the molecule. Examples include water and alcohols, especially methanol.
The term "mild oxidizing agent" refers to, for example, t-butyl hydroperoxide.
The term "oxysalt" refers to a salt containing oxygen ([ H ]3O+]) As salts of cations.
The term "ammonium salt" refers to a salt containing ammonium ([ NH ]4 +]) As salts of cations.
The term "pharmaceutically acceptable salt" refers to salts suitable for use in contact with the tissues of humans and animals. Examples of suitable salts with inorganic and organic acids are, but not limited to: acetic acid, citric acid, formic acid, fumaric acid, hydrochloric acid, lactic acid, maleic acid, malic acid, methanesulfonic acid, nitric acid, phosphoric acid, p-toluenesulfonic acid, succinic acid, sulfuric acid, tartaric acid, trifluoroacetic acid, and the like. The terms "pharmaceutically acceptable carrier" and "pharmaceutically acceptable auxiliary substance" refer to carriers and auxiliary substances such as diluents or excipients that are compatible with the other ingredients of the formulation.
The term "pharmaceutical composition" includes a product comprising the specified ingredients in predetermined amounts or proportions, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Preferably it comprises a product comprising one or more active ingredients, and optionally a carrier comprising inert ingredients, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from decomposition of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
The term "inhibitor" refers to a compound that competes with, reduces or prevents binding of a particular receptor to a particular ligand for that particular receptor or reduces or prevents inhibition of the function of a particular protein.
The term "half maximal inhibitory concentration" (IC)50) Refers to the concentration of a particular compound required to obtain 50% inhibition of a biological process in vitro. Can be combined with IC50Value-to-number conversion to pIC50Value (-log IC)50) Where larger values represent exponentially increasing potential. IC (integrated circuit)50The values are not absolute values but depend on the test conditions, such as the concentrations employed. Can be combined with IC50The values were converted to absolute inhibition constants (Ki) using the Cheng-Prusoff equation (biochem. Pharmacol (1973) 22: 3099). The term "inhibition constant" (Ki) refers to the absolute binding affinity of a particular inhibitor for a receptor. It is measured using a competitive binding assay and is equal to the concentration at which a particular inhibitor would occupy 50% of the receptor if no competing ligand (e.g., radioligand) were present. The Ki value can be converted numerically to a pKi value (-logKi), with larger values indicating exponentially increasing potential.
By "therapeutically effective amount" is meant an amount of a compound that, when administered to a subject for the treatment of a disease state, is sufficient to effect such treatment for the disease state. The "therapeutically effective amount" will vary depending on the compound, the disease state being treated, the severity of the disease being treated, the age and relative health of the subject, the route and form of administration, the judgment of the attending physician or veterinarian, and other factors.
The terms "as defined herein" and "as described herein" when referring to a variable incorporate by reference the broadest definition of the variable as well as preferred, more preferred and most preferred definitions, if any.
The terms "treating", "contacting" and "reacting" when referring to a chemical reaction mean adding or mixing two or more reagents under suitable conditions to produce the indicated and/or desired product. It is to be understood that the reaction that produces the indicated and/or desired product may not necessarily result directly from the combination of the two reagents that were initially charged, i.e., one or more intermediates that ultimately lead to the formation of the indicated and/or desired product may be produced in the mixture. The term "aromatic" refers to the conventional concept of aromaticity as defined in the literature, in particular as defined in IUPAC-Complex of Chemical technology, 2nd, A.D.McNaught & A.Wilkinson (Eds).
The term "pharmaceutically acceptable excipient" refers to any ingredient used in formulating pharmaceutical products that is not therapeutically active and is non-toxic, such as disintegrants, binders, fillers, solvents, buffers, tonicity agents, stabilizers, antioxidants, surfactants, or lubricants.
The compounds of formula I may also be solvated, for example by hydration. The solvation may be effected during the preparation process or may for example be brought about by the hygroscopic properties of the initially anhydrous form of the compound of formula I (hydration). The term "pharmaceutically acceptable salts" also includes physiologically acceptable solvates.
"isomers" are compounds having the same molecular formula but differing in nature or in the order of their atomic bonding or in the arrangement of their atoms in space. Isomers differing in the arrangement of their atoms in space are referred to as "stereoisomers". Stereoisomers that are not mirror images of each other are referred to as "diastereomers", stereoisomers that are non-superimposable mirror images are referred to as "enantiomers", and sometimes are referred to as optical isomers. The carbon atom linking the four different substituents is called a "chiral center". When a chiral carbon is present in a chemical structure, it is intended that the structure include all stereoisomers associated with that chiral carbon.
All individual embodiments may be combined.
The invention relates to compounds of formula I
Wherein
R1Is H or F;
R2is C1-7-an alkyl group; and is
R3Is- (C ═ O) -R4Or R5Wherein
R4Is selected from C3-7-cycloalkyl-C1-7-alkoxy-, C3-7-cycloalkyl-C2-7-alkynyl-, C1-7-alkoxy-C2-7-alkynyl-, unsubstituted heteroaryl, unsubstituted C3-7-cycloalkyl and C1-7Heteroaryl substituted with one substituent of the group consisting of-alkyl-S-, or
R4Is heteroaryl substituted with one halogen and one amide group;
R5is C3-7-cycloalkyl-C1-7-alkyl-;
or a pharmaceutically acceptable salt thereof.
A particular embodiment are compounds of formula I as described herein, wherein
R1Is F;
R2is Me; and is
R3Is- (C ═ O) -R4Wherein
R4Is selected from C3-7-cycloalkyl-C1-7-alkoxy-, C3-7-cycloalkyl-C2-7-alkynyl-, C1-7-alkoxy-C2-7-alkynyl-substituted pyridyl with one substituent of the group, or
R4Is pyridyl substituted with one F and one amide group;
or a pharmaceutically acceptable salt thereof.
A particular embodiment are compounds of formula I, as described herein, wherein R is1Is F, R2Is Me, R3Is- (C ═ O) -R4And R is4Is 3-fluoro-5-amide-pyridin-2-yl, 6- (cyclopropylmethoxy) -pyridin-2-yl, 5- (cyclopropylethynyl) -pyridin-2-yl or 5- (3-methoxyprop-1-ynyl) -pyridin-2-yl.
A particular embodiment are compounds of formula I, as described herein, wherein R is1Is F.
A particular embodiment are compounds of formula I, as described herein, wherein R is1Is H.
A particular embodiment are compounds of formula I, as described herein, wherein R is2Is Me.
A particular embodiment are compounds of formula I, as described herein, wherein R is2Is Et.
A particular embodiment are compounds of formula I, as described herein, wherein R is3Is C3-7-cycloalkyl-C1-7-alkyl-.
A particular embodiment are compounds of formula I, as described herein, wherein R is3Is cyclopropyl-CH2-。
A particular embodiment are compounds of formula I as described herein, which are (5R, 6R) -5- [5- (cyclopropylmethyl-amino) -2-fluoro-phenyl]-6-fluoro-5-methyl-2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-amine.
A particular embodiment are compounds of formula I, as described herein, wherein R is3Is- (C ═ O) -R4。
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is pyridyl substituted with halogen and one amido group.
A specific embodiment isA compound of formula I as described herein, wherein R4Is pyridyl substituted with one F and one amide group.
A particular embodiment are compounds of formula I as described herein which are (R) -N2- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -3-fluoropyridine-2, 5-dicarboxamide formate.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is selected from C3-7-cycloalkyl-C1-7-alkoxy-, C3-7-cycloalkyl-C2-7-alkynyl-, C1-7-alkoxy-C2-7-alkynyl-, unsubstituted heteroaryl and unsubstituted C3-7-pyridyl substituted with one substituent of the group consisting of cycloalkyl.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is selected from C3-7-cycloalkyl-C1-7-alkoxy-substituted pyridyl with one substituent of the group consisting of alkyl.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is selected from C3-7-cycloalkyl-C2-7-alkynyl-substituted pyridyl with one substituent of the group.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is selected from C1-7-alkoxy-C2-7-alkynyl-substituted pyridyl with one substituent of the group.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is a pyridyl group substituted with one substituent selected from the group consisting of unsubstituted heteroaryl groups.
One particular embodimentIs a compound of formula I as described herein, wherein R is4Is pyridyl substituted with one substituent selected from the group consisting of unsubstituted thiazol-2-yl.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is selected from unsubstituted C3-7-pyridyl substituted with one substituent of the group consisting of cycloalkyl.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is a pyridyl group substituted with one substituent selected from the group consisting of unsubstituted cyclopropyl groups.
A particular embodiment is a compound of formula I as described herein, selected from the group consisting of:
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -6- (cyclopropylmethoxy) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (thiazol-2-yl) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (cyclopropylethynyl) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5-cyclopropylpyridinecarboxamide formate salt, and
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (3-methoxyprop-1-ynyl) pyridinecarboxamide formate.
A particular embodiment are compounds of formula I as described herein which are (R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -6- (cyclopropylmethoxy) pyridinecarboxamide formate.
A particular embodiment are compounds of formula I as described herein which are (R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5- (thiazol-2-yl) pyridinecarboxamide formate.
A particular embodiment are compounds of formula I as described herein which are (R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5- (cyclopropylethynyl) pyridinecarboxamide formate.
A particular embodiment are compounds of formula I as described herein which are (R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5-cyclopropylpyridinecarboxamide formate.
A particular embodiment are compounds of formula I as described herein which are (R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5- (3-methoxyprop-1-ynyl) pyridinecarboxamide formate.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is a quilt C1-7-alkyl-S-substituted pyrazinyl.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is pyrazinyl substituted by methyl-S-.
A particular embodiment are compounds of formula I as described herein which are (R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5- (methylthio) pyrazine-2-carboxamide formate salt.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is a quilt C3-7-cycloalkyl-substituted oxazolyl.
A particular embodiment are compounds of formula I, as described herein, wherein R is4Is an oxazolyl group substituted with a cyclopropyl group.
A particular embodiment are compounds of formula I as described herein which are (R) -N- (3- (3-amino-5-ethyl-6, 6-difluoro-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5-Cyclopropyloxazole-4-carboxamide.
A particular embodiment is a compound of formula I as described herein, selected from the group consisting of:
(R) -N2- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -3-fluoropyridine-2, 5-dicarboxamide formate salt,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -6- (cyclopropylmethoxy) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (cyclopropylethynyl) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (3-methoxyprop-1-ynyl) pyridinecarboxamide formate, and
(5R, 6R) -5- [5- (cyclopropylmethyl-amino) -2-fluoro-phenyl]-6-fluoro-5-methyl-2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-amine.
A particular embodiment is a compound of formula I as described herein, selected from the group consisting of:
(R) -N2- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -3-fluoropyridine-2, 5-dicarboxamide formate salt,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -6- (cyclopropylmethoxy) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (cyclopropylethynyl) pyridinecarboxamide formate salt, and
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (3-methoxyprop-1-ynyl) pyridinecarboxamide formate.
A particular embodiment is a process for the preparation of a compound of formula I as defined herein, which process comprises:
a) reacting an amine of formula II with a carboxylic acid of formula III in the presence of a coupling agent under basic conditions or with the aid of a triazine derivative to obtain a compound of formula Ia
Wherein R is1、R2And R4Such as bookThe terms are defined herein, or alternatively,
b) reacting an amine of formula II with a compound of formula IV in the presence of acetic acid and sodium triacetoxyborohydride to obtain a compound of formula Ib,
wherein R is hydrogen or C1-7-alkyl, and wherein R1、R2、R3And R5As defined herein.
The invention also relates to compounds of formula I as described herein, obtainable according to the process as described herein before.
A particular embodiment of the invention relates to a compound of formula I as defined herein and a pharmaceutically acceptable carrier and/or excipient.
A particular embodiment of the invention relates to a compound of formula I as defined herein for use as a medicament.
A particular embodiment of the present invention relates to compounds of formula I as defined herein for use in the therapeutic and/or prophylactic treatment of alzheimer's disease and/or type II diabetes.
A particular embodiment of the present invention relates to compounds of formula I as defined herein for use in the therapeutic and/or prophylactic treatment of alzheimer's disease.
A particular embodiment of the present invention relates to compounds of formula I as defined herein for use in the therapeutic and/or prophylactic treatment of type2 diabetes.
Particular embodiments of the present invention relate to compounds of formula I as defined herein for use as therapeutically active substances for the therapeutic and/or prophylactic treatment of Amyotrophic Lateral Sclerosis (ALS), arterial thrombosis, autoimmune/inflammatory diseases, cancer such as breast cancer, cardiovascular diseases such as myocardial infarction and stroke, dermatomyositis, down syndrome, gastrointestinal diseases, glioblastoma multiforme, graves ' disease, huntington's disease, Inclusion Body Myositis (IBM), inflammatory responses, kaposi's sarcoma, costermann's disease, lupus erythematosus, macrophage myofasciitis, juvenile idiopathic arthritis, granulomatous arthritis, malignant melanoma, multiple myeloma, rheumatoid arthritis, sjogren's syndrome, spinocerebellar ataxia type 1, spinocerebellar ataxia type 7, juvenile idiopathic arthritis, rheumatoid arthritis, and rheumatoid arthritis, Wheeler's disease or wilson's disease.
A particular embodiment of the present invention relates to a compound of formula I as defined herein for use in the preparation of a medicament for the therapeutic and/or prophylactic treatment of alzheimer's disease and/or type2 diabetes.
A particular embodiment of the present invention relates to a compound of formula I as defined herein for use in the preparation of a medicament for the therapeutic and/or prophylactic treatment of alzheimer's disease.
A particular embodiment of the present invention relates to a compound of formula I as defined herein for use in the preparation of a medicament for the therapeutic and/or prophylactic treatment of type2 diabetes.
A particular embodiment of the present invention relates to a method for the therapeutic and/or prophylactic treatment of diseases which can be ameliorated by the inhibition of BACE1 and/or BACE2 activity, in particular for the treatment of alzheimer's disease and type2diabetes, which method comprises administering to a human or animal a therapeutically active amount of a compound of formula I as described herein.
A particular embodiment of the present invention relates to a compound of formula I as defined herein for use in the manufacture of a medicament for use in the therapeutic and/or prophylactic treatment of alzheimer's disease and/or type2 diabetes.
A particular embodiment of the present invention relates to compounds of formula I as defined herein for use as therapeutically active substances for the therapeutic and/or prophylactic treatment of: amyotrophic Lateral Sclerosis (ALS), arterial thrombosis, autoimmune/inflammatory diseases, cancer such as breast cancer, cardiovascular diseases such as myocardial infarction and stroke, dermatomyositis, down's syndrome, gastrointestinal diseases, glioblastoma multiforme, graves' disease, huntington's disease, Inclusion Body Myositis (IBM), inflammatory responses, kaposi's sarcoma, costerman's disease, lupus erythematosus, macrophage myofascitis, juvenile idiopathic arthritis, granulomatous arthritis, malignant melanoma, multiple myeloma, rheumatoid arthritis, sjogren's syndrome, spinocerebellar ataxia type 1, spinocerebellar ataxia type 7, whipple's disease, or wilson's disease.
A particular embodiment of the present invention relates to a compound of formula I as defined herein for the preparation of a medicament for the therapeutic and/or prophylactic treatment of alzheimer's disease and/or type2 diabetes.
A particular embodiment of the present invention relates to a method for the therapeutic and/or prophylactic treatment of diseases which can be ameliorated by the inhibition of BACE1 and/or BACE2 activity, in particular for the treatment of alzheimer's disease and type2diabetes, which method comprises administering to a human or animal a therapeutically active amount of a compound of formula I according to any one of claims 1 to 17.
Particular embodiments of the present invention relate to and a method for the therapeutic and/or prophylactic treatment of diseases which can be ameliorated by the inhibition of BACE1 and/or BACE2 activity, in particular for the treatment of Amyotrophic Lateral Sclerosis (ALS), arterial thrombosis, autoimmune/inflammatory diseases, cancer such as breast cancer, cardiovascular diseases such as myocardial infarction and stroke, dermatomyositis, Down syndrome, gastrointestinal diseases, glioblastoma multiforme, Graves ' disease, Huntington's disease, Inclusion Body Myositis (IBM), inflammatory reactions, Kaposi's sarcoma, Cowster's disease, lupus erythematosus, macrophage myofasciitis, juvenile idiopathic arthritis, granulomatous arthritis, malignant melanoma, multiple myeloma, rheumatoid arthritis, Sjogren's syndrome, spinocerebellar ataxia type 1, spinocerebellar ataxia type 7, Whipple's disease or Wilson's disease, the method comprising administering a therapeutically active amount of a compound of formula I according to any one of claims 1 to 17 to a human being or animal.
As indicated above, the compounds of formula I of the present invention are useful in preserving and restoring β -cell function and stimulating insulin secretion in diabetic patients as well as in non-diabetic patients with impaired glucose tolerance or in a pre-diabetic state. They may be used to treat type 1 diabetes or to delay or prevent the need for insulin therapy in type2 diabetic patients. The compounds of formula I are further useful for ameliorating hyperinsulinemia (hyperinsulinemia), which commonly occurs in diabetic or pre-diabetic patients, and for reducing the risk associated with metabolic syndrome, which may also be useful for treating vascular diseases such as hypertension.
Thus, the expression "a disease which can be ameliorated by the inhibition of BACE2 activity" refers to diseases such as metabolic and cardiovascular diseases, especially diabetes, more especially type2diabetes, gestational diabetes, impaired fasting glucose, impaired glucose tolerance, insulin resistance, pre-diabetes, metabolic syndrome, type 1 diabetes, diabetic complications including diabetic nephropathy, diabetic retinopathy and diabetic neuropathy, chronic kidney disease, dyslipidemia, atherosclerosis, myocardial infarction, hypertension, and other metabolic and cardiovascular diseases.
In particular, the expression 'a disease which can be ameliorated by the inhibition of BACE2 activity' relates to diabetes, in particular type2diabetes, impaired glucose tolerance, pre-diabetes, metabolic syndrome and hypertension. More specifically, the expression 'a disease associated with inhibition of BACE2 activity' refers to diabetes, especially type2 diabetes.
The compounds of formula I may contain more than one asymmetric center and may therefore exist as: racemic compounds, racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. Additional asymmetric centers may be present depending on the nature of the different substituents on the molecule. Each such asymmetric center will independently produce two optical isomers, and all possible optical isomers and diastereomers, both in admixture and as pure or partially purified compounds, are intended to be included within the scope of the present invention. The present invention is intended to encompass all such isomeric forms of these compounds. The independent synthesis of these diastereomers or their chromatographic separation may be achieved as is well known in the art by appropriate modification of the methods disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates, which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds can be separated to separate the individual enantiomers. Separation can be carried out by methods known in the art, such as the coupling of a racemic mixture of compounds with an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.
In more detail, the compounds of formula I according to the invention can be prepared by the methods and procedures given below. Some typical procedures for preparing compounds of formula I will be in scheme A (R)12H, Br or NO2) As described in (1).
Sulfenimides of general formula a2 can be prepared in analogy to t.p.tang and j.a.ellman, j.org.chem.1999, 64, 12 by condensation of aryl ketones and sulfenamides, e.g. alkyl sulfenamides, most preferably (R) - (+) -tert-butyl sulfenamide in the presence of lewis acids such as titanium (IV) alkoxides, more preferably titanium (IV) ethoxide, in solvents such as ethers, e.g. diethyl ether or more preferably THF.
The conversion of sulfinylimine A2 to sulfinamide ester A3 is performed stereoselectively via a chiral directing group as described by Tang and Ellman. The sulfinimide A2 can be reacted with a titanium alkoxide which is produced, for example, from an alkyl acetate, preferably ethyl acetate, LDA and triisopropoxytitanium chloride, at low temperatures, preferably at-78 ℃ in a solvent such as an ether, for example diethyl ether or more preferably THF. Alternatively the sulfonamido ester A3 can be prepared from the sulfinylimine a2 by reaction of a bromoacetate derivative and zinc dust, optionally in the presence of copper (I) chloride, in a solvent such as an ether, for example diethyl ether or more preferably THF, at a temperature of 0 to 70 ℃, preferably 23 ℃.
The sulfonamido ester A3 can be reduced to the alcohol a4 by reducing the ethyl ester with a basic hydride, preferably lithium borohydride or lithium aluminium hydride, in a solvent such as an ether, e.g. diethyl ether or more preferably THF.
Alkylation of alcohol A4 to nitrile A5 may be carried out with a suitable weak base, preferably silver (I) oxide, in a solvent such as THF or CH2Cl2More preferably CH2Cl2In the presence of an alkylation catalyst such as tetrabutylammonium iodide.
Hydrolysis of the chiral directing group in nitrile a5 gives the aminonitrile a6, which can be achieved using a mineral acid, for example sulfuric acid or preferably hydrochloric acid, in a solvent such as an ether, for example diethyl ether or more preferably 1, 4-dioxane.
AminooxazazepineA7 can be prepared by reaction of an aminonitrile A6 and trimethylaluminum in a solvent such as xylene, preferably toluene.
Scheme A
Introduction of the nitro group into A7 to give A8 is best carried out at low temperature, preferably at 0 ℃ according to standard procedures involving sulfuric and nitric acids.
AminooxazazepineReduction of the nitro group in A8 to aniline a9 can be accomplished by hydrogenation using a catalyst such as Pd/C in a protic solvent, such as an alcohol, preferably ethanol or methanol.
The amide coupling of aniline a9 and a carboxylic acid to give amide Ia can be carried out with a carbodiimide, for example DCC or EDCI, in a solvent such as dichloromethane or, in particular, with a triazine derivative, 4- (4, 6-dimethoxy-1, 3, 5-triazin-2-yl) -4-methyl-morpholine chloride in an alcohol, in particular methanol.
The target amines Ib can be prepared via reductive amination of aniline a9 and a carbonyl compound with a borohydride reducing agent, e.g. sodium borohydride, preferably sodium triacetoxyborohydride and a weak acid, e.g. acetic acid, in a solvent such as tetrahydrofuran or dichloromethane.
The compounds of formula I may form pharmaceutically acceptable salts. The term "pharmaceutically acceptable salt" refers to salts that retain the biological effectiveness and properties of the free base or free acid, which are not biologically or otherwise undesirable. The pharmaceutically acceptable salts of the compounds of formula I are preferably acid addition salts with physiologically compatible inorganic acids, such as hydrochloric acid, sulfuric acid, sulfurous acid or phosphoric acid, or with organic acids, such as methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, formic acid, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, lactic acid, trifluoroacetic acid, citric acid, fumaric acid, maleic acid, malonic acid, tartaric acid, benzoic acid, cinnamic acid, mandelic acid, succinic acid or salicylic acid. In addition, pharmaceutically acceptable salts can be prepared by addition of an inorganic or organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium and the like. Salts derived from organic bases include, but are not limited to, salts of the following bases: primary, secondary and tertiary amines; substituted amines, including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, lysine, arginine, N-ethylpiperidine, piperidine, polyamine (polymine) resins, and the like. The compounds of formula I may also exist in the form of zwitterions. Particular pharmaceutically acceptable salts of the compounds of formula I are acid addition salts such as the hydrochloride, formate or trifluoroacetate salt. In particular formate (salt of formic acid).
The corresponding pharmaceutically acceptable salts formed with the acids may be obtained by standard methods known to those skilled in the art, for example by dissolving a compound of formula I in a suitable solvent such as, for example, dioxane or THF, and adding an appropriate amount of the corresponding acid. The product can usually be isolated by filtration or by chromatography. The conversion of a compound of formula I into a pharmaceutically acceptable salt with a base can be carried out by treating such a compound with such a base. One possible way of forming such salts is, for example, by reacting 1/n equivalent of a base salt such as, for example, M (OH)nWhere M ═ metal or ammonium cations and n ═ the number of hydroxide anions, are added to a solution of the compound in a suitable solvent (e.g. ethanol, ethanol-water mixtures, tetrahydrofuran-water mixtures) and the solvent is removed by evaporation or freeze-drying. Particular salts are the hydrochloride, formate and trifluoroacetate salts.
In case their preparation is not described in the examples, the compounds of formula I as well as all intermediate products may be prepared according to similar methods or according to the methods given herein. The starting materials are commercially available, known in the art or can be prepared by methods known in the art or similar thereto.
It will be appreciated that the compounds of formula I in the present invention may be derivatised at functional groups to provide derivatives which are capable of conversion back to the parent compound in vivo.
Pharmacological testing
The compounds of formula I and their pharmaceutically acceptable salts possess valuable pharmacological properties. It has been found that the compounds of the present invention are associated with the inhibition of BACE1 and/or BACE2 activity. The compounds were investigated according to the tests given below.
Cytological a β reduction assay:
human HEK293 cells stably transfected with a vector expressing cDNA of the human APP wt gene (APP695) were used to assess potency of compounds in cytological assays. Cells were plated in 96-well microtiter plates, in cell culture medium (Iscove, supplemented with 10% (v/v) fetal calf serum, glutamine, penicillin/streptomycin), to about 80% confluence, and compounds were added at 10x concentration in 1/10 volumes of medium without FCS containing 8% DMSO (final concentration of DMSO is maintained at 0.8% v/v). In a humidified incubator at 37 ℃ and 5% CO2After 18-20 hours of incubation, the culture medium supernatant was harvested for determination of the concentration of A.beta.40. 96-well ELISA plates (e.g., NuncMaxisorb) were coated with monoclonal antibodies that specifically recognize the C-terminal end of A.beta.40 (Brockhaus et al, NeuroReport9, 1481-1486; 1998). After blocking non-specific binding sites with, e.g., 1% BSA and washing, the culture supernatant is added at an appropriate dilution with horseradish peroxidase-conjugated a β detection antibody (e.g., antibody 4G8, Senetek, Maryland Heights, MO) and incubated for 5 to 7 hours. The wells of the microtiter plate were then washed thoroughly with Tris-buffered saline containing 0.05% Tween20 and the assay was washed with tetramethylbenzidine/H in citrate buffered saline2O2And (4) developing color. In using a volume of 1N H2SO4After termination of the reaction, the reaction was measured at 450nm wavelength on an ELISA reader. The concentration of a β in the culture supernatant was calculated from a standard curve obtained with known amounts of pure a β peptide.
BACE inhibition assay by measuring cellular TMEM27 lysis:
this assay uses the principle of inhibiting human TMEM27 in the Ins1e rat cell line from being cleaved by endogenous cellular BACE2 and shed from the cell surface into the culture medium (which is then detected in an ELISA assay). Inhibition of BACE2 prevents cleavage and shedding in a dose-related manner.
The stable cell line "INS-TMEM 27" represents a cell line with a doxycyclineAn INS1 e-derived cell line that induced expression (using the TetOn system) of full-length hTMEM27 in an insulin-dependent manner. Throughout the experiment, cells were cultured in RPMI1640+ Glutamax (Invitrogen) penicillin/streptomycin, 10% fetal bovine serum, 100mM pyruvate, 5mM beta-mercaptoethanol, 100 microgram/ml G418, and 100 microgram/ml hygromycin and incubated in standard CO2The cells were grown in non-adherent culture at 37 ℃ in an incubator.
INS-TMEM27 cells were seeded in 96-well plates. After 2 days of culture, BACE2 inhibitor was added at the concentration range required for the assay, and doxycycline was added to a final concentration of 500ng/ml after two further hours of culture. Cells were further incubated for 46 hours and supernatants were harvested for detection of exfoliated TMEM 27.
An ELISA assay (using a pair of mouse anti-human TMEM27 antibodies, directed against the extracellular domain of TMEM 27) was used for detection of TMEM27 in culture. EC for BACE2 inhibition was calculated for each inhibitor concentration using ELISA readings using standard curve fitting software such as XLFit for Excel spreadsheet program50。
Preferred compounds according to formula I have an inhibitory activity (IC) in the above assay of preferably 5nM to 50. mu.M, more preferably 5nM to 1. mu.M50)。
For example, the following compounds showed the following IC in the above assay50The value:
pharmaceutical composition
The compounds of formula I and the pharmaceutically acceptable salts can be used as therapeutically active substances, for example in the form of pharmaceutical preparations. The pharmaceutical preparations can be administered orally, for example, in the form of tablets, coated tablets, dragees, hard and soft gelatine capsules, solutions, emulsions or suspensions. However, administration may also be effected rectally, for example in the form of suppositories, or parenterally, for example in the form of ampoules.
The compounds of formula I and their pharmaceutically acceptable salts can be processed with pharmaceutically inert, inorganic or organic carriers for the preparation of pharmaceutical preparations. For example, lactose, corn starch or derivatives thereof, talc, stearic acid or its salts and the like can be used as carriers for tablets, coated tablets, dragees and hard gelatine capsules. Suitable carriers for soft gelatine capsules are, for example, vegetable oils, waxes, fats, and semi-solid and liquid polyols and the like. However, depending on the nature of the active ingredient, no carriers are often required in the case of soft gelatin capsules. Suitable carriers for the preparation of solutions and syrups are, for example, water, polyols, glycerol, vegetable oils and the like. Suitable carriers for suppositories are, for example, natural or hardened oils, waxes, fats and semi-liquid or liquid polyols and the like.
In addition, the pharmaceutical preparations can contain pharmaceutically acceptable auxiliary substances such as preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for varying the osmotic pressure, buffers, masking agents or antioxidants. They may also contain other therapeutically valuable substances.
Medicaments containing a compound of formula I or a pharmaceutically acceptable salt thereof and a therapeutically inert carrier are also an object of the present invention, as are processes for their preparation, which comprise bringing one or more compounds of formula I and/or pharmaceutically acceptable salts thereof and, if desired, one or more other therapeutically valuable substances into a galenical administration form together with one or more therapeutically inert carriers.
The dosage can vary within wide limits and must, of course, be adjusted to the individual requirements in each particular case. In the case of oral administration, the dosage for adults may vary from about 0.01 mg/day to about 1000 mg/day, in particular from about 1 to 500 mg/day, of a compound of formula I or of a corresponding amount of a pharmaceutically acceptable salt thereof. Depending on the severity of the disease and the precise pharmacokinetic profile of the compound, the daily dose may be administered in a single dose or in divided doses, and furthermore, when desired, the upper limit may also be exceeded.
The following examples illustrate the invention, but are not intended to be limiting and are intended to be exemplary only. Examples of compositions according to the invention are:
example A
Tablets having the following composition were manufactured in the usual manner:
| composition (I) | mg/tablet |
| 5 | 25 | 100 | 500 | |
| A compound of formula I | 5 | 25 | 100 | 500 |
| Lactose anhydrous DTG | 125 | 105 | 30 | 150 |
| Sta-Rx1500 | 6 | 6 | 6 | 60 |
| Microcrystalline cellulose | 30 | 30 | 30 | 450 |
| Magnesium stearate | 1 | 1 | 1 | 1 |
| Total of | 167 | 167 | 167 | 831 |
Table 1: possible tablet compositions
The manufacturing process comprises the following steps:
1. ingredients 1, 2, 3 and 4 were mixed and granulated with pure water.
2. The granules were dried at 50 ℃.
3. The particles are passed through a suitable milling apparatus.
4. Add ingredient 5 and mix for three minutes; compressed on a suitable press.
Example B-1
Capsules having the following composition were prepared:
table 2: possible capsule ingredient composition
The manufacturing process comprises the following steps:
1. ingredients 1, 2 and 3 were mixed in a suitable mixer for 30 minutes.
2. Ingredients 4 and 5 were added and mixed for 3 minutes.
3. Filling into suitable capsules.
The compound of formula I, lactose and corn starch are first mixed in a mixer and then mixed in a mill. The mixture was returned to the mixer, talc was added thereto and mixed thoroughly. The mixture is filled by machine into suitable capsules, for example hard gelatin capsules.
Example B-2
Soft gelatin capsules were prepared having the following composition:
| composition (I) | mg/capsule |
| A compound of formula I | 5 |
| Yellow wax | 8 |
| Hydrogenated soybean oil | 8 |
| Partially hydrogenated vegetable oils | 34 |
| Soybean oil | 110 |
| Total of | 165 |
Table 3: possible soft gelatin capsule ingredient compositions
| Composition (I) | mg/capsule |
| Gelatin | 75 |
| Glycerin 85% | 32 |
| Karion83 | 8 (Dry matter) |
| Titanium dioxide | 0.4 |
| Iron oxide yellow | 1.1 |
| Total of | 116.5 |
Table 4: possible soft gelatin capsule compositions
Manufacturing process
The compound of formula I is dissolved in a warm melt of the other ingredients and the mixture is filled into soft gelatin capsules of appropriate size. The filled soft gelatin capsules were processed according to the general procedure.
Example C
Suppositories of the following composition were prepared:
| composition (I) | mg/suppository |
| A compound of formula I | 15 |
| Suppository block | 1285 |
| Total of | 1300 |
Table 5: possible suppository composition
Manufacturing process
The suppository blocks were melted in a glass or steel vessel, mixed well and cooled to 45 ℃. Thereupon, the finely powdered compound of formula I is added thereto and stirred until it is completely dispersed. The mixture is poured into suppository molds of appropriate size, left to cool, after which the suppositories are removed from the molds and individually packaged in wax paper or metal foil.
Example D
The injection with the following composition is prepared:
| composition (I) | mg/injection |
| A compound of formula I | 3 |
| Polyethylene glycol 400 | 150 |
| Acetic acid | Appropriate amount to pH5.0 |
| Water for injection | To 1.0ml |
Table 6: possible injection composition
Manufacturing process
The compound of formula I is dissolved in a mixture of polyethylene glycol 400 and water for injection (part). The pH was adjusted to 5.0 by acetic acid. The volume was adjusted to 1.0ml by adding the balance of water. The solution was filtered, filled into vials with the appropriate excess and sterilized.
Example E
Sachets of the following composition were made:
| composition (I) | mg/sachet |
| A compound of formula I | 50 |
| Lactose, fine powder | 1015 |
| Microcrystalline cellulose (AVICEL PH102) | 1400 |
| Sodium carboxymethylcellulose | 14 |
| Polyvinylpyrrolidone K30 | 10 |
| Magnesium stearate | 10 |
| Fragrance additive | 1 |
| Total of | 2500 |
Table 7: possible sachet composition
Manufacturing process
The compound of formula I is mixed with lactose, microcrystalline cellulose and sodium carboxymethylcellulose and granulated with a mixture of polyvinylpyrrolidone in water. The granules were mixed with magnesium stearate and flavor additives and filled into sachets.
Examples
And (4) summarizing:
MS: mass Spectrometry (MS) was measured with either positive or negative ion spray (ISP or ISN) on a Perkin-Elmer SCIEX API300 or electron bombardment (EI, 70eV) on a Finnigan MAT SSQ7000 spectrometer.
Abbreviations:
DCC ═ N, N' -diisopropyl-carbodiimide, DCE ═ 1, 2-dichloroethane, DCM ═ dichloromethane, DIEA ═ diisopropylethylamine, DMAc ═ dimethylacetamide, DMAP ═ 4-dimethylaminopyridine, DMF ═ N, n-dimethylformamide, DMSO, dimethyl sulfoxide, EDCI N- (3-dimethylaminopropyl) -N' -ethyl-carbodiimide hydrochloride, HATU 1- [ bis (dimethylamino) methylene ] -1H-1, 2, 3-triazolo [4, 5-b ] pyridine-3-oxide, HCl hydrogen chloride, HPLC high performance liquid chromatography, LDA lithium diisopropylamide, MS mass spectrometry, NMR nuclear magnetic resonance, TEA triethylamine, TBME t-butyl methyl ether, and THF tetrahydrofuran.
The following examples are provided to illustrate the present invention. They should not be considered as limitations on the scope of the invention, but merely as representatives thereof.
Synthesis of intermediate 1- (2-fluoro-5-nitro-phenyl) -propan-1-one A1A
To a solution of 1- (2-fluoro-phenyl) -propan-1-one (99mmol) in concentrated sulfuric acid (80ml) cooled to-30 ℃ fuming nitric acid (8ml) was slowly added over 20 minutes and the solution was stirred at-30 ℃ for 15 minutes. The mixture was slowly poured into a stirred mixture of 200ml of water and 400g of ice. The aqueous phase was extracted with ethyl acetate and the organic layer was again extracted with water and NaHCO31M aqueous solution. The organic layer was washed with Na2SO4Dried over silica, evaporated and the residue purified by chromatography on silica using a mixture of heptane and ethyl acetate as eluent to afford pure nitro intermediate J. Ms (isp): 198.1[ M + H ] M/z]+。
Synthesis of intermediate sulfinylimine A2
General procedure
To a solution of (R) - (+) -tert-butylsulfinamide (66mmol) in THF (350ml) were added successively ketone A1(72.6mmol) and titanium (IV) ethoxide (132mmol) and the solution was stirred at reflux temperature for 5 h. The mixture was cooled to 22 ℃, treated with brine (400ml), the suspension stirred for 10 minutes and filtered over celite (dicalite). The layers were separated, the aqueous layer was extracted with ethyl acetate, the combined organic layers were washed with water, dried and concentrated in vacuo. The residue was purified by chromatography on silica using cyclohexane/ethyl acetate to give pure sulfinimide a 2.
Intermediate A2A
Starting from 1- (2-fluorophenyl) -ethanone, the product (R) -2-methyl-propane-2-sulfinic acid [1- (2-fluorophenyl) - (E) -ethylene is obtained]Amides as light brown oils. Ms (isp): 242.3[ M + H ] M/z]+。
Intermediate A2B
Starting from 1- (2-fluoro-phenyl) -propan-1-one, the product 2-methyl-propane-2-sulfinic acid [1- (2-fluoro-phenyl) -propan- (E) -ylidene]Amides, as pale yellow oils. MS: m/z 256.2[ M + H ]]+。
Synthesis of intermediate sulfonamido ester A3
General procedure:
in a dry apparatus, a suspension of freshly activated zinc powder (1.63g, 24.9mmol) in dry THF (70ml) was heated to reflux under an inert atmosphere. A solution of sulfinimide A2(24.9mmol) and bromoacetate (24.9mmol) in dry THF (15ml) was added dropwise over a period of 15 minutes, and the suspension was heated to reflux for 5 h. The cooled mixture was brought to saturated NH4Partition between aqueous Cl and ethyl acetate and dry the organic layerAnd evaporated. The crude material was purified by flash chromatography using heptane/ethyl acetate to give sulfonamido ester a 3.
Intermediates A3A and A3B
Starting from (R) -2-methyl-propane-2-sulfinic acid [1- (2-fluorophenyl) - (E) -ethylene]-amide and ethyl 2-bromo-2-fluoroacetate, to obtain the faster eluting minor isomer ethyl (2S, 3R) -3- ((R) -1, 1-dimethylethylsulphideneamino) -2-fluoro-3- (2-fluorophenyl) butanoate (intermediate A3A) as a dark brown oil. Ms (isp): m/z 348.2[ M + H ]]+。
The second fraction contained the slower eluting major isomer ethyl (2R, 3R) -3- ((R) -1, 1-dimethylethylenesulfonamido) -2-fluoro-3- (2-fluorophenyl) butanoate (intermediate A3B) as a brown oil. Ms (isp): m/z 348.2[ M + H ]]+。
Intermediate A3C
Starting from (R) -2-methyl-propane-2-sulfinic acid [1- (2-fluorophenyl) - (E) -ethylene]-amide, obtaining the product ethyl (R) -2, 2-difluoro-3- (2-fluoro-phenyl) -3- ((R) -2-methyl-propane-2-sulfinylamino) -butyrate as a pale yellow oil. MS: 366.1[ M + H ] M/z]+。
Intermediate A3D
Starting from 2-methyl-propane-2-sulfinic acid [1- (2-fluoro-phenyl) -prop- (E) -ylidene]-amide to obtain the product (R) -ethyl 2, 2-difluoro-3- (2-fluoro-phenyl) -3- ((R) -2-methyl-propane-2-sulfinylamino) -pentanoate as colorless oil. MS: 380.2[ M + H ] M/z]+。
Synthesis of intermediate sulfonamidol A4
General procedure:
a solution of sulfonamido ester A3(12.7mmol) in dry THF (50ml) was treated with lithium borohydride (25.3mmol) at 0 deg.C and stirring was continued for 4h at 0 deg.C. The reaction mixture was quenched by the addition of acetic acid (2ml) and water (50ml), extracted with ethyl acetate, and the organic layer was dried and evaporated. The residue was purified by chromatography on silica using a mixture of n-heptane and ethyl acetate to give the pure intermediate sulfinamidol a 4.
Intermediate A4A
Starting from ethyl (2R, 3R) -3- ((R) -1, 1-dimethylethylethylenesulfonamido) -2-fluoro-3- (2-fluorophenyl) butanoate, the product (R) -2-methyl-propane-2-sulfinic acid [ (1R, 2R) -2-fluoro-1- (2-fluoro-phenyl) -3-hydroxy-1-methyl-propyl-butyric acid is obtained]Amide, as a pale red crystal. Ms (isp): 306.1[ M + H ] M/z]+。
Intermediate A4B
Starting from (R) -2, 2-difluoro-3- (2-fluoro-phenyl) -3- ((R) -2-methyl-propane-2-sulfinylamino) -butyric acid ethyl ester, the product 2-methyl-propane-2-sulfinic acid [ (R) -2, 2-bisFluoro-1- (2-fluoro-phenyl) -3-hydroxy-1-methyl-propyl]Amide as a white solid. Ms (isp): 324.2[ M + H ] M/z]+。
Intermediate A4C
Starting from (R) -2, 2-difluoro-3- (2-fluoro-phenyl) -3- ((R) -2-methyl-propane-2-sulfinylamino) -pentanoic acid ethyl ester, the product 2-methyl-propane-2-sulfinic acid [ (R) -1-ethyl-2, 2-difluoro-1- (2-fluoro-phenyl) -3-hydroxy-propyl is obtained]Amide as a white solid. Ms (isp): 338.1[ M + H ] M/z]+。
Intermediate sulfinamide nitrile A5
General procedure:
to a solution of sulfinamidol A4(4.1mmol) in dichloromethane (23ml) was added 2-bromoacetonitrile (6.2mmol), silver (I) oxide (1.9g) and tetrabutylammonium iodide (0.30g) successively at 22 ℃ and stirring was continued for 2 h. The suspension was filtered and the filtrate was taken up with saturated NaHCO3The aqueous solution was washed, the organic layer was dried and evaporated to give crude sulfinamide nitrile a5, which was used without further purification.
Intermediate A5A
Starting from (R) -2-methyl-propane-2-sulfinic acid [ (1R, 2R) -2-fluoro-1- (2-fluoro-phenyl) -3-hydroxy-1-methyl-propyl]-amide to obtain the product (R) -N- ((2R, 3R) -4- (cyanomethoxy) -3-fluoro-2- (2-fluorophenyl) but-2-yl) -2-methylpropane-2-sulfinamide as a pale yellow oil. Ms (isp): 345.2[ M + H ] M/z]+。
Intermediate A5B
Starting from 2-methyl-propane-2-sulfinic acid [ (R) -2, 2-difluoro-1- (2-fluoro-phenyl) -3-hydroxy-1-methyl-propyl]Amide to obtain the product 2-methyl-propane-2-sulfinic acid [ (R) -3-cyanomethoxy-2, 2-difluoro-1- (2-fluoro-phenyl) -1-methyl-propyl]Amides, as pale yellow oils. Ms (isp): 363.2[ M + H ] M/z]+。
Intermediate A5C
Starting from 2-methyl-propane-2-sulfinic acid [ (R) -1-ethyl-2, 2-difluoro-1- (2-fluoro-phenyl) -3-hydroxy-propyl]Amide, obtaining the product 2-methyl-propane-2-sulfinic acid [ (R) -3-cyanomethoxy-1-ethyl-2, 2-difluoro-1- (2-fluoro-phenyl) -propyl]Amides, as pale yellow oils. Ms (isp): 377.3[ M + H ] M/z]+。
Synthesis of intermediate aminonitrile A6
General procedure:
a solution of sulfoximine A5(4.25mmol) in 1, 4-dioxane (20ml) was treated with a solution of HCl in 1, 4-dioxane (4M, 5.3ml) and stirring was continued for 1h at 22 ℃. The mixture was diluted with ethyl acetate and saturated Na2CO3The aqueous solution was washed, the organic layer was dried and evaporated. The crude material was purified on silica using n-heptane/ethyl acetate to give pure aminonitrile a 6.
Intermediate A6A
Starting from (R) -N- ((2R, 3R) -4- (cyanomethoxy) -3-fluoro-2- (2-fluorophenyl) but-2-yl) -2-methylpropane-2-sulfinamide, the product 2- ((2R, 3R) -3-amino-2-fluoro-3- (2-fluorophenyl) butoxy) acetonitrile was obtained as a pale yellow oil. Ms (isp): m/z 241.1[ M + H ]]+。
Intermediate A6B
Starting from 2-methyl-propane-2-sulfinic acid [ (R) -3-cyanomethoxy-2, 2-difluoro-1- (2-fluoro-phenyl) -1-methyl-propyl]Amide to obtain the product [ (R) -3-amino-2, 2-difluoro-3- (2-fluoro-phenyl) -butoxy]Acetonitrile as a light yellow oil. Ms (isp): 259.1[ M + H ] M/z]+。
Intermediate A6C
Starting from 2-methyl-propane-2-sulfinic acid [ (R) -3-cyanomethoxy-1-ethyl-2, 2-difluoro-1- (2-fluoro-phenyl) -propyl]Amide to obtain the product [ (R) -3-amino-2, 2-difluoro-3- (2-fluoro-phenyl) -pentyloxy group]Acetonitrile as a light yellow oil. MS: 273.1[ M + H ] M/z]+。
Intermediate 1, 4-oxaza
Synthesis of A7
General procedure:
to a solution of aminonitrile A6(2.20mmol) in toluene (38ml) was added AlMe at 22 deg.C3Solution in toluene (2M, 1.2ml) and the mixture was heated to 80 ℃ for 1 h. The mixture was cooled to 0 ℃ and saturated Na was added2CO3The aqueous solution was diluted and the aqueous layer was extracted with ethyl acetate. The combined organic layers were dried, evaporated and the residue was taken up in NH2Purification on silica by chromatography using n-heptane/ethyl acetate to give pure 1, 4-oxazaA7。
Intermediate A7A
Starting from 2- ((2R, 3R) -3-amino-2-fluoro-3- (2-fluorophenyl) butoxy) acetonitrile, the product (5R, 6R) -6-fluoro-5- (2-fluorophenyl) -5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza is obtained-3-amine as a light yellow solid. Ms (isp): m/z 241.2[ M + H ]]+。
Intermediate A7B
Starting from [ (R) -3-amino-2, 2-difluoro-3- (2-fluoro-phenyl) -butoxy]-acetonitrile to obtain the product (R) -6, 6-difluoro-5- (2-fluoro-phenyl) -5-methyl-2, 5, 6, 7-tetrahydro- [1, 4[ ] -]Oxazazem-3-ylamine as a white solid. Ms (isp): 259.1[ M + H ] M/z]+。
Intermediate A7C
Starting from [ (R) -3-amino-2, 2-difluoro-3- (2-fluoro-phenyl) -pentyloxy]-acetonitrile to obtain the product (R) -5-ethyl-6, 6-difluoro-5- (2-fluoro-phenyl) -2, 5, 6, 7-tetrahydro- [1, 4-]Oxazazem-3-ylamine as a brown oil. MS: 273.1[ M + H ] M/z]+。
Intermediate nitrobenzene A8
General procedure:
to the intermediate 1, 4-oxazaA solution of A7(1.2mmol) in sulfuric acid (5.0ml) was added red fuming nitric acid (1.9mmol) at 0 ℃ over a period of 20 minutes and stirring was continued for 30 minutes. The solution was slowly added dropwise to ice/water (60ml), the pH was adjusted to 9 by addition of 4N aqueous NaOH and extracted with ethyl acetate. The organic layer was dried, evaporated and the residue was washed with silica-NH2Purified by chromatography using n-heptane/ethyl acetate to give nitrobenzene A8.
Intermediate A8A
Starting from (5R, 6R) -6-fluoro-5- (2-fluorophenyl) -5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxa-nHetero compound-3-amine to obtain the product (5R, 6R) -6-fluoro-5- (2-fluoro-5-nitro-phenyl) -5-methyl-2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-ylamine as a pale yellow solid. Ms (isp): 286.2[ M + H ] M/z]+。
Intermediate A8B
Starting from (R) -6, 6-difluoro-5- (2-fluoro-phenyl) -5-methyl-2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-ylamine to obtain the product (R) -6, 6-difluoro-5- (2-fluoro-5-nitro-phenyl) -5-methyl-2, 5, 6, 7-tetrahydro- [1, 4%]Oxazazem-3-ylamine as a white solid. Ms (isp): 304.1[ M + H ] M/z]+。
Intermediate A8C
Starting from (R) -5-ethyl-6, 6-difluoro-5- (2-fluoro-phenyl) -2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-ylamine to obtain the product (R) -5-ethyl-6, 6-difluoro-5- (2-fluoro-5-nitro-phenyl) -2, 5, 6, 7-tetrahydro-[1,4]Oxazazem-3-ylamine as a pale yellow solid. MS: 318.1[ M + H ] M/z]+。
Synthesis of intermediate aniline A9
General procedure:
a suspension of intermediate nitrobenzene A8(1.0mmol) in ethanol (9ml) and Pd/C (10%, 100mg) was hydrogenated at 22 ℃ and atmospheric pressure for 2 h. The suspension was filtered and the residue was evaporated to give crude aniline a10, which was used without further purification.
Intermediate A9A
Starting from (5R, 6R) -6-fluoro-5- (2-fluoro-5-nitro-phenyl) -5-methyl-2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-ylamine to obtain the product (5R, 6R) -5- (5-amino-2-fluorophenyl) -6-fluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-3-amine as a light yellow solid. Ms (isp): m/z 256.3[ M + H ]]+。
Intermediate A9B
Starting from (R) -6, 6-difluoro-5- (2-fluoro-5-nitro-phenyl) -5-methyl-2, 5, 6, 7-tetrakisHydrogen- [1, 4 ]]Oxazazem-3-ylamine to obtain the product (R) -5- (5-amino-2-fluoro-phenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro- [1, 4%]Oxazazem-3-ylamine as a white solid. Ms (isp): 274.1[ M + H ] M/z]+。
Intermediate A9C
Starting from (R) -5-ethyl-6, 6-difluoro-5- (2-fluoro-5-nitro-phenyl) -2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-ylamine to obtain the product (R) -5- (5-amino-2-fluoro-phenyl) -5-ethyl-6, 6-difluoro-2, 5, 6, 7-tetrahydro- [1, 4%]Oxazazem-3-ylamine as a pale yellow solid. MS: 288.1[ M + H ] M/z]+。
Synthesis of amide Ia from Aniline A9
General procedure:
to a solution of the acid (0.16mmol) in MeOH (1ml) was added 4- (4, 6-dimethoxy-1, 3, 5-triazin-2-yl) -4-methyl-morpholine chloride (0.19mmol) at 22 deg.C and stirring continued for 30 min at 0 deg.C. A solution of aniline A9(0.15mmol) in MeOH (2ml) was added to the mixture and stirring was continued for 2h at 0 ℃. The mixture was saturated with Na2CO3The aqueous solution was diluted, MeOH evaporated and the aqueous solution extracted with ethyl acetate. Drying the organic layer, evaporating and leaving the residue in the form of a preparationPurification on a HPLCRP18 column using a gradient of water/HCOOH (99.9: 0.1) - > MeOH to give the formate, or water/NEt3(99.9∶0.1)->CH3Gradient purification of CN to give the free base of amide Ia.
Synthesis of amine Ib from aniline A9 by reductive amination
General procedure:
to a solution of aniline A9(0.1mmol) in dichloromethane (0.7ml) was added successively carbonyl compound (0.11mmol), acetic acid (0.2mmol) and sodium triacetoxyborohydride (0.14mmol) at 22 ℃ and stirring of the mixture was continued for 18 h. The mixture was diluted with water (1ml) and the organic layer was washed with saturated NaHCO3The aqueous solution was washed, dried and evaporated. The residue is washed with silica-NH2The column was purified by chromatography using dichloromethane to give amine Ib.
Example 1
(R) -N2- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -3-fluoropyridine-2, 5-dicarboxamide formate salt
(R) -5- (5-amino-2-fluorophenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaCoupling of-3-amine (intermediate A9B) and 5-carbamoyl-3-fluoro-pyridine-2-carboxylic acid (prepared according to Hori, a. et al, international patent application publication No. WO 2009151098) yielded the title compound as an off-white amorphous. Ms (isp): 440.2[ M + H ] M/z]+。
Example 2
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -6- (cyclopropylmethoxy) pyridinecarboxamide formate salt
(R) -5- (5-amino-2-fluorophenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaCoupling of-3-amine (intermediate A9B) and 6-cyclopropylmethoxy-pyridine-2-carboxylic acid yielded the title compound as an off-white amorphous form. Ms (isp): 449.2[ M + H ] M/z]+。
6-cyclopropylmethoxy-pyridine-2-carboxylic acid was obtained as follows:
a solution of 6-chloro-pyridine-2-carboxylic acid (9.46mmol) in anhydrous dimethylsulfoxide (5ml) was treated with cyclopropyl-methanol (14.1mmol) followed by treatment with powdered potassium hydroxide (37.8 mmol). The reaction mixture was then irradiated in a microwave oven at 100 ℃ for 90 minutes. For work-up, the reaction mixture was quenched with aqueous citric acid (10%, pH4-5) before extraction with ethyl acetate (5 × 30ml) followed by extraction with a mixture of methanol and dichloromethane (20%; 5 × 100 ml). The combined organic layers were washed with brine (200ml), dried and evaporated under reduced pressure. Lyophilization of the residue yielded 6-chloro-pyridine-2-carboxylic acid as a brown solid (48% of theory) ms (isp): m/z 195.0[ M + H ]]+。
Example 3
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (thiazol-2-yl) pyridinecarboxamide formate salt
(R) -5- (5-amino-2-fluorophenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaCoupling of-3-amine (intermediate A9B) and 5-thiazol-2-yl-pyridine-2-carboxylic acid (prepared according to Suzuki, y, et al, international patent application publication No. WO 2009091016) yielded the title compound as an off-white amorphous. Ms (isp): 462.2[ M + H ] M/z]+。
Example 4
(R) -N- (3- (3-amino-5-ethyl-6, 6-difluoro-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5-cyclopropyloxazole-4-carboxamide
(R) -5- (5-amino-2-fluoro-phenyl) -5-ethyl-6, 6-difluoro-2, 5, 6, 7-tetrahydro- [1, 4]OxazazemCoupling of (E) -3-ylamine (intermediate A9C) and 5-cyclopropyl-oxazole-4-carboxylic acidThe title compound was produced as a white solid. MS: 423.2[ M + H ] M/z]+。
Example 5
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (methylthio) pyrazine-2-carboxamide formate salt
(R) -5- (5-amino-2-fluorophenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaThe coupling of-3-amine (intermediate A9B) and 5-methylthio-pyrazine-2-carboxylic acid (prepared according to Suzuki, y, et al, international patent application publication No. WO 2009091016) yielded the title compound as an off-white solid. Ms (isp): m/z 426.1[ M + H ]]+。
Example 6
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (cyclopropylethynyl) pyridinecarboxamide formate salt
(R) -5- (5-amino-2-fluorophenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaCoupling of-3-amine (intermediate A9B) and 5-cyclopropylethynyl-pyridine-2-carboxylic acid (prepared according to Suzuki, y, et al, international patent application publication No. WO 2009091016) yielded the title compound as an off-white solid. Ms (isp): 443.3[ M + H ] M/z]+。
Example 7
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5-cyclopropylpyridinecarboxamide formate salt
(R) -5- (5-amino-2-fluorophenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaCoupling of-3-amine (intermediate A9B) and 5-cyclopropyl-pyridine-2-carboxylic acid (prepared according to Suzuki, y, et al, international patent application publication No. WO 2009091016) yielded the title compound as an amorphous light yellow material. Ms (isp): m/z 419.2[ M + H ]]+。
Example 8
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (3-methoxyprop-1-ynyl) picolinoylAmine formate salt
(R) -5- (5-amino-2-fluorophenyl) -6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaCoupling of-3-amine (intermediate A9B) and 5- (3-methoxy-prop-1-ynyl) -pyridine-2-carboxylic acid (prepared according to Suzuki, y, et al, international patent application publication No. WO 2009091016) yielded the title compound as a white solid. Ms (isp): 410.2[ M + H ] M/z]+。
Example 9
(5R, 6R) -5- (5- (cyclopropylmethylamino) -2-fluorophenyl) -6-fluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-3-amines
(5R, 6R) -5- (5-amino-2-fluorophenyl) -6-fluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxazaReductive amination of the-3-amine (intermediate A9A) and cyclopropanecarboxaldehyde gave the title compound as a colorless solid. MS: 310.4[ M + H ] M/z]+。
Claims (26)
1. A compound of formula I
Wherein
R1Is H or F;
R2is C1-7-an alkyl group; and is
R3Is- (C ═ O) -R4Or R5Wherein
R4Is heteroaryl substituted with one substituent selected from the group consisting of: c3-7-cycloalkyl-C1-7-alkoxy-, C3-7-cycloalkyl-C2-7-alkynyl-, C1-7-alkoxy-C2-7-alkynyl-, unsubstituted heteroaryl, unsubstituted C3-7-cycloalkyl and C1-7-alkyl-S-, or
R4Is heteroaryl substituted with one halogen and one amide group;
R5is C3-7-cycloalkyl-C1-7-alkyl-;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, wherein R1Is F.
3. The compound of claim 1, wherein R1Is H.
4. The compound of any one of claims 1-3, wherein R2Is Me.
5. The compound of any one of claims 1-3, wherein R2Is Et.
6. The compound of any one of claims 1-5, wherein R3Is C3-7-cycloalkyl-C1-7-alkyl-.
7. The compound of any one of claims 1-5, wherein R3Is cyclopropyl-CH2-。
8. The compound of claim 7, which is (5R, 6R) -5- [5- (cyclopropylmethyl-ammonia)2-fluoro-phenyl radical]-6-fluoro-5-methyl-2, 5, 6, 7-tetrahydro- [1, 4]Oxazazem-3-amine.
9. The compound of any one of claims 1-5, wherein R3Is- (C ═ O) -R4。
10. The compound of claim 9, wherein R4Is pyridyl substituted with halogen and one amido group.
11. The compound of claim 9 which is (R) -N2- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -3-fluoropyridine-2, 5-dicarboxamide formate.
12. The compound of claim 9, wherein R4Is selected from C3-7-cycloalkyl-C1-7-alkoxy-, C3-7-cycloalkyl-C2-7-alkynyl-, C1-7-alkoxy-C2-7-alkynyl-, unsubstituted heteroaryl and unsubstituted C3-7-pyridyl substituted with one substituent of the group consisting of cycloalkyl.
13. The compound of claim 12, selected from the group consisting of:
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -6- (cyclopropylmethoxy) picolinamide formate salt,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (thiazol-2-yl) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (cyclopropylethynyl) pyridinecarboxamide formate,
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5-cyclopropylpyridinecarboxamide formate salt, and
(R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza)-5-yl) -4-fluorophenyl) -5- (3-methoxyprop-1-ynyl) pyridinecarboxamide formate.
14. The compound of claim 9, wherein R4Is a quilt C1-7-alkyl-S-substituted pyrazinyl.
15. The compound of claim 14, which is (R) -N- (3- (3-amino-6, 6-difluoro-5-methyl-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5- (methylthio) pyrazine-2-carboxamide formate salt.
16. The compound of claim 9, wherein R4Is a quilt C3-7-cycloalkyl-substituted oxazolyl.
17. The compound of claim 16, which is (R) -N- (3- (3-amino-5-ethyl-6, 6-difluoro-2, 5, 6, 7-tetrahydro-1, 4-oxaza-5-yl) -4-fluorophenyl) -5-cyclopropyloxazole-4-carboxamide.
18. A process for the preparation of a compound of formula I as defined in any one of claims 1 to 21, which process comprises:
a) reacting an amine of formula II with a carboxylic acid of formula III in the presence of a coupling agent under basic conditions or with the aid of a triazine derivative to obtain a compound of formula Ia
Wherein R is1、R2And R4As defined in claim 1, or alternatively,
b) reacting an amine of formula II with a compound of formula IV in the presence of acetic acid and sodium triacetoxyborohydride to obtain a compound of formula Ib
Wherein R is hydrogen or C1-7-alkyl, and wherein R1、R2、R3And R5As defined in claim 1.
19. A pharmaceutical composition comprising a compound of formula I according to any one of claims 1 to 17 and a pharmaceutically acceptable carrier and/or excipient.
20. A compound of formula I according to any one of claims 1 to 17 for use as a medicament.
21. A compound of formula I according to any one of claims 1 to 17 for use in the therapeutic and/or prophylactic treatment of alzheimer's disease and/or type2 diabetes.
22. A compound of formula I according to any one of claims 1-17 for use as therapeutically active substance for the therapeutic and/or prophylactic treatment of: amyotrophic Lateral Sclerosis (ALS), arterial thrombosis, autoimmune/inflammatory diseases, cancer such as breast cancer, cardiovascular diseases such as myocardial infarction and stroke, dermatomyositis, down's syndrome, gastrointestinal diseases, glioblastoma multiforme, graves' disease, huntington's disease, Inclusion Body Myositis (IBM), inflammatory responses, kaposi's sarcoma, costerman's disease, lupus erythematosus, macrophage myofascitis, juvenile idiopathic arthritis, granulomatous arthritis, malignant melanoma, multiple myeloma, rheumatoid arthritis, sjogren's syndrome, spinocerebellar ataxia type 1, spinocerebellar ataxia type 7, whipple's disease, or wilson's disease.
23. Use of a compound of formula I according to any one of claims 1 to 17 for the preparation of a medicament for the therapeutic and/or prophylactic treatment of alzheimer's disease and/or type2 diabetes.
24. A method for the therapeutic and/or prophylactic treatment of diseases which can be ameliorated by the inhibition of BACE1 and/or BACE2 activity, particularly for the treatment of alzheimer's disease and type2diabetes, which method comprises administering to a human or animal a therapeutically active amount of a compound of formula I according to any one of claims 1 to 17.
25. A method for the therapeutic and/or prophylactic treatment of a disease which can be ameliorated by the inhibition of BACE1 and/or BACE2 activity, in particular the treatment of Amyotrophic Lateral Sclerosis (ALS), arterial thrombosis, autoimmune/inflammatory diseases, cancer such as breast cancer, cardiovascular diseases such as myocardial infarction and stroke, dermatomyositis, Down syndrome, gastrointestinal diseases, glioblastoma multiforme, Graves ' disease, Huntington's disease, Inclusion Body Myositis (IBM), inflammatory responses, Kaposi's sarcoma, Coasteman's disease, lupus erythematosus, macrophage myofasciitis, juvenile idiopathic arthritis, granulomatous arthritis, malignant melanoma, multiple myeloma, rheumatoid arthritis, Sjogren's syndrome, spinocerebellar ataxia 1, spinocerebellar ataxia 7, Whipple's disease or Wilson's disease, the method comprising administering a therapeutically active amount of a compound of formula I according to any one of claims 1 to 17 to a human being or animal.
26. The invention as hereinbefore described.
Applications Claiming Priority (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP11155051.3 | 2011-02-18 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| HK1191328A true HK1191328A (en) | 2014-07-25 |
Family
ID=
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US8815841B2 (en) | 1,4-Oxazepines as BACE1 and/or BACE2 inhibitors | |
| JP5823503B2 (en) | 2,5,6,7-tetrahydro- [1,4] oxazepin-3-ylamine or 2,3,6,7-tetrahydro- [1,4] oxazepin-5-ylamine compound | |
| US9115098B2 (en) | 1,3-oxazines as BACE1 and/or BACE2 inhibitors | |
| EP2729464B1 (en) | Cyclopropyl-fused-1,3-thiazepines as bace 1 and/or bace 2 inhibitors | |
| CA2837252A1 (en) | Halogen-alkyl-1,3 oxazines as bace1 and/or bace2 inhibitors | |
| HK1191328A (en) | 1,4-oxazepines as bace1 and/or bace2 inhibitors | |
| TW201307307A (en) | 1,3-oxazines as BACE1 and/or BACE2 inhibitors | |
| HK1193593A (en) | Cyclopropyl-fused-1,3-thiazepines as bace 1 and/or bace 2 inhibitors | |
| NZ614545B2 (en) | 1,3 oxazines as bace1 and/or bace2 inhibitors | |
| HK1179617B (en) | 2,5,6,7-tetrahydro-[1,4]oxazepin-3-ylamine or 2,3,6,7-tetrahydro-[1,4]oxazepin-5-ylamine compounds | |
| HK1191943A (en) | 1,3 oxazines as bace1 and/or bace2 inhibitors | |
| HK1193600A (en) | Halogen-alkyl-1,3 oxazines as bace1 and/or bace2 inhibitors | |
| NZ617507B2 (en) | Halogen-alkyl-1,3 oxazines as bace1 and/or bace2 inhibitors |