WO2012061972A1 - Selective glycosidase inhibitors and uses thereof - Google Patents

Selective glycosidase inhibitors and uses thereof Download PDF

Info

Publication number
WO2012061972A1
WO2012061972A1 PCT/CN2010/078528 CN2010078528W WO2012061972A1 WO 2012061972 A1 WO2012061972 A1 WO 2012061972A1 CN 2010078528 W CN2010078528 W CN 2010078528W WO 2012061972 A1 WO2012061972 A1 WO 2012061972A1
Authority
WO
WIPO (PCT)
Prior art keywords
pyrano
tetrahydro
thiazole
diol
methyl
Prior art date
Application number
PCT/CN2010/078528
Other languages
French (fr)
Inventor
Kun Liu
Ernest J. Mceachern
Changwei Mu
Harold G. Selnick
David J. Vocadlo
Yaode Wang
Zhongyong Wei
Yuanxi Zhou
Yongbao Zhu
Original Assignee
Alectos Therapeutics Inc.
Merck Sharp & Dohme Corp.
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 Alectos Therapeutics Inc., Merck Sharp & Dohme Corp. filed Critical Alectos Therapeutics Inc.
Priority to PCT/CN2010/078528 priority Critical patent/WO2012061972A1/en
Priority to PCT/CN2011/080691 priority patent/WO2012062157A1/en
Priority to TW100140566A priority patent/TW201249848A/en
Priority to EP11840448.2A priority patent/EP2637503B1/en
Priority to KR1020137014710A priority patent/KR101922266B1/en
Priority to US13/883,811 priority patent/US8901087B2/en
Priority to ES11840448.2T priority patent/ES2600028T3/en
Priority to RU2013126531/04A priority patent/RU2592285C2/en
Priority to BR112013011441A priority patent/BR112013011441B8/en
Priority to AU2011326186A priority patent/AU2011326186A1/en
Priority to CA2816798A priority patent/CA2816798C/en
Priority to MX2013005118A priority patent/MX336611B/en
Priority to JP2013537923A priority patent/JP5965407B2/en
Priority to PCT/US2011/059668 priority patent/WO2012064680A1/en
Priority to ARP110104166A priority patent/AR083798A1/en
Publication of WO2012061972A1 publication Critical patent/WO2012061972A1/en
Priority to AU2016202186A priority patent/AU2016202186B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • 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
    • A61P25/28Drugs 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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

Definitions

  • This application relates to compounds which selectively inhibit glycosidases and uses thereof.
  • a wide range of cellular proteins, both nuclear and cytoplasmic, are post- translationally modified by the addition of the monosaccharide 2-acetamido-2-deoxy-B-D- glucopyranoside ( ⁇ - ⁇ -acetylglucosamine) which is attached via an O-glycosidic linkage.1
  • This modification is generally referred to as O-linked N-acetylglucosamine or O-GlcNAc.
  • the enzyme responsible for post-translationally linking ⁇ - ⁇ -acetylglucosamine (GlcNAc) to specific serine and threonine residues of numerous nucleocytoplasmic proteins is O-GlcNAc transferase (OGT).2-5
  • O-GlcNAc transferase O-GlcNAc transferase2-5
  • a second enzyme, known as 0-GlcNAcase6,7 removes this post- translational modification to liberate proteins making the O-GlcNAc-modification a dynamic cycle occurring several times during the lifetime of a protein.8
  • O-GlcNAc-modified proteins regulate a wide range of vital cellular functions including, for example, transcription, 9- 12 proteasomal degradation, 13 and cellular signaling.14
  • O-GlcNAc is also found on many structural proteins.15-17 For example, it has been found on a number of cytoskeletal proteins, including neurofilament proteins, 18, 19 synapsins,6,20 synapsin-specific clathrin assembly protein AP-3,7 and ankyrinG.14
  • O- GlcNAc modification has been found to be abundant in the brain.21,22 it has also been found on proteins clearly implicated in the etiology of several diseases including Alzheimer's disease (AD) and cancer.
  • AD Alzheimer's disease
  • AD and a number of related tauopathies including Downs' syndrome, Pick's disease, Niemann-Pick Type C disease, and amyotrophic lateral sclerosis (ALS) are characterized, in part, by the development of neurofibrillary tangles (FTs).
  • FTs neurofibrillary tangles
  • PHFs paired helical filaments
  • tau Normally tau stabilizes a key cellular network of microtubules that is essential for distributing proteins and nutrients within neurons.
  • tau becomes hyperphosphorylated, disrupting its normal functions, forming PHFs and ultimately aggregating to form FTs.
  • O-GlcNAc is a dynamic modification that can be removed and reinstalled several times during the lifespan of a protein.
  • O-GlcNAcase has been mapped to a chromosomal locus that is linked to AD.7,42 Hyperphosphorylated tau in human AD brains has markedly lower levels of O-GlcNAc than are found in healthy human brains.21 Very recently, it has been shown that O-GlcNAc levels of soluble tau protein from human brains affected with AD are markedly lower than those from healthy brain.21 Furthermore, PHF from diseased brain was suggested to lack completely any O- GlcNAc modification whatsoever.21 The molecular basis of this hypoglycosylation of tau is not known, although it may stem from increased activity of kinases and/or dysfunction of one of the enzymes involved in processing O-GlcNAc.
  • Neurons do not store glucose and therefore the brain relies on glucose supplied by blood to maintain its essential metabolic functions. Notably, it has been shown that within brain, glucose uptake and metabolism decreases with aging.43 Within the brains of AD patients marked decreases in glucose utilization occur and are thought to be a potential cause of neurodegeneration.44 The basis for this decreased glucose supply in AD brain45-47 i s thought to stem from any of decreased glucose transport,48,49 impaired insulin signaling, 50,51 a nd decreased blood flow.52
  • OGT recognizes many of its substrates54,55 and binding partners41,56 through its tetratricopeptide repeat (TPR) domains.57,58 As described above, 0-GlcNAcase6,7 removes this post-translational modification to liberate proteins making the O-GlcNAc-modification a dynamic cycle occurring several times during the lifetime of a protein.8 O-GlcNAc has been found in several proteins on known phosphorylation sites, 10,37,38,59 including tau and neurofilaments.60 Additionally, OGT shows unusual kinetic behaviour making it extraordinarly sensitive to intracellular UDP-GlcNAc substrate concentrations and therefore glucose supply.41
  • TPR tetratricopeptide repeat
  • therapeutically useful intervention61 comes from recent studies showing that when transgenic mice harbouring human tau are treated with kinase inhibitors, they do not develop typical motor defects33 and, in another case,32 show decreased levels of insoluble tau.
  • Recent studies63 support the therapeutic potential of small-molecule O- GlcNAcase inhibitors to limit tau hyperphosphorylation for treatment of AD and related tauopathies.
  • the O-GlcNAcase inhibitor thiamet-G has been implicated in the reduction of tau phosphorylation in cultured PC- 12 cells at pathologically relevant sites.
  • oral administration of thiamet-G to healthy Sprague-Dawley rats has been implicated in reduced phosphorylation of tau at Thr231, Ser396 and Ser422 in both rat cortex and hippocampus.63 .
  • O-GlcNAcase O- glycoprotein 2-acetamido-2-deoxy-P-D-glucopyranosidase
  • O-GlcNAcase is a member of family 84 of glycoside hydrolases that includes enzymes from organisms as diverse as prokaryotic pathogens to humans (for the family classification of glycoside hydrolases see Coutinho, P.M. & Henrissat, B.
  • O-GlcNAcase acts to hydrolyse O-GlcNAc off of serine and threonine residues of post-translationally modified proteins.1,6,7,80,81 Consistent with the presence of O-GlcNAc on many intracellular proteins, the enzyme O- GlcNAcase appears to have a role in the etiology of several diseases including type II diabetes, 14,82 AD, 16,21,83 a nd cancer.22,84 Although O-GlcNAcase was likely isolated earlier on, 18, 19 about 20 years elapsed before its biochemical role in acting to cleave O- GlcNAc from serine and threonine residues of proteins was understood.6 More recently O- GlcNAcase has been cloned,7 partially characterized,20 and suggested to have additional activity as a histone acet
  • HEXA and HEXB encode enzymes catalyzing the hydrolytic cleavage of terminal ⁇ - ⁇ -acetylglucosamine residues from glycoconjugates.
  • the gene products of HEXA and HEXB predominantly yield two dimeric isozymes,
  • Hexosaminidase A a heterodimeric isozyme
  • Hexosaminidase B a homodimeric isozyme
  • the two subunits, a- and B- bear a high level of sequence identity. Both of these enzymes are classified as members of family 20 of glycoside hydrolases and are normally localized within lysosomes.
  • glycosidases87-90 As a result of the biological importance of these ⁇ -N-acetyl-glucosaminidases, small molecule inhibitors of glycosidases87-90 have received a great deal of attention,91 both as tools for elucidating the role of these enzymes in biological processes and in developing potential therapeutic applications.
  • the control of glycosidase function using small molecules offers several advantages over genetic knockout studies including the ability to rapidly vary doses or to entirely withdraw treatment.
  • NAG-thiazoline has been found to be a potent inhibitor of family 20 hexosaminidases,90, 109 and more recently, the family 84 O-GlcNAcases.108 Despite its potency, a downside to using NAG-thiazoline in a complex biological context is that it lacks selectivity and therefore perturbs multiple cellular processes.
  • PUGNAc is another compound that suffers from the same problem of lack of selectivity, yet has enjoyed use as an inhibitor of both human 0-GlcNAcase6, 110 and the family 20 human ⁇ -hexosaminidases.111
  • This molecule developed by Vasella and coworkers, was found to be a potent competitive inhibitor of the ⁇ -N-acetyl-glucosaminidases from Canavalia ensiformis, Mucor rouxii, and the ⁇ -hexosaminidase from bovine kidney.88 it has been demonstrated that administration of PUGNAc in a rat model of trauma hemorrhage decreases circulating levels of the pro-inflammatory cytokines TNF-a and IL-6.112 It has also been shown that administration of PUGNAc in a cell-based model of lymphocyte activation decreases production of the cytokine IL-2.113 Recent studies have indicated that PUGNAc can be used in an animal model to reduce myocardial infarct size
  • occlusions are particularly significant.
  • elevation of O-GlcNAc levels by administration of PUGNAc, an inhibitor of O-GlcNAcase, in a rat model of trauma hemorrhage improves cardiac function.112, 115 i n addition, elevation of O-GlcNAc levels by treatment with PUGNAc in a cellular model of ischemia/reperfusion injury using neonatal rat ventricular myocytes improved cell viability and reduced necrosis and apoptosis compared to untreated cells.116
  • the invention is directed to compounds for selectively inhibiting glycosidases, uses of the compounds and pharmaceutical compositions including the compounds, and methods of treating diseases and disorders related to deficiency or overexpression of O- GlcNAcase, and/or accumulation or deficiency of O-GlcNAc.
  • the invention encompasses a compound of Formula (I) or a pharmaceutically acceptable salt thereof:
  • each R is independently H or C(0)CH3;
  • Rl and R2 are independently selected from the group consisting of: H, Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl and -(03 ⁇ 4) ⁇ - cyclobutyl wherein n is 0, 1, 2, 3 or 4; or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine, pyrrolidine or piperidine, said Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl, -(CH2)n-cyclobutyl, azetidine, pyrrolidine or piperidine optionally substituted from one up to the maximum number of substituents with fluoro or methyl;
  • R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2- Salkynyl, C3-6cycloalkyl, aryl and heteroaryl, each optionally substituted from one up
  • the invention encompasses a genus of compounds of Formula (la) or a pharmaceutically acceptable salt thereof:
  • Rl and R2 are independently selected from the group consisting of: H, Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl and -(CH2)n-cyclobutyl wherein n is 0, 1, 2, 3 or 4; or
  • Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine, pyrrolidine or piperidine, said Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl, - (CH2)n-cyclobutyl, azetidine, pyrrolidine or piperidine optionally substituted from one up to the maximum number of substituents with fluoro or methyl;
  • R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2-8alkynyl, C3-6cycloalkyl, aryl and heteroaryl, each optionally substituted from one up to the maximum number of substituents with fluoro and OH;
  • R4 is selected from the group consisting of: H, F, Ci-8alkyl, C2-8alkenyl and C2- 8alkynyl, each excluding hydrogen optionally substituted from one up to the maximum number of substituents with fluoro and OH
  • the invention encompasses a first sub-genus of compounds of Formula (la) wherein: Rl and R2 are independently Ci-4alkyl; R3 is Ci-6alkyl; R4 is selected from the group consisting of: H and Ci-6alkyl; and R5 is OH.
  • the invention further encompasses compounds of Formula (la) wherein: Rl and R2 are independently methyl or ethyl; R3 is methyl or ethyl; and R4 is selected from the group consisting of: H, methyl and ethyl.
  • the invention encompasses a second sub-genus of compounds of Formula (la) wherein:R3 and R4 and the carbon atom to which they are attached may join together to form a 3 to 7-membered carbocyclic or heterocyclic ring, said 3 to 7-membered carbocyclic or heterocyclic ring optionally containing a double bond and optionally substituted from one up to the maximum number of substituents with fluoro and OH.
  • the invention encompasses a third sub-genus of compounds of Formula (la) wherein: Rl and R2 are independently selected from the group consisting of: H, Ci-6alkyl and cyclopropylmethyl; or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine or pyrrolidine, said Ci- 6alkyl, cyclopropylmethyl, azetidine or pyrrolidine optionally substituted with 1 to 3 substituents selected from fluoro and methyl; R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2-8alkynyl and C3-6cycloalkyl, each optionally substituted with 1 to 3 substituents selected from fluoro and OH; and R4 is selected from the group consisting of: H, F, Ci-8alkyl, C2-8alkenyl and C2-8alkynyl, each excluding hydrogen optionally substituted with 1 to 3 substituent
  • the invention encompasses a fourth sub-genus of compounds of Formula (la) wherein R3 is CF3 and R5 is OH.
  • the invention also encompasses the compounds that follow or pharmaceutically acceptable salts thereof.
  • the invention also encompasses a pharmaceutical composition comprising the compound of Formula (I) or (la) in combination with a pharmaceutically acceptable carrier.
  • the invention also encompasses a method of selectively inhibiting O- GlcNAcase in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula (I) or (la), or a pharmaceutically acceptable salt thereof.
  • the invention also encompasses a method of elevating the level of O-GlcNAc in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula (I) or (la), or a pharmaceutically acceptable salt thereof.
  • the invention also encompasses a method of treating a condition that is modulated by O-GlcNAcase, in a subject in need thereof, the method comprising
  • An aspect of the invention encompasses this method wherein the condition is selected from one or more of the group consisting of an inflammatory disease, an allergy, asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias, delayed-type hypersensitivity, atherosclerosis, interstitial lung disease (ILD), idiopathic pulmonary fibrosis, ILD associated with rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, systemic sclerosis, Sjogren's syndrome, polymyositis or dermatomyositis, systemic anaphylaxis or hypersensitivity response, drug allergy, insect sting allergy, autoimmune disease, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Guillain-Barre syndrome, systemic lup
  • the invention also encompasses a method of treating a condition selected from the group consisting of a neurodegenerative disease, a tauopathy, cancer and stress, in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula (I) or (la), or a pharmaceutically acceptable salt thereof.
  • An aspect of the invention encompasses this method wherein the condition is selected from one or more of the group consisting of Alzheimer's disease, Amyotrophic lateral sclerosis (ALS), Amyotrophic lateral sclerosis with cognitive impairment (ALSci), Argyrophilic grain dementia, Bluit disease, Corticobasal degeneration (CBD), Dementia pugilistica, Diffuse neurofibrillary tangles with calcification, Down's syndrome, Familial British dementia, Familial Danish dementia, Frontotemporal dementia with parkinsonism linked to
  • chromosome 17 FTDP-17
  • Gerstmann-Straussler-Scheinker disease Guadeloupean parkinsonism
  • Hallevorden-Spatz disease neurodegeneration with brain iron accumulation type 1
  • Multiple system atrophy Myotonic dystrophy
  • Niemann-Pick disease type C
  • Pallido-ponto-nigral degeneration Parkinsonism-dementia complex of Guam, Pick's disease (PiD), Post-encephalitic parkinsonism (PEP), Prion diseases (including Creutzfeldt- Jakob Disease (CJD), Variant Creutzfeldt- Jakob Disease (vCJD), Fatal Familial Insomnia, and Kuru), Progressive supercortical gliosis, Progressive supranuclear palsy (PSP), Richardson's syndrome, Subacute sclerosing panencephalitis, Tangle-only dementia, Huntington's disease, Parkinson's disease, Schizophrenia, Mild Cognitive Impairment (MCI),
  • the stress is a cardiac disorder.
  • the cardiac disorder is selected from one or more of the group consisting of ischemia; hemorrhage; hypovolemic shock; myocardial infarction; an interventional cardiology procedure; cardiac bypass surgery; fibrinolytic therapy; angioplasty; and stent placement.
  • the compounds of the invention are capable of inhibiting an O-glycoprotein 2-acetamido-2-deoxy-P-D-glucopyranosidase (O-GlcNAcase).
  • the O-GlcNAcase is a mammalian O-GlcNAcase, such as a rat, mouse or human O-GlcNAcase.
  • the ⁇ -hexosaminidase is a mammalian ⁇ -hexosaminidase, such as a rat, mouse or human ⁇ -hexosaminidase.
  • a compound that "selectively" inhibits an O-GlcNAcase is a compound that inhibits the activity or biological function of an O- GlcNAcase, but does not substantially inhibit the activity or biological function of a ⁇ - hexosaminidase.
  • a selective inhibitor of an O-GlcNAcase is a compound that inhibits the activity or biological function of an O- GlcNAcase, but does not substantially inhibit the activity or biological function of a ⁇ - hexosaminidase.
  • GlcNAcase selectively inhibits the cleavage of 2-acetamido-2-deoxy ⁇ -D-glucopyranoside (O-GlcNAc) from polypeptides.
  • a selective inhibitor of an O- GlcNAcase selectively binds to an O-GlcNAcase.
  • a selective inhibitor of an O-GlcNAcase inhibits hyperphosphorylation of a tau protein and/or inhibits formations of FTs.
  • a selective inhibitor of an O-GlcNAcase elevates or enhances O-GlcNAc levels e.g., O-GlcNAc-modified polypeptide or protein levels, in cells, tissues, or organs (e.g., in brain, muscle, or heart (cardiac) tissue) and in animals.
  • O-GlcNAc levels e.g., O-GlcNAc-modified polypeptide or protein levels
  • O-GlcNAc-modified polypeptide or protein levels in cells, tissues, or organs (e.g., in brain, muscle, or heart (cardiac) tissue) and in animals.
  • elevating or “enhancing” is meant an increase by any value between 10% and 90%), or of any integer value between 30% and 60%, or over 100%), or an increase by 1-fold, 2-fold, 5-fold, 10-fold, 15-fold, 25-fold, 50-fold, 100-fold or more.
  • a selective inhibitor of an O-GlcNAcase exhibits a selectivity ratio, as described herein, in the range 10 to 100000, or in the range 100 to 100000, or in the range 1000 to 100000, or at least 10, 20, 50, 100, 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 6000, 7000, 10,000, 25,000, 50,000, 75,000, or any value within or about the described range.
  • the compounds of the present invention elevate O-GlcNAc levels on O- GlcN Ac-modified polypeptides or proteins in vivo specifically via interaction with an O- GlcNAcase enzyme, and are effective in treating conditions which require or respond to inhibition of O-GlcNAcase activity.
  • the compounds of the present invention are useful as agents that produce a decrease in tau phosphorylation and NFT formation.
  • the compounds are therefore useful to treat Alzheimer's disease and related tauopathies.
  • the compounds are thus capable of treating Alzheimer's disease and related tauopathies by lowering tau phosphorylation and reducing NFT formation as a result of increasing tau O-GlcNAc levels.
  • the compounds produce an increase in levels of O-GlcNAc modification on O-GlcNAc-modified
  • polypeptides or proteins are therefore useful for treatment of disorders responsive to such increases in O-GlcNAc modification; these disorders include without limitation neurodegenerative, inflammatory, cardiovascular, and immunoregulatory diseases.
  • the compounds are also useful as a result of other biological activites related to their ability to inhibit the activity of glycosidase enzymes.
  • the compounds of the invention are valuable tools in studying the physiological role of O- GlcNAc at the cellular and organismal level.
  • the invention provides methods of enhancing or elevating levels of protein O-GlcNAc modification in animal subjects, such as, veterinary and human subjects. In alternative embodiments, the invention provides methods of selectively inhibiting an O-GlcNAcase enzyme in animal subjects, such as, veterinary and human subjects. In alternative embodiments, the invention provides methods of inhibiting phosphorylation of tau polypeptides, or inhibiting formation of NFTs, in animal subjects, such as, veterinary and human subjects.
  • a compound refers to one or more of such compounds
  • the enzyme includes a particular enzyme as well as other family members and equivalents thereof as known to those skilled in the art.
  • the term "compound” or “compounds” refers to the compounds discussed herein and includes precursors and derivatives of the compounds, including acyl-protected derivatives, and pharmaceutically acceptable salts of the compounds, precursors, and derivatives.
  • the invention also includes prodrugs of the compounds, pharmaceutical compositions including the compounds and a pharmaceutically acceptable carrier, and pharmaceutical compositions including prodrugs of the compounds and a pharmaceutically acceptable carrier.
  • all of the compounds of the invention contain at least one chiral center.
  • the formulations, preparation, and compositions including compounds according to the invention include mixtures of stereoisomers, individual stereoisomers, and enantiomeric mixtures, and mixtures of multiple stereoisomers.
  • the compound may be supplied in any desired degree of chiral purity.
  • Alkyl refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing no unsaturation and including, for example, from one to ten carbon atoms, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms, and which is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, the alkyl group may be optionally substituted by one or more
  • alkenyl refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one double bond and including, for example, from two to ten carbon atoms, such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms, and which is attached to the rest of the molecule by a single bond or a double bond.
  • the alkenyl group may be optionally substituted by one or more substituents as described herein. Unless stated otherwise specifically herein, it is understood that the substitution can occur on any carbon of the alkenyl group.
  • Alkynyl refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond and including, for example, from two to ten carbon atoms. Unless stated otherwise specifically in the specification, the alkenyl group may be optionally substituted by one or more substituents as described herein.
  • Aryl means mono- or bicyclic aromatic rings containing only carbon atoms, including for example, 6-14 members.
  • aryl include phenyl, naphthyl, indanyl, indenyl, tetrahydronaphthyl, 2,3-dihydrobenzofuranyl, dihydrobenzopyranyl, 1,4- benzodioxanyl, and the like.
  • aryl is meant to include aryl groups optionally substituted by one or more substituents as described herein.
  • Heteroaryl refers to a single or fused aromatic ring group containing one or more heteroatoms in the ring, for example N, O, S, including for example, 5-14 members, such as 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 members.
  • heteroaryl groups include furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, isoxazole, isothiazole, 1,2,3-oxadiazole, 1,2,3-triazole, 1,2,4-triazole, 1,3,4-thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, 1,3,5-triazine, imidazole, benzimidazole, benzoxazole, benzothiazole, indolizine, indole, isoindole, benzofuran, benzothiophene, IH-indazole, purine, 4H-quinolizine, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,8- naphthyridine, pteridine. Unless stated otherwise specifically herein,
  • Cycloalkyl refers to a stable monovalent monocyclic, bicyclic or tricyclic hydrocarbon group consisting solely of carbon and hydrogen atoms, having for example from 3 to 15 carbon atoms, and which is saturated and attached to the rest of the molecule by a single bond. Unless otherwise stated specifically herein, the term “cycloalkyl” is meant to include cycloalkyl groups which are optionally substituted as described herein.
  • 3 to 7-membered carbocyclic or heterocyclic ring means a monocylic carbon ring of 3 to 7 atoms or a monocyclic ring of 3 to 7 atoms containing one or more heterotaoms selected from O, N and S.
  • Optional or “optionally” means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not.
  • optionally substituted alkyl means that the alkyl group may or may not be substituted and that the description includes both substituted alkyl groups and alkyl groups having no substitution. Examples of optionally substituted alkyl groups include, without limitation, methyl, ethyl, propyl, etc.
  • optionally substituted alkyl and alkenyl groups include Ci-6 alkyls or alkenyls.
  • Halo refers to bromo, chloro, fluoro, iodo, etc.
  • suitable halogens include fluorine or chlorine.
  • optionally substituted carbonyl groups, or sulfonyl groups include optionally substituted forms of such groups formed from various hydrocarbyls such as alkyl, alkenyl and 5- to 6-membered monocyclic aromatic group (e.g., phenyl, pyridyl, etc.), as described herein.
  • the invention provides methods of treating conditions that are modulated, directly or indirectly, by an O-GlcNAcase enzyme or by O-GlcNAc-modified protein levels, for example, a condition that is benefited by inhibition of an O-GlcNAcase enzyme or by an elevation of O-GlcNAc-modified protein levels.
  • Such conditions include, without limitation, glaucoma, schizophrenia, tauopathies, such as Alzheimer's disease, neurodegenerative diseases, cardiovascular diseases, diseases associated with inflammation, diseases associated with immunosuppression and cancers.
  • the compounds of the invention are also useful in the treatment of diseases or disorders related to deficiency or over-expression of O-GlcNAcase or accumulation or depletion of O-GlcNAc, or any disease or disorder responsive to glycosidase inhibition therapy.
  • diseases and disorders include, but are not limited to, glaucoma, schizophrenia, neurodegenerative disorders, such as Alzheimer's disease (AD), or cancer.
  • diseases and disorders may also include diseases or disorders related to the accumulation or deficiency in the enzyme OGT.
  • a method of protecting or treating target cells expressing proteins that are modified by O-GlcNAc residues, the dysregulation of which modification results in disease or pathology is also included.
  • the term "treating" as used herein includes treatment, prevention, and amelioration.
  • the invention provides methods of enhancing or elevating levels of protein O-GlcNAc modification in animal subjects, such as, veterinary and human subjects.
  • This elevation of O-GlcNAc levels can be useful for the prevention or treatment of Alzheimer's disease; prevention or treatment of other neurodegenerative diseases (e.g. Parkinson's disease, Huntington's disease); providing neuroprotective effects; preventing damage to cardiac tissue; and treating diseases associated with inflammation or immunosuppression.
  • the invention provides methods of selectively inhibiting an O-GlcNAcase enzyme in animal subjects, such as veterinary and human subjects.
  • the invention provides methods of inhibiting phosphorylation of tau polypeptides, or inhibiting formation of FTs, in animal subjects, such as, veterinary and human subjects. Accordingly, the compounds of the invention may be used to study and treat AD and other tauopathies.
  • the methods of the invention are effected by administering a compound according to the invention to a subject in need thereof, or by contacting a cell or a sample with a compound according to the invention, for example, a pharmaceutical composition comprising a therapeutically effective amount of the compound according to Formula (I) or (la). More particularly, they are useful in the treatment of a disorder in which the regulation of O-GlcNAc protein modification is implicated, or any condition as described herein.
  • Disease states of interest include Alzheimer's disease (AD) and related
  • the compounds may be used to block hyperphosphorylation of tau by maintaining elevated levels of O-GlcNAc on tau, thereby providing therapeutic benefit.
  • the effectiveness of the compounds in treating pathology associated with the accumulation of toxic tau species may be confirmed by testing the ability of the compounds to block the formation of toxic tau species in established cellularl 18-120 and/or transgenic animal models of disease.32,33
  • Tauopathies that may be treated with the compounds of the invention include: Alzheimer's disease, Amyotrophic lateral sclerosis (ALS), Amyotrophic lateral sclerosis with cognitive impairment (ALSci), Argyrophilic grain dementia, Bluit disease, Corticobasal degeneration (CBD), Dementia pugilistica, Diffuse neurofibrillary tangles with calcification, Down's syndrome, Familial British dementia, Familial Danish dementia, Frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), Gerstmann-Straussler-Scheinker disease, Guadeloupean parkinsonism, Hallevorden-Spatz disease (neurodegeneration with brain iron accumulation type 1), Multiple system atrophy, Myotonic dystrophy, Niemann- Pick disease (type C), Pallido-ponto-nigral degeneration, Parkinsonism-dementia complex of Guam, Pick's disease (PiD), Post-encephalitic parkinsonism (PE
  • the compounds of this invention are also useful in the treatment of conditions associate with tissue damage or stress, stimulating cells, or promoting differentiation of cells. Accordingly, in some embodiments, the compounds of this invention may be used to provide therapeutic benefit in a variety of conditions or medical procedures involving stress in cardiac tissue, including but not limited to: ischemia; hemorrhage; hypovolemic shock; myocardial infarction; an interventional cardiology procedure; cardiac bypass surgery; fibrinolytic therapy; angioplasty; and stent placement.
  • the effectiveness of the compounds in treating pathology associated with cellular stress may be confirmed by testing the ability of the compounds to prevent cellular damage in established cellular stress assays, 105, 1 16, 1 17 a nd to prevent tissue damage and promote functional recovery in animal models of ischemia- reperfusion,70, 1 14 and trauma-hemorrhage.72, 1 12, 1 15
  • Compounds that selectively inhibit O-GlcNAcase activity may be used for the treatment of diseases that are associated with inflammation, including but not limited to, inflammatory or allergic diseases such as asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias, delayed-type
  • ILD interstitial lung disease
  • autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Guillain-Barre syndrome, systemic lupus erythematosus, myastenia gravis, glomerulonephritis, autoimmune thyroiditis, graft rejection, including allograft rejection or graft-versus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e.g., necrotizing, cutaneous, and hypersensitivity vasculitis); eosin
  • compounds that affects levels of protein O-GlcNAc modification may be used for the treatment of diseases associated with immunosuppression, such as in individuals undergoing chemotherapy, radiation therapy, enhanced wound healing and burn treatment, therapy for autoimmune disease or other drug therapy (e.g., corticosteroid therapy) or combination of conventional drugs used in the treatment of autoimmune diseases and graft/transplantation rejection, which causes immunosuppression; or immunosuppression due to congenital deficiency in receptor function or other causes.
  • the compounds of the invention may be useful for treatment of neurodegenerative diseases, including Parkinson's disease and Huntington's disease. Other conditions that may be treated are those triggered, affected, or in any other way correlated with levels of O-GlcNAc post-translational protein modification.
  • graft rejection in particular but not limited to solid organ transplants, such as heart, lung, liver, kidney, and pancreas transplants (e.g. kidney and lung allografts); cancer, in particular but not limited to cancer of the breast, lung, prostate, pancreas, colon, rectum, bladder, kidney, ovary; as well as non-Hodgkin's lymphoma and melanoma; epilepsy, pain, fibromyalgia, or stroke, e.g., for neuroprotection following a stroke.
  • compositions including compounds according to the invention, or for use according to the invention are contemplated as being within the scope of the invention.
  • pharmaceutical compositions including an effective amount of a compound of Formula (I) or (la) are provided.
  • the compounds of Formula (I) or (la) and their pharmaceutically acceptable salts, stereoisomers, solvates, and derivatives are useful because they have pharmacological activity in animals, including humans.
  • the compounds according to the invention are stable in plasma, when administered to a subject.
  • compounds according to the invention, or for use according to the invention may be provided in combination with any other active agents or pharmaceutical compositions where such combined therapy is useful to modulate O- GlcNAcase activity, for example, to treat neurodegenerative, inflammatory, cardiovascular, or immunoregulatory diseases, or any condition described herein.
  • compounds according to the invention, or for use according to the invention may be provided in combination with one or more agents useful in the prevention or treatment of Alzheimer's disease. Examples of such agents include, without limitation,
  • -acetylcholine esterase inhibitors such as Aricept® (Donepezil), Exelon® (Rivastigmine), Razadyne® (Razadyne ER®, Reminyl®, Nivalin®, Galantamine), Cognex® (Tacrine), Dimebon, Huperzine A, Phenserine, Debio-9902 SR (ZT-1 SR), Zanapezil (TAK0147), ganstigmine, NP7557, etc.; - MDA receptor antagonists such as Namenda® (Axura®, Akatinol®, Ebixa®, Memantine), Dimebon, SGS-742, Neramexane, Debio-9902 SR (ZT-1 SR), etc.; gamma- seer etase inhibitors and/or modulators such as FlurizanTM (Tarenflurbil, MPC-7869, R-flurbipro
  • -beta-secretase inhibitors such as ATG-Z1, CTS-21166, etc.
  • -alpha-secretase activators such as NGX267, etc;
  • -amyloid- ⁇ aggregation and/or fibrillization inhibitors such as AlzhemedTM (3APS, Tramiprosate, 3-amino-l-propanesulfonic acid), AL-108, AL-208, AZD-103, PBT2, Cereact, ONO-2506PO, PPI-558, etc.;
  • -tau aggregation inhibitors such as methylene blue, etc.
  • microtubule stabilizers such as AL-108, AL-208, paclitaxel, etc.
  • -RAGE inhibitors such as TTP488, etc.
  • -5-HTla receptor antagonists such as Xaliproden, Lecozotan, etc.
  • -5-HT4 receptor antagonists such as PRX-03410, etc.
  • -kinase inhibitors such as SRN-003-556, amfurindamide, LiCl, AZD1080, NP031112, SAR-502250, etc.
  • -humanized monoclonal anti- ⁇ antibodies such as Bapineuzumab (AAB- 001), LY2062430, RN1219, ACU-5A5, etc.;
  • -amyloid vaccines such as AN- 1792, ACC-001
  • -neuroprotective agents such as Cerebrolysin, AL-108, AL-208, Huperzine A, etc.;
  • -L-type calcium channel antagonists such as MEM- 1003, etc.
  • -nicotinic receptor antagonists such as AZD3480, GTS-21, etc.
  • -nicotinic receptor agonists such as MEM 3454, Nefiracetam, etc.
  • PPAR peroxisome proliferator-activated receptor
  • PDE4 -phosphodiesterase IV
  • -hormone replacement therapy such as estrogen (Premarin), etc.
  • -monoamine oxidase (MAO) inhibitors such as NS2330, Rasagiline
  • -AMP A receptor modulators such as Ampalex (CX 516), etc.
  • nerve growth factors or NGF potentiators such as CERE-110 (AAV-NGF), T-588, T- 817MA, etc.
  • luteinizing hormone LH
  • VP-4896 leuoprolide
  • -GABA receptor modulators such as AC-3933, NGD 97-1, CP-457920, etc.; benzodiazepine receptor inverse agonists such as SB-737552 (S-8510), AC-3933, etc.;
  • -noradrenaline-releasing agents such as T-588, T-817MA, etc.
  • combination of compounds according to the invention, or for use according to the invention, with Alzheimer's agents is not limited to the examples described herein, but includes combination with any agent useful for the treatment of Alzheimer's disease.
  • Combination of compounds according to the invention, or for use according to the invention, and other Alzheimer's agents may be administered separately or in conjunction.
  • the administration of one agent may be prior to, concurrent to, or subsequent to the administration of other agent(s).
  • the compounds may be supplied as "prodrugs" or protected forms, which release the compound after administration to a subject.
  • the compound may carry a protective group which is split off by hydrolysis in body fluids, e.g., in the bloodstream, thus releasing the active compound or is oxidized or reduced in body fluids to release the compound.
  • a “prodrug” is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention.
  • the term “prodrug” refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable.
  • a prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention.
  • Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a subject.
  • prodrug is also meant to include any covalently bonded carriers which release the active compound of the invention in vivo when such prodrug is
  • Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention.
  • Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively.
  • Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and acetamide, formamide, and benzamide derivatives of amine functional groups in the compounds of the invention and the like.
  • Compounds according to the invention, or for use according to the invention can be provided alone or in combination with other compounds in the presence of a liposome, an adjuvant, or any pharmaceutically acceptable carrier, diluent or excipient, in a form suitable for administration to a subject such as a mammal, for example, humans, cattle, sheep, etc. If desired, treatment with a compound according to the invention may be combined with more traditional and existing therapies for the therapeutic indications described herein.
  • Compounds according to the invention may be provided chronically or intermittently.
  • “Chronic” administration refers to administration of the compound(s) in a continuous mode as opposed to an acute mode, so as to maintain the initial therapeutic effect (activity) for an extended period of time. "Intermittent” administration is treatment that is not consecutively done without interruption, but rather is cyclic in nature. The terms "administration,”
  • “Pharmaceutically acceptable carrier, diluent or excipient” includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier that has been approved, for example, by the United States Food and Drug Administration or other governmental agency as being acceptable for use in humans or domestic animals.
  • compositions in accordance with this invention may comprise a salt of such a compound, preferably a physiologically acceptable salt, which are known in the art.
  • a salt of such a compound preferably a physiologically acceptable salt, which are known in the art.
  • “pharmaceutically acceptable salt” as used herein means an active ingredient comprising compounds of Formula (I) or (la) used in the form of a salt thereof, particularly where the salt form confers on the active ingredient improved pharmacokinetic properties as compared to the free form of the active ingredient or other previously disclosed salt form.
  • a “pharmaceutically acceptable salt” includes both acid and base addition salts.
  • a “pharmaceutically acceptable acid addition salt” refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid,
  • a “pharmaceutically acceptable base addition salt” refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of 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, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine,
  • glucosamine methylglucamine, theobromine, purines, piperazine, piperidine,
  • N-ethylpiperidine N-ethylpiperidine, polyamine resins and the like.
  • pharmaceutically acceptable salt encompasses all acceptable salts including but not limited to acetate, lactobionate, benzenesulfonate, laurate, benzoate, malate, bicarbonate, maleate, bisulfate, mandelate, bitartarate, mesylate, borate,
  • polygalacturonate polygalacturonate, gluconate, salicylate, glutame, stearate, glycollylarsanilate, sulfate, hexylresorcinate, subacetate, hydradamine, succinate, hydrobromide, tannate, hydrochloride, tartrate, hydroxynaphthoate, teoclate, iodide, tosylate, isothionate, triethiodide, lactate, panoate, valerate, and the like.
  • Pharmaceutically acceptable salts of the compounds of the present invention can be used as a dosage for modifying solubility or hydrolysis characteristics, or can be used in sustained release or prodrug formulations.
  • pharmaceutically acceptable salts of the compounds of this invention may include those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc, and from bases such as ammonia, ethylenediamine, N-methyl-glutamine, lysine, arginine, ornithine, choline,
  • compositions will typically include one or more carriers acceptable for the mode of administration of the preparation, be it by injection, inhalation, topical administration, lavage, or other modes suitable for the selected treatment.
  • Suitable carriers are those known in the art for use in such modes of administration.
  • Suitable pharmaceutical compositions may be formulated by means known in the art and their mode of administration and dose determined by the skilled practitioner.
  • a compound may be dissolved in sterile water or saline or a pharmaceutically acceptable vehicle used for administration of non-water soluble compounds such as those used for vitamin K.
  • the compound may be administered in a tablet, capsule or dissolved in liquid form.
  • the table or capsule may be enteric coated, or in a formulation for sustained release.
  • Many suitable formulations are known, including, polymeric or protein microparticles encapsulating a compound to be released, ointments, gels, hydrogels, or solutions which can be used topically or locally to administer a compound.
  • a sustained release patch or implant may be employed to provide release over a prolonged period of time.
  • Many techniques known to skilled practitioners are described in Remington: the Science & Practice of Pharmacy by Alfonso Gennaro, 20th e d., Williams & Wilkins, (2000).
  • Formulations for parenteral administration may, for example, contain excipients, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated naphthalenes.
  • Biocompatible, biodegradable lactide polymer
  • lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds.
  • Other potentially useful parenteral delivery systems for modulatory compounds include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
  • Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.
  • the compounds or pharmaceutical compositions according to the present invention may be administered by oral or non-oral, e.g., intramuscular, intraperitoneal, intravenous, intracisternal injection or infusion, subcutaneous injection, transdermal or transmucosal routes.
  • compounds or pharmaceutical compositions in accordance with this invention or for use in this invention may be administered by means of a medical device or appliance such as an implant, graft, prosthesis, stent, etc.
  • Implants may be devised which are intended to contain and release such compounds or compositions.
  • An example would be an implant made of a polymeric material adapted to release the compound over a period of time.
  • the compounds may be administered alone or as a mixture with a pharmaceutically acceptable carrier e.g., as solid formulations such as tablets, capsules, granules, powders, etc.; liquid formulations such as syrups, injections, etc.; injections, drops, suppositories, pessaryies.
  • a pharmaceutically acceptable carrier e.g., as solid formulations such as tablets, capsules, granules, powders, etc.; liquid formulations such as syrups, injections, etc.; injections, drops, suppositories, pessaryies.
  • a pharmaceutically acceptable carrier e.g., as solid formulations such as tablets, capsules, granules, powders, etc.; liquid formulations such as syrups, injections, etc.; injections, drops, suppositories, pessaryies.
  • compounds or pharmaceutically acceptable carrier e.g., as solid formulations such as tablets, capsules, granules, powders, etc.;
  • compositions in accordance with this invention or for use in this invention may be any suitable compositions in accordance with this invention or for use in this invention.
  • inhalation spray nasal, vaginal, rectal, sublingual, or topical routes and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
  • the compounds of the invention may be used to treat animals, including mice, rats, horses, cattle, sheep, dogs, cats, and monkeys. However, compounds of the invention can also be used in other organisms, such as avian species (e.g., chickens). The compounds of the invention may also be effective for use in humans.
  • the term "subject” or alternatively referred to herein as "patient” is intended to be referred to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment. However, the compounds, methods and pharmaceutical compositions of the present invention may be used in the treatment of animals.
  • a "subject” may be a human, non-human primate, rat, mouse, cow, horse, pig, sheep, goat, dog, cat, etc.
  • the subject may be suspected of having or at risk for having a condition requiring
  • an “effective amount” of a compound according to the invention includes a therapeutically effective amount or a prophylactically effective amount.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as inhibition of an O-GlcNAcase, elevation of O-GlcNAc levels, inhibition of tau phosphorylation, or any condition described herein.
  • a therapeutically effective amount of a compound may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as inhibition of an O-GlcNAcase, elevation of O-GlcNAc levels, inhibition of tau phosphorylation, or any condition described herein.
  • a prophylactic dose is used in subjects prior to or at an earlier stage of disease, so that a prophylactically effective amount may be less than a therapeutically effective amount.
  • a suitable range for therapeutically or prophylactically effective amounts of a compound may be any integer from 0.1 nM-0.1M, 0.1 nM-0.05M, 0.05 ⁇ -15 ⁇ or 0.01 ⁇ -10 ⁇ .
  • an appropriate dosage level will generally be about 0.01 to 500 mg per kg subject body weight per day, and can be
  • the dosage level will be about 0.1 to about 250 mg/kg per day. It will be understood that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound used, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the patient undergoing therapy.
  • dosage values may vary with the severity of the condition to be alleviated. For any particular subject, specific dosage regimens may be adjusted over time according to the individual need and the professional judgement of the person
  • Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected by medical practitioners.
  • the amount of active compound(s) in the composition may vary according to factors such as the disease state, age, sex, and weight of the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Toxicity of the compounds of the invention can be determined using standard techniques, for example, by testing in cell cultures or experimental animals and determining the therapeutic index, i.e., the ratio between the LD50 (the dose lethal to 50% of the population) and the LD100 (the dose lethal to 100% of the population). In some circumstances however, such as in severe disease conditions, it may be necessary to administer substantial excesses of the compositions
  • the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature.
  • the present invention is meant to include all suitable isotopic variations of the compounds of generic Formula (I) or (la).
  • different isotopic forms of hydrogen (H) include protium (IF!), deuterium (2H) and tritium (3 ⁇ 4).
  • Protium is the predominant hydrogen isotope found in nature.
  • Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples.
  • Isotopically-enriched compounds within generic Formula (I) or (la) can be prepared without undue
  • a compound of Formula (I) or (la) may be used in screening assays for compounds which modulate the activity of glycosidase enzymes, preferably the O-
  • test compound is any naturally- occurring or artificially-derived chemical compound. Test compounds may include, without limitation, peptides, polypeptides, synthesised organic molecules, naturally occurring organic molecules, and nucleic acid molecules.
  • a test compound can "compete" with a known compound such as a compound of Formula (I) or (la) by, for example, interfering with inhibition of O-GlcNAcase-dependent cleavage of O-GlcNAc or by interfering with any biological response induced by a compound of Formula (I) or (la).
  • a test compound can exhibit any value between 10% and 200%, or over 500%), modulation when compared to a compound of Formula (I) or (la) or other reference compound.
  • a test compound may exhibit at least any positive or negative integer from 10%> to 200%) modulation, or at least any positive or negative integer from 30% to 150%) modulation, or at least any positive or negative integer from 60%> to
  • a compound that is a negative modulator will in general decrease modulation relative to a known compound, while a compound that is a positive modulator will in general increase modulation relative to a known compound.
  • test compounds are identified from large libraries of both natural products or synthetic (or semi- synthetic) extracts or chemical libraries according to methods known in the art.
  • Those skilled in the field of drug discovery and development will understand that the precise source of test extracts or compounds is not critical to the method(s) of the invention. Accordingly, virtually any number of chemical extracts or compounds can be screened using the exemplary methods described herein. Examples of such extracts or compounds include, but are not limited to, plant-, fungal-, prokaryotic- or animal-based extracts, fermentation broths, and synthetic compounds, as well as modification of existing compounds.
  • Synthetic compound libraries are commercially available.
  • libraries of natural compounds in the form of bacterial, fungal, plant, and animal extracts are commercially available from a number of sources, including Biotics (Sussex, UK), Xenova (Slough, UK), Harbor Branch Oceanographic Institute (Ft. Pierce, FL, USA), and
  • compounds shown to be useful agents for treatment are chemically modified according to methods known in the art.
  • Compounds identified as being of therapeutic, prophylactic, diagnostic, or other value may be subsequently analyzed using a suitable animal model, as described herein on known in the art.
  • the compounds are useful in the development of animal models for studying diseases or disorders related to deficiencies in O-GlcNAcase, over- expression of O-GlcNAcase, accumulation of O-GlcNAc, depletion of O-GlcNAc, and for studying treatment of diseases and disorders related to deficiency or over-expression of O- GlcNAcase, or accumulation or depletion of O-GlcNAc.
  • diseases and disorders include neurodegenerative diseases, including Alzheimer's disease, and cancer.
  • EDC l-(3-Dimethylaminopropyl)-3-ethlycarbodiimide
  • TBAB tetra-n-butylammonium bromide
  • TBAF tetra- «-butylammonium fluoride
  • THF tetrahydrofuran Synthesis of intermediate ( 5) l,3,4,6-tetra-0-acetyl-2-deoxy-2-isothiocyanate-P-D-glucopyranose (5)
  • reaction mixture was treated with TFA (1.6 g, 14 mmol) overnight at room temperature.
  • TFA saturated sodium bicarbonate
  • the reaction mixture was washed with saturated sodium bicarbonate (50 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 1 % MeOH in dichloromethane to give compound 6 as yellow oil (1.65 g, 85 %).
  • reaction solution was treated with trifluoroacetic acid (130 g, 1.34 mol) overnight at room temperature.
  • the reaction mixture was washed with saturated sodium bicarbonate (300 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 1 % MeOH in dichloromethane to give compound 14 as a yellow oil (50 g, 84 %).
  • reaction mixture was condensed to give a residue, which was purified by Prep-HPLC with the following conditions [(3#-Agilent 1200 detect prep HPLC): Column, C18, 19* 150mm, 5um; mobile phase, water with 0.03% ammonia and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV220nm] to give compound 32 (Example 3) as a white solid (67.9 mg, 78 %).
  • Prep-HPLC with the following conditions [(3#-Agilent 1200 detect prep HPLC): Column, C18, 19* 150mm, 5um; mobile phase, water with 0.03% ammonia and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV220nm] to give compound 32 (Example 3) as a white solid (67.9 mg, 78 %).
  • reaction mixture was condensed to give a residue, which was purified by Prep-HPLC with the following conditions [(3#- Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19*50mm 5um; mobile phase, WATER with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min; Detector, UV 220nm)] to give compound 35 (Example 4) as a white solid (TFA salt, 238 mg, 92 %).
  • Prep-HPLC with the following conditions [(3#- Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19*50mm 5um; mobile phase, WATER with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min; Detector, UV 220nm)] to give compound 35 (Example 4) as a white solid (TFA salt, 238 mg, 92 %).
  • Step 3 (3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-(2-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol (39) : A solution of 2-((3aR,5R,6R,7R,7aR)-2- (dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3, 2- d]thiazol-5-yl)ethanol (100 mg, 0.18 mmol) in dichloromethane (20 mL) was treated with TFA overnight at room temperature.
  • methyltriphenylphosphonium bromide (5.9 g, 16.5 mmol) in THF (70 mL) was treated with n-BuLi (5.9 ml , 2.5 M in THF) for 30 min at 0 °C, and followed by addition of
  • reaction mixture was quenched by sat. aqueous Na2S2C"3 solution (20 mL) and sat. aqueous NaHC03 solution (20 mL), and extracted with dichloromethane (2 x 20 mL).
  • dichloromethane (2 x 20 mL).
  • the combined, organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by silica gel column, eluted with 2 - 3 % methanol in dichloromethane to give compound 33 as a yellow syrup (400 mg, 88 %).
  • reaction was quenched by sat. aqueous Na2CC"3 solution (30 mL), and extracted with dichloromethane (2 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by silica gel column, eluted with 1 - 2 % methanol in dichloromethane to give compound 50 as a yellow syrup (240 mg, 85 %).
  • reaction mixture was concentrated under reduced pressure to give a residue, which was purified by Prep-HPLC under the following conditions [(Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19 * 50 mm 5 urn; mobile phase, water with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV 220nm] to afford compound 51 (Example 9) as a white solid (30 mg, 24 %).
  • Prep-HPLC Prep-HPLC under the following conditions [(Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19 * 50 mm 5 urn; mobile phase, water with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV 220nm] to afford compound 51 (Example 9) as a white solid (30 mg, 24 %).
  • Example 10 and 11 (3aR,5S,6S,7R,7aR)-2-(dimethylamino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol and (3aR,5S,6S,7R,7aR)-2- (dimethylamino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol
  • Step 3 l-((3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(methylamino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)cyclopentanol .
  • a mixture of compound 58 (0.35 g, 0.67 mmol) and Pd/C (0.10 g) in methanol (20 mL) was stirred under hydrogen atmosphere for 20 hours at room temperature.
  • reaction mixture was filtered and the filtrate was concentrated under vacuum to afford a residue, which was purified by a silica gel column with 30 % - 40 % ethyl acetate in petroleum ether to afford crude compound 59 as a yellow oil (300 mg), which was employed in the next step without further purification.
  • reaction mixture was quenched by saturated aqueous Na2SC"3 (5 mL) ; extracted with dichloromethane (3 x 15 mL), dried over magnesium sulfate, and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 20 % ethyl acetate in petroleum ether to provide compound 72 as a yellow syrup (0.4 g, 67 %).
  • Prep-HPLC Column, 19* 150 mm; mobile phase, WATER with 0.03 % H 3 H 2 0 and CH 3 CN (10 % CH 3 CN up to 45 % in 10 min); Detector, 220 nm.] to give the title compound 83 (Example 144) as a yellow solid (8.1 mg, 28 %).
  • Example 145 was prepared in a similar fashion to Example 144.
  • Example 145 was prepared in a similar fashion to Example 144.
  • dichloromethane (10 mL) was treated with DAST (262 mg, 1.63 mmol) overnight at room temperature, then quenched by saturated aqueous Na 2 C0 3 (10 mL), extracted with dichloromethane (3 x 10 mL). The combined organic layers were concentrated under vacuum to give crude compound 89 as a yellow foam (100 mg), which was dissolved into dichloromethane (5 mL) and treated with TFA (0.5 mL) for 3 hours at room temperature.
  • Enzymatic reactions are carried out in a reaction containing 50mM NaH2PC"4, 100 mM NaCl and 0.1% BSA (pH 7.0) using 2 mM 4-Methylumbelliferyl N-acetyl- ⁇ - ⁇ - glucosaminide dihydrate (Sigma M2133) dissolved in ddH20, as a substrate.
  • the amount of purified human O-GlcNAcase enzyme used in the reaction is 0.7 nM.
  • Test compound of varying concentrations is added to the enzyme prior to initiation of the reaction. The reaction is performed at room temperature in a 96-well plate and is initiated with the addition of substrate.
  • the production of fluorescent product is measured every 60 sec for 45 min with a Tecan Infinite M200 plate-reader with excitation at 355nM and emission detected at 460nM, with 4-Methylumbelliferone (Sigma M1381) used to produce a standard curve.
  • the slope of product production is determined for each concentration of compound tested and plotted, using standard curve fitting algorithms for sigmoidal dose response curves. The values for a four parameter logistic curve fit of the data are determined.
  • Ki values are determined using the Cheng-Prusoff equation; the Km of O- GlcNacase for substrate is 0.2 mM.
  • Examples 1 to 148 were tested in the above described assay and exhibited Kj values for inhibition of O-GlcNAcase in the range 0.1 nM - 10 ⁇ .
  • Enzymatic reactions are carried out in a reaction containing 50mM NaH2PC"4, 100 mM NaCl and 0.1% BSA (pH 7.0) using 2 mM 4-Methylumbelliferyl N-acetyl- ⁇ - ⁇ - glucosaminide dihydrate (Sigma M2133) dissolved in ddH20, as a substrate.
  • the amount of purified human ⁇ hexosaminidase enzyme used in the reaction is 24 nM.
  • Test compound of varying concentrations is added to the enzyme prior to initiation of the reaction. The reaction is performed at room temperature in a 96-well plate and is initiated with the addition of substrate.
  • the production of fluorescent product is measured every 60 sec for 45 min with a Tecan Infinite M200 plate-reader with excitation at 355nM and emission detected at 460nM, with 4-Methylumbelliferone (Sigma M1381) used to produce a standard curve.
  • the slope of product production is determined for each concentration of compound tested and plotted, using standard curve fitting algorithms for sigmoidal dose response curves. The values for a four parameter logistic curve fit of the data are determined.
  • Ki values are determined using the Cheng-Prusoff equation..
  • the selectivity ratio for inhibition of O-GlcNAcase over ⁇ -hexosaminidase is defined here as:
  • Ki ⁇ -hexosaminidase)/Kl (O-GlcNAcase) In general, the compounds described herein exhibit a selectivity ratio range of about 10 to 100000. Thus, many compounds of the invention exhibit high selectivity for inhibition of O-GlcNAcase over ⁇ -hexosaminidase.
  • O-GlcNAcase which removes O-GlcNAc from cellular proteins, results in an increase in the level of O-GlcNAcylated protein in cells.
  • An increase in O- GlcNAcylated protein can be measured by an antibody, such as RL-2, that binds to O- GlcNAcylated protein.
  • the amount of O-GlcNAcylated protein:RL2 antibody interaction can be measured by enzyme linked immunosorbant assay (ELISA) procedures.
  • tissue culture cell lines expressing endogenous levels of O- GlcNAcase, can be utilized; examples include rat PC-12, and human U-87, or SK-N-SH cells.
  • Cells are plated in 96-well plates with approximately 10,000 cells / well.
  • Compounds to be tested are dissolved in DMSO, either 2 or 10 mM stock solution, and then diluted with DMSO and water in a two-step process using a Tecan workstation. Cells are treated with diluted compounds for 24 hours (5.4 ⁇ . into 200 ⁇ . 1 well volume) to reach a final concentration of inhibitor desired to measure a compound concentration dependent response, typically, ten 3 fold dilution steps, starting at 10 ⁇ are used to determine a concentration response curve.
  • the media from compound treated cells is removed, the cells are washed once with phosphate buffered saline (PBS) and then lysed for 5 minutes at room temperature in 50 ⁇ . of Phosphosafe reagent (Novagen Inc, Madison, WI) with protease inhibitors and PMSF.
  • PBS phosphate buffered saline
  • the cell lysate is collected and transferred to a new plate, which is then either coated to assay plates directly or frozen -80°C until used in the ELISA procedure. If desired, the total protein concentration of samples is determined using 20 ⁇ , of the sample using the BCA method.
  • the ELISA portion of the assay is performed in a black Maxisorp 96-well plate that is coated overnight at 4°C with 100 ⁇ , /well of the cell lysate (1 : 10 dilution of the lysate with PBS containing protease inhibitors, phosphatase inhibitors, and PMSF.
  • the following day the wells are washed 3 times with 300 ⁇ , /well of Wash buffer (Tris-buffered saline with 0.1% Tween 20).
  • the wells are blocked with 100 ⁇ , /well Blocking buffer (Tris buffered saline w/0.05% Tween 20 and 2.5% Bovine serum albumin). Each well is then washed two times with 300 ul/well of wash buffer.
  • the anti O-GlcNAc antibody RL-2 (Abeam, Cambridge, MA), diluted 1 : 1000 in blocking buffer, is added at 100 ul/well.
  • the plate is sealed and incubated at 37°C for 2 hr with gentle shaking. The wells are then washed 3 -times with 300 ul/well wash buffer.
  • HRP horse-radish peroxidase
  • To detect the amount of RL-2 bound horse-radish peroxidase (HRP) conjugated goat anti-mouse secondary antibody (diluted 1 :3000 in blocking buffer) is added at 100 /well.
  • the plate is incubated for 60 min at 37°C with gentle shaking. Each wells is then washed 3 -times with 300 ul/well wash buffer.
  • the detection reagent is added, 100 ⁇ .
  • Amplex Ultra RED reagent prepared by adding 30 of 10 mM Amplex Ultra Red stock solution to 10 ml PBS with 18 ⁇ . 3% hydrogen peroxide, H2O2. The detection reaction is incubated for 15 minutes at room temperature and then read with excitation at 530 nm and emission at 590 nm.
  • the amount of O-GlcNAcylated protein, as detected by the ELISA assay, is plotted for each concentration of test compound using standard using standard curve fitting algorithms for sigmoidal dose response curves.
  • the values for a four parameter logistic curve fit of the data are determined, with the inflection point of the curve being the potency value for the test compound.

Abstract

The invention is directed to compounds for selectively inhibiting glycosidases, uses of the compounds and pharmaceutical compositions including the compounds, and methods of treating diseases and disorders related to deficiency or overexpression of O-GlcNAcase, and/or accumulation or deficiency of O-GlcNAc.

Description

SELECTIVE GLYCOSIDASE INHIBITORS AND USES THEREOF
FIELD OF THE INVENTION
This application relates to compounds which selectively inhibit glycosidases and uses thereof.
BACKGROUND OF THE INVENTION
A wide range of cellular proteins, both nuclear and cytoplasmic, are post- translationally modified by the addition of the monosaccharide 2-acetamido-2-deoxy-B-D- glucopyranoside (β-Ν-acetylglucosamine) which is attached via an O-glycosidic linkage.1 This modification is generally referred to as O-linked N-acetylglucosamine or O-GlcNAc. The enzyme responsible for post-translationally linking β-Ν-acetylglucosamine (GlcNAc) to specific serine and threonine residues of numerous nucleocytoplasmic proteins is O-GlcNAc transferase (OGT).2-5 A second enzyme, known as 0-GlcNAcase6,7 removes this post- translational modification to liberate proteins making the O-GlcNAc-modification a dynamic cycle occurring several times during the lifetime of a protein.8
O-GlcNAc-modified proteins regulate a wide range of vital cellular functions including, for example, transcription, 9- 12 proteasomal degradation, 13 and cellular signaling.14 O-GlcNAc is also found on many structural proteins.15-17 For example, it has been found on a number of cytoskeletal proteins, including neurofilament proteins, 18, 19 synapsins,6,20 synapsin-specific clathrin assembly protein AP-3,7 and ankyrinG.14 O- GlcNAc modification has been found to be abundant in the brain.21,22 it has also been found on proteins clearly implicated in the etiology of several diseases including Alzheimer's disease (AD) and cancer.
For example, it is well established that AD and a number of related tauopathies including Downs' syndrome, Pick's disease, Niemann-Pick Type C disease, and amyotrophic lateral sclerosis (ALS) are characterized, in part, by the development of neurofibrillary tangles ( FTs). These FTs are aggregates of paired helical filaments (PHFs) and are composed of an abnormal form of the cytoskeletal protein "tau". Normally tau stabilizes a key cellular network of microtubules that is essential for distributing proteins and nutrients within neurons. In AD patients, however, tau becomes hyperphosphorylated, disrupting its normal functions, forming PHFs and ultimately aggregating to form FTs. Six isoforms of tau are found in the human brain. In AD patients, all six isoforms of tau are found in NFTs, and all are markedly hyperphosphorylated.23, 24 Tau in healthy brain tissue bears only 2 or 3 phosphate groups, whereas those found in the brains of AD patients bear, on average, 8 phosphate groups.25, 26 A clear parallel between NFT levels in the brains of AD patients and the severity of dementia strongly supports a key role for tau dysfunction in AD.27,28 The precise causes of this hyperphosphorylation of tau remain elusive.
Accordingly, considerable effort has been dedicated toward: a) elucidating the molecular physiological basis of tau hyperphosphorylation;29 and b) identifying strategies that could limit tau hyperphosphorylation in the hope that these might halt, or even reverse, the progression of Alzheimer's disease30-33 Thus far, several lines of evidence suggest that up- regulation of a number of kinases may be involved in hyperphosphorylation of tau,21,34,35 although very recently, an alternative basis for this hyperphosphorylation has been advanced.21
In particular, it has recently emerged that phosphate levels of tau are regulated by the levels of O-GlcNAc on tau. The presence of O-GlcNAc on tau has stimulated studies that correlate O-GlcNAc levels with tau phosphorylation levels. The recent interest in this field stems from the observation that O-GlcNAc modification has been found to occur on many proteins at amino acid residues that are also known to be phosphorylated.36-38
Consistent with this observation, it has been found that increases in phosphorylation levels result in decreased O-GlcNAc levels and conversely, increased O-GlcNAc levels correlate with decreased phosphorylation levels.39 This reciprocal relationship between O-GlcNAc and phosphorylation has been termed the "Yin- Yang hypothesis"40 and has gained strong biochemical support by the recent discovery that the enzyme OGT4 forms a functional complex with phosphatases that act to remove phosphate groups from proteins.41 Like phosphorylation, O-GlcNAc is a dynamic modification that can be removed and reinstalled several times during the lifespan of a protein. Suggestively, the gene encoding O-GlcNAcase has been mapped to a chromosomal locus that is linked to AD.7,42 Hyperphosphorylated tau in human AD brains has markedly lower levels of O-GlcNAc than are found in healthy human brains.21 Very recently, it has been shown that O-GlcNAc levels of soluble tau protein from human brains affected with AD are markedly lower than those from healthy brain.21 Furthermore, PHF from diseased brain was suggested to lack completely any O- GlcNAc modification whatsoever.21 The molecular basis of this hypoglycosylation of tau is not known, although it may stem from increased activity of kinases and/or dysfunction of one of the enzymes involved in processing O-GlcNAc. Supporting this latter view, in both PC- 12 neuronal cells and in brain tissue sections from mice, a nonselective N- acetylglucosamindase inhibitor was used to increase tau O-GlcNAc levels, whereupon it was observed that phosphorylation levels decreased.21 The implication of these collective results is that by maintaining healthy O-GlcNAc levels in AD patients, such as by inhibiting the action of O-GlcNAcase, one should be able to block hyperphosphorylation of tau and all of the associated effects of tau hyperphosphorylation, including the formation of FTs and downstream effects. However, because the proper functioning of the β-hexosaminidases is critical, any potential therapeutic intervention for the treatment of AD that blocks the action of O-GlcNAcase would have to avoid the concomitant inhibition of both hexosaminidases A and B.
Neurons do not store glucose and therefore the brain relies on glucose supplied by blood to maintain its essential metabolic functions. Notably, it has been shown that within brain, glucose uptake and metabolism decreases with aging.43 Within the brains of AD patients marked decreases in glucose utilization occur and are thought to be a potential cause of neurodegeneration.44 The basis for this decreased glucose supply in AD brain45-47 is thought to stem from any of decreased glucose transport,48,49 impaired insulin signaling, 50,51 and decreased blood flow.52
In light of this impaired glucose metabolism, it is worth noting that of all glucose entering into cells, 2-5% is shunted into the hexosamine biosynthetic pathway, thereby regulating cellular concentrations of the end product of this pathway, uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) 53 UDP-GlcNAc is a substrate of the nucleocytoplasmic enzyme O-GlcNAc transferase (OGT),2-5 which acts to post- translationally add GlcNAc to specific serine and threonine residues of numerous
nucleocytoplasmic proteins. OGT recognizes many of its substrates54,55 and binding partners41,56 through its tetratricopeptide repeat (TPR) domains.57,58 As described above, 0-GlcNAcase6,7 removes this post-translational modification to liberate proteins making the O-GlcNAc-modification a dynamic cycle occurring several times during the lifetime of a protein.8 O-GlcNAc has been found in several proteins on known phosphorylation sites, 10,37,38,59 including tau and neurofilaments.60 Additionally, OGT shows unusual kinetic behaviour making it exquisitely sensitive to intracellular UDP-GlcNAc substrate concentrations and therefore glucose supply.41
Consistent with the known properties of the hexosamine biosynthetic pathway, the enzymatic properties of OGT, and the reciprocal relationship between O-GlcNAc and phosphorylation, it has been shown that decreased glucose availability in brain leads to tau hyperphosphorylation.44 Therefore the gradual impairment of glucose transport and metabolism, whatever its causes, leads to decreased O-GlcNAc and hyperphosphorylation of tau (and other proteins). Accordingly, the inhibition of O-GlcNAcase should compensate for the age related impairment of glucose metabolism within the brains of health individuals as well as patients suffering from AD or related neurodegenerative diseases.
These results suggest that a malfunction in the mechanisms regulating tau O- GlcNAc levels may be vitally important in the formation of FTs and associated
neurodegeneration. Good support for blocking tau hyperphosphorylation as a
therapeutically useful intervention61 comes from recent studies showing that when transgenic mice harbouring human tau are treated with kinase inhibitors, they do not develop typical motor defects33 and, in another case,32 show decreased levels of insoluble tau.
These studies provide a clear link between lowering tau phosphorylation levels and alleviating AD-like behavioural symptoms in a murine model of this disease. Indeed, pharmacological modulation of tau hyperphosphorylation is widely recognized as a valid therapeutic strategy for treating AD and other neurodegenerative disorders.6
Recent studies63 support the therapeutic potential of small-molecule O- GlcNAcase inhibitors to limit tau hyperphosphorylation for treatment of AD and related tauopathies. Specifically, the O-GlcNAcase inhibitor thiamet-G has been implicated in the reduction of tau phosphorylation in cultured PC- 12 cells at pathologically relevant sites.63 Moreover, oral administration of thiamet-G to healthy Sprague-Dawley rats has been implicated in reduced phosphorylation of tau at Thr231, Ser396 and Ser422 in both rat cortex and hippocampus.63 .
There is also a large body of evidence indicating that increased levels of O- GlcNAc protein modification provides protection against pathogenic effects of stress in cardiac tissue, including stress caused by ischemia, hemorrhage, hypervolemic shock, and calcium paradox. For example, activation of the hexosamine biosynthetic pathway (HBP) by administration of glucosamine has been demonstrated to exert a protective effect in animals models of ischemia/reperfusion, 64-70 trauma hemorrhaged 1-73 hypervolemic shock,74 and calcium paradox.64,75 Moreover, strong evidence indicates that these cardioprotective effects are mediated by elevated levels of protein O-GlcNAc modification.64,65, 67,70,72,75- 78 There is also evidence that the O-GlcNAc modification plays a role in a variety of neurodegenerative diseases, including Parkinson's disease and Huntington's disease.79 Humans have three genes encoding enzymes that cleave terminal β-Ν-acetyl- glucosamine residues from glycoconjugates. The first of these encodes the enzyme O- glycoprotein 2-acetamido-2-deoxy-P-D-glucopyranosidase (O-GlcNAcase). O-GlcNAcase is a member of family 84 of glycoside hydrolases that includes enzymes from organisms as diverse as prokaryotic pathogens to humans (for the family classification of glycoside hydrolases see Coutinho, P.M. & Henrissat, B. (1999) Carbohydrate-Active Enzymes server at URL: http://afmb.cnrs-mrs.fr/CAZY/.27,28 O-GlcNAcase acts to hydrolyse O-GlcNAc off of serine and threonine residues of post-translationally modified proteins.1,6,7,80,81 Consistent with the presence of O-GlcNAc on many intracellular proteins, the enzyme O- GlcNAcase appears to have a role in the etiology of several diseases including type II diabetes, 14,82 AD, 16,21,83 and cancer.22,84 Although O-GlcNAcase was likely isolated earlier on, 18, 19 about 20 years elapsed before its biochemical role in acting to cleave O- GlcNAc from serine and threonine residues of proteins was understood.6 More recently O- GlcNAcase has been cloned,7 partially characterized,20 and suggested to have additional activity as a histone acetyltransferase.20 However, little was known about the catalytic mechanism of this enzyme.
The other two genes, HEXA and HEXB, encode enzymes catalyzing the hydrolytic cleavage of terminal β-Ν-acetylglucosamine residues from glycoconjugates. The gene products of HEXA and HEXB predominantly yield two dimeric isozymes,
hexosaminidase A and hexosaminidase B, respectively. Hexosaminidase A (αβ), a heterodimeric isozyme, is composed of an a- and a B-subunit. Hexosaminidase B (ββ), a homodimeric isozyme, is composed of two B-subunits. The two subunits, a- and B-, bear a high level of sequence identity. Both of these enzymes are classified as members of family 20 of glycoside hydrolases and are normally localized within lysosomes. The proper functioning of these lysosomal β-hexosaminidases is critical for human development, a fact that is underscored by the tragic genetic illnesses, Tay-Sach's and Sandhoff diseases which stem from a dysfunction in, respectively, hexosaminidase A and hexosaminidase B.85 These enzymatic deficiencies cause an accumulation of glycolipids and glycoconjugates in the lysosomes resulting in neurological impairment and deformation. The deleterious effects of accumulation of gangliosides at the organismal level are still being uncovered.86
As a result of the biological importance of these β-N-acetyl-glucosaminidases, small molecule inhibitors of glycosidases87-90 have received a great deal of attention,91 both as tools for elucidating the role of these enzymes in biological processes and in developing potential therapeutic applications. The control of glycosidase function using small molecules offers several advantages over genetic knockout studies including the ability to rapidly vary doses or to entirely withdraw treatment.
However, a major challenge in developing inhibitors for blocking the function of mammalian glycosidases, including O-GlcNAcase, is the large number of functionally related enzymes present in tissues of higher eukaryotes. Accordingly, the use of non-selective inhibitors in studying the cellular and organismal physiological role of one particular enzyme is complicated because complex phenotypes arise from the concomitant inhibition of such functionally related enzymes. In the case of B-N-acetylglucosaminidases, existing compounds that act to block O-GlcNAcase function are non-specific and act potently to inhibit the lysosomal β-hexosaminidases.
A few of the better characterized inhibitors of β-N-acetyl-glucosaminidases which have been used in studies of O-GlcNAc post-translational modification within both cells and tissues are streptozotocin (STZ), 2'-methyl-a-D-glucopyrano-[2, l-<JJ-A2'-thiazoline (NAG-thiazoline) and O-(2-acetamido-2-deoxy-D-glucopyranosylidene)amino N- phenylcarbamate (PUGNAc).14,92-95
STZ has long been used as a diabetogenic compound because it has a particularly detrimental effect on B-islet cells.96 STZ exerts its cytotoxic effects through both the alkylation of cellular DNA96,97 as well as the generation of radical species including nitric oxide.98 The resulting DNA strand breakage promotes the activation of poly(ADP-ribose) polymerase (PARP)99 with the net effect of depleting cellular NAD+ levels and, ultimately, leading to cell death.100, 101 Other investigators have proposed instead that STZ toxicity is a consequence of the irreversible inhibition of O-GlcNAcase, which is highly expressed within β-islet cells.92, 102 This hypothesis has, however, been brought into question by two independent research groups.103, 104 Because cellular O-
GlcNAc levels on proteins increase in response to many forms of cellular stress 105 it seems possible that STZ results in increased O-GlcNAc-modification levels on proteins by inducing cellular stress rather than through any specific and direct action on O-GlcNAcase. Indeed, Hanover and coworkers have shown that STZ functions as a poor and somewhat selective inhibitor of O-GlcNAcasel06 and although it has been proposed by others that STZ acts to irreversibly inhibit O-GlcNAcase, 107 there has been no clear demonstration of this mode of action. Recently, it has been shown that STZ does not irreversibly inhibit O-GlcNAcase.108
NAG-thiazoline has been found to be a potent inhibitor of family 20 hexosaminidases,90, 109 and more recently, the family 84 O-GlcNAcases.108 Despite its potency, a downside to using NAG-thiazoline in a complex biological context is that it lacks selectivity and therefore perturbs multiple cellular processes.
PUGNAc is another compound that suffers from the same problem of lack of selectivity, yet has enjoyed use as an inhibitor of both human 0-GlcNAcase6, 110 and the family 20 human β-hexosaminidases.111 This molecule, developed by Vasella and coworkers, was found to be a potent competitive inhibitor of the β-N-acetyl-glucosaminidases from Canavalia ensiformis, Mucor rouxii, and the β-hexosaminidase from bovine kidney.88 it has been demonstrated that administration of PUGNAc in a rat model of trauma hemorrhage decreases circulating levels of the pro-inflammatory cytokines TNF-a and IL-6.112 It has also been shown that administration of PUGNAc in a cell-based model of lymphocyte activation decreases production of the cytokine IL-2.113 Recent studies have indicated that PUGNAc can be used in an animal model to reduce myocardial infarct size after left coronary artery
occlusions.114 Of particular significance is the fact that elevation of O-GlcNAc levels by administration of PUGNAc, an inhibitor of O-GlcNAcase, in a rat model of trauma hemorrhage improves cardiac function.112, 115 in addition, elevation of O-GlcNAc levels by treatment with PUGNAc in a cellular model of ischemia/reperfusion injury using neonatal rat ventricular myocytes improved cell viability and reduced necrosis and apoptosis compared to untreated cells.116
More recently, it has been suggested that the selective O-GlcNAcase inhibitor NButGT exhibits protective activity in cell-based models of ischemia/reperfusion and cellular stresses, including oxidative stress.117 This study suggests the use of O-GlcNAcase inhibitors to elevate protein O-GlcNAc levels and thereby prevent the pathogenic effects of stress in cardiac tissue.
International patent applications PCT/CA2006/000300, filed 1 March 2006, published under No. WO 2006/092049 on 8 September 2006; PCT/CA2007/001554, filed 31 August 2007, published under No. WO 2008/025170 on 6 March 2008;
PCT/CA2009/001087, filed 31 July 2009, published under No. WO 2010/012106 on 4 Februrary 2010; PCT/CA2009/001088, filed 31 July 2009, published under WO
2010/012107 on 4 Februrary 2010; and PCT/CA2009/001302, filed 16 September 2009, published under WO 2010/037207 on 8 April 2010, describe selective inhibitors of O- GlcNAcase.
SUMMARY OF THE INVENTION
The invention is directed to compounds for selectively inhibiting glycosidases, uses of the compounds and pharmaceutical compositions including the compounds, and methods of treating diseases and disorders related to deficiency or overexpression of O- GlcNAcase, and/or accumulation or deficiency of O-GlcNAc.
DETAILED DESCRIPTION
The invention encompasses a compound of Formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000009_0001
(I) wherein: each R is independently H or C(0)CH3; Rl and R2 are independently selected from the group consisting of: H, Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl and -(0¾)η- cyclobutyl wherein n is 0, 1, 2, 3 or 4; or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine, pyrrolidine or piperidine, said Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl, -(CH2)n-cyclobutyl, azetidine, pyrrolidine or piperidine optionally substituted from one up to the maximum number of substituents with fluoro or methyl; R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2- Salkynyl, C3-6cycloalkyl, aryl and heteroaryl, each optionally substituted from one up to the maximum number of substituents with fluoro and OH; R4 is selected from the group consisting of: H, F, Ci-8alkyl, C2-8alkenyl and C2-8alkynyl, each excluding hydrogen optionally substituted from one up to the maximum number of substituents with fluoro and OH; or R3 and R4 and the carbon atom to which they are attached may join together to form vinyl or a 3 to 7-membered carbocyclic or heterocyclic ring, said 3 to 7-membered carbocyclic or heterocyclic ring optionally containing a double bond and optionally substituted from one up to the maximum number of substituents with fluoro and OH; and R5 is selected from H, F and OH; with the proviso that when R4 is F then R5 is other than OH.
The invention encompasses a genus of compounds of Formula (la) or a pharmaceutically acceptable salt thereof:
Figure imgf000010_0001
(la) wherein Rl and R2 are independently selected from the group consisting of: H, Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl and -(CH2)n-cyclobutyl wherein n is 0, 1, 2, 3 or 4; or
Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine, pyrrolidine or piperidine, said Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl, - (CH2)n-cyclobutyl, azetidine, pyrrolidine or piperidine optionally substituted from one up to the maximum number of substituents with fluoro or methyl; R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2-8alkynyl, C3-6cycloalkyl, aryl and heteroaryl, each optionally substituted from one up to the maximum number of substituents with fluoro and OH; R4 is selected from the group consisting of: H, F, Ci-8alkyl, C2-8alkenyl and C2- 8alkynyl, each excluding hydrogen optionally substituted from one up to the maximum number of substituents with fluoro and OH; or R3 and R4 and the carbon atom to which they are attached may join together to form vinyl or a 3 to 7-membered carbocyclic or heterocyclic ring, said 3 to 7-membered carbocyclic or heterocyclic ring optionally containing a double bond and optionally substituted from one up to the maximum number of substituents with fluoro and OH; and R5 is selected from H, F and OH; with the proviso that when R4 is F then R5 is other than OH.
Within the genus, the invention encompasses a first sub-genus of compounds of Formula (la) wherein: Rl and R2 are independently Ci-4alkyl; R3 is Ci-6alkyl; R4 is selected from the group consisting of: H and Ci-6alkyl; and R5 is OH. Within the first subgenus, the invention further encompasses compounds of Formula (la) wherein: Rl and R2 are independently methyl or ethyl; R3 is methyl or ethyl; and R4 is selected from the group consisting of: H, methyl and ethyl.
Also within the genus, the invention encompasses a second sub-genus of compounds of Formula (la) wherein:R3 and R4 and the carbon atom to which they are attached may join together to form a 3 to 7-membered carbocyclic or heterocyclic ring, said 3 to 7-membered carbocyclic or heterocyclic ring optionally containing a double bond and optionally substituted from one up to the maximum number of substituents with fluoro and OH.
Also within the genus, the invention encompasses a third sub-genus of compounds of Formula (la) wherein: Rl and R2 are independently selected from the group consisting of: H, Ci-6alkyl and cyclopropylmethyl; or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine or pyrrolidine, said Ci- 6alkyl, cyclopropylmethyl, azetidine or pyrrolidine optionally substituted with 1 to 3 substituents selected from fluoro and methyl; R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2-8alkynyl and C3-6cycloalkyl, each optionally substituted with 1 to 3 substituents selected from fluoro and OH; and R4 is selected from the group consisting of: H, F, Ci-8alkyl, C2-8alkenyl and C2-8alkynyl, each excluding hydrogen optionally substituted with 1 to 3 substituents selected from fluoro and OH; or R3 and R4 and the carbon atom to which they are attached may join together to form a 3- to 6-membered carbocyclic ring optionally containing a double bond and optionally substituted with 1 to 3 substituents selected from fluoro and OH.
Also within the genus, the invention encompasses a fourth sub-genus of compounds of Formula (la) wherein R3 is CF3 and R5 is OH.
The invention also encompasses the compounds that follow or pharmaceutically acceptable salts thereof.
The invention also encompasses a pharmaceutical composition comprising the compound of Formula (I) or (la) in combination with a pharmaceutically acceptable carrier.
The invention also encompasses a method of selectively inhibiting O- GlcNAcase in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula (I) or (la), or a pharmaceutically acceptable salt thereof.
The invention also encompasses a method of elevating the level of O-GlcNAc in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula (I) or (la), or a pharmaceutically acceptable salt thereof.
The invention also encompasses a method of treating a condition that is modulated by O-GlcNAcase, in a subject in need thereof, the method comprising
administering to the subject an effective amount of a compound of Formula (I) or (la), or a pharmaceutically acceptable salt thereof. An aspect of the invention encompasses this method wherein the condition is selected from one or more of the group consisting of an inflammatory disease, an allergy, asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias, delayed-type hypersensitivity, atherosclerosis, interstitial lung disease (ILD), idiopathic pulmonary fibrosis, ILD associated with rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, systemic sclerosis, Sjogren's syndrome, polymyositis or dermatomyositis, systemic anaphylaxis or hypersensitivity response, drug allergy, insect sting allergy, autoimmune disease, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Guillain-Barre syndrome, systemic lupus erythematosus, myastenia gravis, glomerulonephritis, autoimmune thyroiditis, graft rejection, allograft rejection, graft-versus-host disease, inflammatory bowel disease, Crohn's disease, ulcerative colitis, spondyloarthropathy, scleroderma, psoriasis, T-cell mediated psoriasis, inflammatory dermatosis, dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria, vasculitis, necrotizing, cutaneous, and hypersensitivity vasculitis, eosinphilic myotis, eosiniphilic fasciitis, solid organ transplant rejection, heart transplant rejection, lung transplant rejection, liver transplant rejection, kidney transplant rejection, pancreas transplant rejection, kidney allograft, lung allograft, epilepsy, pain, fibromyalgia, stroke,
neuroprotection.
The invention also encompasses a method of treating a condition selected from the group consisting of a neurodegenerative disease, a tauopathy, cancer and stress, in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula (I) or (la), or a pharmaceutically acceptable salt thereof. An aspect of the invention encompasses this method wherein the condition is selected from one or more of the group consisting of Alzheimer's disease, Amyotrophic lateral sclerosis (ALS), Amyotrophic lateral sclerosis with cognitive impairment (ALSci), Argyrophilic grain dementia, Bluit disease, Corticobasal degeneration (CBD), Dementia pugilistica, Diffuse neurofibrillary tangles with calcification, Down's syndrome, Familial British dementia, Familial Danish dementia, Frontotemporal dementia with parkinsonism linked to
chromosome 17 (FTDP-17), Gerstmann-Straussler-Scheinker disease, Guadeloupean parkinsonism, Hallevorden-Spatz disease (neurodegeneration with brain iron accumulation type 1), Multiple system atrophy, Myotonic dystrophy, Niemann-Pick disease (type C), Pallido-ponto-nigral degeneration, Parkinsonism-dementia complex of Guam, Pick's disease (PiD), Post-encephalitic parkinsonism (PEP), Prion diseases (including Creutzfeldt- Jakob Disease (CJD), Variant Creutzfeldt- Jakob Disease (vCJD), Fatal Familial Insomnia, and Kuru), Progressive supercortical gliosis, Progressive supranuclear palsy (PSP), Richardson's syndrome, Subacute sclerosing panencephalitis, Tangle-only dementia, Huntington's disease, Parkinson's disease, Schizophrenia, Mild Cognitive Impairment (MCI), Neuropathy
(including peripheral neuropathy, autonomic neuropathy, neuritis, and diabetic neuropathy), or Glaucoma. Another aspect of the invention encompasses this method wherein the stress is a cardiac disorder. In another aspect, the cardiac disorder is selected from one or more of the group consisting of ischemia; hemorrhage; hypovolemic shock; myocardial infarction; an interventional cardiology procedure; cardiac bypass surgery; fibrinolytic therapy; angioplasty; and stent placement. The compounds of the invention are capable of inhibiting an O-glycoprotein 2-acetamido-2-deoxy-P-D-glucopyranosidase (O-GlcNAcase). In some embodiments, the O-GlcNAcase is a mammalian O-GlcNAcase, such as a rat, mouse or human O-GlcNAcase. In some embodiments, the β-hexosaminidase is a mammalian β-hexosaminidase, such as a rat, mouse or human β-hexosaminidase.
Compounds of the invention selectively inhibit the activity of a mammalian O- GlcNAcase over a mammalian β-hexosaminidase. A compound that "selectively" inhibits an O-GlcNAcase is a compound that inhibits the activity or biological function of an O- GlcNAcase, but does not substantially inhibit the activity or biological function of a β- hexosaminidase. For example, in some embodiments, a selective inhibitor of an O-
GlcNAcase selectively inhibits the cleavage of 2-acetamido-2-deoxy^-D-glucopyranoside (O-GlcNAc) from polypeptides. In some embodiments, a selective inhibitor of an O- GlcNAcase selectively binds to an O-GlcNAcase. In some embodiments, a selective inhibitor of an O-GlcNAcase inhibits hyperphosphorylation of a tau protein and/or inhibits formations of FTs. By "inhibits," "inhibition" or "inhibiting" means a decrease by any value between 10% and 90%, or of any integer value between 30% and 60%, or over 100%, or a decrease by 1-fold, 2-fold, 5-fold, 10-fold or more. It is to be understood that the inhibiting does not require full inhibition. In some embodiments, a selective inhibitor of an O-GlcNAcase elevates or enhances O-GlcNAc levels e.g., O-GlcNAc-modified polypeptide or protein levels, in cells, tissues, or organs (e.g., in brain, muscle, or heart (cardiac) tissue) and in animals. By "elevating" or "enhancing" is meant an increase by any value between 10% and 90%), or of any integer value between 30% and 60%, or over 100%), or an increase by 1-fold, 2-fold, 5-fold, 10-fold, 15-fold, 25-fold, 50-fold, 100-fold or more. In some embodiments, a selective inhibitor of an O-GlcNAcase exhibits a selectivity ratio, as described herein, in the range 10 to 100000, or in the range 100 to 100000, or in the range 1000 to 100000, or at least 10, 20, 50, 100, 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 6000, 7000, 10,000, 25,000, 50,000, 75,000, or any value within or about the described range.
The compounds of the present invention elevate O-GlcNAc levels on O- GlcN Ac-modified polypeptides or proteins in vivo specifically via interaction with an O- GlcNAcase enzyme, and are effective in treating conditions which require or respond to inhibition of O-GlcNAcase activity. In some embodiments, the compounds of the present invention are useful as agents that produce a decrease in tau phosphorylation and NFT formation. In some embodiments, the compounds are therefore useful to treat Alzheimer's disease and related tauopathies. In some embodiments, the compounds are thus capable of treating Alzheimer's disease and related tauopathies by lowering tau phosphorylation and reducing NFT formation as a result of increasing tau O-GlcNAc levels. In some embodiments, the compounds produce an increase in levels of O-GlcNAc modification on O-GlcNAc-modified
polypeptides or proteins, and are therefore useful for treatment of disorders responsive to such increases in O-GlcNAc modification; these disorders include without limitation neurodegenerative, inflammatory, cardiovascular, and immunoregulatory diseases. In some embodiments, the compounds are also useful as a result of other biological activites related to their ability to inhibit the activity of glycosidase enzymes. In alternative embodiments, the compounds of the invention are valuable tools in studying the physiological role of O- GlcNAc at the cellular and organismal level.
In alternative embodiments, the invention provides methods of enhancing or elevating levels of protein O-GlcNAc modification in animal subjects, such as, veterinary and human subjects. In alternative embodiments, the invention provides methods of selectively inhibiting an O-GlcNAcase enzyme in animal subjects, such as, veterinary and human subjects. In alternative embodiments, the invention provides methods of inhibiting phosphorylation of tau polypeptides, or inhibiting formation of NFTs, in animal subjects, such as, veterinary and human subjects.
As will be appreciated by a person skilled in the art, Formula (I) above may also be represented alternatively as follows:
Figure imgf000016_0001
As used herein the singular forms "a", "and", and "the" include plural referents unless the context clearly dictates otherwise. For example, "a compound" refers to one or more of such compounds, while "the enzyme" includes a particular enzyme as well as other family members and equivalents thereof as known to those skilled in the art.
Throughout this application, it is contemplated that the term "compound" or "compounds" refers to the compounds discussed herein and includes precursors and derivatives of the compounds, including acyl-protected derivatives, and pharmaceutically acceptable salts of the compounds, precursors, and derivatives. The invention also includes prodrugs of the compounds, pharmaceutical compositions including the compounds and a pharmaceutically acceptable carrier, and pharmaceutical compositions including prodrugs of the compounds and a pharmaceutically acceptable carrier.
In some embodiments, all of the compounds of the invention contain at least one chiral center. In some embodiments, the formulations, preparation, and compositions including compounds according to the invention include mixtures of stereoisomers, individual stereoisomers, and enantiomeric mixtures, and mixtures of multiple stereoisomers. In general, the compound may be supplied in any desired degree of chiral purity.
"Alkyl" refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing no unsaturation and including, for example, from one to ten carbon atoms, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms, and which is attached to the rest of the molecule by a single bond. Unless stated otherwise specifically in the specification, the alkyl group may be optionally substituted by one or more
substituents as described herein. Unless stated otherwise specifically herein, it is understood that the substitution can occur on any carbon of the alkyl group.
"Alkenyl" refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one double bond and including, for example, from two to ten carbon atoms, such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms, and which is attached to the rest of the molecule by a single bond or a double bond. Unless stated otherwise specifically in the specification, the alkenyl group may be optionally substituted by one or more substituents as described herein. Unless stated otherwise specifically herein, it is understood that the substitution can occur on any carbon of the alkenyl group.
"Alkynyl" refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond and including, for example, from two to ten carbon atoms. Unless stated otherwise specifically in the specification, the alkenyl group may be optionally substituted by one or more substituents as described herein.
"Aryl" means mono- or bicyclic aromatic rings containing only carbon atoms, including for example, 6-14 members. Examples of aryl include phenyl, naphthyl, indanyl, indenyl, tetrahydronaphthyl, 2,3-dihydrobenzofuranyl, dihydrobenzopyranyl, 1,4- benzodioxanyl, and the like. Unless stated otherwise specifically herein, the term "aryl" is meant to include aryl groups optionally substituted by one or more substituents as described herein.
"Heteroaryl" refers to a single or fused aromatic ring group containing one or more heteroatoms in the ring, for example N, O, S, including for example, 5-14 members, such as 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 members. Examples of heteroaryl groups include furan, thiophene, pyrrole, oxazole, thiazole, imidazole, pyrazole, isoxazole, isothiazole, 1,2,3-oxadiazole, 1,2,3-triazole, 1,2,4-triazole, 1,3,4-thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, 1,3,5-triazine, imidazole, benzimidazole, benzoxazole, benzothiazole, indolizine, indole, isoindole, benzofuran, benzothiophene, IH-indazole, purine, 4H-quinolizine, quinoline, isoquinoline, cinnoline, phthalazine, quinazoline, quinoxaline, 1,8- naphthyridine, pteridine. Unless stated otherwise specifically herein, the term "heteroaryl" is meant to include heteroaryl groups optionally substituted by one or more substituents as described herein.
"Cycloalkyl" refers to a stable monovalent monocyclic, bicyclic or tricyclic hydrocarbon group consisting solely of carbon and hydrogen atoms, having for example from 3 to 15 carbon atoms, and which is saturated and attached to the rest of the molecule by a single bond. Unless otherwise stated specifically herein, the term "cycloalkyl" is meant to include cycloalkyl groups which are optionally substituted as described herein.
The term "3 to 7-membered carbocyclic or heterocyclic ring" means a monocylic carbon ring of 3 to 7 atoms or a monocyclic ring of 3 to 7 atoms containing one or more heterotaoms selected from O, N and S.
"Optional" or "optionally" means that the subsequently described event of circumstances may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. For example, "optionally substituted alkyl" means that the alkyl group may or may not be substituted and that the description includes both substituted alkyl groups and alkyl groups having no substitution. Examples of optionally substituted alkyl groups include, without limitation, methyl, ethyl, propyl, etc. and including cycloalkyls such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, etc.; examples of optionally substituted alkenyl groups include allyl, crotyl, 2-pentenyl, 3-hexenyl, 2-cyclopentenyl, 2-cyclohexenyl, 2-cyclopentenylmethyl, 2-cyclohexenylmethyl, etc. In some embodiments, optionally substituted alkyl and alkenyl groups include Ci-6 alkyls or alkenyls.
"Halo" refers to bromo, chloro, fluoro, iodo, etc. In some embodiments, suitable halogens include fluorine or chlorine.
Examples of optionally substituted carbonyl groups, or sulfonyl groups include optionally substituted forms of such groups formed from various hydrocarbyls such as alkyl, alkenyl and 5- to 6-membered monocyclic aromatic group (e.g., phenyl, pyridyl, etc.), as described herein.
Therapeutic Indications
The invention provides methods of treating conditions that are modulated, directly or indirectly, by an O-GlcNAcase enzyme or by O-GlcNAc-modified protein levels, for example, a condition that is benefited by inhibition of an O-GlcNAcase enzyme or by an elevation of O-GlcNAc-modified protein levels. Such conditions include, without limitation, glaucoma, schizophrenia, tauopathies, such as Alzheimer's disease, neurodegenerative diseases, cardiovascular diseases, diseases associated with inflammation, diseases associated with immunosuppression and cancers. The compounds of the invention are also useful in the treatment of diseases or disorders related to deficiency or over-expression of O-GlcNAcase or accumulation or depletion of O-GlcNAc, or any disease or disorder responsive to glycosidase inhibition therapy. Such diseases and disorders include, but are not limited to, glaucoma, schizophrenia, neurodegenerative disorders, such as Alzheimer's disease (AD), or cancer. Such diseases and disorders may also include diseases or disorders related to the accumulation or deficiency in the enzyme OGT. Also included is a method of protecting or treating target cells expressing proteins that are modified by O-GlcNAc residues, the dysregulation of which modification results in disease or pathology. The term "treating" as used herein includes treatment, prevention, and amelioration.
In alternative embodiments, the invention provides methods of enhancing or elevating levels of protein O-GlcNAc modification in animal subjects, such as, veterinary and human subjects. This elevation of O-GlcNAc levels can be useful for the prevention or treatment of Alzheimer's disease; prevention or treatment of other neurodegenerative diseases (e.g. Parkinson's disease, Huntington's disease); providing neuroprotective effects; preventing damage to cardiac tissue; and treating diseases associated with inflammation or immunosuppression.
In alternative embodiments, the invention provides methods of selectively inhibiting an O-GlcNAcase enzyme in animal subjects, such as veterinary and human subjects.
In alternative embodiments, the invention provides methods of inhibiting phosphorylation of tau polypeptides, or inhibiting formation of FTs, in animal subjects, such as, veterinary and human subjects. Accordingly, the compounds of the invention may be used to study and treat AD and other tauopathies.
In general, the methods of the invention are effected by administering a compound according to the invention to a subject in need thereof, or by contacting a cell or a sample with a compound according to the invention, for example, a pharmaceutical composition comprising a therapeutically effective amount of the compound according to Formula (I) or (la). More particularly, they are useful in the treatment of a disorder in which the regulation of O-GlcNAc protein modification is implicated, or any condition as described herein. Disease states of interest include Alzheimer's disease (AD) and related
neurodegenerative tauopathies, in which abnormal hyperphosphorylation of the microtubule- associated protein tau is involved in disease pathogenesis. In some embodiments, the compounds may be used to block hyperphosphorylation of tau by maintaining elevated levels of O-GlcNAc on tau, thereby providing therapeutic benefit.
The effectiveness of the compounds in treating pathology associated with the accumulation of toxic tau species (for example, Alzheimer's disease and other tauopathies) may be confirmed by testing the ability of the compounds to block the formation of toxic tau species in established cellularl 18-120 and/or transgenic animal models of disease.32,33
Tauopathies that may be treated with the compounds of the invention include: Alzheimer's disease, Amyotrophic lateral sclerosis (ALS), Amyotrophic lateral sclerosis with cognitive impairment (ALSci), Argyrophilic grain dementia, Bluit disease, Corticobasal degeneration (CBD), Dementia pugilistica, Diffuse neurofibrillary tangles with calcification, Down's syndrome, Familial British dementia, Familial Danish dementia, Frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), Gerstmann-Straussler-Scheinker disease, Guadeloupean parkinsonism, Hallevorden-Spatz disease (neurodegeneration with brain iron accumulation type 1), Multiple system atrophy, Myotonic dystrophy, Niemann- Pick disease (type C), Pallido-ponto-nigral degeneration, Parkinsonism-dementia complex of Guam, Pick's disease (PiD), Post-encephalitic parkinsonism (PEP), Prion diseases (including Creutzfeldt- Jakob Disease (CJD), Variant Creutzfeldt- Jakob Disease (vCJD), Fatal Familial Insomnia, and Kuru), Progressive supercortical gliosis, Progressive supranuclear palsy (PSP), Richardson's syndrome, Subacute sclerosing panencephalitis, Tangle-only dementia, and Glaucoma.
The compounds of this invention are also useful in the treatment of conditions associate with tissue damage or stress, stimulating cells, or promoting differentiation of cells. Accordingly, in some embodiments, the compounds of this invention may be used to provide therapeutic benefit in a variety of conditions or medical procedures involving stress in cardiac tissue, including but not limited to: ischemia; hemorrhage; hypovolemic shock; myocardial infarction; an interventional cardiology procedure; cardiac bypass surgery; fibrinolytic therapy; angioplasty; and stent placement. The effectiveness of the compounds in treating pathology associated with cellular stress (including ischemia, hemorrhage, hypovolemic shock, myocardial infarction, and other cardiovascular disorders) may be confirmed by testing the ability of the compounds to prevent cellular damage in established cellular stress assays, 105, 1 16, 1 17 and to prevent tissue damage and promote functional recovery in animal models of ischemia- reperfusion,70, 1 14 and trauma-hemorrhage.72, 1 12, 1 15
Compounds that selectively inhibit O-GlcNAcase activity may be used for the treatment of diseases that are associated with inflammation, including but not limited to, inflammatory or allergic diseases such as asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias, delayed-type
hypersensitivity, atherosclerosis, interstitial lung disease (ILD) (e.g., idiopathic pulmonary fibrosis, or ILD associated with rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, systemic sclerosis, Sjogren's syndrome, polymyositis or
dermatomyositis); systemic anaphylaxis or hypersensitivity responses, drug allergies, insect sting allergies; autoimmune diseases, such as rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Guillain-Barre syndrome, systemic lupus erythematosus, myastenia gravis, glomerulonephritis, autoimmune thyroiditis, graft rejection, including allograft rejection or graft-versus-host disease; inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis; spondyloarthropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and inflammatory dermatoses such as dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria; vasculitis (e.g., necrotizing, cutaneous, and hypersensitivity vasculitis); eosinphilic myotis, eosiniphilic fasciitis; and cancers.
In addition, compounds that affects levels of protein O-GlcNAc modification may be used for the treatment of diseases associated with immunosuppression, such as in individuals undergoing chemotherapy, radiation therapy, enhanced wound healing and burn treatment, therapy for autoimmune disease or other drug therapy (e.g., corticosteroid therapy) or combination of conventional drugs used in the treatment of autoimmune diseases and graft/transplantation rejection, which causes immunosuppression; or immunosuppression due to congenital deficiency in receptor function or other causes. The compounds of the invention may be useful for treatment of neurodegenerative diseases, including Parkinson's disease and Huntington's disease. Other conditions that may be treated are those triggered, affected, or in any other way correlated with levels of O-GlcNAc post-translational protein modification. It is expected that the compounds of this invention may be useful for the treatment of such conditions and in particular, but not limited to, the following for which a association with O-GlcNAc levels on proteins has been established: graft rejection, in particular but not limited to solid organ transplants, such as heart, lung, liver, kidney, and pancreas transplants (e.g. kidney and lung allografts); cancer, in particular but not limited to cancer of the breast, lung, prostate, pancreas, colon, rectum, bladder, kidney, ovary; as well as non-Hodgkin's lymphoma and melanoma; epilepsy, pain, fibromyalgia, or stroke, e.g., for neuroprotection following a stroke.
Pharmaceutical & Veterinary Compositions. Dosages. And Administration
Pharmaceutical compositions including compounds according to the invention, or for use according to the invention, are contemplated as being within the scope of the invention. In some embodiments, pharmaceutical compositions including an effective amount of a compound of Formula (I) or (la) are provided.
The compounds of Formula (I) or (la) and their pharmaceutically acceptable salts, stereoisomers, solvates, and derivatives are useful because they have pharmacological activity in animals, including humans. In some embodiments, the compounds according to the invention are stable in plasma, when administered to a subject.
In some embodiments, compounds according to the invention, or for use according to the invention, may be provided in combination with any other active agents or pharmaceutical compositions where such combined therapy is useful to modulate O- GlcNAcase activity, for example, to treat neurodegenerative, inflammatory, cardiovascular, or immunoregulatory diseases, or any condition described herein. In some embodiments, compounds according to the invention, or for use according to the invention, may be provided in combination with one or more agents useful in the prevention or treatment of Alzheimer's disease. Examples of such agents include, without limitation,
-acetylcholine esterase inhibitors (AChEIs) such as Aricept® (Donepezil), Exelon® (Rivastigmine), Razadyne® (Razadyne ER®, Reminyl®, Nivalin®, Galantamine), Cognex® (Tacrine), Dimebon, Huperzine A, Phenserine, Debio-9902 SR (ZT-1 SR), Zanapezil (TAK0147), ganstigmine, NP7557, etc.; - MDA receptor antagonists such as Namenda® (Axura®, Akatinol®, Ebixa®, Memantine), Dimebon, SGS-742, Neramexane, Debio-9902 SR (ZT-1 SR), etc.; gamma- seer etase inhibitors and/or modulators such as FlurizanTM (Tarenflurbil, MPC-7869, R-flurbiprofen), LY450139, MK 0752, E2101, BMS-289948, BMS-299897, BMS-433796, LY-411575, GSI-136, etc.;
-beta-secretase inhibitors such as ATG-Z1, CTS-21166, etc.;
-alpha-secretase activators, such as NGX267, etc;
-amyloid-β aggregation and/or fibrillization inhibitors such as AlzhemedTM (3APS, Tramiprosate, 3-amino-l-propanesulfonic acid), AL-108, AL-208, AZD-103, PBT2, Cereact, ONO-2506PO, PPI-558, etc.;
-tau aggregation inhibitors such as methylene blue, etc.;
microtubule stabilizers such as AL-108, AL-208, paclitaxel, etc.;
-RAGE inhibitors, such as TTP488, etc.;
-5-HTla receptor antagonists, such as Xaliproden, Lecozotan, etc.;
-5-HT4 receptor antagonists, such as PRX-03410, etc.;
-kinase inhibitors such as SRN-003-556, amfurindamide, LiCl, AZD1080, NP031112, SAR-502250, etc.
-humanized monoclonal anti-Αβ antibodies such as Bapineuzumab (AAB- 001), LY2062430, RN1219, ACU-5A5, etc.;
-amyloid vaccines such as AN- 1792, ACC-001
-neuroprotective agents such as Cerebrolysin, AL-108, AL-208, Huperzine A, etc.;
-L-type calcium channel antagonists such as MEM- 1003, etc.;
-nicotinic receptor antagonists, such as AZD3480, GTS-21, etc.; -nicotinic receptor agonists, such as MEM 3454, Nefiracetam, etc.;
-peroxisome proliferator-activated receptor (PPAR) gamma agonists such as Avandia® (Rosglitazone), etc.;
-phosphodiesterase IV (PDE4) inhibitors, such as MK-0952, etc.;
-hormone replacement therapy such as estrogen (Premarin), etc.; -monoamine oxidase (MAO) inhibitors such as NS2330, Rasagiline
(Azilect®), TVP-1012, etc.;
-AMP A receptor modulators such as Ampalex (CX 516), etc.; nerve growth factors or NGF potentiators, such as CERE-110 (AAV-NGF), T-588, T- 817MA, etc.;
-agents that prevent the release of luteinizing hormone (LH) by the pituitary gland, such as leuoprolide (VP-4896), etc.;
-GABA receptor modulators such as AC-3933, NGD 97-1, CP-457920, etc.; benzodiazepine receptor inverse agonists such as SB-737552 (S-8510), AC-3933, etc.;
-noradrenaline-releasing agents such as T-588, T-817MA, etc.
It is to be understood that combination of compounds according to the invention, or for use according to the invention, with Alzheimer's agents is not limited to the examples described herein, but includes combination with any agent useful for the treatment of Alzheimer's disease. Combination of compounds according to the invention, or for use according to the invention, and other Alzheimer's agents may be administered separately or in conjunction. The administration of one agent may be prior to, concurrent to, or subsequent to the administration of other agent(s).
In alternative embodiments, the compounds may be supplied as "prodrugs" or protected forms, which release the compound after administration to a subject. For example, the compound may carry a protective group which is split off by hydrolysis in body fluids, e.g., in the bloodstream, thus releasing the active compound or is oxidized or reduced in body fluids to release the compound. Accordingly, a "prodrug" is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound of the invention. Thus, the term "prodrug" refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject in need thereof, but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood.
The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a subject.
The term "prodrug" is also meant to include any covalently bonded carriers which release the active compound of the invention in vivo when such prodrug is
administered to a subject. Prodrugs of a compound of the invention may be prepared by modifying functional groups present in the compound of the invention in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound of the invention. Prodrugs include compounds of the invention wherein a hydroxy, amino or mercapto group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy, free amino or free mercapto group, respectively. Examples of prodrugs include, but are not limited to, acetate, formate and benzoate derivatives of alcohol and acetamide, formamide, and benzamide derivatives of amine functional groups in the compounds of the invention and the like.
A discussion of prodrugs may be found in "Smith and Williams' Introduction to the Principles of Drug Design," H.J. Smith, Wright, Second Edition, London (1988);
Bundgard, H., Design of Prodrugs (1985), pp. 7-9, 21-24 (Elsevier, Amsterdam); The
Practice of Medicinal Chemistry, Camille G. Wermuth et al, Ch 31, (Academic Press, 1996); A Textbook of Drug Design and Development, P. Krogsgaard-Larson and H. Bundgaard, eds. Ch 5, pgs 113 191 (Harwood Academic Publishers, 1991); Higuchi, T., et al.,
"Pro-drugs as Novel Delivery Systems," A.C.S. Symposium Series, Vol. 14; or in
Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, all of which are incorporated in full by reference herein.
Compounds according to the invention, or for use according to the invention, can be provided alone or in combination with other compounds in the presence of a liposome, an adjuvant, or any pharmaceutically acceptable carrier, diluent or excipient, in a form suitable for administration to a subject such as a mammal, for example, humans, cattle, sheep, etc. If desired, treatment with a compound according to the invention may be combined with more traditional and existing therapies for the therapeutic indications described herein.
Compounds according to the invention may be provided chronically or intermittently.
"Chronic" administration refers to administration of the compound(s) in a continuous mode as opposed to an acute mode, so as to maintain the initial therapeutic effect (activity) for an extended period of time. "Intermittent" administration is treatment that is not consecutively done without interruption, but rather is cyclic in nature. The terms "administration,"
"administrable," or "administering" as used herein should be understood to mean providing a compound of the invention to the subject in need of treatment.
"Pharmaceutically acceptable carrier, diluent or excipient" includes without limitation any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier that has been approved, for example, by the United States Food and Drug Administration or other governmental agency as being acceptable for use in humans or domestic animals.
The compounds of the present invention may be administered in the form of pharmaceutically acceptable salts. In such cases, pharmaceutical compositions in accordance with this invention may comprise a salt of such a compound, preferably a physiologically acceptable salt, which are known in the art. In some embodiments, the term
"pharmaceutically acceptable salt" as used herein means an active ingredient comprising compounds of Formula (I) or (la) used in the form of a salt thereof, particularly where the salt form confers on the active ingredient improved pharmacokinetic properties as compared to the free form of the active ingredient or other previously disclosed salt form.
A "pharmaceutically acceptable salt" includes both acid and base addition salts. A "pharmaceutically acceptable acid addition salt" refers to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as acetic acid, trifluoroacetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid,
p-toluenesulfonic acid, salicylic acid, and the like.
A "pharmaceutically acceptable base addition salt" refers to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of 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, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine,
glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine,
N-ethylpiperidine, polyamine resins and the like.
Thus, the term "pharmaceutically acceptable salt" encompasses all acceptable salts including but not limited to acetate, lactobionate, benzenesulfonate, laurate, benzoate, malate, bicarbonate, maleate, bisulfate, mandelate, bitartarate, mesylate, borate,
methylbromide, bromide, methylnitrite, calcium edetate, methylsulfate, camsylate, mucate, carbonate, napsylate, chloride, nitrate, clavulanate, N-methylglucamine, citrate, ammonium salt, dihydrochloride, oleate, edetate, oxalate, edisylate, pamoate (embonate), estolate, palmitate, esylate, pantothenate, fumarate, phosphate/diphosphate, gluceptate,
polygalacturonate, gluconate, salicylate, glutame, stearate, glycollylarsanilate, sulfate, hexylresorcinate, subacetate, hydradamine, succinate, hydrobromide, tannate, hydrochloride, tartrate, hydroxynaphthoate, teoclate, iodide, tosylate, isothionate, triethiodide, lactate, panoate, valerate, and the like.
Pharmaceutically acceptable salts of the compounds of the present invention can be used as a dosage for modifying solubility or hydrolysis characteristics, or can be used in sustained release or prodrug formulations. Also, pharmaceutically acceptable salts of the compounds of this invention may include those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc, and from bases such as ammonia, ethylenediamine, N-methyl-glutamine, lysine, arginine, ornithine, choline,
Ν,Ν'-dibenzylethylene-diamine, chloroprocaine, diethanolamine, procaine,
N-benzylphenethyl-amine, diethylamine, piperazine, tris(hydroxymethyl)aminomethane, and tetramethylammonium hydroxide.
Pharmaceutical formulations will typically include one or more carriers acceptable for the mode of administration of the preparation, be it by injection, inhalation, topical administration, lavage, or other modes suitable for the selected treatment. Suitable carriers are those known in the art for use in such modes of administration.
Suitable pharmaceutical compositions may be formulated by means known in the art and their mode of administration and dose determined by the skilled practitioner. For parenteral administration, a compound may be dissolved in sterile water or saline or a pharmaceutically acceptable vehicle used for administration of non-water soluble compounds such as those used for vitamin K. For enteral administration, the compound may be administered in a tablet, capsule or dissolved in liquid form. The table or capsule may be enteric coated, or in a formulation for sustained release. Many suitable formulations are known, including, polymeric or protein microparticles encapsulating a compound to be released, ointments, gels, hydrogels, or solutions which can be used topically or locally to administer a compound. A sustained release patch or implant may be employed to provide release over a prolonged period of time. Many techniques known to skilled practitioners are described in Remington: the Science & Practice of Pharmacy by Alfonso Gennaro, 20th ed., Williams & Wilkins, (2000). Formulations for parenteral administration may, for example, contain excipients, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated naphthalenes. Biocompatible, biodegradable lactide polymer,
lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Other potentially useful parenteral delivery systems for modulatory compounds include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops, or as a gel.
The compounds or pharmaceutical compositions according to the present invention may be administered by oral or non-oral, e.g., intramuscular, intraperitoneal, intravenous, intracisternal injection or infusion, subcutaneous injection, transdermal or transmucosal routes. In some embodiments, compounds or pharmaceutical compositions in accordance with this invention or for use in this invention may be administered by means of a medical device or appliance such as an implant, graft, prosthesis, stent, etc. Implants may be devised which are intended to contain and release such compounds or compositions. An example would be an implant made of a polymeric material adapted to release the compound over a period of time. The compounds may be administered alone or as a mixture with a pharmaceutically acceptable carrier e.g., as solid formulations such as tablets, capsules, granules, powders, etc.; liquid formulations such as syrups, injections, etc.; injections, drops, suppositories, pessaryies. In some embodiments, compounds or pharmaceutical
compositions in accordance with this invention or for use in this invention may be
administered by inhalation spray, nasal, vaginal, rectal, sublingual, or topical routes and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.
The compounds of the invention may be used to treat animals, including mice, rats, horses, cattle, sheep, dogs, cats, and monkeys. However, compounds of the invention can also be used in other organisms, such as avian species (e.g., chickens). The compounds of the invention may also be effective for use in humans. The term "subject" or alternatively referred to herein as "patient" is intended to be referred to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment. However, the compounds, methods and pharmaceutical compositions of the present invention may be used in the treatment of animals. Accordingly, as used herein, a "subject" may be a human, non-human primate, rat, mouse, cow, horse, pig, sheep, goat, dog, cat, etc. The subject may be suspected of having or at risk for having a condition requiring
modulation of O-GlcNAcase activity.
An "effective amount" of a compound according to the invention includes a therapeutically effective amount or a prophylactically effective amount. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as inhibition of an O-GlcNAcase, elevation of O-GlcNAc levels, inhibition of tau phosphorylation, or any condition described herein. A therapeutically effective amount of a compound may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as inhibition of an O-GlcNAcase, elevation of O-GlcNAc levels, inhibition of tau phosphorylation, or any condition described herein. Typically, a prophylactic dose is used in subjects prior to or at an earlier stage of disease, so that a prophylactically effective amount may be less than a therapeutically effective amount. A suitable range for therapeutically or prophylactically effective amounts of a compound may be any integer from 0.1 nM-0.1M, 0.1 nM-0.05M, 0.05 ηΜ-15μΜ or 0.01 ηΜ-10μΜ. In alternative embodiments, in the treatment or prevention of conditions which require modulation of O-GlcNAcase activity, an appropriate dosage level will generally be about 0.01 to 500 mg per kg subject body weight per day, and can be
administered in singe or multiple doses. In some embodiments, the dosage level will be about 0.1 to about 250 mg/kg per day. It will be understood that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound used, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the patient undergoing therapy.
It is to be noted that dosage values may vary with the severity of the condition to be alleviated. For any particular subject, specific dosage regimens may be adjusted over time according to the individual need and the professional judgement of the person
administering or supervising the administration of the compositions. Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that may be selected by medical practitioners. The amount of active compound(s) in the composition may vary according to factors such as the disease state, age, sex, and weight of the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. In general, compounds of the invention should be used without causing substantial toxicity, and as described herein, the compounds exhibit a suitable safety profile for therapeutic use. Toxicity of the compounds of the invention can be determined using standard techniques, for example, by testing in cell cultures or experimental animals and determining the therapeutic index, i.e., the ratio between the LD50 (the dose lethal to 50% of the population) and the LD100 (the dose lethal to 100% of the population). In some circumstances however, such as in severe disease conditions, it may be necessary to administer substantial excesses of the compositions
In the compounds of generic Formula (I) or (la), the atoms may exhibit their natural isotopic abundances, or one or more of the atoms may be artificially enriched in a particular isotope having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number predominantly found in nature. The present invention is meant to include all suitable isotopic variations of the compounds of generic Formula (I) or (la). For example, different isotopic forms of hydrogen (H) include protium (IF!), deuterium (2H) and tritium (¾). Protium is the predominant hydrogen isotope found in nature. Enriching for deuterium may afford certain therapeutic advantages, such as increasing in vivo half-life or reducing dosage requirements, or may provide a compound useful as a standard for characterization of biological samples. Isotopically-enriched compounds within generic Formula (I) or (la) can be prepared without undue
experimentation by conventional techniques well known to those skilled in the art or by processes analogous to those described in the Schemes and Examples herein using appropriate isotopically-enriched reagents and/or intermediates.
Other Uses and Assays
A compound of Formula (I) or (la) may be used in screening assays for compounds which modulate the activity of glycosidase enzymes, preferably the O-
GlcNAcase enzyme. The ability of a test compound to inhibit O-GlcNAcase-dependent cleavage of O-GlcNAc from a model substrate may be measured using any assays, as described herein or known to one of ordinary skill in the art. For example, a fluoresence or UV-based assay known in the art may be used. A "test compound" is any naturally- occurring or artificially-derived chemical compound. Test compounds may include, without limitation, peptides, polypeptides, synthesised organic molecules, naturally occurring organic molecules, and nucleic acid molecules. A test compound can "compete" with a known compound such as a compound of Formula (I) or (la) by, for example, interfering with inhibition of O-GlcNAcase-dependent cleavage of O-GlcNAc or by interfering with any biological response induced by a compound of Formula (I) or (la).
Generally, a test compound can exhibit any value between 10% and 200%, or over 500%), modulation when compared to a compound of Formula (I) or (la) or other reference compound. For example, a test compound may exhibit at least any positive or negative integer from 10%> to 200%) modulation, or at least any positive or negative integer from 30% to 150%) modulation, or at least any positive or negative integer from 60%> to
100%) modulation, or any positive or negative integer over 100%) modulation. A compound that is a negative modulator will in general decrease modulation relative to a known compound, while a compound that is a positive modulator will in general increase modulation relative to a known compound.
In general, test compounds are identified from large libraries of both natural products or synthetic (or semi- synthetic) extracts or chemical libraries according to methods known in the art. Those skilled in the field of drug discovery and development will understand that the precise source of test extracts or compounds is not critical to the method(s) of the invention. Accordingly, virtually any number of chemical extracts or compounds can be screened using the exemplary methods described herein. Examples of such extracts or compounds include, but are not limited to, plant-, fungal-, prokaryotic- or animal-based extracts, fermentation broths, and synthetic compounds, as well as modification of existing compounds. Numerous methods are also available for generating random or directed synthesis (e.g., semi-synthesis or total synthesis) of any number of chemical compounds, including, but not limited to, saccharide-, lipid-, peptide-, and nucleic acid-based compounds. Synthetic compound libraries are commercially available. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant, and animal extracts are commercially available from a number of sources, including Biotics (Sussex, UK), Xenova (Slough, UK), Harbor Branch Oceanographic Institute (Ft. Pierce, FL, USA), and
PharmaMar, MA, USA. In addition, natural and synthetically produced libraries are produced, if desired, according to methods known in the art, e.g., by standard extraction and fractionation methods. Furthermore, if desired, any library or compound is readily modified using standard chemical, physical, or biochemical methods.
When a crude extract is found to modulate inhibition of O-GlcNAcase- dependent cleavage of O-GlcNAc, or any biological response induced by a compound of Formula (I) or (la), further fractionation of the positive lead extract is necessary to isolate chemical constituents responsible for the observed effect. Thus, the goal of the extraction, fractionation, and purification process is the careful characterization and identification of a chemical entity within the crude extract having O-GlcNAcase- inhibitory activities. The same assays described herein for the detection of activities in mixtures of compounds can be used to purify the active component and to test derivatives thereof. Methods of fractionation and purification of such heterogeneous extracts are known in the art. If desired, compounds shown to be useful agents for treatment are chemically modified according to methods known in the art. Compounds identified as being of therapeutic, prophylactic, diagnostic, or other value may be subsequently analyzed using a suitable animal model, as described herein on known in the art.
In some embodiments, the compounds are useful in the development of animal models for studying diseases or disorders related to deficiencies in O-GlcNAcase, over- expression of O-GlcNAcase, accumulation of O-GlcNAc, depletion of O-GlcNAc, and for studying treatment of diseases and disorders related to deficiency or over-expression of O- GlcNAcase, or accumulation or depletion of O-GlcNAc. Such diseases and disorders include neurodegenerative diseases, including Alzheimer's disease, and cancer.
Various alternative embodiments and examples of the invention are described herein. These embodiments and examples are illustrative and should not be construed as limiting the scope of the invention.
Examples
The following examples are intended to illustrate embodiments of the invention and are not intended to be construed in a limiting manner.
Abbreviations
DCM = dichloromethane
DMAP = 4-dimethylaminopyridine
DMF = N,N-dimethylformamide
DMP = Dess-Martin periodinane
EDC = l-(3-Dimethylaminopropyl)-3-ethlycarbodiimide
hydrochloride
BS = N-bromosuccinimide
TBAB = tetra-n-butylammonium bromide
TBAF = tetra-«-butylammonium fluoride
TEMPO = 2,2,6,6-tetramethyl-piperidin-l-oxy free radical
TFA = 2,2,2-trifluoroacetic acid
THF = tetrahydrofuran Synthesis of intermediate ( 5) l,3,4,6-tetra-0-acetyl-2-deoxy-2-isothiocyanate-P-D-glucopyranose (5)
Figure imgf000034_0001
(2S,3R,4R,5S,6R)-6-(acetoxymethyl)-3-amino-tetrahydro-2H-pyran-2,4,5-triyl triacetate hydrochloride (4) was prepared from compound 1 according to publication: D. Silva etc. J. Org. Chem., 1999, 64(16), 5926-5929.
Figure imgf000035_0001
(2S,3R,4R,5S,6R)-6-(acetoxymethyl)-3-isothiocyanato-tetrahydro-2H-pyran-2,4,5-triyl triacetate ( 5) was prepared from compound 4 according to publication: M.V. Gonzalez etc. Carbohydrate Research, 1986, 154, 49-62.
Example 1
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000035_0002
Scheme II
Figure imgf000035_0003
Step 1
Figure imgf000036_0001
(3aR,5R,6S,7R,7aR)-5-(Acetoxymethyl)-2-(dimethylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diyl diacetate (6): To a solution of (3R,4R,5S,6R)-6- (acetoxymethyl)-3-isothiocyanato-tetrahydro-2H-pyran-2,4,5-triyl triacetate (2 g, 5.14 mmol) in dichloromethane (20 mL) was added dimethylamine hydrochloride (460 mg, 5.64 mmol) and triethylamine (675 mg, 6.68 mmol) at 5-10 °C. After stirred for 3 h, the reaction mixture was treated with TFA (1.6 g, 14 mmol) overnight at room temperature. The reaction mixture was washed with saturated sodium bicarbonate (50 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 1 % MeOH in dichloromethane to give compound 6 as yellow oil (1.65 g, 85 %). (ES, m/z): [M+H]+ 374.9; iH MR (300 MHz, CDCI3) δ 6.24 - 6.26 (d, J= 6.6 Hz, 1H), 5.31 - 5.43 (m, 1H), 4.94 - 4.99 (m, 1H), 4.34 - 4.38 (t, J= 10.8 Hz, 1H), 4.16 - 4.22 (m, 2H), 4.38 - 4.39 (m, 1H), 3.02 (s, 6H), 2.06 - 2.12 (m ,9H).
Step 2
Figure imgf000036_0002
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-(hydroxymethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol (7): To a solution of (3aR,5R,6S,7R,7aR)-5- (acetoxymethyl)-2-(dimethylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diyl diacetate (1.65 g, 4.41 mmol) in methanol (20 mL) was added potassium carbonate (25 mg, 0.18 mmol). The resulting mixture was stirred overnight at room temperature to yield a solid This was collected by filtration, washed with cold methanol and dried. The product 7 was obtained as a light yellow solid (1.05 g, 94 %). (ES, m/z): [M+H]+ 248.9; lH MR (300 MHz, CDCI3) δ 6.33 - 6.35 (d, J= 6.3 Hz, 1H), 4.29 - 4.33 (t, J= 6.0 Hz 1H), 4.16 (s, 1H), 3.76 - 3.89 (m, 2H), 3.70 (s, 2H), 3.03 (s, 6H).
Step 3
Figure imgf000037_0001
7 8
(3aR,5R,6S,7R,7aR)-5-((tert-Butyldimethylsilyloxy)methyl)-2-(dimethylamino)- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol (8): To a solution of
(3 aR, 5R, 6 S, 7R, 7aR)-2-(dimethylamino)-5 -(hydroxymethyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol (1 g, 4.03 mmol), DMAP (49.2 mg, 0.40 mmol) and triethylamine (611 mg, 6.05 mmol) in DMF (50mL) was added tert- butylchlorodimethylsilane (665 mg, 4.43 mmol). After stirred overnight at 50 °C, the resulting mixture was concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 2 -5 % MeOH in dichloromethane to give compound 8 as a yellow solid (1.0 g, 65 %). (ES, m/z): [M+H]+ 263.0; lH MR (300 MHz, CDCI3) δ 6.33 - 6.35 (d, J= 6.3 Hz, 1H), 4.35 - 4.39 (t, J= 5.7 Hz, 1H), 4.18 - 4.21 (t, J= 4.5 Hz, 1H), 3.81 - 3.84 (m, 3H), 3.62 - 3.67 (m, 1H), 3.05 (s, 6H), 0.93 (s, 9H), 0.11 (s, 6H).
Step 4
Figure imgf000037_0002
8 9
(3aR,5R,6S,7R,7aR)-5-((tert-Butyldimethylsilyloxy)methyl)-6,7-bis(4- methoxybenzyloxy)-N,N-dimethyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2- amine (9): To a solution of (3aR,5R,6S,7R,7aR)-5-((tert-butyldimethylsilyloxy)methyl)-2- (dimethylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol (1 g, 2.76 mmol) in DMF (20 mL) was added sodium hydride (568 mg, 16.6 mmol, 70 %) at 15 °C, and followed by addition of l-(bromomethyl)-4-methoxybenzene (2.22 g, 11.0 mmol). The resulting solution was stirred for 3 h at room temperature, quenched by addition of cold water (50 mL), and extracted with dichloromethane (3 x 50 mL). The combined organic layers were dried over anhydrous magnesium sulfate and concentrated to give a residue, which was purified by silica gel column, eluted with 10 -25 % ethyl acetate in petroleum ether to give compound 9 as a yellow oil (1.2 g, 64 %). (ES, m/z): [M+H]+ 603.1; lH
NMR (300 MHz, CDCI3) δ 7.22 - 7.35 (m, 4H), 6.84 - 6.92 (m. 4H), 6.27 - 6.29 (d, 7= 6. Hz, 1H), 4.60 - 4.76 (m, 4H), 4.36 - 4.43 (m, 2H), 4.10 - 4.17 (m, 2H), 3.81 (s, 6H), 3.72 (m, 1H), 3.61 ( m, 1H), 2.99 (s, 6H), 0.83 (s, 9H), 0.07 (s, 6H).
Step 5
Figure imgf000038_0001
9 10
((3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano [3,2-d] thiazol-5-yl)methanol (10): (3aR,5R,6S,7R,7aR)-6,7- bis(4-methoxybenzyloxy)-5 -((tert-butyldimethylsilyloxy)methyl)-N,N-dimethyl-5 , 6, 7, 7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-amine (9.5 g, 15.8 mmol) in THF (100 mL) was treated with TBAF (8.27 g, 31. 6 mmol) overnight at room temperature. The resulting solution was diluted with brine (200 mL), extracted with ethyl acetate (2 x200 mL), and dried over anhydrous magnesium sulfate. After removal of solvents, the residue was purified by silica gel column, eluted with 1 -2.5 % MeOH in dichloromethane to give compound 10 as a yellow oil (7.0 g, 86 %). (ES, m/z): [M+H]+ 489.0; lH NMR (300 MHz, CDCI3) δ 7.32 - 7.62 (m, 2H), 7.22 - 7.28 (m, 2H), 6.85 - 6.91 (m, 4H), 6.26 - 6.28 (d, 7= 6.6 Hz, 1H), 4.52 - 4.73 (m, 4H), 4.31 - 4.34 (d, 7= 11.4 Hz, 1H), 4.23 (s, 1H), 3.81 (s, 6H), 3.53 - 3.76 (m, 4H), 3.01 (m, 6H), 1.78 - 1.82 (m, 1H).
Step 6
Figure imgf000038_0002
10 11 (3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-5-carbaldehyde (11): To a mixture of
((3aR,5R,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(dimethylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-5-yl)methanol (500 mg, 1.0 mmol), TBAB (16.5 mg, 0.05 mmol), KHCO3 (461 mg, 4.6 mmol) and TEMPO (8 mg, 0.05 mmol) in dichloromethane (25 mL) and H2O (5 mL) was added BS (201 mg, 1.13 mmol) at 15 °C. After stirred for 30 min, the reaction mixture was quenched by saturated Na2SC"3 (5 mL). The organic layer was dried over anhydrous magnesium sulfate and condensed to provide a residue, which was purified by silica gel column, eluted with 20 -30 % ethyl acetate in dichloromethane to give compound 11 as a yellow syrup (320 mg, 75 % pure). (ES, m/z): [M+H]+ 487.0. lH MR (300 MHz, CDCI3) δ 9.61 (s, 1H), 7.22 - 7.34 (m, 4H), 6.83 - 6.92 (m, 4H), 6.11 - 6.13 (d, J= 6.0 Hz, 1H), 4.17 - 4.67 (m, 8H), 3.83 (s, 6H), 3.00 - 3.04 (s, 6H).
Step
Figure imgf000039_0001
11 12
l-((3aR,5R,6S,7R,7aR)-6,7-Bis(4-methoxybenzyloxy)-2-(dimethylamino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanol (12): To a solution of
(3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(dimethylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-5-carbaldehyde (260 mg, 0.53 mmol) in THF (10 mL) was added methylmagnesium bromide (0.3 mL, 3M in THF). After stirred for 2 h at room temperature, the reaction mixture was quenched with sat. LjCl (aq, 20 mL), extracted with ethyl acetate (3 x 20 mL). The combined organic layers were dried over anhydrous magnesium sulfate, concentrated under vacuum to give a residue, which was purified by silica gel column, eluted with 1 -2 % MeOH in dichloromethane to give compound 12 as a yellow syrup (250 mg, 74 %, two diastereomers, faster moving one: slower moving one = 1 : 5). (ES, m/z):
[M+H]+ 503.0; lH NMR (300 MHz, CDCI3) δ 7.26 - 7.35 (m, 2H), 7.22 - 7.28 (m, 2H),
6.85 - 6.91 (m, 4H), 6.29 - 6.31 (d, J= 6.9 Hz, 1H), 4.52 - 4.73 (m, 4H), 4.31 - 4.34 (d , J = 11.4 Hz, 1H), 4.23 (s, 1H), 3.81 (s, 6H), 3.53 - 3.76 (m, 4H), 3.01 (m, 6H), 1.19 - 1.21 (d , J = 6.6 Hz, 3H). tep 8
Figure imgf000040_0001
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-((R)-l-hydroxyethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol (13a,) and (3aR,5R,6S,7R,7aR)-2-(dimethylamino)- 5-((S)-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol (13, Example 1): A solution of l-((3aR,5R,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2- (dimethylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanol ( 315 mg, 0.63 mmol, a 1 :5 mixture of two diastereomers) in dichloromethane (20 mL) was treated with TFA (2 mL) for 1 h at room temperature. The reaction mixture was concentrated under vacuum to give a residue, which was purified by Prep-HPLC under the following conditions [(3#- Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19*50mm 5um; mobile phase, WATER with 0.03 % H4OH and CH3CNQO % CH3CN up to 45 % in 10 min); Detector, UV 220nm] to afford the faster eluting isomer as a white solid (13a, 6.9 mg, 4.2 %): (ES, m/z): [M+H]+ 262.9; lH MR ( 300 MHz, CDCI3 ) δ 6.21 - 6.23 (d, J= 6.6 Hz, 1H), 4.12 - 4.16 (m, 1H), 3.92 - 3.99 (m, 2H), 3.48 - 3.59 (m, 2H), 2.9 (s, 6H), 1.07 - 1.10 (d, J= 6.6 Hz, 3H); and the slower eluting isomer as white solid (13, Example 1, 52.8 mg, 32 %): (ES, m/z): [M+H]+ 262.9; iH MR (slower eluting isomer, 300 MHz, CDCI3) δ 6.18 - 6.21 (d, J = 6.6 Hz, 1H), 4.13 - 4.16 (m, 1H), 3.95 -3.98 (m, 1H), 3.84 - 3.88 (m, 1H), 3.58 - 3.63 (m, 1H), 3.21 - 3.26 (m, 1H), 2.89 (s, 6H), 1.11 - 1.13 (d, J= 6.6 Hz, 3H). Example 2
(3aR, 5R, 6S,7R,7aR)-5-((S)-l-hydroxyethyl)-2-(propylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000041_0001
Scheme III
Boc20,MeOH λ 2rFAPaDCMamine AcO-V"N ΗΟ-γ"Ν
OAc OAc OH
1
Figure imgf000041_0002
21 22
Figure imgf000041_0003
(3aR,5R,6S,7R,7aR)-5-(Acetoxymethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diyl diacetate (14): To a solution of (3R,4R,5S,6R)-6- (acetoxymethyl)-3-isothiocyanato-tetrahydro-2H-pyran-2,4,5-triyl triacetate (59 g, 151 mmol) in dichloromethane (500 mL) was added propan-1 -amine (9.4 g, 159 mmol) dropwise with stirring at 0 oc. After stirred for 1 h, the reaction solution was treated with trifluoroacetic acid (130 g, 1.34 mol) overnight at room temperature. The reaction mixture was washed with saturated sodium bicarbonate (300 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 1 % MeOH in dichloromethane to give compound 14 as a yellow oil (50 g, 84 %). (ES, m/z): [M+H]+ 388.9; iH MR (300 MHz, CDCI3) δ 6.14 - 6.16 (d, J = 6.6 Hz, 1H), 5.33 - 5.35 (m, 1H), 5.05 - 5.19 (m, 1H), 4.85 -4.89 (m, 1H), 4.27 - 4.30 (m, 1H), 4.08 (s, 2H), 3.75 - 3.81 (m, 1H), 3.06 - 3.28 (m, 2H), 1.97 - 2.04 (m ,9H), 1.49 - 1.58 (m, 2H), 0.82 - 0.91 (m, 3H).
Step 2
Figure imgf000042_0001
(3aR,5R,6S,7R,7aR)-5-(Hydroxymethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol (15): To a solution of (3aR,5R,6S,7R,7aR)-5- (acetoxymethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diyl diacetate (100 g, 257 mmol) in methanol (800 mL) was added potassium carbonate (18 g, 130 mmol). The resulting solution was stirred overnight at 40 oc and then cooled down to 0 oC to yield a solid This was collected by filtration, washed with cold methanol and dried. The product 15 was obtained as a white solid (50 g, 74 %). (ES, m/z): [M+H]+ 262.9; lH MR (300 MHz, D20) δ 6.17 - 6.19 (d, J= 6.3 Hz, 1H), 4.07 - 4.10 (m, 1H), 3.93 - 3.96 (m, 1H), 3.71 - 3.76 (m, 1H), 3.45 - 3.69 (m, 3H), 3.04 - 3.13 (m , 2H), 1.39 - 1.51 (m, 2H), 0.82 - 0.91 (m, 3H). Step
Figure imgf000043_0001
15 16
tert-Butyl (3aR,5R,6S,7R,7aR)-6,7-dihydroxy-5-(hydroxymethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-2-yl(propyl)carbamate (16): To a solution of
(3 aR, 5R, 6 S, 7R, 7aR)-5 -(hydroxymethyl)-2-(propylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol (67.6 g, 258 mmol) in methanol (800 mL) was added
(Boc)20 (83 g, 384 mmol). After stirred for 3 h at room temperature, the resulting mixture was concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 1 % MeOH in dichloromethane to give compound 16 as yellow oil (53 g, 46 %). (ES, m/z): [M+H]+ 363.0. lH MR (300 MHz, CDCI3) δ 6.11 - 6.14 (d, J= 6.6 Hz, 1H), 4.16 - 4.26 (m, 2H), 3.75 - 3.96 (m, 6H), 3.58 - 3.63 (m, 1H), 1.50 - 1.57 (m, 12H), 0.89 - 0.94 (m, 3H).
Step 4
Figure imgf000043_0002
16 17 tert-Butyl (3aR,5R,6S,7R,7aR)-5-((tert-butyldimethylsilyloxy)methyl)-6,7-dihydroxy- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(propyl)carbamate (17): To a stirred solution of tert-butyl (3aR,5R,6S,7R,7aR)-6,7-dihydroxy-5-(hydroxymethyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(propyl)carbamate (45 g, 124 mmol), DMAP (760 mg, 6 mmol) and triethylamine (25.1 g, 248 mmol) in dichloromethane (300 mL) was added tert-butylchlorodimethylsilane (22.4 g, 149 mmol). After stirred for 6 h at room temperature, the mixture was condensed to provide a residue, which was purified by silica gel column, eluted with 10 -30 % ethyl acetate in petroleum ether to give compound 17 as a yellow oil (43 g. 65 %). (ES, m/z): [M+H]+ 477.1; lH NMR (300 MHz, CDCI3) δ 6.12 - 6.14 (d, J = 6.3 Hz, 1H), 4.40 - 4.45 (m, 1H), 3.84 - 3.96 (m, 4H), 3.64 - 3.71 (m, 1H), 3.13 - 3.30 (m, 3H), 1.67 - 1.74 (m , 2H), 1.53 (s, 9H), 0.93 - 0.98 (m, 3H), 0.92 (s ,9H), 0.12 (s, 6H).
Step 5
Figure imgf000044_0001
tert-Butyl (3aR,5R,6S,7R,7aR)-5-((tert-butyldimethylsilyloxy)methyl)-6,7-bis(4- methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2- yl(propyl)carbamate (18): To a solution of tert-butyl (3aR,5R,6S,7R,7aR)-5-((tert- butyldimethylsilyloxy)methyl)-6,7-dihy
2-yl(propyl)carbamate (20 g, 42 mmol) in DMF (300 mL) was added sodium hydride (10 g, 417 mmol) at 15 °C in portions, followed by addition of l-(bromomethyl)-4-methoxybenzene (33.8 g, 168 mmol). The resulting solution was stirred for 1.5 h at room temperature, quenched by addition of cold water (100 mL), and extracted with dichloromethane (5x100 mL). The combined organic layers were dried over anhydrous magnesium sulfate and concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 10 -25 % ethyl acetate in petroleum ether to give compound 18 as a yellow oil (20 g, 64.8 %). (ES, m/z): [M+H]+ 717.1; iH MR (300 MHz, CDCI3) δ 7.32 - 7.35 (d, J = 8.7 Hz, 2H), 7.19 - 7.22 (d, J= 8.7 Hz, 2H), 6.83 - 6.92 (m. 4H), 6.05 - 6.07 (d, J= 6.9 Hz, 1H), 4.57 - 4.74 (m, 3H), 4.30 - 4.39 (m, 2H), 4.14 - 4.17 (t, J= 4.2 Hz , 1H), 3.68 - 3.84 (m, 11H), 3.41 - 3.45 (m, 1H), 1.67 - 1.74 (m , 2H), 1.53 (s, 9H), 0.89 (s, 9H), 0.84 (m, 3H) , 0.08 (s, 6H).
Step 6
Figure imgf000044_0002
tert-Butyl (3aR,5R,6S,7R,7aR)-5-(hydroxymethyl)-6,7-bis(4-methoxybenzyloxy)- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(propyl)carbamate (19): To a solution of tert-butyl (5R,6S,7R)-6,7-bis(4-methoxybenzyloxy)-5-((tert- butyldimethylsilyloxy)methyl)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2- yl(propyl)carbamate (19 g, 26.50 mmol) in tetrahydrofuran (100 mL) was treated with TBAF (13.8 g, 53 mmol) overnight at room temperature. The resulting solution was diluted with brine (200 mL), extracted with ethyl acetate (2 x200 mL), and dried over anhydrous magnesium sulfate. After removal of solvents, the residue was purified by silica gel column, eluted with 10-30 % ethyl acetate in petroleum ether to give compound 19 as a yellow oil (14.5 g, 91 %). (ES, m/z): [M+H]+ 603.0; iH MR (300 MHz, CDCI3) δ 7.32 - 7.35 (d, J = 8.7 Hz, 2H), 7.17 - 7.20 (d, J= 8.7 Hz, 2H), 6.83 - 6.92 (m. 4H), 6.04 - 6.06 (d, J= 6.9 Hz, 1H), 4.64 - 4.66 (m, 2H), 4.40 - 4.48 (m, 1H), 4.17 - 4.38 (m, 3H), 3.72 - 3.83 (m, 9H), 3.45 - 3.60 (m, 2H), 3.43 -. 3.45 (m, 1H), 1.58 - 1.63 (m , 2H), 1.53 (s, 9H), 0.78 - 0.83 (t , J= 7.2 Hz ,3H).
Step 7
Figure imgf000045_0001
19 20
tert-Butyl (3aR,5S,6S,7R,7aR)-5-formyl-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(propyl)carbamate (20) : To a mixture of tert- butyl (5R,6S,7R)-6,7-bis(4-methoxybenzyloxy)-5-(hydroxymethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-yl(propyl) carbamate (2 g, 3.32 mmol), KHCO3 (1.49bg, 15 mmol), TBAB (53 mg, 0.16 mmol) and TEMPO (26 mg, 0.17 mmol) in dichloromethane (60 mL) and H2O (12 mL) was added BS (592 mg, 3.33 mmol) at 15 °C. After stirred for 30 min, the reaction mixture was quenched by saturated Na2SC"3 (10 mL). The organic layer was dried over anhydrous magnesium sulfate and condensed to provide a residue, which was purified by silica gel column, eluted with 20 -30 % AcOEt in dichloromethane to give compound 20 as a yellow syrup (1.2 g, purity 60 %). (ES, m/z): [M+H] + 601.1. lH MR (300 MHz, CDCI3) δ 9.61 (s, 1H), 7.23 - 7.28 (m, 4H), 6.82 - 6.86 (m, 4H), 6.19 - 6.21 (d, J= 6.9 Hz, 1H), 4.60 - 4.67 (m, 2H), 4.41 - 4.55 (m, 3H), 4.02 - 4.03 (m, 1H), 3.88 - 3.93 (m, 1H), 3.68 - 3.81 (m, 9H), 1.46 - 1.53 (m, 11H), 0.83 - 0.86 (t, J= 7.5 Hz, 3H).
Step 8
Figure imgf000046_0001
20 21
tert-Butyl (3aR,5R,6S,7R,7aR)-l-hydroxyethyl)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a tetrahyd ro-3aH-pyrano[3,2-d]thiazol-2-yl(propyl)carbamate (21): To a solution oftert- butyl (3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-5-formyl-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-yl(propyl)carbamate (600 mg, 1.00 mmol, 60 % purity) in
tetrahydrofuran (20 mL) was added methylmagnesium bromide (1 mL, 3M in THF, 3 mmol). After stirred for 10 min at r.t, the reaction was quenched with sat. H4Cl (aq, 20 mL), extracted with dichloromethane (4x10 mL ). The combined organic layers were dried over anhydrous magnesium sulfate, and concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 30 % AcOEt in dichloromethane to give compound 21 as a yellow syrup (300 mg, 81 %, two diastereomers, faster moving one: slower moving one = 1 : 3). (ES, m/z): [M+H]+ 617.0; iH MR (300 MHz, CDCI3) δ 7.16 - 7.72 (m, 5H), 6.83 - 6.93 (m, 4H), 6.05 - 6.08 (m, 1H), 4.67 (s, 2H), 4.30 - 4.44 (m, 2H), 4.27 - 4.35 (m, 2H), 3.67 - 3.95 (m, 10H), 3.20 - 3.31 (m, 1H), 1.71 - 1.76 (m, 1H), 1.53 - 1.55 (m , 14H), 0.78 - 0.83 (m ,3H).
Step 9
Figure imgf000046_0002
21 22
(3aR, 5R, 6S,7R,7aR)-5-((S)-l-Hydroxyethyl)-2-(propylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol (22): A solution of tert-butyl (3aR,5R,6S,7R,7aR)- 6, 7-bis(4-methoxybenzyloxy)- 1 -hydroxyethyl)-5 , 6, 7, 7a-tetrahydro-3 aH-pyrano [3 ,2- d]thiazol-2-yl(propyl)carbamate (300 mg, 0.49 mmol, a mixture of two diastereomers, faster moving one: slower moving one = 1 : 3) in 20 mL dichloromethane was treated with 2 mL TFA. After stirred overnight at room temperature, the resulting mixture was concentrated under vacuum to give a crude product, which was purified by Prep-HPLC with the following conditions [(Agilent 1200 detecl), Column, 19* 150mm; mobile phase, WATER WITH 0.03% H4OH and CH3CN (10% CH3CN up to 45 % in 10 min); Detector, 254nm 220nm] to afford compound 22 (Example 2) as white solid (TFA salt, 143.1 mg, 75 %). (ES, m/z): [M+H]+ 277.0; lH MR (300MHz, D2O): δ. 6.49 - 6.45 (m, 1H), 4.14 - 4.69 (m, 1H) , 3.86 - 3.99 (m, 2H), 3.59 - 3.66 (m, 1H), 3.32 - 3.36 (m, 1H), 3.24 - 3.30 (m, 2H), 1.47 - 1.59 (m, 3H), 1.10 - 1.13 (m, 3H).
Example 3 (3aR,5S,6S,7R,7aR)-5-(2-Hydroxypropan-2-yl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-di
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000048_0002
23
4
(3aR,5R,6S,7R,7aR)-5-(Acetoxymethyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diyl diacetate (23): To a solution of (3R,4R,5S,6R)-2,4,5- triacetoxy-6-(acetoxymethyl)-tetrahydro-2H-pyran-3-aminium chloride (100 g, 261 mmol) in CH3CN (1 L) was added methyl isothiocyanate (21 g, 287 mmol), triethylamine (29 g, 287 mmol). After stirred for 12 h at 60 °C, the resulting solution was treated with TFA (110 g, 0.96 mol) at room temperature overnight, and then washed with saturated sodium bicarbonate (1 L). The organic layer was dried over magnesium sulfate and concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 1 % MeOH in dichloromethane to give compound 23 as a yellow oil (150 g, 87 %). (ES, m/z):
[M+H]+ 360.9; lH NMR (300MHz, CDCI3) δ 6.31 - 6.33 (d, J= 6.6 Hz, IH), 5.41 - 5.48 (t, J= 8.4 Hz, 1H), 4.97 - 5.00 (m, 1H), 4.31 - 4.37 (t, J= 5.7 Hz, 1H), 4.21 (m, 2H), 3.97 (m, 1H), 2.99 (s, 3H), 2.00 - 2.20 (m, 9H).
Step 2
Figure imgf000049_0001
(3aR,5R,6S,7R,7aR)-5-(Hydroxymethyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol(24): A solution of (3aR,5R,6S,7R,7aR)-5-(acetoxymethyl)- 2-(methylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diyl diacetate (150 g, 417 mmol) in methanol (1 L) was treated with potassium carbonate (11.5 g, 83 mmol). The resulting mixture was stirred overnight at room temperature to yield a solid. This was collected by filtration, washed with cold methanol and dried. The product 24 was obtained as a yellow solid (85 g, 87 %). (ES, m/z): [M+H]+ 235.1; lH MR (300MHz, D2O) δ 6.14 - 6.16 (d, J= 6.3 Hz, 1H), 4.03 - 4.07 (m, 1H), 3.89 - 3.92 (m, 1H), 3.65 - 3.70 (m, 1H), 3.48 - 3.56 (m, 2H), 3.41 - 3.45 (m, 1H), 2.69 (s, 3H).
Step 3
Figure imgf000049_0002
24 25
tert-Butyl (3aR,5R,6S,7R,7aR)-6,7-dihydroxy-5-(hydroxymethyl)-5,6,7,7a-tetrahydro-
3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (25): A solution of
(3aR,5R,6S,7R,7aR)-5-(hydroxymethyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol (85 g, 363 mmol) in methanol (600 mL) was treated with
B0C2O (117.7 g, 540 mmol) and triethylamine (73.3 g, 726 mmol). The resulting solution was stirred overnight at 45 °C, and then concentrated under vacuum to give a residue, which was purified by silica gel column, eluted with 2.5 % methanol in dichloromethane to give compound 25 as a yellow solid (90 g, 74 %). (ES, m/z): [M+H]+ 334.8; lH NMR (300MHz, CDCI3) δ 6.14 - 6.17 (d, J = 6.9 Hz, 1H), 4.19 - 4.23 (t, J = 6.3 Hz, 1H), 4.10 - 4.14 (t, J = 5.4 Hz, 1H), 3.79 - 3.84 (m, 3H), 3.60 - 3.64 (m, 2H), 3.14 (s, 3H), 1.55 (s, 9H).
Step 4
Figure imgf000050_0001
25 26
tert-Butyl (3aR,5R,6S,7R,7aR)-5-((tert-butyldimethylsilyloxy)methyl)-6,7-dihydroxy- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (26): To a mixture of tert-butyl (3aR,5R,6S,7R,7aR)-6,7-dihydroxy-5-(hydroxymethyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (30 g, 90 mmol), DMAP (550 mg, 4.5 mmol) and triethylamine (18.2 g, 180 mmol) in dichloromethane (200 mL) was added tert-butylchlorodimethylsilane (16.3 g, 108 mmol) at 0 °C. The resulting solution was stirred for 4 h at room temperature, and then concentrated under vacuum to provide a residue, which was purified by silica gel column, eluted with 1 % methanol in
dichloromethane to give compound 26 as a white solid (20 g, 50 %). (ES, m/z): [M+H]+ 449.0; iH MR (300MHz, CDCI3) δ 6.10 - 6.12 (d, J = 6.9 Hz, 1H), 4.15 - 4.24 (m, 2H), 3.83 - 3.97 (m, 5H), 3.60 (m, 1H), 2.58 - 2.66 (m, 2H), 1.55 (s, 9H), 1.18 (m, 3H), 0.91 (s, 9H), 0.09 (s, 6H).
Step 5
Figure imgf000050_0002
26 27
tert-Butyl (3aR,5R,6S,7R,7aR)-5-((tert-butyldimethylsilyloxy)methyl)-6,7-bis(4- methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano [3,2-d] thiazol-2- yl(methyl)carbamate (27): To a solution of (3aR,5R,6S,7R,7aR)-5-((tert- butyldimethylsilyloxy)methyl)-6,7-dihydroxy-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol- 2-yl(methyl)carbamate (20 g, 45 mmol) in DMF (150 mL) was added sodium hydride (10.7 g, 446 mmol) in portion at 0 °C, and followed by addition of l-(bromomethyl)-4- methoxybenzene (36 g, 179 mmol). The resulting solution was stirred for 2 h at room temperature, quenched with cold water (200 mL), and extracted with ethyl acetate (3 x 500 mL). The combined organic layers were washed with brine (5 x 200 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to afford a residue, which was purified by silica gel column, eluted with 10 % ethyl acetate in petroleum ether to give compound 27 as a yellow liquid (21 g, 68 %). (ES, m/z): [M+H]+ 689.1. lH NMR
(300MHz, CDCI3) δ 7.31 - 7.37 (m, 2H), 7.22 - 7.28 (m, 2H), 6.10 - 6.12 (d, J= 6.9 Hz, 1H) , 4.60 - 4.74 (m, 4H), 4.20 - 4.47 (m, 4H), 3.82 (s, 6H), 3.69 (s, 3H), 3.33 - 3.37 (m, 2H), 1.56 (s, 9H), 0.89 (s, 9H) , 0.05 (s , 6H).
Step 6
Figure imgf000051_0001
27 28
tert-Butyl (3aR,5R,6S,7R,7aR)-5-(hydroxymethyl)-6,7-bis(4-methoxybenzyloxy)- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (28): A solution of (3aR,5R,6S,7R,7aR)-5-((tert-butyldimethylsilyloxy)methyl)-6,7-bis(4- methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (1.54 g, 2.24 mmol) in THF (30 mL) was treated with TBAF (1.17 g, 4.5 mmol) overnight at room temperature. The resulting solution was diluted with brine (20 mL), extracted with ethyl acetate (2 x 50 mL), and dried over anhydrous magnesium sulfate. After removal of solvents, the residue was purified by silica gel column, eluted with 20 % ethyl acetate in petroleum ether to give compound 28 as a yellow solid (0.8 g, 62 %). (ES, m/z): [M+H]+ 575.2, lH NMR (300MHz, CDCI3) δ 7.33 - 7.36 (d, J= 7.8 Hz, 2H), 7.20 - 7.22 (d, J = 8.1 Hz, 2H), 6.08 - 6.10 (d, J= 6.3 Hz, 1H) , 4.27 - 4.71(m, 6H), 3.82 (s, 6H), 3.48 - 3.70 (m, 4H), 3.33 (s, 3H), 1.76 - 1.80 (t, J= 7.2 Hz, 1H), 1.54 (s, 9H). Step 7
Figure imgf000052_0001
28 29
(3aR,5S,6S,7R,7aR)-2-(tert-Butoxycarbonyl(methyl)amino)-6,7-bis(4- methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano [3,2-d] thiazole-5-carboxylic acid (29): A mixture of tert-butyl (3aR,5R,6S,7R,7aR)-5-(hydroxymethyl)-6,7-bis(4- methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (1 g, 1.74 mmol), KHCO3 (780 mg, 7.8 mmol), TBAB (28 mg, 0.09 mmol) and TEMPO (14 mg, 0.09 mmol) in dichloromethane(30 mL) and H2O (6 mL) was treated with BS (620 mg, 3.5 mmol) overnight at room temperature. The reaction mixture was adjusted to acidic (pH at 3) with hydrochloric acid, and then extracted with dichloromethane(3 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under vacuum to provide a residue , which was purified by a silica gel column, eluted with 20-50 % ethyl acetate in petroleum ether to give compound 29 as a white solid (600 mg, 58 %). (ES, m/z): [M+H]+ 589.0, lH MR (300MHz, CDCI3) δ 7.25 - 7.32 (m , 2H), 6.85
- 6.90 (m , 2H), 6.08 - 6.10 (d, J= 6.3 Hz, 1H) , 4.50 - 4.62 (m, 5H), 4.22 - 4.29 (m, 2H), 3.97 (m, 1H), 3.81(s, 6H), 3.34 (s, 3H), 1.54 (s, 9H).
Step 8
Figure imgf000052_0002
29 30
(3aR,5S,6S,7R,7aR)-Methyl 6,7-bis(4-methoxybenzyloxy)-2-(tert-butoxycarbonyl)- 5,6,7,7a-tetrahydro-3aH-pyrano [3,2-d] thiazole-5-carboxylate (30): A solution of (3aR,5S,6S,7R,7aR)-2-(tert-butoxycarbonyl(methyl)amino)-6,7-bis(4-methoxybenzyloxy)- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-5-carboxylic acid (600 mg, 1.02 mmol) in methanol (30 mL) was treated with EDC (392 mg, 2.04 mmol) for 2 h at room temperature. After removal of solvents, the residue was purified by a silica gel column, eluted with 20 % ethyl acetate in petroleum ether to give compound 30 (510 mg, 83 %). (ES, m/z): [M+H]+ 603.0, iH MR (300MHz, CDCI3) δ 7.22 - 7.29 (m , 4H), 6.83 - 6.91 (m, 4H), 6.07 - 6.09 (d, J= 5.7 Hz, 1H), 4.48 - 4.61 (m , 4H), 4.30 - 4.31 (m, 1H), 4.22 - 4.29 (m, 2H), 4.17 - 4.19 (t, J= 4.8 Ηζ, ΙΗ), 3.82 (s, 6H), 3.67 (s, 3H), 3.30 (s, 3H), 1.54 (s, 9H).
Ste 9
Figure imgf000053_0001
30 31 : R=Boc
31 a: R=H
tert-Butyl (3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-5-(2-hydroxypropan-2- yl)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (31) and 2- ((3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(methylamino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)propan-2-ol (31a): Compound 30 (510 mg) in THF (20 mL) was treated with methylmagnesium chloride (1 mL, 3M in THF) for 2 h at room temperature. The reaction was quenched with brine (20 mL), and extracted with dichloromethane (3 x 20 mL). The combined organic layers were dried over anhydrous magnesium sulfate and concentrated under vacuum to provide a residue, which was purified by a silica gel column, eluted with 20 % ethyl acetate in petroleum ether to give compound 31 as a yellow oil (308 mg, 57 %). (ES, m/z): [M+H]+ 603.3; lH NMR (300MHz, CDCI3) δ 7.34 - 7.37 (d , J= 8.4 Hz, 2H), 7.19 - 7.22 (d, J= 8.4 Hz, 2H), 6.90 - 6.93 (dd, J= 2.1 Hz, 1.8 Hz, 2H), 6.83 - 6.86 (dd, J = 2.1 Hz, 1.8 Hz, 2H), 6.15 - 6.18 (d, J = 7.2 Hz, 1H) , 4.68 (s, 2H), 4.35 - 4.48 (m,3H),4.23 - 4.26 (d, J= 4.5 Ηζ, ΙΗ), 3.86 (s, 6H), 3.26 (s, 3H), 3.21 (d, J= 8.1 Hz, 1H), 1.54 (s, 9H); and then eluted with 1% MeOH in dichloromethane to give 31a as a white solid (152 mg, 31 %). (ES, m/z): [M+H]+ 503.3; lH NMR (300MHz, CDCI3) δ 7.28 - 7.34 (m , 4H), 6.85 - 6.92 (m , 4H), 6.34 - 6.37 (d, J= 7.2 Hz, 1H) , 4.49 - 4.68 (m, 5H), 4.25 - 4.29 (m, 2H), 3.76 - 3.86 (m, 8H), 3.34 - 3.37 (m, 1H), 3.90 (s, 3H). Ste 10
Figure imgf000054_0001
(3aR,5S,6S,7R,7aR)-5-(2-Hydroxypropan-2-yl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol (32): A solution of tert-butyl (3aR,5S,6S,7R,7aR)-5- (2-hydroxypropan-2-yl)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazol-2-yl(methyl)carbamate (200 mg, 0.33 mmol) in DCM (20 mL) was treated with trifluoroacetic acid (2 mL) overnight at room temperature. The reaction mixture was condensed to give a residue, which was purified by Prep-HPLC with the following conditions [(3#-Agilent 1200 detect prep HPLC): Column, C18, 19* 150mm, 5um; mobile phase, water with 0.03% ammonia and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV220nm] to give compound 32 (Example 3) as a white solid (67.9 mg, 78 %). (ES, m/z): [M+H]+ 263.1; lH MR (300 MHz, D2O) δ 6.20 - 6.22 (d, J= 6.3 Hz, 1H), 4.31 (m, 1H), 4.28 - 4.30 (m, 1H), 3.68 - 3.71 (m, 1H), 3.14 - 3.17 (d, 2H), 2.71 (s, 3H), 1.10 (s, 3H), 1.07 (s, 3H).
Example 4
(3aR,5S,6S,7R,7aR)-2-(dimethylamino)-5-(2-hydroxypropan-2-yl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000054_0002
Scheme V
Figure imgf000055_0001
33
l-((3aR,5S,6S,7R,7aR)-2-(dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanone (33): A solution of 1- ((3aR,5R,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(dimethylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-5-yl)ethanol (700 mg, 1.39 mmol, a mixture of diastereomers: faster moving one: slower moving one = 1 : 5) in dichloromethane (25 mL) was treated with DMP (1.18 g, 2.78 mmol) for 2 h at room temperature. The reaction mixture was quenched by saturated Na2S204 (25 mL) and saturated NaHC03 (25 mL), extracted with
dichloromethane(2 x 50 mL). The combined organic layers were concentrated under vacuum to provide a residue, which was purified by a silica gel column with 10 % AcOEt in dichloromethane to give compound 33 as a yellow oil (550 mg, 71 %). (ES, m/z): [M+H]+ 500.9; lH MR (300 MHz, CDCI3) δ 7.24 - 7.30 (m, 4H), 6.83 - 6.90 (m, 4H), 6.27 - 6.29 (d, J= 6.3 Hz, 1H), 3.80 - 4.60 (m, 14H), 3.02 (s, 6H), 2.16 (s, 3H). Step 2
Figure imgf000056_0001
33 34
2-((3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)propan-2-ol (34): A solution of 1- ((3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(dimethylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-5-yl)ethanone (500 mg, 1.00 mmol) in THF (20 mL) was treated with methylmagnesium bromide (1 mL, 3M in THF) for 1 h at room temperature, then quenched by brine (50 mL), and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were dried over anhydrous magnesium sulfate and concentrated under vacuum to provide a residue, which was purified by a silica gel column with 1 % methanol in dichloromethane to give compound 34 as a yellow oil (480 mg, 84 %). (ES, m/z): [M+H]+ 516.9; iH MR (300MHz, CDCI3) δ 7.33 - 7.36 (d, J= 8.7 Hz, 2H), 6.83 - 6.94 (m, 4H), 6.32 - 6.34 (d, J= 6.9 Hz, 1H) , 4.47 - 4.71 (m, 5H), 4.28 - 4.36 (m, 2H), 3.82 (s, 6H), 3.36 - 3.49 (m , 1H), 3.29 (s, 6H), 1.18 (s, 6H).
Step 3
Figure imgf000056_0002
34 35
(3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-5-(2-hydroxypropan-2-yl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol (35 as its TFA salt): A solution of 2- ((3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(dimethylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-5-yl)propan-2-ol (480 mg, 0.93 mmol) in dichloromethane (20 mL) was treated with TFA (2 mL) for 1 h at room temperature. The reaction mixture was condensed to give a residue, which was purified by Prep-HPLC with the following conditions [(3#- Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19*50mm 5um; mobile phase, WATER with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min; Detector, UV 220nm)] to give compound 35 (Example 4) as a white solid (TFA salt, 238 mg, 92 %). (ES, m/z): [M+H]+ 276.9, lH NMR (300MHz, CDCI3) δ 6.46 - 6.48 (d, J= 7.5 Hz, 1H), 4.46 - 4.49 (m, 1H), 4.08 - 4.10 (m, 1H), 3.81 - 3.87 (m , 1H), 3.28 - 3.30 (d, J= 8.4 Hz, 1H), 3.18 (s, 3H), 3.12 (s, 3H), 1.07 - 1.20 (m, 6H).
Example 5
(3aR,5R,6S,7R,7aR)-2-(DimethyIamino)-5-(2-hydroxyethyI)-5,6,7,7a-tetrahyd pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000057_0001
Step l
Figure imgf000057_0002
11 37
(3aR,5R,6R,7R,7aR,E)-6,7-Bis(4-methoxybenzyloxy)-5-(2-methoxyvinyl)-N,N- dimethyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-amine (37) : A solution of Ph3PCH20CH3Br (2.0 g, 5.90 mmol) in THF (30 mL) was treated with t-BuOK (645 mg,
5.76 mmol) at -10 °C for 30 min, and followed by addition of (3aR, 5S, 6S, 7R, 7aR)-2- (dimethylamino)-6, 7-bis(4-methoxybenzyloxy)-5, 6, 7, 7a-tetrahydro-3aH-pyrano [3, 2-d] thiazole-5-carbaldehyde (700 mg, 1.37 mmol). After stirred for 3 h at 0°C, the resulting solution was quenched with ice-water (20 mL), and extracted with ethyl acetate (2 x 20 mL). The combined organic layers were dried over anhydrous magnesium sulfate and concentrated to give a residue, which was purified by a silica gel column, eluted with 10 - 50 % ethyl acetate in petroleum ether to give the product 37 as a yellow oil (100 mg, 14 %). (ES, m/z)[M+H]+ 515.0; 1H MR (300 MHz, CDCI3) δ 7.15 - 7.35 (m, 4H), 6.84 - 6.98 (m, 4H), 6.45 (d, J= 7.8 Hz, 1H ), 6.28 (d, J= 6.3 Hz, 1H), 5.21 (m, 1H), 4.45 - 4.89 (m, 4H), 4.18 (t, J= 5.7 Hz, 1H), 3.94 (t, J= 4.8 Hz, 1H), 3.87 - 3.91 (m, 1H), 3.78 (s, 6H), 3.56 - 3.65 (m, 1H), 3.52 (s, 3H), 2.92 (s, 6H).
Step 2
Figure imgf000058_0001
37 38
2-((3aR,5R,6R,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano [3,2-d] thiazol-5-yl)ethanol (38) : To a cold solution of 37
(100 mg, 0.18 mmol) in acetonitrile (20 mL) at -5 °C was added a solution of Hg(OAc)2
(68 mg, 0.21 mmol) in water (10 mL) dropwise. After stirred at 0 °C for 4 h, the reaction mixture was treated with potassium iodide (40 mg, 0.24 mmol) in water (1.6 mL) for 10 min. The reaction solution was quenched with water, extracted with dichloromethane (40 mL), and dried over MgS04. Removal of solvents gave crude 2-((3aR,5R,6R,7R,7aR)- 6,7-bis(4-methoxybenzyloxy)-2-(dimethylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazol-5-yl)acetaldehyde. To a solution of the crude product in dry THF (20 mL) was added NaBH4 (16 mg, 0.42 mmol) at 0 °C in portions. After stirred at 0 °C overnight, the reaction solution was quenched with water (25 mL) and extracted with ethyl acetate (2 x 20 mL). The combined organic phases were washed with saturated aqueous H4CI, dried over MgS04, and concentrated to give a residue, which was purified by silica gel chromatography, eluted with 20 - 50 % ethyl acetate in petroleum ether to give the product 38 as a light yellow oil (50 mg, 48 %).(ES, m/z)[M+H]+ 503.0; 1H MR (300 MHz, CDCI3) δ 7.15 - 7.35 (m, 4H), 6.84 - 6.98 (m, 4H), 6.28 (d, J= 6.3 Hz, 1H), 4.45 - 4.89 (m, 4H), 4.18 (t, J= 5.7 Hz, 1H), 3.94 (t, J= 4.8 Hz, 1H), 3.87 - 3.91 (m, 1H), 3.78 (s, 6H), 3.56 - 3.65 (m, 3H), 2.92 (s, 6H), 1.89 - 1.98 (m, 1H), 1.62 - 1.73 (m, 1H).
Step 3
Figure imgf000059_0001
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-(2-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol (39) : A solution of 2-((3aR,5R,6R,7R,7aR)-2- (dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3, 2- d]thiazol-5-yl)ethanol (100 mg, 0.18 mmol) in dichloromethane (20 mL) was treated with TFA overnight at room temperature. Removal of solvents provided a residue, which was purified by Prep-HPLC with the following conditions: (Agilent 1200 prep HPLC: Column, Sun fire prep. CI 8; mobile phase, water with 0.03 % trifiuoroacetic acid and CH3CN (10 % up to 20 % in time 10); Detector, 220 nm.) to give the product 39 (Example 5) as a white solid (5.0 mg, 11 %). (ES, m/z)[M+H]+ 263.0; 1H MR (300 MHz, D2O) δ 6.29 (d, J= 6.3 Hz, 1H), 4.18 (t, J= 5.7 Hz, 1H), 3.98 (t, J= 4.8 Hz, 1H), 3.56 - 3.65 (m, 3H), 3.39 - 3.43 (m, 1H), 2.92 (s, 6H), 1.89 - 1.98 (m, 1H), 1.62 - 1.72 (m, 1H).
Example 6 & 1
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-ethyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6,7-diol and (3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-vinyl-5,6,7,7a- tetrahydro-3aH-pyrano [3,2-d] thiazole-6,7-diol
Figure imgf000059_0002
Scheme VII
Figure imgf000060_0001
Step l
Figure imgf000060_0002
11 40
(3aR,5R,6R,7R,7aR)-6,7-Bis(4-methoxybenzyloxy)-N,N-dimethyl-5-vinyl-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-amine (40) : To a suspension of
methyltriphenylphosphonium bromide (5.9 g, 16.5 mmol) in THF (70 mL) was treated with n-BuLi (5.9 ml , 2.5 M in THF) for 30 min at 0 °C, and followed by addition of
(3aR,5S,6S,7R,7aR)-2-(dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-5-carbaldehyde (1.6 g, 3.29 mmol) in THF (10 mL). After stirred overnight at room temperature, the reaction mixture was quenched with water (50 mL), extracted with dichloromethane (3 x 50 mL). The organic phases was combined and dried over MgSO-i^ concentrated under vacuum to give a residue, which was purified by a silica gel column, eluted with 30 % ethyl acetate in petroleum ether to give compound 40 as a yellow oil (520 mg, purity 57 % by LC-MS). This material was used in the next step without further purification. (ES, m/z): [M+H]+ 485.0.
Step 2
Figure imgf000061_0001
40 41
(3aR,5R,6R,7R,7aR)-5-Ethyl-6,7-bis(4-methoxybenzyloxy)-N,N-dimethyl-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-amine (41) : A solution of above crude compound 2 (300 mg, purity 57 % by LC - MS) was dissolved into ethanol (20 mL) and treated with Ranney Ni (200 mg) under ¾ atmosphere overnight at room temperature.
After filtration, the organic solution was concentrated to give crude compound 41 (260 mg, purity 54 % by LC-MS), which was used in the next step without further purification. (ES, /// r): [M+H]+ 487.0.
Step 3
Figure imgf000061_0002
41 42
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-ethyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6,7-diol (42) : A solution of above crude product 41 (260 mg, purity 54 % by LC - MS) in dichloromethane (10 mL) was treated with TFA (1 mL) for 4 hours at room temperature. The solution was adjusted to pH at 8 with NH4OH, and then condensed to give a crude product, which was purified by Prep-HPLC with the following conditions (Agilent 1200 prep HPLC: Column, SunFire Prep C18, 19*50mm 5um; mobile phase, WATER with 0.03 %NH40H and CH3CNQO %CH3CN up to 45 % in 10 min; Detector, UV 220nm) to provide compound 42 (Example 6) as a white solid (38 mg). (ES, m/z): [M+H]+ 247.0. 1H NMR (300 MHz, D2O) 5 6.18 - 6.21 (d, J = 6.6 Hz, 1H), 4.09 - 4.13 (t, 7 = 6.0 Hz, 1H), 3.91 - 3.94 (t, J= 4.8 Hz, 1H), 3.42 - 3.44 (m, 2H), 2.92 (s, 6H), 1.69 - 1.77 (m, 1H), 1.40 - 1.50 (m, 1H), 0.84 - 0.89 (t, J= 7.5 Hz, 3H).
Figure imgf000062_0001
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-vinyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6,7-diol (43) : A solution of above crude product 40 (220 mg, purity 57 % by LC-MS) in dichloromethane (10 mL) was treated with TFA (1 mL) for 4 hours at room temperature. The solution was adjusted to pH at 8 with H4OH, and then condensed to give a crude product, which was purified by Prep-HPLC with the following conditions (Agilent 1200 prep HPLC: Column, SunFire Prep C18, 19*50mm 5um; mobile phase, WATER with 0.03 % H4OH and CH3CNQO % CH3CN up to 45 % in 10 min; Detector, UV 220nm) to provide compound 43 (Example 7) as a white solid (35.7 mg). (ES, m/z): [M+H]+ 245.0. 1H MR (300 MHz, D2O) δ 6.20 - 6.22 (d, J= 6.3 Hz, 1H), 5.79 - 5.87 (m, 1H), 5.30 - 5.38 (m, 2H), 3.98 - 4.08 (m, 2H), 3.87 - 3.91 (t, J= 6.0 Hz, 1H), 3.46 - 3.52 (m, 1H), 2.93 (s, 6H).
Example 8
(3aR,5R,6S,7R,7aR)-5-Ethyl-2-(methylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6,7-diol
Figure imgf000062_0002
Scheme VIII
Figure imgf000063_0001
44 45 46
Figure imgf000063_0002
47
Step l
Figure imgf000063_0003
44 45 tert-Butyl (3aR,5R,6R,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-5-vinyl-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (45) : A suspension of methyltriphenylphosphonium bromide (1.85 g, 5.18 mmol) in tetrahydrofuran (35 mL) was treated with n-BuLi (1.9 ml, 2.5M) for 30 min at 0 °C, and followed by addition of tert-butyl (3aR,5S,6S,7R,7aR)-5-formyl-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (0.6 g, 1.05 mmol) in THF (5 mL). The resulting solution was stirred overnight at room temperature, quenched by water (25 mL), and extracted with dichloromethane (3 x 25 ml). The combined organic layers were dried over magnesium sulfate and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluted with 20 % ethyl acetate in petroleum ether to give crude compound 45 as a yellow oil (220 mg, purity 50 % by LC-MS). (ES, m/z): [M+H]+ 571.0. Step 2
Figure imgf000064_0001
45 46 tert-Butyl (3aR,5R,6R,7R,7aR)-5-ethyl-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(methyl)carbamate (46) : A solution of above crude compound 45 (220 mg, purity 50 %) was dissolved into ethanol (15 mL) and treated with Ranney Ni (100 mg) under ¾ atmosphere overnight at room temperature. After filtration, the organic solution was concentrated to give crude compound 46 (200 mg, purity 48 % by LC-MS), which was used in the next step without further purification. (ES, m/z): [M+H]+ 573.0.
Step 3
Figure imgf000064_0002
(3aR,5R,6S,7R,7aR)-5-Ethyl-2-(methylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6,7-diol (47) : A solution of above crude product 46 (200 mg, purity 48 % by LC-MS) in dichloromethane (10 mL) was treated with TFA (1 mL) for 4 hours at room temperature. The solution was adjusted to pH at 8 with NH4OH, and then condensed to give a crude product, which was purified by Prep-HPLC with the following conditions (Agilent 1200 prep HPLC: Column, SunFire Prep C18, 19*50mm 5um; mobile phase, WATER with 0.03 % NH4OH and CH3CNQO % CH3CN up to 45 % in 10 min; Detector, UV 220nm) to provide the title compound 47 (Example 8) as a white solid (7.6 mg). (ES, m/z): [M+H]+ 233.0. 1H NMR (300 MHz, D2O) δ 6.18 - 6.20 (d, J= 6.3 Hz, 1H), 4.09 - 4.13 (t, J= 6.0 Hz, 1H), 3.92 - 3.95 (t, J= 4.8 Hz, 1H), 3.36 - 3.45 (m, 2H), 2.75 (s, 3H), 1.65 - 1.75 (m, 1H), 1.38 - 1.48 (m, 1H), 0.81 - 0.86 (t, J= 7.5 Hz, 3H). Example 9
(3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-5-((S)-l-fluoroethyl)-5,6,7,7a-tetrahyd pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000065_0001
50 51
Step 1
Figure imgf000065_0002
l-((3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano [3,2-d] thiazol-5-yl)ethanone (33) : A solution of 1- ((3aR,5R,6S,7R,7aR)-2-(dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-5-yl) ethanol (450 mg, 0.90 mmol, a mixture of diastereomers, faster eluting isomer: slower eluting isomer = 1 : 5 by HPLC and 1HNMR) in dichloromethane (20 mL) was treated with Dess-Martin reagent (760 mg, 1.80 mmol) for 2 hours at room temperature. The reaction mixture was quenched by sat. aqueous Na2S2C"3 solution (20 mL) and sat. aqueous NaHC03 solution (20 mL), and extracted with dichloromethane (2 x 20 mL). The combined, organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by silica gel column, eluted with 2 - 3 % methanol in dichloromethane to give compound 33 as a yellow syrup (400 mg, 88 %). (ES, m/z): [M+H]+ 501.0; 1H NMR (300 MHz, CDCI3) δ 7.25 - 7.30 (m, 4H), 6.83 - 6.90 (m, 4H), 6.29 - 6.31 (d, J= 6.0 Hz, 1H), 4.26 - 4.64 (m, 6H), 3.81 - 3.98 (m, 8H), 2.96 - 3.02 (m, 6H), 2.17 (s, 3H).
Step 2
Figure imgf000066_0001
(R)-l-((3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanol (48) and (S)-l-
((3aR,5R,6S,7R,7aR)-2-(dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanol (48a): A solution of 1- ((3aR,5S,6S,7R,7aR)-2-(dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanone (400 mg, 0.80 mmol) in methanol (10 mL) was treated with NaBH4 (150 mg, 3.95 mmol) for 1 hour at room temperature. The reaction was quenched by water (10 mL), and extracted with ethyl acetate (3 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by Chiral- HPLC, eluted with 20 % ethanol in hexane to give compound 48 as a light yellow oil (slower eluting isomer by Chiral-HPLC, 270 mg, 67 %). (ES, m/z): [M+H]+ 503.0; 1H NMR (300 MHz, CDCI3) δ 7.22 - 7.36 (m, 4H), 6.84 - 6.92 (m, 4H), 6.34 (broad, 1H), 4.53 - 4.71 (m, 4H), 4.25 - 4.35 (m, 2H), 3.76 - 3.94 (m, 8H), 3.45 - 3.49 (dd, J= 4.2 Hz, 4.5 Hz, 1H), 3.01 (s, 6H), 1.19 - 1.21 (d , J= 6.0 Hz, 1H), 1.11 - 1.13 (d , J= 6.0 Hz, 3H); and compound 48a as a light yellow oil (faster eluting isomer by Chiral-HPLC, 113 mg, 28 %).(ES, m/z): [M+H]+ 503.0; 1H MR (300 MHz, CDCI3) δ 7.32 - 7.35 (d, J= 8.4 Hz, 2H), 7.24 - 7.27 (d, J = 8.7 Hz, 2H), 6.83 - 6.91 (m, 4H), 6.31 - 6.33 (d, J= 6.6 Hz, 1H), 4.60 - 4.71 (m, 2H), 4.53 - 4.57 (m, 2H), 4.25 - 4.35 (m, 2H), 3.85 - 3.87 (m, 7H), 3.73 - 3.75 (d, J= 8.7 Hz, 1H), 3.32 - 3.36 (dd, J= 3.6 Hz, 3.3 Hz, 1H), 3.01 (s, 6H), 1.92 - 1.94 (d , J= 7.5 Hz, 1H), 1.19 - 1.21 (d , 7= 6.3 Hz, 3H).
Step 3
Figure imgf000067_0001
(3aR,5S,6S,7R,7aR)-5-((S)-l-Fluoroethyl)-6,7-bis(4-methoxybenzyloxy)-N,N-dimethyl- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-amine (50) : A solution of (R)-l- ((3aR,5R,6S,7R,7aR)-2-(dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-5-yl)ethanol (280 mg, 0.56 mmol) in dichloromethane (10 mL) was treated with DAST (900 mg, 5.59 mmol) at -78 °C for 10 min and at 0 °C for 1 hr. The reaction was quenched by sat. aqueous Na2CC"3 solution (30 mL), and extracted with dichloromethane (2 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by silica gel column, eluted with 1 - 2 % methanol in dichloromethane to give compound 50 as a yellow syrup (240 mg, 85 %). (ES, m/z): [M+H]+ 505.0; 1H MR (300 MHz, CDCI3) δ 7.22 - 7.43 (m, 4H), 6.83 - 6.94 (m, 4H), 6.38 - 6.50 (m, 1H), 4.58 - 4.78 (m, 6H), 3.75 - 3.90 (m, 8H), 3.46 - 3.50 (m, 1H), 3.05 - 3.18 (m, 6H), 1.27 - 1.36 (m, 3H).
Step 4
Figure imgf000067_0002
50 51
(3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-5-((S)-l-fluoroethyl)-5,6,7,7a-tetrahyd pyrano[3,2-d]thiazole-6,7-diol (51) : A solution of (3aR,5S,6S,7R,7aR)-5-((S)-l- fluoroethyl)-6,7-bis(4-methoxybenzyloxy)-N,N-dimethyl-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-amine (240 mg, 0.48 mmol) in dichloromethane (20 mL) was treated with TFA (2 mL) for 1 h at room temperature. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by Prep-HPLC under the following conditions [(Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19 * 50 mm 5 urn; mobile phase, water with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV 220nm] to afford compound 51 (Example 9) as a white solid (30 mg, 24 %). (ES, m/z): [M+H]+ 265.0; 1H MR (300 MHz, D2O) δ 6.23 - 6.25 (d, J= 6.0 Hz, 1H), 4.93 - 4.99 (m, 0.5H), 4.77 - 4.84 (m, 0.5H), 4.17 (t, J= 6.0 Hz, 1H), 4.00 (t, J= 6.0 Hz, 1H), 3.71 - 3.74 (m, 1H), 3.36 - 3.49 (m, 1H), 2.94 (s, 1H), 1.25 - 1.35 (dd, J= 6.6 Hz, 6.3 Hz, 3H).
Example 10 and 11 (3aR,5S,6S,7R,7aR)-2-(dimethylamino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol and (3aR,5S,6S,7R,7aR)-2- (dimethylamino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000068_0001
Scheme X
Figure imgf000069_0001
Step l
Figure imgf000069_0002
l-((3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)-2,2,2-trifluoroethanol (52, mixture of two diastereomers): To a stirred mixture of TBAF (107 mg, 0.41 mmol) and 4 A molecule sieves in THF (20 mL) was added a solution of (3aR,5S,6S,7R,7aR)-2-(dimethylamino)-6,7- bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-5-carbaldehyde (1) (400 mg, 0.82 mmol) and TMS-CF3 (230 mg, 1.64 mmol) in THF (5 mL) at 0 °C. After stirring for 4 hours at 0 °C, the reaction was quenched by brine (30 mL), and extracted with ethyl acetate (3 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by silica gel column, eluted with 2 - 3 % methanol in dichloromethane to give compound 52 as a yellow syrup (300 mg, 65 %, a mixture of diastereomers, faster eluting isomer : slower moving isomer = 1 : 2 by Chiral-HPLC). (ES, m/z): [M+H]+ 557.0; 1H MR (300 MHz, CDCI3) δ 7.20 - 7.35 (m, 4H), 6.85 - 6.92 (m, 4H), 6.25 - 6.27 (d, J = 6.6 Hz, 1H), 4.56 4.69 (m, 5H), 4.30 - 4.36 (m, 2H), 3.82 - 3.83 (m, 8H), 2.99 - 3.00 (m, 6H).
Step 2
Figure imgf000070_0001
(3aR,5S,6S,7R,7aR)-2-(dimethylamino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol (53) and (3aR,5S,6S,7R,7aR)-2- (dimethylamino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol (54) : A solution of l-((3aR,5R,6S,7R,7aR)-2- (dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazol-5-yl)-2,2,2-trifluoroethanol (52, a mixture of two diastereomers from previous step) (400 mg, 0.72 mmol) in dichloromethane (20 mL) was treated with TFA (2 mL) for 1 h at room temperature. The reaction mixture was concentrated under reduced pressure to give a residue, which was purified by Prep-HPLC under the following conditions [(Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19 * 50 mm 5 um; mobile phase, water with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV 220nm] to afford compound 53 (Example 10) as a white solid (faster eluting isomer, 62 mg, 27 %): (ES, m/z): [M+H]+ 316.9; 1H MR (300 MHz, D2O) δ 6.19 - 6.21 (d, J = 6.6 Hz, 1H), 4.22 - 4.27 (m, 1H), 4.04 (t, J = 6.6 Hz, 1H), 3.86 - 3.90 (m, 1H), 3.71 - 3.76 (m, 1H), 2.93 (s, 6H); Compound 54 (Example 11) as a white solid (slower eluting isomer, 55 mg, 24 %).(ES, m/z): [M+H]+ 316.9; 1H NMR (300 MHz, D2O) δ 6.25 - 6.27 (d, J = 6.3 Hz, 1H), 4.26 - 4.34 (m, 1H), 4.10 (t, J= 6.0 Hz, 1H), 3.94 (t, J= 5.4 Hz, 1H), 3.72 - 3.82 (m, 1H), 2.92 (s, 6H).
Example 12
(3aR,5S,6S,7R,7aR)-5-(l,l-Difluoroethyl)-2-(dimethylamino)-5,6,7,7a-tetrahyd pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000071_0001
SchemeXI
Figure imgf000071_0002
11 55
(S)-l-((3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanol (55) : A solution of 1- ((3aR,5S,6S,7R,7aR)-2-(dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-5-yl)ethanone (280 mg, 0.56 mmol) in dichloromethane (10 mL) was treated with (diethylamino) sulfur trifluoride (451 g, 2.80 mol) and ethanol (5.2 mg, 0.11 mmol) overnight at room temperature. The reaction was then quenched by addition of sat. aqueous sodium carbonate solution (50 mL), and extracted with dichloromethane (3 x 20 mL). The combined organic layers were dried over anhydrous magnesium sulfate and condensed under vacuum to give a crude product 55 (140 mg), which was used in the next step without further purification. (ES, m/z) [M+H]+ 523.0; 1H MR (300 MHz, CD3CI) δ 1H MR (300 MHz, CDCI3) δ 7.24 - 7.30 (m, 4H), 6.84 - 6.95 (m, 4H), 6.30 - 6.32 (m, 1H), 4.57 - 4.62 (m, 4H), 4.27 - 4.35 (m, 2H), 3.81 - 3.91 (m, 8H), 2.99 - 3.01 (m, 6H), 1.56 - 1.62 (m, 3H). Step 2
Figure imgf000072_0001
55 56 l-((3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanone (56) : A solution of crude product 2 (140 mg, 0.27 mmol, 1.00 equiv) in dichloromethane (10 mL) was treated with TFA (1 mL) for 2 hrs at room temperature, and then condensed o give a residue, which was purified by Prep-HPLC with the following conditions [(Prep-HPLC): Column, 19* 150mm; mobile phase, Water with 0.03% ammonia and CH3CN (10 % CH3CN up to 35 % in 10 min); Detector, 220nm.] to give the title compound 56 (Example 12) as a white solid (76.3 mg, 40 %). (ES, m/z) [M+H]+ 282.9; 1H NMR (300 MHz, D2O) δ 6.23 (d, J= 6.6 Hz, 1H), 4.35 (t, J= 5.1 Hz, 1H), 4.14 (t, J= 2.7 Hz, 1H), 3.91 - 3.95 (m, 1H), 3.57 - 3.67 (m, 1H), 2.94 (s, 6H), 1.59 (t, J= 19.8 Hz, 3H).
The following examples were synthesized according to procedures analogous to the schemes and examples outlined above.
Table 1
Figure imgf000072_0002
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
isomer)
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
OH 5 , 6, 7, 7a-tetrahydro-3 aH-
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
isomer)
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Examples 137 and 138
(3aR,5S,6S,7R,7aR)-5-(l-hydroxycyclopentyl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol and (3aR,5S,6S,7R,7aR)-5-(l-hydroxycyclopent-3- enyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000090_0002
Figure imgf000090_0003
Figure imgf000091_0001
4-((3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(methylamino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)hepta-l,6-dien-4-ol . A solution of
(3aR,5S,6S,7R,7aR)-methyl 2-(tert-butoxycarbonyl(methyl)amino)-6,7-bis(4-metho xybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-5-carboxylate (1.0 g, 1.66 mmol) in THF (15 mL) was treated with allylmagnesium chloride (8.0 mL, 0.5 M in THF) for 1 hour at room temperature The reaction was quenched by aqueous H4CI (20 mL), and extracted with ethyl acetate (2 x 30 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to give a residue, which was purified by a silica gel column with 1 % methanol in dichloromethane to afford the title compound as a yellow oil (0.75 g, 81 %). (ES, m/z) [M+H]+ 555.0; 1H MR (300 MHz, CDCI3) δ 7.23 - 7.35 (m, 4H), 6.84 - 6.93 (m, 4H), 6.34 - 6.37 (d, J= 6.9 Hz, 1H), 5.86 - 5.92 (m, 2H), 5.06 - 5.11 (m, 4H), 4.65 - 4.66 (m, 2H), 4.50 - 4.54 (m, 2H), 4.26 - 4.33 (m, 2H), 3.96 - 3.99 (d, J= 8.4 Hz, 1H), 3.81 - 3.83 (m, 6H), 3.51 - 3.54 (d, J= 8.4 Hz, 1H), 2.92 (s, 3H), 2.22 - 2.31 (m, 4H).
Figure imgf000091_0002
l-((3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(methylamino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)cyclopent-3-enol . To a solution of compound 57 (0.60 g, 1.08 mmol) in dichloromethane (10 mL) was added Grubbs' catalyst (0.19 g, 0.22 mmol) under nitrogen atmosphere. After stirring for 12 hours at room temperature, the reaction mixture was concentrated under reduced pressure to afford a residue, which was purified by a silica gel column with 30 % - 40 % ethyl acetate in petroleum ether to afford compound 58 as a gray oil (0.35 g, 61 %). (ES, m/z) [M+H]+ 527.0; 1H MR (300 MHz, CDCI3) δ 7.24 - 7.35 (m, 4H), 6.85 - 6.92 (m, 4H), 6.34 - 6.36 (d, J= 6.9 Hz, 1H), 5.66 - 5.67 (m, 2H), 4.49 - 4.65 (m, 4H), 4.28 - 4.31 (m, 2H), 3.90 - 3.93 (d, J= 8.4 Hz, 1H), 3.81 - 3.82 (m, 6H), 3.55 - 3.57 (d, J= 8.4 Hz, 1H), 2.92 (s, 3H), 2.58 - 2.78 (m, 2H), 2.23 - 2.34 (m, 2H).
Step 3
Figure imgf000092_0001
l-((3aR,5S,6S,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-2-(methylamino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)cyclopentanol . A mixture of compound 58 (0.35 g, 0.67 mmol) and Pd/C (0.10 g) in methanol (20 mL) was stirred under hydrogen atmosphere for 20 hours at room temperature. The reaction mixture was filtered and the filtrate was concentrated under vacuum to afford a residue, which was purified by a silica gel column with 30 % - 40 % ethyl acetate in petroleum ether to afford crude compound 59 as a yellow oil (300 mg), which was employed in the next step without further purification.
[M+H]+ 529.0.
Step 4
Figure imgf000092_0002
(3aR,5S,6S,7R,7aR)-5-(l-hydroxycyclopentyl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol : A solution of the above compound 59 (100 mg) in dichloromethane (5 mL) was treated with TFA (0.5 mL) for 4 hours at room temperature. The reaction solution was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions [(Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19 * 50 mm 5 urn; mobile phase, ¾0 with 0.03 % H4OH and CH3CN (10 %
CH3CN up to 45 % in 10 min); Detector, UV 220 nm] to afford compound 60 (Example 137) as a white solid (5.0 mg). (ES, m/z): [M+H]+ 289.0;1H NMR (300 MHz, D2O) δ 6.22 - 6.24 (d, J= 6.9 Hz, 1H), 4.29 - 4.32 (m, 1H), 4.12 - 4.14 (m, 1H), 3.80 - 3.83 (m, 1H), 3.30 - 3.33 (d, J= 8.4 Hz, 1H), 2.74 (s, 3H), 1.51 -1.66 (m, 8H).
Figure imgf000093_0001
(3aR,5S,6S,7R,7aR)-5-(l-hydroxycyclopent-3-enyl)-2-(methylamino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol : A solution of compound 58 (100 mg, 0.19 mmol)) in dichloromethane (5 mL) was treated with TFA (0.5 mL) for 4 hours at room temperature. The reaction solution was concentrated under reduced pressure. The residue was purified by Prep-HPLC with the following conditions [(Agilent 1200 prep HPLC):
Column, SunFire Prep C18, 19 * 50 mm 5 urn; mobile phase, ¾0 with 0.03 % H4OH and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV 220 nm] to afford compound 61 (Example 138) as a white solid (15.0 mg, 27 %). (ES, m/z): [M+H]+ 287.0; 1H NMR (300 MHz, D2O) δ 6.25 - 6.28 (d, J= 6.9 Hz, 1H), 5.62 - 5.64 (m, 2H), 4.31 - 4.37 (m, 1H), 4.13 - 4.15 (m, 1H), 3.76 - 3.81 (m, 1H), 3.44 - 3.47 (d, J= 8.4 Hz, 1H), 2.76 (s, 3H), 2.58 - 2.64 (m, 2H), 2.16 - 2.31 (m, 2H).
Examples 139 and 140
(3aR,5S,6S,7R,7aR)-5-(2-fluoropropan-2-yl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol and (3aR,5R,6S,7R,7aR) -2- (methylamino)-5- (prop-l-en-2-yl)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol.
Figure imgf000094_0001
Scheme XIII
Figure imgf000094_0002
Step l
Figure imgf000094_0003
31 a 62 63
(3aR,5S,6S,7R,7aR)-5-(2-fluoropropan-2-yl)-6,7-bis(4-methoxybenzyloxy)-N-methyl- 5,6,7,7a-tetrahydro-3aH-pyrano [3,2-d] thiazol-2-amine and (3aR,5R,6R,7R,7aR)-6,7- bis(4-methoxybenzyloxy)-N-methyl-5-(prop-l-en-2-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-amine. To a solution of 2-((3aR,5S,6S,7R,7aR)-6,7-bis(4- methoxybenzyloxy)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-5- yl)propan-2-ol (450.0 mg, 0.90 mmol) in dichloromethane (20 mL) was added DAST (720.0 mg, 4.47 mmol) at -78°C. After stirred for 1 hour at 0 °C, the reaction was quenched by aqueous potassium carbonate, extracted with dichloromethane (2 x 50 mL), dried over anhydrous sodium sulfate, and concentrated under reduced pressure to afford a mixture of 62 and 63 as a yellow oil (400.0 mg), which was used for next step without further purification: (ES, /// r) [M+H]+ 505.0 and (ES, m/z) [M+H]+ 485.0. Step 2
Figure imgf000095_0001
62 63 64 65
(3aR,5S,6S,7R,7aR)-5-(2-fluoropropan-2-yl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol and (3aR,5R,6S,7R,7aR) -2- (methylamino)-5- (prop-l-en-2-yl)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol. A solution of the above crude products (400.0 mg) in dichloromethane (20 mL) was treated with trifluoroacetic acid (2 mL) for 2 hours at room temperature. Removal of volatiles gave a residue, which was purified by Prep-HPLC with the following conditions [(Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19 * 50 mm 5 urn; mobile phase, ¾0 with 0.03 % TFA and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, UV 220 nm] to afford compound 64 (Example 139) as its TFA salt as a white solid (50.0 mg). (ES, m/z): [M+H]+ 265.0;1H NMR (300 MHz, CD3OD) δ 6.56 - 6.58 (d, J= 7.2 Hz, 1H), 4.48 - 4.49 (m, 1H), 4.12 - 4.14 (m, 1H), 3.92 - 3.94 (m, 1H), 3.44 - 3.51 (m, 1H), 3.07 (s, 3H), 1.45 - 1.47 (d, J = 6.3 Hz, 3H), 1.37 - 1.39 (d, J= 6.6 Hz, 3H); and compound 65 (Example 140) as its TFA salt as a white solid (17.8 mg). (ES, m/z): [M+H]+ 245.0; 1H NMR (300 MHz, D2O) δ 6.57 - 6.61 (m, 1H), 5.10 - 5.11 (m, 2H), 4.09 - 4.19 (m, 2H), 3.84 - 3.94 (m, 1H), 3.58 - 3.66 (m, 1H), 2.97 (s, 3H), 1.71 (s, 3H).
Example 141
(3aR,5R,6S,7R,7aR)-2-(2-Fluoroethylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000095_0002
Scheme XIV
Figure imgf000096_0001
Step 1
Figure imgf000096_0002
(3aR,5R,6S,7R,7aR)-5-(Acetoxymethyl)-2-(2-fluoroethylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diyl diacetate (66): A solution of
(2S,3R,4R,5S,6R)-6-(acetoxymethyl)-3-isothiocyanato-tetrahydro-2H-pyran-2,4,5-triyl triacetate (30 g, 77 mmol) in dichloromethane (100 mL) was added 2-fluoroethylamine hydrochloride (8.4 g, 84 mmol) and triethylamine (11.7 mL, 115 mmol). The resulting solution was stirred for 1 hour at room temperature, and then condensed to give a light yellow foam, which was dissolved into dichloromethane (100 mL) and treated with 2,2,2- trifluoroacetic acid (75.6 g, 663 mmol) overnight at room temperature. The reaction mixture was quenched by saturated aqueous sodium carbonate (500 mL), washed with sodium chloride (3 x 50 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 50 % ethyl acetate in petroleum ether to provide compound 66 as a light yellow oil (24 g, 70 %). (ES, m/z) [M+H]+ 393.0; 1H MR (300 MHz, CDCI3) δ 6.27 - 6.29 (d, J= 6.60 Hz, 1H), 5.42 - 5.44 (t, J= 2.7 Hz, 1H), 4.95 - 4.99 (dd, J= 1.8 Hz, 9.5 Hz, 1H), 4.70 - 4.73 (m, 1H), 4.54 - 4.65 (m, 1H), 4.37 - 4.39 (t, J= 0.9 Hz, 1H), 4.16 - 4.17 (m, 2H), 3.81 - 3.87 (m, 1H), 3.56 - 3.71 (m, 2H), 2.09 (s, 3H), 2.11 (s, 3H), 2.14 (s, 3H).
Ste 2
Figure imgf000097_0001
(3aR,5R,6S,7R,7aR)-2-(2-Fluoroethylamino)-5-(hydroxymethyl)-5,6,7,7a-tetrahydro-
3aH-pyrano[3,2-d]thiazole-6,7-diol (67): A solution of (3aR,5R,6S,7R,7aR)-5-
(acetoxymethyl)-2-(2-fluoroethylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7- diyl diacetate (29 g, 74 mmol) in methanol (200 mL) was treated with potassium carbonate
(1 g, 7 mmol) overnight at room temperature. The solids were collected by filtration and washed with dichloromethane (3 x 50 mL) to afford compound 67 as a white solid (18 g,
92 %). (ES, m/z) [M+H]+ 266.9; 1H MR (300 MHz, D2O) δ 6.21 - 6.23 (d, J= 6.3 Hz,
1H), 4.55 - 4.59 (m, 1H), 4.40 - 4.44 (m, 1H), 4.10 - 4.14 (t, J= 5.7 Hz, 1H), 3.95 - 3.98 (t,
J= 5.4 Hz, 1H), 3.70 - 3.75 (m, 1H), 3.40 - 3.62 (m, 5H).
Step 3
Figure imgf000097_0002
tert-Butyl (3aR,5R,6S,7R,7aR)-6,7-dihydroxy-5-(hydroxymethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(2-fluoroethyl)carbamateb (68): To a solution of (3aR,5R,6S,7R,7aR)-2-(2-fluoroethylamino)-5-(hydroxymethyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol (13.7 g, 51 mmol) in methanol (100 mL) was added di-tert-butyl dicarbonate (22 g, 102 mmol) and triethylamine (8.6 mL). After stirred overnight at room temperature, the resulting mixture was concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 20 % methanol in dichloromethane to give compound 68 as a white syrup (16 g, 85 %). (ES, m/z) [M+H]+ 367.0; 1H MR (300 MHz, CDCI3) δ 6.18 - 6.20 (d, J= 6.3 Hz, 1H), 4.70 - 4.76 (m, 1H), 4.55 - 4.60 (m, 1H), 4.43 - 4.48 (m, 1H), 4.27 - 4.30 (m, 1H), 3.86 - 3.98 (m, 3H), 3.71 - 3.79 (m, 2H), 3.49 - 3.62 (m, 2H), 1.57 (s, 9H).
Step 4
Figure imgf000098_0001
69 tert-Butyl (3aR,5R,6S,7R,7aR)-5-((tert-butyldimethylsilyloxy)methyl)-6,7-dihydroxy- 5,6,7,7a-tetrahydro-3aH-pyrano [3,2-d] thiazol-2-yl(2-fluoroethyl)carbamate (69) :
To a solution of tert-butyl (3aR,5R,6S,7R,7aR)-6,7-dihydroxy-5-(hydroxymethyl)- 5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(2-fluoroethyl)carbamate (16 g, 43.7 mmol) in dichloromethane (100 mL) was added tert-butylchlorodimethylsilane (7.6 g, 50.7 mmol), 4-dimethylaminopyridine (530 mg, 4.34 mmol) and triethylamine (6.6 g, 65 mmol). After stirred for 6 h at room temperature, the resulting mixture was concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 20 % petroleum in ethyl acetate to give compound 69 as pale yellow oil (10 g, 48 %). (ES, m/z) [M+H]+ 481.0; 1H MR (300 MHz, CDCI3) δ 6.17 - 6.19 (d, J= 6.3 Hz, 1H), 4.42 - 4.74 (m, 3H), 3.52 - 4.26 (m, 7H), 1.56 (s, 9H), 0.92 (s, 9H), 0.10 (s, 6H).
Step 5
Figure imgf000098_0002
tert-Butyl (3aR,5R,6S,7R,7aR)-6,7-bis(benzyloxy)-5-((tert- butyldimethylsilyloxy)methyl)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(2- fluoroethyl)carbamate (70): A solution of tert-butyl (3aR,5R,6S,7R,7aR)-5-((tert- butyldimethylsilyloxy)methyl)-6,7-dihy
2-yl(2-fluoroethyl)carbamate (2.0 g, 4.2 mmol) in N,N-dimethylformamide (30 mL) was treated with sodium hydride (510 mg, 21 mmol) for 10 min at 10 °C in a nitrogen
atmosphere, and followed by addition of l-(bromomethyl)benzene (2.2 g, 13 mmol). After stirred for 15 min, the reaction was quenched with saturated aqueous H4CI and extracted with dichloromethane (3 x 30 mL). The organic layers were dried over anhydrous magnesium sulfate and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 1.5 - 3 % ethyl acetate in petroleum ether to provide compound 70 as a yellow oil (0.7 g, 25 %). (ES, m/z) [M+H]+ 660.8; 1H MR (300 MHz, CDCI3) δ 7.28 - 7.43 (m, 10H), 6.11 - 6.13 (d, J= 6.3 Hz, 1H), 4.60 - 4.74 (m, 4H), 4.30 - 4.41 (m, 6H), 3.73 - 3.75 (m, 3H), 3.37 - 3.44 (m, 1H), 1.54 (s, 9H), 0.90 (s, 9H), 0.06 (s, 6H).
Step 6
Figure imgf000099_0001
70 71 tert-Butyl (3aR,5R,6S,7R,7aR)-6,7-bis(benzyloxy)-5-(hydroxymethyl)-5,6,7,7a- tetrahydro-3aH-pyrano [3,2-d] thiazol-2-yl(2-fluoroethyl)carbamate (71): A solution of tert-butyl (3aR,5R,6S,7R,7aR)-6,7-bis(benzyloxy)-5-((tert- butyldimethylsilyloxy)methyl)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(2- fluoroethyl)carbamate (1.36 g, 2.06 mmol) in THF (30 mL) was treated with
tetrabutylammonium fluoride (1.1 g, 4.2 mmol) for 2 hrs at room temperature. The reaction mixture was quenched with ¾0 (10 mL), extracted with ethyl acetate (3 x 50 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 10 % - 20 % ethyl acetate in petroleum ether to provide compound 71 as a pale yellow oil (600 mg, 53 %). (ES, m/z) [M+H]+ 546.8; 1H NMR (300 MHz, CDCI3) δ 7.24 - 7.43 (m, 10H), 6.09 - 6.11 (d, J= 4.2 Hz, 1H), 4.60 - 4.66 (m, 2H), 4.50 - 4.54 (m, 1H), 4.46 - 4.47 (m, 1H), 4.40 - 4.45 (m, 2H), 4.27 - 4.39 (m, 2H), 4.12 - 4.22 (m, 2H), 3.72 - 3.79 (m, 1H), 3.67 - 3.71 (m, 1H), 3.55 - 3.63 (m, 1H), 3.40 - 3.45 (m, 1H), 1.54 (s, 9H).
Ste 7
Figure imgf000100_0001
71 72
tert-Butyl (3aR,5S,6S,7R,7aR)-6,7-bis(benzyloxy)-5-formyl-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-yl(2-fluoroethyl)carbamate (72): A mixture oftert-butyl
(3aR,5R,6S,7R,7aR)-6,7-bis(benzyloxy)-5-(hydroxymethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-yl(2-fluoroethyl)carbamate (600 mg, 1.10 mmol), KHCO3 (495 mg, 4.95 mmol), TBAB (18 mg, 0.06 mmol) and TEMPO (9 mg, 0.06 mmol) in dichloromethane (17 mL) and water (2mL) was treated with B S (215 mg, 1.21 mmol) for 15 min at 10 - 15 °C. The reaction mixture was quenched by saturated aqueous Na2SC"3 (5 mL); extracted with dichloromethane (3 x 15 mL), dried over magnesium sulfate, and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 20 % ethyl acetate in petroleum ether to provide compound 72 as a yellow syrup (0.4 g, 67 %). (ES, m/z) [M+H]+ 545.0; 1H MR (300 MHz, CDCI3) δ 9.64 (s, 1H), 7.23 - 7.39 (m, 10H), 6.09 - 6.11 (d, J= 6.3 Hz, 1H), 4.55 - 4.75 (m, 5H), 4.34 - 4.45 (m, 2H), 4.13 - 4.28 (m, 3H), 3.90 - 4.12 (m, 2H), 1.54 (s, 9H).
Step 8
Figure imgf000100_0002
72 73
tert-Butyl (3aR,5R,6S,7R,7aR)-6,7-bis(benzyloxy)-5-(l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(2-fluoroethyl)carbamate (73): To a solution of tert-butyl (3aR,5S,6S,7R,7aR)-6,7-bis(benzyloxy)-5-formyl-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazol-2-yl(2-fluoroethyl)carbamate (400 mg, 0.73 mmol) in THF (20 mL) was added methylmagnesium chloride (1 mL, 3M in THF). After stirred for 3 hours at 0-5 °C, the resulting solution was quenched with saturated aqueous H4CI (10 mL) and extracted with ethyl acetate (3 x 30 mL). The organic layers were dried over anhydrous magnesium sulfate and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 20 % ethyl acetate in petroleum ether to give compound 73 as a yellow oil (0.37 g, 90 %, diasteromers' ratio is 1 :4). (ES, m/z) [M+H]+ 561.0; 1H MR (300 MHz, CDCI3) δ 7.25 - 7.44 (m, 10H), 6.13 - 6.15 (d, J= 6.9 Hz, 1H), 4.73 - 4.78 (m, 2H), 4.54 - 4.68 (m, 2H), 4.28 - 4.49 (m, 4H), 4.07 - 4.25 (m, 2H), 3.75 - 3.97 (m, 2H), 3.12 - 3.17 (m, 1H), 1.54 (s, 9H), 1.22 - 1.24 (d, J= 6.3 Hz, 3H).
Step 9
Figure imgf000101_0001
(3aR,5R,6S,7R,7aR)-2-(2-Fluoroethylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol 2,2,2-trifluoroacetate (74): A solution of tert-butyl (3aR,5R,6S,7R,7aR)-6,7-bis(benzyloxy)-5-(l-hydroxyethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazol-2-yl(2-fluoroethyl)carbamate (250 mg, 0.45 mmol) in
dichloromethane (7 mL) was treated with BCI3 (7 mL, lM in dichloromethane) at -20 - -10 °C for 2 hrs. The reaction was quenched with methanol and concentrated under vacuum to give a crude product (0.25 g), which was purified by Prep-HPLC (Agilent 1200 prep HPLC; Column, X-Bridge 19* 150mm 5um; mobile phase, water with 0.05 % trifluoroacetic acid and CH3CN (10 % up to 20 % in time 10); Detector, 220 nm.) to provide compound 74 (Example 141) as a white solid (55.7 mg, 45 %). (ES, m/z) [M+H]+ 281.0; 1H MR (300 MHz, D2O) δ 6.53 - 6.55 (d, J= 6.9 Hz, 1H), 4.61 - 4.78 (m, 1H), 4.47 - 4.52 (m, 1H), 4.22 - 4.30 (m, 1H), 3.93 - 4.04 (m, 2H), 3.58 - 3.73 (m, 3H), 3.38 - 3.42 (m, 1H), 1.15 - 1.17 (d, J= 6.6 Hz, 3H).
Example 142
(3aR,5R,6S,7R,7aR)-2-(3-Fluoropropylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano [3,2-d] thiazole-6,7-diol
Figure imgf000102_0001
The title compound was synthesized following procedures ananalgous to Example 141.
(ES, m/z) [M+H]+ 295.0; IH MR (300 MHz, D2O) δ 6.51 - 6.57 (m, 1H), 4.60 - 4.67 (m, 1H), 4.41 - 4.45 (m, 1H), 4.22 - 4.30 (m, 1H), 3.96 - 4.00 (m, 2H), 3.58 - 3.65 (m, 4H), 1.89 - 2.07 (m, 2H), 1.15 - 1.19 (m, 3H).
Example 143
(3aR,5R,6S,7R,7aR)-5-(3-Hydroxypropyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000102_0002
Scheme XV
Figure imgf000102_0003
44 76
LiAIH4,THF
Figure imgf000102_0004
78 79 Ste 1
Ρ
Figure imgf000103_0001
(E)-Ethyl 3-((3aR,5R,6R,7R,7aR)-2-(tert-butoxycarbonyl(methyl)amino)-6,7-bis(4- methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano [3,2-d] thiazol-5-yl)acrylate (76) : A solution of 44 (420 mg, 0.51 mmol) in toluene (20 mL) was treated with
(carbethoxymethylene)triphenylphosphorane (280 mg, 0.81 mmol) overnight at 80 °C. The reaction mixture was concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 10 % ethyl acetate in petroleum ether to provide compound 76 as a light yellow solid (231 mg, 70 %). (ES, m/z) [M+H]+ 643.3; 1H NMR (300 MHz, CDCI3) δ 7.31 - 7.34 (d, J= 8.4 Hz, 2H), 7.20 - 7.22 (d, J= 8.7 Hz, 2H), 6.85 - 6.96 (m, 5H), 6.02 - 6.08 (m, 2H), 4.63 - 4.71 (m, 2H), 4.53 - 4.59 (m, 1H), 4.35 - 4.41 (m, 1H), 4.16 - 4.35 (m, 4H), 3.98 -3.99 (m, 1H), 3.82 (s, 6H), 3.48 - 3.52 (m, 1H), 3.20 (s, 3H), 1.54 (s, 9H), 1.26 - 1.31 (t, J= 6.9 Hz, 3H).
Step 2
Figure imgf000103_0002
Ethyl 3-((3aR,5R,6R,7R,7aR)-2-(tert-butoxycarbonyl(methyl)amino)-6,7-bis(4- methoxybenzyloxy)-5,6,7,7a-tetrahydro-3aH-pyrano [3,2-d] thiazol-5-yl)propanoate (77) :
A mixture of compound 76 (100 mg, 0.16 mmol) and Raney Ni (20 mg) in methanol (10 mL) was stirred under hydrogen atmosphere overnight at room temperature. After removal of solids, the solution was concentrated under vacuum to give 77 as a yellow oil (80 mg, 76 %). (ES, m/z) [M+H]+ 645.3; 1H NMR (300 MHz, CDC13) δ 7.34 - 7.35 (d, J= 6.6 Hz, 2H), 7.19 - 7.20 (d, J= 6.6 Hz, 2H), 6.84 - 6.93 (m, 4H), 6.02 - 6.04 (d, J= 6.9 Hz, 1H), 4.60 - 4.71 (m, 2H), 4.49 - 4.52 (d, J= 8.1 Hz, 1H), 4.37 - 4.40 (m, 1H), 4.27 - 4.30 (d, J= 8.1 Hz, 1H), 4.22 - 4.24 (m, 1H), 4.07 - 4.14 (m, 2H), 3.82 (s, 6H), 3.80 - 3.81 (m, 1H), 3.40 - 3.43 (m, 1H), 3.10 (s, 3H), 2.25 - 2.45 (m, 2H), 2.01 - 2.09 (m, 1H), 1.66 - 1.74 (m, 1H), 1.54 (s, 9H), 1.22 - 1.29 (t, J= 6.9 Hz, 3H).
Step 3
Figure imgf000104_0001
77
Figure imgf000104_0002
79
(3aR,5R,6S,7R,7aR)-5-(3-Hydroxypropyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol (79): A solution of compound 77 (150 mg, 0.23 mmol) in THF (10 mL) was treated with LiAlH4 (26.6 mg, 0.70 mmol) for 3 h at room temperature. The reaction mixture was then quenched by addition of saturated aqueous H4C1 (20 mL), extracted with dichloromethane (3 x 30 mL), and dried over anhydrous magnesium sulfate. The filtrate was concentrated under vacuum to give crude compound 78 as a yellow oil (90 mg), which was dissolved into dichloromethane (10 mL) and treated with TFA (1 mL) overnight at room temperature. Removal of volatiles gave a residue, which was purified by Prep-HPLC with the following conditions: [(Agilent 1200 prep HPLC: Column, Sun fire prep. C18; mobile phase, water with 0.03 % trifiuoroacetic acid and CH3CN (10 % up to 20 % in time 10); Detector, 220 nm.)] to give the title compound 79 (Example 143) as a white solid (10.8 mg, 23 %). (ES, m/z) [M+H]+ 263.1; 1H MR (300 MHz, D20) 5 6.18 - 6.20 (d, J= 6.3 Hz, 1H), 4.08 - 4.12 (m, 1H), 3.89 - 3.93 (m, 1H), 3.36 - 3.51 (m, 4H), 2.74 - 2.76 (m, 3H), 1.68 - 1.76 (m, 1H), 1.33 - 1.62 (m, 3H).
Example 144
(3aR,5R,6S,7R,7aR)-2-(Ethylamino)-5-(2-hydroxyethyl)-5,6,7,7a-tetrahydr( pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000104_0003
Scheme XVI
Figure imgf000105_0001
80 81
Figure imgf000105_0002
Step 1
Figure imgf000105_0003
80 81 tert-Butyl (3aR,5R,6R,7R,7aR)-6,7-bis(4-methoxybenzyloxy)-5-((E)-2-methoxyvinyl)-
5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazol-2-yl(ethyl)carbamate (81): A solution of Ph3PCH2OCH3Cl (613 mg, 1.79 mmol) in THF (20 mL) was treated with potassium t-butoxide (191 mg, 1.71 mmol) at 0 °C in a nitrogen atmosphere for 30 min, and followed by addition of 80 (500 mg, 0.85 mmol) in THF (5 mL). The resulting solution was stirred for 3 hours at 25 °C, and then quenched by saturated aqueous H4C1 (20 mL), extracted with dichloromethane (3 x 20 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to give a residue, which was purified by a silica gel column, eluting with 30 % ethyl acetate in petroleum ether to give compound 81 as a light yellow solid (200 mg, 38 %). (ES, m/z) [M+H]+ 615.3. Step 2
Figure imgf000106_0001
(3aR,5R,6S,7R,7aR)-2-(Ethylamino)-5-(2-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol A solution of compound 81 (200 mg, 0.33 mmol) in CH3CN (10 mL) and water (10 mL) was treated with Hg(OAc)2 (128 mg, 0.40 mmol) for 4 hours at -5 °C, and followed by addition of KI (277 mg, 1.67 mmol) and NaBH4 (50.67 mg, 1.33 mmol) sequentially. The resulting mixture was stirred overnight at 0 °C, and then quenched by saturated aqueous H4C1 (20 mL), extracted with dichloromethane (3 x 20 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous magnesium sulfate, and condensed to give crude compound 82 as a yellow foam, which was dissolved into dichloromethane (10 mL) and treated with TFA (1 mL) overnight at room temperature. Removal of volatiles provided a residue, which was purified by Prep-HPLC with the following conditions [(Prep-HPLC): Column, 19* 150 mm; mobile phase, WATER with 0.03 % H3H20 and CH3CN (10 % CH3CN up to 45 % in 10 min); Detector, 220 nm.] to give the title compound 83 (Example 144) as a yellow solid (8.1 mg, 28 %). (ES, m/z) [M+H]+ 263.1; 1H MR (300 MHz, D20) δ 6.18 - 6.20 (d, J= 6.3 Hz, 1H), 4.06 - 4.10 (t, J = 5.4 Hz, 1H), 3.87 - 3.91 (t, J= 3.9 Hz, 1H), 3.50 - 3.64 (m, 3H), 3.38 - 3.42 (m, 1H), 3.10 - 3.22 (m, 2H), 1.88 - 1.99 (s, 1H), 1.65 - 1.69 (m, 1H), 0.96 - 1.05 (t, J= 6.9 Hz, 3H).
The following Example 145 was prepared in a similar fashion to Example 144. Example 145
Figure imgf000107_0003
Example 146
(3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-5-(2-fluoropropan-2-yl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000107_0001
85 86 87
Figure imgf000107_0002
(S)-l-((3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanol (12): A solution of 11 (600 mg, 1.23 mmol) in THF (10 mL) was treated with methylmagnesium chlorine (0.82 mL, 3M in THF) for 2 hours at room temperature, and then quenched with saturated aqueous H4C1 (20 mL), extracted with ethyl acetate (3 x 20 mL). The combined organic layers were dried over anhydrous magnesium sulfate and concentrated under vacuum to give a residue, which was purified by silica gel column, eluting with 1 - 2 % methanol in dichloromethane to give compound (12) as a yellow syrup (500 mg, 81 %, diastereomers' ratio is ~ 1 :4). (ES, m/z): [M+H]+ 503.0. 1H MR (300 MHz, CDC13) δ 7.23 - 7.35 (m, 4H), 6.85 - 6.92 (m, 4H), 6.30 - 6.32 (d, J = 6.6 Hz, 1H), 4.52 - 4.67 (m, 4H), 4.33 - 4.37 (m, 2H), 3.81 - 3.84 (m, 8H), 3.35 - 3.50 (m, 1H), 2.99 - 3.02 (m, 6H), 1.11 - 1.21 (m, 3H).
Step 2
Figure imgf000108_0001
12 84 l-((3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)ethanone (84): A solution of 12 (450 mg, 0.90 mmol) in dichloromethane (20 mL) was treated with Dess-Martin reagent (760 mg,
1.80 mmol) for 2 hours at room temperature, and then quenched by saturated aqueous Na2S203 (20 mL) and saturated aqueous NaHC03 (20 mL), extracted with dichloromethane (2 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by silica gel column, eluting with 2 - 3 % methanol in dichloromethane to give compound (84) as a yellow syrup (400 mg, 88 %). (ES, m/z): [M+H]+ 501.0. 1H MR (300 MHz, CDC13) δ 7.25 - 7.30 (m, 4H), 6.83 - 6.90 (m, 4H), 6.29 - 6.31 (d, J= 6.0 Hz, 1H), 4.26 - 4.64 (m, 6H),
3.81 - 3.98 (m, 8H), 2.96 - 3.02 (m, 6H), 2.17 (s, 3H). Step 3
Figure imgf000109_0001
84 85
2-((3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazol-5-yl)propan-2-ol (85): A solution of compound 84 (100 mg, 0.2 mmol) in THF (10 mL) was treated with methylmagnesium chlorine (0.5 mL, 3M in THF) for 2 hours at room temperature, then quenched with saturated aqueous H4C1 (20 mL), extracted with ethyl acetate (3 x 20 mL). The combined organic layers were dried over anhydrous magnesium sulfate, concentrated under vacuum to give a residue, which was purified by silica gel column, eluting with 1 - 2 % methanol in dichloromethane to give compound (85) as a yellow syrup (100 mg, 97 %). (ES, m/z): [M+H]+ 517.0. 1H MR (300 MHz, CDCls) δ 7.22 - 7.36 (m, 4H), 6.83 - 6.92 (m, 4H), 6.32 - 6.34 (d, J= 6.9 Hz, 1H), 4.25 - 4.70 (m, 7H), 3.81 (s, 6H), 3.33 - 3.36 (d, J= 8.7 Hz, 1H), 2.98 (s, 6H), 1.18 (s, 6H).
Step 4
Figure imgf000109_0002
(3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-5-(2-fluoropropan-2-yl)-5,6,7,7a-tetrahydro- 3aH-pyrano [3,2-d] thiazole-6,7-diol (87) : A solution of 85 ( 100 mg, 0.19 mmol) in dichloromethane (10 mL) was treated with DAST (156 mg, 0.97 mmol) at -78 °C for 30 min. The reaction mixture was allowed to warm up to room temperature gradually. After stirring for another 30 min, the reaction was quenched with saturated aqueous Na2C03 (10 mL) and extracted with dichloromethane (3 x 10 mL). The combined organic layers were dried over anhydrous MgS04 and concentrated under reduced pressure to give crude compound 86 as a yellow oil (100 mg), which was dissolved into dichloromethane (10 mL) and treated with TFA (1 mL) for 1 hour at room temperature. Removal of volatiles gave a residue, which was purified by Prep-HPLC with the following conditions [(Agilent 1200 Detecl Prep-HPLC): Column, SunFire Prep C18; mobile phase, water with 0.03% ammonia and CH3CN; Detector, UV220nm] to afFord the title compound 87 (Example 146) as a white solid (34.8 mg, 65 %). (ES, m/z): [M+H]+ 279.0; 1H NMR (300 MHz, D20) δ 6.21 - 6.23 (d, J= 6.6 Hz, 1H), 4.33 - 4.36 (t, J= 6.3 Hz, 1H), 4.14 - 4.16 (t, J= 2.7 Hz, 1H), 3.78 - 3.81 (m, 1H), 3.30 - 3.39 (m, 1H), 2.91 (s, 6H), 1.34 (s, 3H) 1.27 (s, 3H).
Example 147
(3aR,5S,6S,7R,7aR)-5-(l,l-Difluoroethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000110_0001
SCHEME XVIII
tert-Butyl (3aR,5S,6S,7R,7aR)-5-acetyl-6,7-bis(4-methoxybenzyloxy)-5,6,7,7a- tetrahydro-3aH-pyrano [3,2-d] thiazol-2-yl(propyl)carbamate (88) : A solution of 21 (450 mg, 0.72 mmol) in dichloromethane (10 mL) was treated with Dess- martin reagent (780 mg, 1.83 mmol) for 1 hour at room temperature, then quenched by saturated aqueous Na2S203 (20 mL) and saturated aqueous NaHC03 aqueous (20 mL), extracted with dichloromethane (2 x 20 mL). The combined organic layers were dried over anhydrous sodium sulfate and concentrated under reduced pressure to provide a residue, which was purified by 30 % ethyl acetate in petroleum ether to give compound 88 as light yellow oil (210 mg, 47 %). (ES, m/z): [M+H]+ 615.0. 1H MR (300 MHz, CDC13) δ 7.21 - 7.31 (m, 4H), 6.84 - 6.92 (m, 4H), 6.05 - 6.07 (d, J= 6.9 Hz, 1H), 4.26 - 4.64 (m, 6H), 3.87 - 3.92 (m, 2H), 3.80 (s, 6H), 3.70 - 3.79 (m, 2H), 2.21 (s, 3H), 1.60 - 1.70 (m, 2H), 1.25 - 1.30 (t, 7= 7.5 Hz, 3H).
Step 2
Figure imgf000111_0001
(3aR,5S,6S,7R,7aR)-5-(l,l-Difluoroethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano [3,2-d] thiazole-6,7-diol (90): A solution of 88 (200 mg, 0.33 mmol) in
dichloromethane (10 mL) was treated with DAST (262 mg, 1.63 mmol) overnight at room temperature, then quenched by saturated aqueous Na2C03 (10 mL), extracted with dichloromethane (3 x 10 mL). The combined organic layers were concentrated under vacuum to give crude compound 89 as a yellow foam (100 mg), which was dissolved into dichloromethane (5 mL) and treated with TFA (0.5 mL) for 3 hours at room temperature. Removal of volatiles gave a residue, which was purified by Prep-HPLC with the following conditions [(Agilent 1200 prep HPLC): Column, SunFire Prep C18, 19 * 50 mm 5 urn; mobile phase, H20 with 0.03 % H4OH and CH3CN (10% CH3CN up to 45 % in 10 min); Detector, UV 220 nm] to give the title compound 90 (Example 147) as a yellow solid (21.8 mg, 21.8 %). (ES, m/z) [M+H]+ 297.0; 1H NMR (300 MHz, D20) δ 6.20 - 6.22 (d, J = 6.6 Hz, 1H), 4.33 - 4.36 (t, J = 4.2 Hz, 1H), 4.15 - 4.18 (t, J = 3.0 Hz, 1H), 3.93 - 3.97 (m, 1H), 3.56 - 3.66 (m, 1H), 3.10 - 3.14 (t, 7 = 6.9 Hz, 2H), 1.43 -1.68 (m, 5H), 0.81 - 0.86 (t, 7 = 7.5 Hz, 3H).
The following compound was prepared in a manner analogous to Example
147 .
Example 148
(3aR,5S,6S,7R,7aR)-5-(l, l-difluoroethyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol
Figure imgf000112_0001
Biological Activity
Assay for determination of Κτ values for inhibition of O-GlcNAcase activity Experimental procedure for kinetic analyses
Enzymatic reactions are carried out in a reaction containing 50mM NaH2PC"4, 100 mM NaCl and 0.1% BSA (pH 7.0) using 2 mM 4-Methylumbelliferyl N-acetyl-β-ϋ- glucosaminide dihydrate (Sigma M2133) dissolved in ddH20, as a substrate. The amount of purified human O-GlcNAcase enzyme used in the reaction is 0.7 nM. Test compound of varying concentrations is added to the enzyme prior to initiation of the reaction. The reaction is performed at room temperature in a 96-well plate and is initiated with the addition of substrate. The production of fluorescent product is measured every 60 sec for 45 min with a Tecan Infinite M200 plate-reader with excitation at 355nM and emission detected at 460nM, with 4-Methylumbelliferone (Sigma M1381) used to produce a standard curve. The slope of product production is determined for each concentration of compound tested and plotted, using standard curve fitting algorithms for sigmoidal dose response curves. The values for a four parameter logistic curve fit of the data are determined.
Ki values are determined using the Cheng-Prusoff equation; the Km of O- GlcNacase for substrate is 0.2 mM.
Examples 1 to 148 were tested in the above described assay and exhibited Kj values for inhibition of O-GlcNAcase in the range 0.1 nM - 10 μΜ.
Assay for determination of Κτ values for inhibition of β-hexosaminidase activity Experimental procedure for kinetic analyses
Enzymatic reactions are carried out in a reaction containing 50mM NaH2PC"4, 100 mM NaCl and 0.1% BSA (pH 7.0) using 2 mM 4-Methylumbelliferyl N-acetyl-β-ϋ- glucosaminide dihydrate (Sigma M2133) dissolved in ddH20, as a substrate. The amount of purified human β hexosaminidase enzyme used in the reaction is 24 nM. Test compound of varying concentrations is added to the enzyme prior to initiation of the reaction. The reaction is performed at room temperature in a 96-well plate and is initiated with the addition of substrate. The production of fluorescent product is measured every 60 sec for 45 min with a Tecan Infinite M200 plate-reader with excitation at 355nM and emission detected at 460nM, with 4-Methylumbelliferone (Sigma M1381) used to produce a standard curve. The slope of product production is determined for each concentration of compound tested and plotted, using standard curve fitting algorithms for sigmoidal dose response curves. The values for a four parameter logistic curve fit of the data are determined.
Ki values are determined using the Cheng-Prusoff equation..
When tested in this assay, many of the compounds described herein exhibit ¾ values for inhibition of β-hexosaminidase in the range 10 nM to greater than 100 μΜ.
The selectivity ratio for inhibition of O-GlcNAcase over β-hexosaminidase is defined here as:
Ki ^-hexosaminidase)/Kl (O-GlcNAcase) In general, the compounds described herein exhibit a selectivity ratio range of about 10 to 100000. Thus, many compounds of the invention exhibit high selectivity for inhibition of O-GlcNAcase over β-hexosaminidase.
Assay for determination of cellular activity for compounds that inhibit O-GlcNAcase activity
Inhibition of O-GlcNAcase, which removes O-GlcNAc from cellular proteins, results in an increase in the level of O-GlcNAcylated protein in cells. An increase in O- GlcNAcylated protein can be measured by an antibody, such as RL-2, that binds to O- GlcNAcylated protein. The amount of O-GlcNAcylated protein:RL2 antibody interaction can be measured by enzyme linked immunosorbant assay (ELISA) procedures.
A variety of tissue culture cell lines, expressing endogenous levels of O- GlcNAcase, can be utilized; examples include rat PC-12, and human U-87, or SK-N-SH cells. Cells are plated in 96-well plates with approximately 10,000 cells / well. Compounds to be tested are dissolved in DMSO, either 2 or 10 mM stock solution, and then diluted with DMSO and water in a two-step process using a Tecan workstation. Cells are treated with diluted compounds for 24 hours (5.4 μΐ. into 200 μΐ. 1 well volume) to reach a final concentration of inhibitor desired to measure a compound concentration dependent response, typically, ten 3 fold dilution steps, starting at 10 μΜ are used to determine a concentration response curve. To prepare a cell lysate, the media from compound treated cells is removed, the cells are washed once with phosphate buffered saline (PBS) and then lysed for 5 minutes at room temperature in 50 μΐ. of Phosphosafe reagent (Novagen Inc, Madison, WI) with protease inhibitors and PMSF. The cell lysate is collected and transferred to a new plate, which is then either coated to assay plates directly or frozen -80°C until used in the ELISA procedure. If desired, the total protein concentration of samples is determined using 20 μΐ, of the sample using the BCA method.
The ELISA portion of the assay is performed in a black Maxisorp 96-well plate that is coated overnight at 4°C with 100 μΐ, /well of the cell lysate (1 : 10 dilution of the lysate with PBS containing protease inhibitors, phosphatase inhibitors, and PMSF. The following day the wells are washed 3 times with 300 μΐ, /well of Wash buffer (Tris-buffered saline with 0.1% Tween 20). The wells are blocked with 100 μΐ, /well Blocking buffer (Tris buffered saline w/0.05% Tween 20 and 2.5% Bovine serum albumin). Each well is then washed two times with 300 ul/well of wash buffer. The anti O-GlcNAc antibody RL-2 (Abeam, Cambridge, MA), diluted 1 : 1000 in blocking buffer, is added at 100 ul/well. The plate is sealed and incubated at 37°C for 2 hr with gentle shaking. The wells are then washed 3 -times with 300 ul/well wash buffer. To detect the amount of RL-2 bound horse-radish peroxidase (HRP) conjugated goat anti-mouse secondary antibody (diluted 1 :3000 in blocking buffer) is added at 100 /well. The plate is incubated for 60 min at 37°C with gentle shaking. Each wells is then washed 3 -times with 300 ul/well wash buffer. The detection reagent is added, 100 μΐ. /well of Amplex Ultra RED reagent (prepared by adding 30 of 10 mM Amplex Ultra Red stock solution to 10 ml PBS with 18 μΐ. 3% hydrogen peroxide, H2O2). The detection reaction is incubated for 15 minutes at room temperature and then read with excitation at 530 nm and emission at 590 nm.
The amount of O-GlcNAcylated protein, as detected by the ELISA assay, is plotted for each concentration of test compound using standard using standard curve fitting algorithms for sigmoidal dose response curves. The values for a four parameter logistic curve fit of the data are determined, with the inflection point of the curve being the potency value for the test compound.
Representative data from the binding and cell-based assays described above are shown in the following table. Certain compounds of the invention exhibited superior potency in one or more of these assays as compared to compounds disclosed in WO
2006/092049 and WO 2008/025170.
Figure imgf000115_0001
Fluorescence-based
Cell-based ELISA
hOGA
Example # EC50 (nM)
Ki (nM)
4 ND 3.1
8 15.1 0.68
11 0.8 0.3
13 ND 0.4
25 9.01 0.70
27 3.23 0.12
40 139.00 7.95
42 8.75 1 .03
47 187.20 6.40
51 22.22 5.1 Fluorescence-based
Cell-based ELISA
hOGA
Example # EC50 (nM)
Ki (nM)
53 9.26 0.7
57 5.90 0.33
61 25.31 2.46
63 2.57 0.22
65 2.7 0.45
67 17.13 1 .25
69 134.10 9.36
71 48.83 9.12
76 3.63 0.18
77 195.80 16.28 Fluorescence-based
Cell-based ELISA
hOGA
Example # EC50 (nM)
Ki (nM)
80 5.31 0.5
82 97.17 5.84
91 11.85 0.53
95 1.05 0.16
96 4.55 0.19
97 2.79 0.17
98 15.28 1.43
104 17.50 3.63
105 14.74 3.4 REFERENCES
1. C. R. Torres, G. W. Hart, J Biol Chem 1984, 259, 3308.
2. R. S. Haltiwanger, G. D. Holt, G. W. Hart, J Biol Chem 1990, 265, 2563. 3. L. K. Kreppel, M. A. Blomberg, G. W. Hart, J Biol Chem 1997, 272, 9308.
4. W. A. Lubas, D. W. Frank, M. Krause, J. A. Hanover, J Biol Chem 1997, 272, 9316.
5. W. A. Lubas, J. A. Hanover, J Biol Chem 2000, 275, 10983.
6. D. L. Dong, G. W. Hart, J Biol Chem 1994, 269, 19321.
7. Y. Gao, L. Wells, F. I. Comer, G. J. Parker, G. W. Hart, J Biol Chem 2001, 276, 9838.
8. E. P. Roquemore, M. R. Chevrier, R. J. Cotter, G. W. Hart, Biochemistry 1996, 35, 3578.
9. S. P. Jackson, R. Tjian, Cell 1988, 55, 125.
10. W. G. Kelly, M. E. Dahmus, G. W. Hart, J Biol Chem 1993, 268, 10416.
11. M. D. Roos, K. Su, J. R. Baker, J. E. Kudlow, Mol Cell Biol 1997, 17, 6472.
12. N. Lamarre- Vincent, L. C. Hsieh- Wilson, J Am Chem Soc 2003, 125, 6612.
13. F. Zhang, K. Su, X. Yang, D. B. Bowe, A. J. Paterson, J. E. Kudlow, Cell 2003, 775, 715.
14. K. Vosseller, L. Wells, M. D. Lane, G. W. Hart, Proc Natl Acad Sci USA 2002, 99, 5313.
15. W. A. Lubas, M. Smith, C. M. Starr, J. A. Hanover, Biochemistry 1995, 34, 1686.
16. L. S. Griffith, B. Schmitz, Biochem Biophys Res Commun 1995, 213, 424. 17. R. N. Cole, G. W. Hart, J Neurochem 1999, 73, 418.
18. I. Braidman, M. Carroll, N. Dance, D. Robinson, Biochem J 1974, 143, 295.
19. R. Ueno, C. S. Yuan, Biochim Biophys Acta 1991, 1074, 79.
20. C. Toleman, A. J. Paterson, T. R. Whisenhunt, J. E. Kudlow, J Biol Chem 2004
21. F. Liu, K. Iqbal, I. Grundke-Iqbal, G. W. Hart, C. X. Gong, Proc Natl Acad Sci USA 2004, 707, 10804.
22. T. Y. Chou, G. W. Hart, Adv Exp Med Biol 2001, 491, 413. 23. M. Goedert, M. G. Spillantini, N. J. Cairns, R. A. Crowther, Neuron 1992, 8, 159.
24. M. Goedert, M. G. Spillantini, R. Jakes, D. Rutherford, R. A. Crowther, Neuron 1989, 3, 519.
25. E. Kopke, Y. C. Tung, S. Shaikh, A. C. Alonso, K. Iqbal, I. Grundke-Iqbal, J
Biol Chem 1993, 268, 24374.
26. H. Ksiezak-Reding, W. K. Liu, S. H. Yen, Brain Res 1992, 597, 209.
27. B. Henrissat, A. Bairoch, Biochem J 1996, 316 ( Pt 2), 695.
28. B. Henrissat, A. Bairoch, Biochem J 1993, 293 (Pt 3), 781.
29. C. X. Gong, F. Liu, I. Grundke-Iqbal, K. Iqbal, J Neural Transm 2005, 112,
813.
30. K. Iqbal, C. Alonso Adel, E. El-Akkad, C. X. Gong, N. Haque, S. Khatoon, I. Tsujio, I. Grundke-Iqbal, J Neural Transm Suppl 2002, 309.
31. K. Iqbal, C. Alonso Adel, E. El-Akkad, C. X. Gong, N. Haque, S. Khatoon, J. J. Pei, H. Tanimukai, I. Tsujio, et al, JMolNeurosci 2003, 20, 425.
32. W. Noble, E. Planel, C. Zehr, V. Olm, J. Meyerson, F. Suleman, K. Gaynor, L. Wang, J. LaFrancois, et al, Proc Natl Acad Sci USA 2005, 102, 6990.
33. S. Le Corre, H. W. Klafki, N. Plesnila, G. Hubinger, A. Obermeier, H.
Sahagun, B. Monse, P. Seneci, J. Lewis, et al, Proc Natl Acad Sci USA 2006, 103, 9673. 34. S. J. Liu, J. Y. Zhang, H. L. Li, Z. Y. Fang, Q. Wang, H. M. Deng, C. X.
Gong, I. Grundke-Iqbal, K. Iqbal, et al, J Biol Chem 2004, 279, 50078.
35. G. Li, H. Yin, J. Kuret, J Biol Chem 2004, 279, 15938.
36. T. Y. Chou, G. W. Hart, C. V. Dang, J Biol Chem 1995, 270, 18961.
37. X. Cheng, G. W. Hart, J Biol Chem 2001, 276, 10570.
38. X. Cheng, R. N. Cole, J. Zaia, G. W. Hart, Biochemistry 2000, 39, 11609.
39. L. S. Griffith, B. Schmitz, Eur J Biochem 1999, 262, 824.
40. K. Kamemura, G. W. Hart, Prog Nucleic Acid Res Mol Biol 2003, 73, 107.
41. L. Wells, L. K. Kreppel, F. I. Comer, B. E. Wadzinski, G. W. Hart, J Biol Chem 2004, 279, 38466.
42. L. Bertram, D. Blacker, K. Mullin, D. Keeney, J. Jones, S. Basu, S. Yhu, M.
G. Mclnnis, R. C. Go, et al, Science 2000, 290, 2302. 43. S. Hoyer, D. Blum-Degen, H. G. Bernstein, S. Engelsberger, J. Humrich, S. Laufer, D. Muschner, A. Thalheimer, A. Turk, et al, Journal of Neural Transmission 1998, 105, 423.
44. C. X. Gong, F. Liu, I. Grundke-Iqbal, K. Iqbal, Journal of Alzheimer s Disease 2006, 9, 1.
45. W. J. Jagust, J. P. Seab, R. H. Huesman, P. E. Valk, C. A. Mathis, B. R. Reed, P. G. Coxson, T. F. Budinger, Journal of Cerebral Blood Flow and Metabolism 1991, 77, 323.
46. S. Hoyer, Experimental Gerontology 2000, 35, 1363.
47. S. Hoyer, in Frontiers in Clinical Neuroscience: Neurodegeneration and
Neuroprotection, Vol. 541, 2004, pp. 135.
48. R. N. Kalaria, S. I. Harik, Journal of Neurochemistry 1989, 53, 1083.
49. I. A. Simpson, K. R. Chundu, T. Davieshill, W. G. Honer, P. Davies, Annals of Neurology 1994, 35, 546.
50. S. M. de la Monte, J. R. Wands, Journal of Alzheimer s Disease 2005, 7, 45.
51. X. W. Zhu, G. Perry, M. A. Smith, Journal of Alzheimer s Disease 2005, 7, 81.
52. J. C. de la Torre, Neurological Research 2004, 26, 517.
53. S. Marshall, W. T. Garvey, R. R. Traxinger, Faseb J 1991, 5, 3031.
54. S. P. Iyer, Y. Akimoto, G. W. Hart, J Biol Chem 2003, 278, 5399.
55. K. Brickley, M. J. Smith, M. Beck, F. A. Stephenson, J Biol Chem 2005, 280, 14723.
56. S. Knapp, C. H. Yang, T. Haimowitz, Tetrahedron Letters 2002, 43, 7101.
57. S. P. Iyer, G. W. Hart, J Biol Chem 2003, 278, 24608.
58. M. Jinek, J. Rehwinkel, B. D. Lazarus, E. Izaurralde, J. A. Hanover, E. Conti,
Nat Struct Mol Biol 2004, 77, 1001.
59. K. Kamemura, B. K. Hayes, F. I. Comer, G. W. Hart, J Biol Chem 2002, 277, 19229.
60. Y. Deng, B. Li, F. Liu, K. Iqbal, I. Grundke-Iqbal, R. Brandt, C.-X. Gong, FASEB J 2007, fj.07.
61. L. F. Lau, J. B. Schachter, P. A. Seymour, M. A. Sanner, Curr Top Med Chem 2002, 2, 395.
62. M. P. Mazanetz, P. M. Fischer, Nature Reviews Drug Discovery 2007, 6, 464. 63. S. A. Yuzwa, M. S. Macauley, J. E. Heinonen, X. Shan, R. J. Dennis, Y. He, G. E. Whitworth, K. A. Stubbs, E. J. McEachern, et al, Nat Chem Biol 2008, 4, 483.
64. P. Bounelis, J. Liu, Y. Pang, J. C. Chatham, R. B. Marchase, Shock 2004, 21 nO Suppl. 2, 58.
65. N. Fulop, V. Champattanachai, R. B. Marchase, J. C. Chatham, Circulation
Research 2005, 97, E28.
66. J. Liu, R. B. Marchase, J. C. Chatham, Faseb Journal 2006, 20, A317.
67. R. Marchase, P. Bounelis, J. Chatham, I. Chaudry, Y. Pang, PCTInt. Appl. WO 2006016904 2006.
68. N. Fulop, P. P. Wang, R. B. Marchase, J. C. Chatham, Journal of Molecular and Cellular Cardiology 2004, 37, 286.
69. N. Fulop, P. P. Wang, R. B. Marchase, J. C. Chatham, Faseb Journal 2005, 19, A689.
70. J. Liu, R. B. Marchase, J. C. Chatham, Journal of Molecular and Cellular Cardiology 2007, 42, 111.
71. L. G. Not, C. A. Brocks, N. Fulop, R. B. Marchase, J. C. Chatham, Faseb Journal 2006, 20, A1471.
72. S. L. Yang, L. Y. Zou, P. Bounelis, I. Chaudry, J. C. Chatham, R. B.
Marchase, Shock 2006, 25, 600.
73. L. Y. Zou, S. L. Yang, P. Bounelis, I. H. Chaudry, J. C. Chatham, R. B.
Marchase, Faseb Journal 2005, 19, A1224.
74. R. B. Marchase, J. Liu, L. Y. Zou, V. Champattanachai, Y. Pang, N. Fulop, P. P. Wang, S. L. Yang, P. Bounelis, et al, Circulation 2004, 110, 1099.
75. J. Liu, Y. Pang, T. Chang, P. Bounelis, J. C. Chatham, R. B. Marchase, Journal of Molecular and Cellular Cardiology 2006, 40, 303.
76. J. Liu, J. C. Chatham, R. B. Marchase, Faseb Journal 2005, 19, A691.
77. T. Nagy, V. Champattanachai, R. B. Marchase, J. C. Chatham, American Journal of Physiology-Cell Physiology 2006, 290, C57.
78. N. Fulop, R. B. Marchase, J. C. Chatham, Cardiovascular Research 2007, 73, 288.
79. T. Lefebvre, C. Guinez, V. Dehennaut, O. Beseme-Dekeyser, W. Morelle, J. C. Michalski, Expert Review of Proteomics 2005, 2, 265.
80. L. Wells, K. Vosseller, G. W. Hart, Science 2001, 291, 2376. 81. J. A. Hanover, FASEB J 2001, 15, 1865.
82. D. A. McClain, W. A. Lubas, R. C. Cooksey, M. Hazel, G. J. Parker, D. C. Love, J. A. Hanover, Proc Natl Acad Sci USA 2002, 99, 10695.
83. P. J. Yao, P. D. Coleman, JNeurosci 1998, 18, 2399.
84. W. H. Yang, J. E. Kim, H. W. Nam, J. W. Ju, H. S. Kim, Y. S. Kim, J. W.
Cho, Nature Cell Biology 2006, 8, 1074.
85. B. Triggs-Raine, D. J. Mahuran, R. A. Gravel, Adv Genet 2001, 44, 199.
86. D. Zhou, J. Mattner, C. Cantu Iii, N. Schrantz, N. Yin, Y. Gao, Y. Sagiv, K. Hudspeth, Y. Wu, et al, Science 2004.
87. G. Legler, E. Lullau, E. Kappes, F. Kastenholz, Biochim Biophys Acta 1991,
1080, 89.
88. M. Horsch, L. Hoesch, A. Vasella, D. M. Rast, Eur J Biochem 1991, 197, 815.
89. J. Liu, A. R. Shikhman, M. K. Lotz, C. H. Wong, Chem Biol 2001, 8, 701. 90. S. Knapp, D. J. Vocadlo, Z. N. Gao, B. Kirk, J. P. Lou, S. G. Withers, J. Am.
Chem. Soc. 1996, 118, 6804.
91. V. H. Lillelund, H. H. Jensen, X. Liang, M. Bols, Chem Rev 2002, 102, 515.
92. R. J. Konrad, I. Mikolaenko, J. F. Tolar, K. Liu, J. E. Kudlow, Biochem J 2001, 356, 31.
93. K. Liu, A. J. Paterson, F. Zhang, J. McAndrew, K. Fukuchi, J. M. Wyss, L.
Peng, Y. Hu, J. E. Kudlow, J Neurochem 2004, 89, 1044.
94. G. Parker, R. Taylor, D. Jones, D. McClain, J Biol Chem 2004, 279, 20636.
95. E. B. Arias, J. Kim, G. D. Cartee, Diabetes 2004, 53, 921.
96. A. Junod, A. E. Lambert, L. Orci, R. Pictet, A. E. Gonet, A. E. Renold, Proc Soc Exp Biol Med 1967, 126, 201.
97. R. A. Bennett, A. E. Pegg, Cancer Res 1981, 41, 2786.
98. K. D. Kroncke, K. Fehsel, A. Sommer, M. L. Rodriguez, V. Kolb-Bachofen, Biol Chem Hoppe Seyler 1995, 376, 179.
99. H. Yamamoto, Y. Uchigata, H. Okamoto, Nature 1981, 294, 284.
100. K. Yamada, K. Nonaka, T. Hanafusa, A. Miyazaki, H. Toyoshima, S. Tarui,
Diabetes 1982, 31, 749.
101. V. Burkart, Z. Q. Wang, J. Radons, B. Heller, Z. Herceg, L. Stingl, E. F.
Wagner, H. Kolb, Nat Med 1999, 5, 314. 102. M. D. Roos, W. Xie, K. Su, J. A. Clark, X. Yang, E. Chin, A. J. Paterson, J. E. Kudlow, Proc Assoc Am Physicians 1998, 110, 422.
103. Y. Gao, G. J. Parker, G. W. Hart, Arch Biochem Biophys 2000, 383, 296.
104. R. Okuyama, M. Yachi, Biochem Biophys Res Commun 2001, 287, 366. 105. N. E. Zachara, N. O'Donnell, W. D. Cheung, J. J. Mercer, J. D. Marth, G. W.
Hart, J Biol Chem 2004, 279, 30133.
106. J. A. Hanover, Z. Lai, G. Lee, W. A. Lubas, S. M. Sato, Arch Biochem Biophys 1999, 362, 38.
107. K. Liu, A. J. Paterson, R. J. Konrad, A. F. Parlow, S. Jimi, M. Roh, E. Chin, Jr., J. E. Kudlow, Mol Cell Endocrinol 2002, 194, 135.
108. M. S. Macauley, G. E. Whitworth, A. W. Debowski, D. Chin, D. J. Vocadlo, J Biol Chem 2005, 280, 25313.
109. B. L. Mark, D. J. Vocadlo, S. Knapp, B. L. Triggs-Raine, S. G. Withers, M. N. James, J Biol Chem 2001, 276, 10330.
110. R. S. Haltiwanger, K. Grove, G. A. Philipsberg, J Biol Chem 1998, 273, 3611.
111. D. J. Miller, X. Gong, B. D. Shur, Development 1993, 118, 1279.
112. L. Y. Zou, S. L. Yang, S. H. Hu, I. H. Chaudry, R. B. Marchase, J. C.
Chatham, Shock 2007, 27, 402.
113. J. B. Huang, A. J. Clark, H. R. Petty, Cellular Immunology 2007, 245, 1. 114. U. J. G. Conference, in US/Japan Glyco 2004 Conference, Honolulu, Hawaii,
2004
115. L. Y. Zou, S. L. Yang, S. H. Hu, I. H. Chaudry, R. B. Marchase, J. C.
Chatham, Faseb Journal 2006, 20, A1471.
116. V. Champattanachai, R. B. Marchase, J. C. Chatham, American Journal of Physiology-Cell Physiology 2007, 292, C 178.
117. V. Champattanachai, R. B. Marchase, J. C. Chatham, American Journal of Physiology-Cell Physiology 2008, 294, CI 509.
118. I. Khlistunova, M. Pickhardt, J. Biernat, Y. P. Wang, E. M. Mandelkow, E. Mandelkow, Current Alzheimer Research 2007, 4, 544.
119. P. Friedhoff, A. Schneider, E. M. Mandelkow, E. Mandelkow, Biochemistry
1998, 37, 10223. 120. M. Pickhardt, Z. Gazova, M. von Bergen, I. Khlistunova, Y. P. Wang, A. Hascher, E. M. Mandelkow, J. Biernat, E. Mandelkow, Journal of Biological Chemistry 2005, 280, 3628.
121. P. H. Liang, W. C. Cheng, Y. L. Lee, H. P. Yu, Y. T. Wu, Y. L. Lin, C. H. Wong, Chembiochem 2006, 7, 165.
122. J. J. Liu, M. M. D. Numa, H. T. Liu, S. J. Huang, P. Sears, A. R. Shikhman, C. H. Wong, Journal of Organic Chemistry 2004, 69, 6273.
123. Y. Takaoka, T. Kajimoto, C. H. Wong, Journal of Organic Chemistry 1993, 58, 4809.
124. T. M. Wrodnigg, A. E. Stutz, S. G. Withers, Tetrahedron Letters 1997, 38,
5463.

Claims

CLAIMS:
1. A compound of Formula (I) or a pharmaceutically acceptable salt thereof:
Figure imgf000126_0001
(I)
wherein:
each R is independently H or C(0)CH3;
R1 and R2 are independently selected from the group consisting of: H, Ci-6alkyl, Ci-
6alkoxy, -(CH2)n-cyclopropyl and -(CH2)n-cyclobutyl wherein n is 0, 1, 2, 3 or 4; or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine, pyrrolidine or piperidine, said Ci-6alkyl, Ci-6alkoxy, -(CH2)n-cyclopropyl,
-(CH2)n-cyclobutyl, azetidine, pyrrolidine or piperidine optionally substituted from one up to the maximum number of substituents with fluoro or methyl;
R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2-8alkynyl, C3- 6cycloalkyl, aryl and heteroaryl, each optionally substituted from one up to the maximum number of substituents with fluoro and OH;
R4 is selected from the group consisting of: H, F, Ci-8alkyl, C2-8alkenyl and C2-8alkynyl, each excluding hydrogen optionally substituted from one up to the maximum number of substituents with fluoro and OH; or R3 and R4 and the carbon atom to which they are attached may join together to form vinyl or a 3 to 7-membered carbocyclic or heterocyclic ring, said 3 to 7-membered carbocyclic or heterocyclic ring optionally containing a double bond and optionally substituted from one up to the maximum number of substituents with fluoro and OH; and
R5 is selected from H, F and OH;
with the proviso that when R4 is F then R5 is other than OH.
2. The compound according to Claim 1 of Formula (la) or a pharmaceutically acceptable salt thereof:
Figure imgf000127_0001
(la).
3. The compound according to claim 2 wherein:
R1 and R2 are independently Ci-4alkyl;
R3 is Ci-6alkyl;
selected from the group consisting of: H and Ci-6alkyl; and
R5 is OH.
4. The compound according to Claim 3 wherein:
R1 and R2 are independently methyl or ethyl;
R3 is methyl or ethyl; and
R4 is selected from the group consisting of: H, methyl and ethyl.
5. The compound according to Claim 2 wherein R3 and R4 and the carbon atom to which they are attached may join together to form a 3 to 7-membered carbocyclic or heterocyclic ring, said 3 to 7-membered carbocyclic or heterocyclic ring optionally containing a double bond and optionally substituted from one up to the maximum number of substituents with fluoro and OH.
6. The compound according to Claim 2, wherein:
R1 and R2 are independently selected from the group consisting of: H, Ci-6alkyl and cyclopropylmethyl; or Rl and R2 may be joined together with the nitrogen atom to which they are attached to form azetidine or pyrrolidine, said Ci-6alkyl, cyclopropylmethyl, azetidine or pyrrolidine optionally substituted with 1 to 3 substituents selected from fluoro and methyl;
R3 is selected from the group consisting of: Ci-8alkyl, C2-8alkenyl, C2-8alkynyl and C3- 6cycloalkyl, each optionally substituted with 1 to 3 substituents selected from fluoro and OH; and R4 is selected from the group consisting of: H, F, Ci-8alkyl, C2-8alkenyl and C2-8alkynyl, each excluding hydrogen optionally substituted with 1 to 3 substituents selected from fluoro and OH; or R3 and R4 and the carbon atom to which they are attached may join together to form a 5-membered carbocyclic ring optionally containing a double bond and optionally substituted with 1 to 3 substituents selected from fluoro and OH.
7. The compound according to Claim 2 wherein R3 is CF3 and R5 is OH.
8. A compound selected from the following group:
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,6S,7R,7aR)-5-((S)-l-hydroxyethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-5-(2-Hydroxypropan-2-yl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol; (3aR,5S,6S,7R,7aR)-2-(dimethylamino)-5-(2-hydroxypropan-2-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-(2-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(Dimethylamino)-5-ethyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6, 7-diol; (3 aR, 5R, 6 S, 7R, 7aR)-2-(dimethylamino)-5 -vinyl-5 , 6, 7, 7a-tetrahydro-3 aH-pyrano [3 ,2- d]thiazole-6, 7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -Ethyl-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH-pyrano [3 ,2- d]thiazole-6, 7-diol
(3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-5-((S)-l-fluoroethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(dimethylamino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(dimethylamino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5 S, 6 S, 7R, 7aR)-5 -( 1 , 1 -Difluoroethyl)-2-(dimethylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -((S)- 1 -hydroxyethyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxyethyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxypropyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxypropyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol; (3 aR, 5R, 6 S, 7R, 7aR)-5 -( 1 -hydroxy-2-methylpropyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -(cyclopropyl(hydroxy)methyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -(hydroxy(pyridin-3 -yl)methyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -(hydroxy(phenyl)methyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxyallyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -((S)- 1 -hydroxyallyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxy-2-methylallyl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -((R)- 1 -hydroxybut-3 -enyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxybut-3-enyl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((R)-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethylamino)-5 -((S)- 1 -hydroxyethyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol; (3aR,5R,6S,7R,7aR)-5-((S)-cyclopropyl(hydroxy)methyl)-2-(ethylamino)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethylamino)-5 -( 1 -hydroxy-2-methylpropyl)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethylamino)-5 -(hydroxy(phenyl)methyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((R)-l-hydroxyallyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((S)-l-hydroxyallyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-(2-hydroxypropan-2-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((R)-l-hydroxy-2-methylallyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((R)-l-hydroxybutyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethylamino)-5 -((S)- 1 -hydroxybutyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((R)-l-hydroxypentyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((S)-l-hydroxypentyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol; (3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((R)-l-hydroxybut-3-enyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-((S)-l-hydroxybut-3-enyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-5-(2-hydroxypropan-2-yl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxyallyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxybutyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxybutyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxypentyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxypentyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-(l-hydroxypropyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxybut-3-enyl)-2-(propylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxybut-3-enyl)-2-(propylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol; (3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxyethyl)-2-(methoxy(methyl)amino)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -((S)- 1 -hydroxyethyl)-2-(methoxy(methyl)amino)-5 , 6, 7, 7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-((R)-l-hydroxyethyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-((S)-l-hydroxyethyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethyl(methyl)amino)-5-((S)-l-hydroxybut-3-enyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethyl(methyl)amino)-5-((R)-l-hydroxybut-3-enyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5 S, 6 S, 7R, 7aR)-2-(ethyl(methyl)amino)-5 -(2-hydroxypropan-2-yl)-5 , 6, 7, 7a-tetrahydro 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethyl(methyl)amino)-5 -( 1 -hydroxyethyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethyl(methyl)amino)-5-((R)-l-hydroxypropyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethyl(methyl)amino)-5 -( 1 -hydroxypropyl)-5 , 6, 7, 7a-tetrahydro-3 aH pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((R)-l-hydroxyallyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol; (3 aR, 5R, 6 S, 7R, 7aR)-2-(dimethylamino)-5 -((S)- 1 -hydroxyallyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((S)-l-hydroxypropyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(dimethylamino)-5 -((S)- 1 -hydroxyallyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((R)-l-hydroxy-2-methylallyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((S)-l-hydroxy-2-methylallyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(dimethylamino)-5 -((R)- 1 -hydroxybut-3 -enyl)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((S)-l-hydroxybut-3-enyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((R)-l-hydroxybutyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((S)-l-hydroxybutyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((R)-l-hydroxypentyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((S)-l-hydroxypentyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol; (3 aR, 5R, 6 S, 7R, 7aR)-2-(dimethylamino)-5 -((S)- 1 -hydroxyhexyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((R)-l-hydroxyheptyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(dimethylamino)-5 -((S)- 1 -hydroxyheptyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-((R)-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-((S)-l-hydroxyethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(azetidin-l-yl)-5-(2-hydroxypropan-2-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-((S)-l-hydroxypropyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-((R)-l-hydroxyallyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-((S)-l-hydroxyallyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-((R)-l-hydroxybut-3-enyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-((S)-l-hydroxybut-3-enyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol; (3aR,5S,6S,7R,7aR)-5-(2-hydroxypropan-2-yl)-2-(methyl(propyl)amino)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-(l-hydroxypropyl)-2-(methyl(propyl)amino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -((S)- 1 -hydroxyethyl)-2-(methyl(propyl)amino)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxyallyl)-2-(methyl(propyl)amino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxyallyl)-2-(methyl(propyl)amino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxybut-3-enyl)-2-(methyl(propyl)amino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxybut-3-enyl)-2-(methyl(propyl)amino)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-5-(2-hydroxypropan-2-yl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxyethyl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxypropyl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxypropyl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol; (3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxyallyl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5 S, 6 S, 7R, 7aR)-5 -( 1 -hydroxycyclopentyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-5-(l-hydroxycyclopent-3-enyl)-2-(methylamino)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(methylamino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(ethylamino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(propylamino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(methoxy(methyl)amino)-5-((R)-2,2,2-trifluoro-l-hydroxyethyl)- 5 , 6, 7, 7a-tetrahydro-3 aH-pyrano [3 ,2-d]thiazole-6, 7-diol;
(3aR,5S,6S,7R,7aR)-2-(methylamino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(ethylamino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(propylamino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(methoxy(methyl)amino)-5-((S)-2,2,2-trifluoro-l-hydroxyethyl)- 5 , 6, 7, 7a-tetrahydro-3 aH-pyrano [3 ,2-d]thiazole-6, 7-diol; (3aR,5S,6S,7R,7aR)-2-(ethylamino)-5-((S)-l-fluoroethyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-5-(2-fluoropropan-2-yl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR) -2- (methylamino)-5-(prop-l-en-2-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(2-Fluoroethylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(3-Fluoropropylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-5-((S)-l-fluoroethyl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxybutyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -((S)- 1 -hydroxybutyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxypentyl)-2-(methylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -((S)- 1 -hydroxypentyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxyhexyl)-2-(methylamino)-5, 6,7,7a- tetrahydropyrano[3,2-d]thiazole-6,7-diol; (3 aR, 5R, 6 S, 7R, 7aR)-5 -((S)- 1 -hydroxyhexyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((R)-l-hydroxyethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-((S)-l-hydroxy-2-methylallyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-((R)-l-hydroxy-2-methylallyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-((S)-l-hydroxypropyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-((R)-l-hydroxybut-3-enyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-(l-hydroxybut-3-enyl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(cyclopropylmethylamino)-5-((S)-l-hydroxyallyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(2-cyclopropylethylamino)-5-((R)-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(2-cyclopropylethylamino)-5-((S)-l-hydroxyethyl)-5,6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(2-cyclopropylethylamino)-5-(2-hydroxypropan-2-yl)-5, 6,7,7a- tetrahydro-3aH-pyrano[3,2-d]thiazole-6,7-diol; (3aR,5R,6S,7R,7aR)-2-(ethyl(methyl)amino)-5-((R)-l-hydroxyethyl)-5,6,7,7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethyl(methyl)amino)-5 -((S)- 1 -hydroxybutyl)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(ethyl(methyl)amino)-5 -((S)- 1 -hydroxypentyl)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(dimethylamino)-5-((R)-l-hydroxyhexyl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxybutyl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5R,6S,7R,7aR)-5-((S)-l-hydroxypentyl)-2-(pyrrolidin-l-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5 S, 6 S, 7R, 7aR)-5 -( 1 , 1 -difluoroethyl)-2-(methoxy(methyl)amino)-5 , 6, 7, 7a-tetrahydro- 3aH-pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -ethyl-2-(ethyl(methyl)amino)-5 , 6, 7, 7a-tetrahydro-3 aH-pyrano [3 ,2- d]thiazole-6, 7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-ethyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6, 7-diol;
(3aR,5R,6S,7R,7aR)-2-(azetidin-l-yl)-5-vinyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6, 7-diol;
(3aR,5R,6S,7R,7aR)-5-ethyl-2-(propylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2- d]thiazole-6, 7-diol; (3aR,5R,6S,7R,7aR)-5-ethyl-2-(ethylamino)-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole- 6,7-diol;
(3aR,5R,6S,7R,7aR)-2-(ethylamino)-5-vinyl-5,6,7,7a-tetrahydro-3aH-pyrano[3,2-d]thiazole- 6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -(3 -Hydroxypropyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-2-(Ethylamino)-5 -(2-hydroxyethyl)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3 aR, 5R, 6 S, 7R, 7aR)-5 -(2-hydroxyethyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-2-(Dimethylamino)-5-(2-fluoropropan-2-yl)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol;
(3aR,5S,6S,7R,7aR)-5-(l, l-Difluoroethyl)-2-(propylamino)-5,6,7,7a-tetrahydro-3aH- pyrano[3,2-d]thiazole-6,7-diol; and
(3 aR, 5 S, 6 S, 7R, 7aR)-5 -( 1 , 1 -difluoroethyl)-2-(methylamino)-5 , 6, 7, 7a-tetrahydro-3 aH- pyrano[3,2-d]thiazole-6,7-diol; or a pharmaceutically acceptable salt of any of the foregoing compounds.
9. A pharmaceutical composition comprising the compound of claim 1 or a pharmaceutically acceptable salt thereof in combination with a pharmaceutically acceptable carrier.
10. A method of selectively inhibiting O-GlcNAcase in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof.
11. A method of elevating the level of O-GlcNAc in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof.
12. A method of treating a condition that is modulated by O-GlcNAcase, in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof.
13. The method of claim 12 wherein the condition is selected from one or more of the group consisting of an inflammatory disease, an allergy, asthma, allergic rhinitis, hypersensitivity lung diseases, hypersensitivity pneumonitis, eosinophilic pneumonias, delayed-type hypersensitivity, atherosclerosis, interstitial lung disease (ILD), idiopathic pulmonary fibrosis, ILD associated with rheumatoid arthritis, systemic lupus erythematosus, ankylosing spondylitis, systemic sclerosis, Sjogren's syndrome, polymyositis or
dermatomyositis, systemic anaphylaxis or hypersensitivity response, drug allergy, insect sting allergy, autoimmune disease, rheumatoid arthritis, psoriatic arthritis, multiple sclerosis, Guillain-Barre syndrome, systemic lupus erythematosus, myastenia gravis,
glomerulonephritis, autoimmune thyroiditis, graft rejection, allograft rejection, graft-versus- host disease, inflammatory bowel disease, Crohn's disease, ulcerative colitis,
spondyloarthropathy, scleroderma, psoriasis, T-cell mediated psoriasis, inflammatory dermatosis, dermatitis, eczema, atopic dermatitis, allergic contact dermatitis, urticaria, vasculitis, necrotizing, cutaneous, and hypersensitivity vasculitis, eosinphilic myotis, eosiniphilic fasciitis, solid organ transplant rejection, heart transplant rejection, lung transplant rejection, liver transplant rejection, kidney transplant rejection, pancreas transplant rejection, kidney allograft, lung allograft, epilepsy, pain, fibromyalgia, stroke,
neuroprotection.
14. A method of treating a condition selected from the group consisting of a neurodegenerative disease, a tauopathy, cancer and stress, in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of claim 1, or a pharmaceutically acceptable salt thereof.
15. The method of claim 14 wherein the condition is selected from one or more of the group consisting of Alzheimer's disease, Amyotrophic lateral sclerosis (ALS), Amyotrophic lateral sclerosis with cognitive impairment (ALSci), Argyrophilic grain dementia, Bluit disease, Corticobasal degeneration (CBD), Dementia pugilistica, Diffuse neurofibrillary tangles with calcification, Down's syndrome, Familial British dementia, Familial Danish dementia, Frontotemporal dementia with parkinsonism linked to
chromosome 17 (FTDP-17), Gerstmann-Straussler-Scheinker disease, Guadeloupean parkinsonism, Hallevorden-Spatz disease (neurodegeneration with brain iron accumulation type 1), Multiple system atrophy, Myotonic dystrophy, Niemann-Pick disease (type C), Pallido-ponto-nigral degeneration, Parkinsonism-dementia complex of Guam, Pick's disease (PiD), Post-encephalitic parkinsonism (PEP), Prion diseases (including Creutzfeldt- Jakob Disease (CJD), Variant Creutzfeldt- Jakob Disease (vCJD), Fatal Familial Insomnia, and Kuru), Progressive supercortical gliosis, Progressive supranuclear palsy (PSP), Richardson's syndrome, Subacute sclerosing panencephalitis, Tangle-only dementia, Huntington's disease, Parkinson's disease, Schizophrenia, Mild Cognitive Impairment (MCI), Neuropathy
(including peripheral neuropathy, autonomic neuropathy, neuritis, and diabetic neuropathy), or Glaucoma.
16. The method of claim 14 wherein the stress is a cardiac disorder.
17. The method of claim 16 wherein the cardiac disorder is selected from one or more of the group consisting of ischemia; hemorrhage; hypovolemic shock;
myocardial infarction; an interventional cardiology procedure; cardiac bypass surgery;
fibrinolytic therapy; angioplasty; and stent placement.
PCT/CN2010/078528 2010-11-08 2010-11-08 Selective glycosidase inhibitors and uses thereof WO2012061972A1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
PCT/CN2010/078528 WO2012061972A1 (en) 2010-11-08 2010-11-08 Selective glycosidase inhibitors and uses thereof
PCT/CN2011/080691 WO2012062157A1 (en) 2010-11-08 2011-10-12 Pyrano[3,2-d]thiazol derivatives and uses thereof as selective glycosidase inhibitors
TW100140566A TW201249848A (en) 2010-11-08 2011-11-07 Selective glycosidase inhibitors and uses thereof
RU2013126531/04A RU2592285C2 (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and use thereof
CA2816798A CA2816798C (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof
US13/883,811 US8901087B2 (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof
ES11840448.2T ES2600028T3 (en) 2010-11-08 2011-11-08 Selective glucosidase inhibitors and their uses
EP11840448.2A EP2637503B1 (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof
BR112013011441A BR112013011441B8 (en) 2010-11-08 2011-11-08 SELECTIVE GLYCOSIDASE INHIBITORS COMPOUNDS, PHARMACEUTICAL COMPOSITION, AND USE OF A COMPOUND
AU2011326186A AU2011326186A1 (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof
KR1020137014710A KR101922266B1 (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof
MX2013005118A MX336611B (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof.
JP2013537923A JP5965407B2 (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof
PCT/US2011/059668 WO2012064680A1 (en) 2010-11-08 2011-11-08 Selective glycosidase inhibitors and uses thereof
ARP110104166A AR083798A1 (en) 2010-11-08 2011-11-08 SELECTIVE INHIBITORS OF GLUCOSIDASES AND ITS USES
AU2016202186A AU2016202186B2 (en) 2010-11-08 2016-04-07 Selective glycosidase inhibitors and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2010/078528 WO2012061972A1 (en) 2010-11-08 2010-11-08 Selective glycosidase inhibitors and uses thereof

Publications (1)

Publication Number Publication Date
WO2012061972A1 true WO2012061972A1 (en) 2012-05-18

Family

ID=46050301

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2010/078528 WO2012061972A1 (en) 2010-11-08 2010-11-08 Selective glycosidase inhibitors and uses thereof
PCT/CN2011/080691 WO2012062157A1 (en) 2010-11-08 2011-10-12 Pyrano[3,2-d]thiazol derivatives and uses thereof as selective glycosidase inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/CN2011/080691 WO2012062157A1 (en) 2010-11-08 2011-10-12 Pyrano[3,2-d]thiazol derivatives and uses thereof as selective glycosidase inhibitors

Country Status (1)

Country Link
WO (2) WO2012061972A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012129651A1 (en) * 2011-03-31 2012-10-04 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
WO2013000084A1 (en) * 2011-06-27 2013-01-03 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
EP2655388A1 (en) * 2010-12-23 2013-10-30 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
WO2014105662A1 (en) * 2012-12-24 2014-07-03 Merck Sharp & Dohme Corp. Glycosidase inhibitors and uses thereof
US8962664B2 (en) 2006-08-31 2015-02-24 Simon Fraser University Selective glycosidase inhibitors and uses thereof
US20150152127A1 (en) * 2012-05-08 2015-06-04 Merck Sharp & Dohme Corp. Permeable glycosidase inhibitors and uses thereof
CN104837845A (en) * 2012-10-31 2015-08-12 阿勒克图治疗公司 Glycosidase inhibitors and uses thereof
US9199949B2 (en) 2011-06-27 2015-12-01 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
US9670195B2 (en) 2012-08-31 2017-06-06 Alectos Therapeutics Inc. Glycosidase inhibitors and uses thereof
US9701693B2 (en) 2011-06-27 2017-07-11 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
US9809537B2 (en) 2012-08-31 2017-11-07 Alectos Therapeutics Inc. Glycosidase inhibitors and uses thereof

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9120781B2 (en) 2010-05-11 2015-09-01 Simon Fraser University Selective glycosidase inhibitors and uses thereof
JP5893738B2 (en) 2011-08-25 2016-03-23 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Pyrano [3,2-D] [1,3] thiazole as a glycosidase inhibitor
CN107108601B (en) 2014-08-28 2021-08-20 阿森纽荣股份公司 Glycosidase inhibitors
CN104961778A (en) * 2015-07-22 2015-10-07 中国农业大学 Mannosyl thiazoline compounds, and preparation method and application thereof
EP3419976B1 (en) 2016-02-25 2021-04-07 Asceneuron SA Acid addition salts of piperazine derivatives
US10556902B2 (en) 2016-02-25 2020-02-11 Asceneuron Sa Glycosidase inhibitors
MA43677A (en) 2016-02-25 2018-11-28 Asceneuron Sa GLYCOSIDASE INHIBITORS
US11261183B2 (en) 2016-02-25 2022-03-01 Asceneuron Sa Sulfoximine glycosidase inhibitors
US11213525B2 (en) 2017-08-24 2022-01-04 Asceneuron Sa Linear glycosidase inhibitors
WO2020039029A1 (en) 2018-08-22 2020-02-27 Asceneuron S. A. Spiro compounds as glycosidase inhibitors
US11795165B2 (en) 2018-08-22 2023-10-24 Asceneuron Sa Tetrahydro-benzoazepine glycosidase inhibitors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101595111A (en) * 2006-08-31 2009-12-02 西蒙·弗雷瑟大学 Selective glycosidase inhibitors and uses thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2006220187B2 (en) * 2005-03-01 2012-11-08 Simon Fraser University Selective glycosidase inhibitors, methods of making inhibitors, and uses thereof
EP2322529B1 (en) * 2006-08-31 2017-12-06 Simon Fraser University Selective glycosidase inhibitors and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101595111A (en) * 2006-08-31 2009-12-02 西蒙·弗雷瑟大学 Selective glycosidase inhibitors and uses thereof

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8962664B2 (en) 2006-08-31 2015-02-24 Simon Fraser University Selective glycosidase inhibitors and uses thereof
US9243020B2 (en) 2010-12-23 2016-01-26 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
US9815861B2 (en) 2010-12-23 2017-11-14 Alectos Therapeutics, Inc. Selective glycosidase inhibitors and uses thereof
EP2655388A1 (en) * 2010-12-23 2013-10-30 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
EP2655388A4 (en) * 2010-12-23 2014-06-18 Alectos Therapeutics Inc Selective glycosidase inhibitors and uses thereof
US9718854B2 (en) 2011-03-31 2017-08-01 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
WO2012129651A1 (en) * 2011-03-31 2012-10-04 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
US9409924B2 (en) 2011-06-27 2016-08-09 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
US9701693B2 (en) 2011-06-27 2017-07-11 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
WO2013000084A1 (en) * 2011-06-27 2013-01-03 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
RU2625308C2 (en) * 2011-06-27 2017-07-13 Алектос Терапьютикс Инк. Selective glycosidase inhibitors and their application
AU2012276257B2 (en) * 2011-06-27 2017-05-18 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
US9199949B2 (en) 2011-06-27 2015-12-01 Alectos Therapeutics Inc. Selective glycosidase inhibitors and uses thereof
US20150152127A1 (en) * 2012-05-08 2015-06-04 Merck Sharp & Dohme Corp. Permeable glycosidase inhibitors and uses thereof
US9611275B2 (en) * 2012-05-08 2017-04-04 Alectos Therapeutics, Inc. Permeable glycosidase inhibitors and uses thereof
US9670195B2 (en) 2012-08-31 2017-06-06 Alectos Therapeutics Inc. Glycosidase inhibitors and uses thereof
US9809537B2 (en) 2012-08-31 2017-11-07 Alectos Therapeutics Inc. Glycosidase inhibitors and uses thereof
US9695197B2 (en) 2012-10-31 2017-07-04 Alectos Therapeutics Inc. Glycosidase inhibitors and uses thereof
CN104837845A (en) * 2012-10-31 2015-08-12 阿勒克图治疗公司 Glycosidase inhibitors and uses thereof
EP2935283A4 (en) * 2012-12-24 2016-10-05 Merck Sharp & Dohme Glycosidase inhibitors and uses thereof
WO2014105662A1 (en) * 2012-12-24 2014-07-03 Merck Sharp & Dohme Corp. Glycosidase inhibitors and uses thereof
US9938299B2 (en) 2012-12-24 2018-04-10 Alectos Therapeutics, Inc. Glycosidase inhibitors and uses thereof

Also Published As

Publication number Publication date
WO2012062157A1 (en) 2012-05-18

Similar Documents

Publication Publication Date Title
US9815861B2 (en) Selective glycosidase inhibitors and uses thereof
AU2016202186B2 (en) Selective glycosidase inhibitors and uses thereof
EP2569291B1 (en) Selective glycosidase inhibitors and uses thereof
WO2012061972A1 (en) Selective glycosidase inhibitors and uses thereof
EP2744338B1 (en) Selective glycosidase inhibitors and uses thereof
EP2691407B1 (en) Selective glycosidase inhibitors and uses thereof
US8927507B2 (en) Selective glycosidase inhibitors and uses thereof
AU2011349021A1 (en) Selective glycosidase inhibitors and uses thereof
WO2014032187A1 (en) Glycosidase inhibitors and uses thereof
EP2890679A1 (en) Glycosidase inhibitors and uses thereof
WO2014100934A1 (en) Glycosidase inhibitors and uses thereof
EP2890675A1 (en) Glycosidase inhibitors and uses thereof
CA2890115A1 (en) Glycosidase inhibitors and uses thereof
WO2012129802A1 (en) Pyrano[3,2-d]thiazol derivatives and uses as selective glycosidase inhibitors thereof
US8901087B2 (en) Selective glycosidase inhibitors and uses thereof
WO2012159262A1 (en) Selective glycosidase inhibitors and uses thereof

Legal Events

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

Ref document number: 10859458

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10859458

Country of ref document: EP

Kind code of ref document: A1