Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
N-[4-FLUORO-5-[[(2S,4S)-2-METHYL-4-[(5-METHYL-1,2,4-OXADIAZOL-3-YL)METHOXY]-1-PIPERIDYL]METHYL]THIAZOL-2-YL]ACETAMIDE AS OGA INHIBITOR
Document Type and Number:
WIPO Patent Application WO/2018/140299
Kind Code:
A1
Abstract:
The present invention provides a compound of Formula (I): or a pharmaceutically acceptable salt thereof, and the use of compounds of Formula (I) for treatment of neurodegenerative diseases and disorders, such as Alzheimer's disease.

Inventors:
DREYFUS NICOLAS JACQUES FRANCOIS (US)
LINDSAY-SCOTT PETER JAMES (US)
Application Number:
PCT/US2018/014331
Publication Date:
August 02, 2018
Filing Date:
January 19, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
LILLY CO ELI (US)
International Classes:
A61K31/445; C07D417/14; A61P25/00; A61P25/28
Domestic Patent References:
WO2014159234A12014-10-02
WO2004094380A12004-11-04
Foreign References:
US9120781B22015-09-01
US20160031871A12016-02-04
Other References:
NELSON ET AL., JNEUROPATHOL EXP NEUROL., vol. 71, no. 5, 2012, pages 362 - 381
WILLIAMS ET AL., BRAIN, vol. 130, 2007, pages 1566 - 76
YUZWA ET AL., NAT CHEM BIOL, vol. 4, no. 8, 2008, pages 483 - 490
YUZWA ET AL., NAT CHEM BIOL, vol. 8, 2012, pages 393 - 399
GRAHAM ET AL., NEUROPHARMACOLOGY, vol. 79, 2014, pages 307 - 313
"Remington: The Science and Practice of Pharmacy", 2012, PHARMACEUTICAL PRESS
J. JACQUES ET AL.: "Enantiomers, Racemates, and Resolutions", 1981, JOHN WILEY AND SONS, INC.
E.L. ELIEL; S.H. WILEN: "Stereochemistry of Organic Compounds", 1994, WILEY-INTERSCIENCE
GOULD, P.L.: "Salt selection for basic drugs", INTERNATIONAL JOURNAL OF PHARMACEUTICS, vol. 33, 1986, pages 201 - 217, XP025813036, DOI: doi:10.1016/0378-5173(86)90055-4
BASTIN, R.J. ET AL.: "Salt Selection and Optimization Procedures for Pharmaceutical New Chemical Entities", ORGANIC PROCESS RESEARCH AND DEVELOPMENT, vol. 4, 2000, pages 427 - 435
BERGE, S.M. ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
T. W. GREEN; P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY-INTERSCIENCE
N. MASUDA ET AL., BIOORG MED CHEM, vol. 12, 2004, pages 6171 - 6182
"Characterization of crystalline and partially crystalline solids by X-ray powder diffraction (XRPD) Official", 1 May 2015, article "The U. S. Pharmacopeia 38 - National Formulary 35"
KIM, EUN JU; KANG, DAE OOK; LOVE, DONA C.; HANOVER, JOHN A, CARBOHYDRATE RESEARCH, vol. 341, no. 8, 2006, pages 971 - 982
Attorney, Agent or Firm:
LENTZ, Nelsen L. et al. (US)
Download PDF:
Claims:
WE CLAIM:

1 A compound of the formula:

or a pharmaceutically acceptable salt thereof.

The compound or salt according to claim 1 wherein the methyl at position 2 in the cis configuration relative to the oxygen at position 4 on the piperidine ring:

3. The compound or salt according to either claim 1 or claim 2 wherein the compound is N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazol- 3-yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide.

4. The compound according to claim 3 which is N-[4-fluoro-5-[[(2S,4S)-2- methyl-4-[(5-methyl-l,2,4-oxadiazol-3-yl)methoxy]-l- piperidyl]methyl]thiazol-2-yl]acetamide.

5. The compound according to claim 4 wherein the compound is crystalline.

6. The compound according to claim 5 which is characterized by a peak in the X-ray powder diffraction spectrum, at diffraction angle 2-theta of 12.1° in combination with one or more peaks selected from the group consisting of 15.3°, 21.6°, 22.2°, 22.7°, 23.5°, 24.3°, and 26.8°, with a tolerance for the diffraction angles of 0.2 degrees.

7. A method of treating Alzheimer's disease in a patient, comprising

administering to a patient in need of such treatment an effective amount of a compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof.

8. A method of treating the progression of mild cognitive impairment to

Alzheimer's disease in a patient, comprising administering to a patient in need of such treatment an effective amount of a compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof.

9. A method of treating progressive supranuclear palsy in a patient, comprising administering to a patient in need of such treatment an effective amount of a compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof.

10. A compound or pharmaceutically acceptable salt thereof according to any one of claims 1-6 for use in therapy.

11. A compound or pharmaceutically acceptable salt thereof according to any one of claims 1-6 for use in the treatment of Alzheimer's disease.

12. A compound or pharmaceutically acceptable salt thereof according to any one of claims 1-6 for use in treating the progression of mild cognitive impairment to Alzheimer's disease.

13. A compound or pharmaceutically acceptable salt thereof according to any one of claims 1-6 for use in treating progressive supranuclear palsy.

14. A pharmaceutical composition, comprising a compound or a pharmaceutically acceptable salt thereof according to any one of claims 1-6 with one or more pharmaceutically acceptable carriers, diluents, or excipients.

15. A process for preparing a pharmaceutical composition, comprising admixing a compound or a pharmaceutically acceptable salt thereof according to any one of claims 1-6 with one or more pharmaceutically acceptable carriers, diluents, or excipients.

Description:
N-[4-FLUORO-5-[[(2S,4S)-2-METHYL-4-[(5-METHYL-1 ,2,4-OXADIAZOL-3-YL)METHOXY] -1 -PIPERIDYL]METHYL]THIAZOL-2-YL]ACETAMIDE AS OGA INHIBITOR

The present invention relates to novel 5-methyl-l,2,4-oxadiazol-3-yl compounds, to pharmaceutical compositions comprising the compounds, to methods of using the compounds to treat physiological disorders, and to intermediates and processes useful in the synthesis of the compounds.

The present invention is in the field of treatment of Alzheimer's disease, progressive supranuclear palsy (PSP) and other diseases and disorders involving tau-mediated neurodegeneration, known collectively as tauopathies.

Alzheimer's disease is a devastating neurodegenerative disorder that affects millions of patients worldwide. In view of the currently approved agents on the market which afford only transient, symptomatic benefits to the patient, there is a significant unmet need in the treatment of Alzheimer's disease.

The oligomerization of the microtubule-associated protein tau into filamentous structures such as paired helical filaments (PHFs) and straight or twisted filaments, which give rise to neurofibrillary tangles ( FTs) and neuropil threads (NTs), is one of the defining pathological features of Alzheimer's disease and other tauopathies. The number of NFTs in the brains of individuals with Alzheimer's disease has been found to correlate closely with the severity of the disease, suggesting tau has a key role in neuronal dysfunction and neurodegeneration (Nelson et al., J Neuropathol Exp Neurol, 71(5), 362- 381(2012)). Tau pathology has been shown to correlate with disease duration in PSP; cases with a more aggressive disease course have a higher tau burden than cases with a slower progression. (Williams et al., Brain, 130, 1566-76 (2007)).

Recent studies (Yuzwa et al., Nat Chem Biol, 4(8), 483-490 (2008)) support the therapeutic potential of O-GlcNAcase (OGA) inhibitors to limit tau hyperphosphorylation and aggregation into pathological tau for the treatment of Alzheimer's disease and related tau-mediated neurodegeneration disorders. Specifically, the OGA inhibitor Thiamet-G has been linked in slowing motor neuron loss in the JNPL3 tau mouse model (Yuzwa et al., Nat Chem Biol, 8, 393-399 (2012) and to a reduction in tau pathology and dystrophic neurites in the Tg4510 tau mouse model (Graham et al., Neuropharmacology, 79, 307- 313 (2014)). Accordingly, OGA inhibitors are recognized as a valid therapeutic approach to reduce the accumulation of hyperphosphorylated, pathological forms of tau, such as NFTs and NTs. U.S. Patent No. 9, 120,781 discloses hexahydrobenzooxazole and hexahydrobenzothiazole derivatives which possess OGA inhibitory activity and are further disclosed as useful in treating diseases and disorders related to deficiency or overexpression of OGA, and/or accumulation or deficiency of 2-acetamido-2-deoxy-5B- D-glucopyranoside (O-GlcNAc). In addition, US 2016/0031871 discloses certain glycosidase inhibitors for treating Alzheimer's disease.

OGA inhibitors that are brain penetrant are desired to provide treatments for tau-mediated neurodegeneration disorders, such as Alzheimer's disease and PSP. The present invention provides certain novel compounds that are inhibitors of OGA.

Accordingly, the present invention provides a compound of Formula I:

Formula I

or a pharmaceutically acceptable salt thereof.

In addition, the present invention provides a compound of Formula la:

Formula la

or a pharmaceutically acceptable salt thereof.

The present invention also provides a method of treating Alzheimer's disease in a patient in need of such treatment, comprising administering to the patient an effective amount of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof.

The present invention further provides a method of treating the progression of mild cognitive impairment to Alzheimer's disease in a patient in need of such treatment, comprising administering to the patient an effective amount of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof.

The present invention also provides a method of treating progressive supranuclear palsy in a patient in need of such treatment, comprising administering to the patient an effective amount of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof. The present invention also provides a method of treating tau-mediated neurodegenerative disorders in a patient, comprising administering to a patient in need of such treatment an effective amount of a compound of Formulas I or la, or a

pharmaceutically acceptable salt thereof.

Furthermore, this invention provides a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof for use in therapy, in particular for use in the treatment of Alzheimer's disease or for use in preventing the progression of mild cognitive impairment to Alzheimer's disease. In addition, this invention provides a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof for use in the treatment of progressive supranuclear palsy. The invention also provides a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof for use in treating tau- mediated neurodegenerative disorders.

Even furthermore, this invention provides the use of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of Alzheimer's disease or for preventing the progression of mild cognitive impairment to Alzheimer's disease. In addition, this invention provides the use of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of progressive supranuclear palsy. The invention also provides the use of a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for treating tau-mediated neurodegenerative disorders.

The invention further provides a pharmaceutical composition, comprising a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents, or excipients. The invention further provides a process for preparing a pharmaceutical composition, comprising admixing a compound of Formulas I or la, or a pharmaceutically acceptable salt thereof, with one or more pharmaceutically acceptable carriers, diluents, or excipients. This invention also encompasses novel intermediates and processes for the synthesis of the compounds of Formulas I and la.

Mild cognitive impairment has been defined as a potential prodromal phase of dementia associated with Alzheimer's disease based on clinical presentation and on progression of patients exhibiting mild cognitive impairment to Alzheimer's dementia over time. The term "preventing the progression of mild cognitive impairment to Alzheimer's disease" includes restraining, slowing, stopping, or reversing the progression of mild cognitive impairment to Alzheimer's disease in a patient.

As used herein, the terms "treating" or "to treat" includes restraining, slowing, stopping, or reversing the progression or severity of an existing symptom or disorder.

As used herein, the term "patient" refers to a human.

As used herein, the term "effective amount" refers to the amount or dose of compound of the invention, or a pharmaceutically acceptable salt thereof which, upon single or multiple dose administration to the patient, provides the desired effect in the patient under diagnosis or treatment.

An effective amount can be readily determined by one skilled in the art by the use of known techniques and by observing results obtained under analogous circumstances. In determining the effective amount for a patient, a number of factors are considered, including, but not limited to: the species of patient; its size, age, and general health; the specific disease or disorder involved; the degree of or involvement or the severity of the disease or disorder; the response of the individual patient; the particular compound administered; the mode of administration; the bioavailability characteristics of the preparation administered; the dose regimen selected; the use of concomitant medication; and other relevant circumstances.

The compounds of the present invention are generally effective over a wide dosage range. For example, dosages per day normally fall within the range of about 0.1 to about 15 mg/kg of body weight. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed with acceptable side effects, and therefore the above dosage range is not intended to limit the scope of the invention in any way.

The compounds of the present invention are preferably formulated as

pharmaceutical compositions administered by any route which makes the compound bioavailable, including oral and transdermal routes. Most preferably, such compositions are for oral administration. Such pharmaceutical compositions and processes for preparing same are well known in the art (See, e.g., Remington: The Science and Practice of Pharmacy, L.V. Allen, Editor, 22 nd Edition, Pharmaceutical Press, 2012). The compounds of Formulas I and la, or pharmaceutically acceptable salts thereof are particularly useful in the treatment methods of the invention, but certain

configurations are preferred. The following paragraphs describe such preferred configurations. It will be understood that these preferences are applicable both to the treatment methods and to the compounds of the invention.

Compounds of the present invention include:

Formula la

Formula lb

Formula Ic

Formula Id

and pharmaceutically acceptable salts thereof.

The compound of Formula I wherein the methyl and oxygen substituents on the piperidine ring are in the cis or trans configuration, or pharmaceutically acceptable salt thereof, are included within the scope of the invention, with the cis configuration being preferred. For example, one of ordinary skill in the art will appreciate that the methyl at position 2 is in the cis configuration relative to the oxygen at position 4 as shown in Scheme A below: heme A

Formula la

Formula Ic

In addition, one of ordinary skill in the art will appreciate that the methyl at position 2 is in the trans configuration relative to the oxygen at position 4 as shown Scheme B below:

Scheme B

Formula lb

Formula Id

Compounds wherein the chiral center at position 2 of piperidine ring is in the S- configuration are further preferred. Although the present invention contemplates all individual enantiomers and diasteromers, as well as mixtures of the enantiomers of said compounds, including racemates, the compounds with the absolute configuration as set forth below are particularly preferred:

N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazo l-3-yl)methoxy]-l- piperidyl]methyl]thiazol-2-yl]acetamide, and pharmaceutically acceptable salts thereof; and

N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazo l-3-yl)methoxy]-l- piperidyl]methyl]thiazol-2-yl]acetamide are particularly preferred.

The crystalline form of N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl- 1,2,4- oxadiazol-3-yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acet amide is especially preferred. The crystalline form of N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl- 1,2,4- oxadiazol-3-yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acet amide which is

characterized by a peak in the X-ray powder diffraction spectrum at diffraction angle 2- theta of 12.1° in combination with one or more peaks selected from the group consisting of 15.3°, 21.6°, 22.2°, 22.7°, 23.5°, 24.3°,and 26.8°, with a tolerance for the diffraction angles of 0.2 degrees, is further preferred.

Individual isomers, enantiomers, and diastereomers may be separated or resolved by one of ordinary skill in the art at any convenient point in the synthesis of compounds of the invention, by methods such as selective crystallization techniques or chiral chromatography (See for example, J. Jacques, et al., "Enantiomers, Racemates, and Resolutions" , John Wiley and Sons, Inc., 1981, and E.L. Eliel and S.H. Wilen,"

Stereochemistry of Organic Compounds", Wiley-Interscience, 1994).

A pharmaceutically acceptable salt of the compounds of the invention can be formed, for example, by reaction of an appropriate free base of a compound of the invention and an appropriate pharmaceutically acceptable acid in a suitable solvent under standard conditions well known in the art. The formation of such salts is well known and appreciated in the art. See, for example, Gould, P.L., "Salt selection for basic drugs," International Journal of Pharmaceutics, 33: 201-217 (1986); Bastin, R.J., et al. "Salt Selection and Optimization Procedures for Pharmaceutical New Chemical Entities," Organic Process Research and Development, 4: 427-435 (2000); and Berge, S.M., et al., "Pharmaceutical Salts," Journal of Pharmaceutical Sciences, 66: 1-19, (1977).

The compounds of the present invention, or salts thereof, may be prepared by a variety of procedures known to one of ordinary skill in the art, some of which are illustrated in the schemes, preparations, and examples below. One of ordinary skill in the art recognizes that the specific synthetic steps for each of the routes described may be combined in different ways, or in conjunction with steps from different schemes, to prepare compounds of the invention, or salts thereof. The products of each step in the schemes below can be recovered by conventional methods well known in the art, including extraction, evaporation, precipitation, chromatography, filtration, trituration, and crystallization. In the schemes below, all substituents unless otherwise indicated, are as previously defined. The reagents and starting materials are readily available to one of ordinary skill in the art. Without limiting the scope of the invention, the following schemes, preparations, and examples are provided to further illustrate the invention. In addition, one of ordinary skill in the art appreciates that the compounds of Formulas la, lb, Ic, and Id may be prepared by using starting material with the corresponding stereochemical configuration which can be prepared by one of skill in the art. For example, the Schemes below utilize starting materials with the configuration

corresponding ultimately to Formula la.

Generally, a compound of Formula la may be prepared from a compound of Formula II (Scheme 1). More specifically, a compound of Formula Ila is reductively alkylated with N-(4-fluoro-5-formylthiazol-2-yl)acetamide in the presence of a suitable reducing agent such as sodium triacetoxyborohydride in a suitable solvent to provide a compound of Formula la in a suitable solvent, such as ethyl acetate. N-(4-Fluoro-5- formylthiazol-2-yl)acetamide may be prepared by methods know in the chemical arts as well as methods provided in the following Preparations and Examples.

A compound of Formula Ila may be prepared from a compound of Formula Ilia where Pg is a suitable amine protecting group. More specifically, a compound of

Formula Ila where Pg is tert-butyl carboxylate (t-BOC) is reacted with an acid such as hydrochloric acid or trifluoroacetic acid in a suitable solvent such as dioxane or dichloromethane to provide a compound of Formula Ila. Suitable amine protecting groups are known in the chemical arts and include t-BOC and Cbz as well as those discussed in T. W. Green, P. G. M. Wuts, "Protective Groups in Organic Synthesis" Wiley-Interscience, New York, 1999. Scheme 1

Ilia na la

A compound of Formula Ilia where Pg is a suitable amine protecting group may be prepared from a compound of Formula IVa (Scheme 2). More specifically, a compound of Formula IVa where Pg is tert-butyl carboxylate is reacted with

3-(chloromethyl)-5-methyl-l,2,4-oxadiazole in the presence of a base such as sodium tert-butoxide to provide a compound of Formula Ilia. The reaction is conveniently carried out in a solvent such as acetonitrile or dimethylformamide. A compound of Formula IVa where Pg is tert-butyl carboxylate may be prepared essentially as described in WO 2004/094380 Al . More specifically, a compound of Formula Va is reacted with a reducing agent such as lithium tri(sec-butyl)borohydride in a solvent such as

tetrahydrofuran to provide a compound of Formula IVa where Pg is tert-butyl carboxylate. A compound of Formula Va where Pg is a suitable amine protecting group may be prepared by processes known in the chemical arts including those described in WO 2004/094380 Al . Scheme 2

Va iva Ilia

Preparation 1

Synthesis of tert-butyl N-(4-fluoro-5-formyl-thiazol-2-yl)carbamate.

Cesium fluoride (227 g, 1480 mmol) is added to a solution of tert-butyl N-(4- chloro-5-formyl-thiazol-2-yl)carbamate (38.8 g, 148 mmol; for preparation of tert-butyl N-(4-chloro-5-formyl-thiazol-2-yl)carbamate see for example, N. Masuda, et al., Bioorg Med Chem, 12, 6171-6182 (2004)) in DMSO (776 mL) at room temperature. The reaction mixture is stirred in a 145 °C heating block with an internal temperature of 133 °C for 48 hours, then the mixture is cooled in an ice-water bath. To the mixture is added saturated aqueous sodium bicarbonate solution (500 mL), brine (500 mL) and ethyl acetate (500 mL). The mixture is stirred at room temperature for 10 minutes, then is filtered through diatomaceous earth, washing with ethyl acetate (500 mL). The filtrate is transferred to a separating funnel and the layers are separated, then the aqueous layer is extracted with ethyl acetate (1 L). The combined organics are washed with brine (1 L), then the brine layer is extracted with ethyl acetate (300 mL). The combined organics are dried over sodium sulfate, filtered and concentrated to give a residue. The residue is passed through a pad of silica gel (330 g) eluting with 5% ethyl acetate in dichloromethane (1.5 L) and the filtrate is concentrated to give a residue (24.2 g).

The residue (32.7 g of combined lots, 133 mmol) is dissolved in isopropanol (303 mL), filtered and then is purified by SFC (Supercritical Fluid Chromatography) using an IC column (cellulose polysaccharide derivative: tris (3,5-dichlorophenylcarbamate, 30 x 250mm, 5u) with 10%IPA (no additive) at 180 mL/minute with 3 mL injections. The product-containing fractions are concentrated to give the title compound (16.1 g. MS m/z 247.0 (M+H).

Preparation 2

Synthesis N-(4-fluoro-5-formyl-thiazol-2-yl)acetamide (Method A)

In a jacketed vessel, zinc bromide (91.9 g,408 mmol) is added in one portion to a mixture of tert-butyl N-(4-fluoro-5-formyl-thiazol-2-yl)carbamate (33.5 g, 136 mmol) and dichloromethane (503 mL) at room temperature. The reaction mixture is stirred overnight at an internal temperature of 37 °C, then the jacket temperature is set to -10 °C and tetrahydrofuran (111 mL) is added dropwise over 15 minutes, maintaining an internal temperature below 6 °C. The jacket temperature is then set to -30 °C and pyridine (110 mL, 1360 mmol) is added dropwise over 5 minutes, maintaining an internal temperature below 5 °C. The jacket temperature is set to 0 °C and acetic anhydride (116 mL, 1220 mmol) is added dropwise over 5 minutes. The reaction mixture is stirred overnight at an internal temperature of 37 °C, then is cooled to room temperature and passed through a short pad of diatomaceous earth, eluting with tetrahydrofuran (500 mL). The filtrate is transferred to a flask and the mixture is concentrated to give a residue, which is concentrated from toluene (50 mL). To the residue is added a solution of citric acid monohydrate (57.2 g, 272 mmol) in water (400 mL) and 2-methyltetrahydrofuran (400 mL) and the mixture is stirred at 40 °C for 5 minutes, then is passed through a short pad of diatomaceous earth, eluting with 2-methyltetrahydrofuran (100 mL). The filtrate is transferred to a separating funnel and the layers are separated. The aqueous layer is extracted with 2-methyltetrahydrofuran (2 x 250 mL) and the combined organics are diluted with water (500 mL). To the mixture is added solid sodium bicarbonate portionwise over 5 minutes with stirring until gas evolution ceases. The mixture is transferred to a separating funnel and the layers are separated, then the aqueous layer is extracted with 2-methyltetrahydrofuran (200 mL and 100 mL). The combined organics are dried over sodium sulfate, filtered and concentrated to give a residue, which is diluted with 2-methyltetrahydrofuran (100 mL) and the mixture is passed through a short pad of silica gel (250 g), eluting with 2-methyltetrahydrofuran (2.5 L). The filtrate is concentrated to give a residue which is suspended in a 1 : 1 mixture of dichloromethane and heptane (202 mL). The mixture is stirred at room temperature for 30 minutes and then filtered. The filtered solid is dried under vacuum at 40 °C for 2 hours to give the title compound (18.0 g, 70%). MS m/z 189.0 (M+H).

Alternative synthesis of N-(4-fluoro-5-formyl-thiazol-2-yl)acetamide (Method B).

Add dichloromethane (1325 g, 15.6 mol) to 2-amino-4-chlorothiazole-5- carbaldehyde (100 g, 0.61 mol) and pyridine (194.6 g, 2.46 mol), and cool to 0-5 °C. Add acetic anhydride (188.4 g, 1.85 mol) dropwise, maintaining the temperature at 0-5 °C.

After addition is complete, adjust the temperature to 20-25 °C and stir for 41 hours.

Concentrate under reduced pressure followed by addition of 35% aqueous HC1 (200 mL) and water (1.5 L), maintaining the temperature at less than 40 °C . Cool to 20-25 °C and stir for 18 hours. Filter the mixture and wash the collected solid with water. Dry the solids at 60-65 °C for 24 h to provide N-(4-chloro-5-formylthiazol-2-yl)acetamide (75 g,

0.4 mol).

Under an inert atmosphere, add sulfolane (1000 ml) to the N-(4-chloro-5- formylthiazol-2-yl)acetamide (50 g, 0.244 mol, prepared directly above),

tetramethylammonium chloride (107.1 g, 0.977 mol), and cesium fluoride (370.6 g, 2.44 mmol). Heat to 130 °C and stir for 23 h. HPLC analysis shows 75% conversion with an in situ yield of 45% of the title compound. Alternative synthesis of N-(4-fluoro-5-formyl-thiazol-2-yl)acetamide (Method C).

Add 2-propanol (150 mL) to tetramethylammonium fluoride. tetrahydrate (10.2 g, 109.0 mmol) and concentrate the mixture to 2-3 volumes under vacuum with internal temperature maintained at 70 °C to remove water. Add 2-propanol (200mL) and and concentrate the mixture to 2-3 volumes under vacuum. Repeat two more times. Add DMF (200mL) and concentrate to 2-3 volumes under vacuum. Add THF (200 mL) and concentrate to 2-3 volumes. Repeat two more times. Charge N-(4-chloro-5- formylthiazol-2-yl)acetamide (1.22 g, 5.96 mmol, prepared above in Method B) and DMF (12 ml). Heat to 110 °C and stir for 12 h. Cool reaction mixture to 25 °C. Add 2- methyltetrahydrofuran (40 mL) and water (40 mL). The layers are separated and the aqueous layer was extracted with 2-methyltetrahydrofuran (40 mL). The layers were separated and the combined organic layers were washed with water (20 mL). The layers were separated and the organic layer was concentrated. Add ethyl acetate (20 mL) and water (5 mL). The layers were separated and the organic layer concentrated to remove solvent. Add ethyl acetate (2mL) and heptane (2 mL) and filter. The filtered solid is dried under vacuum at 55 °C for 18 hours to give the title compound as a 93% mixture with N-(4-chloro-5-formylthiazol-2-yl)acetamide.

Preparation 3

Synthesis of tert-butyl (2S,4S)-4-hydroxy-2-methyl-piperidine-l-carboxylate.

To a flask is added tert-butyl (2S)-2-methyl-4-oxo-piperidine-l-carboxylate (50 g, 234.44 mmol) and tetrahydrofuran (500 mL). The mixture is cooled to -65 °C under an atmosphere of nitrogen and lithium tri(sec-butyl)borohydride (304.77 mL, 304.77 mmol; 1 M in tetrahydrofuran) is added dropwise over 45 minutes, maintaining an internal temperature below -60 °C. The reaction mixture is stirred at room temperature for 1 hour, then is cooled to -30 °C. To the reaction mixture is added a mixture of water (25.34 mL) and tetrahydrofuran (100.16 mL), maintaining an internal temperature below -20 °C. An aqueous solution of hydrogen peroxide (118.88 mL, 1.17 mol, 30 wt/wt%) in water (126.70 mL) is added dropwise over 1 hour, maintaining an internal temperature below 10 °C. To the mixture is added aqueous hydrogen chloride solution (46.89 mL, 234.44 mmol, 5 M) and methyl t-butyl ether (1.00 L) and the mixture is warmed to room temperature. The layers are separated and the organic phase is stirred with a solution of sodium metabisulfite (222.84 g, 1.17 mol) in water (500 mL) for 10 minutes at room temperature. The layers are separated and the organic phase is dried over magnesium sulfate and concentrated. The residue is purified by flash chromatography (0-50 % methyl t-butyl ether /isohexane, silica gel) and the product-containing fractions are combined and concentrated to give the title compound (40.4 g, 78%). ES/MS (m/e) 238 (M+Na).

Alternative synthesis of tert-butyl (2S,4S)-4-hydroxy-2-methyl-piperidine-l-carboxylate.

To a glass-lined reactor containing deionized water (460 L), and potassium

dihydrogen phosphate (6.5 kg, 0.41 equiv) at 20 °C is charged DMSO (27.4 kg , 1.0 vol) and D-(+)-glucose monohydrate (28.9 kg, 1.25 equiv). The internal temperature is adjusted to 30 °C, and the pH of the reaction is adjusted to 6.9 by addition of aqueous sodium hydroxide (8%, 15 L, 0.28 equiv). The reactor is charged with tert-butyl (2S)-2- methyl-4-oxo-piperidine-l-carboxylate (24.9 kg, 1.0 equiv (99.1%ee)), and the mixture is agitated at 30 °C for 15 min. Ketoreductase (KRED-130, 250 g, 1% w/w), glucose dehydrogenase (GDH-101, 250 g, 1% w/w), and NADP sodium salt (63 g, 0.25% w/w) are charged directly to the reaction mixture via an open port. The mixture is maintained at a temperature of 30 °C and pH 7.0 ± 0.2 via addition of 8% aqueous NaHC0 3 . After stirring for 16.5 h (99.5% conversion), the reaction is charged with Celite™ (12.5 kg, 50 w/w%) and toluene (125 L, 5 vol). After stirring for 30 min at 30 °C, the mixture is transferred to another 2000 L reactor via an in-line GAF-filter (4 sock) over the period of 1 h. The mixture is allowed to stand 30 min without agitation, the layers are separated, and the aqueous layer is back-extracted with toluene (2 x 125 L). The combined organic layers are filtered (in-line GAF-filter), and the toluene mixture is washed with aqueous sodium chloride solution (25%, 125 L, 5 vol) at 25 °C. The resulting toluene solution is azeotropically dried (partial vacuum, internal temp < 60 °C) to 0.10 w/w% water, and cooled to 20 °C. The mixture is filtered out of the reactor via a cartridge filter into clean drums under positive nitrogen pressure. The reaction mixture is then transferred from the drums into a 500 L glass lined vessel and concentrated under vacuum (< 60 °C) to a target residual volume of 56 L (2.25 vol). n-Heptane (169 kg, 10 vol) is charged at 40 °C, and the mixture is seeded with 25 g of tert-butyl (2S,4S)-4-hydroxy-2-m ethyl -piperi dine- 1- carboxylate. The resulting thick slurry is diluted with additional n-heptane (25 L, 1 vol) and cooled to 16 °C over 4 h. The product is isolated via centrifugation, washing with n-heptane (25 L per spin; 4 spins necessary), yielding 20.3 kg (81%; >99.9% ee) after drying for 11 h in a tray dryer at 30 °C. ES/MS (m/e) 238 (M+Na).

Preparation 4

Synthesis of tert-butyl (2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazol-3- yl)methoxy]piperidine-l-carboxylate.

3-(Chloromethyl)-5-methyl-l,2,4-oxadiazole (43.5 g, 301 mmol) is added to a solution of tert-butyl (2S,4S)-4-hydroxy-2-methyl-piperidine-l-carboxylate (29.5 g, 137 mmol) in acetonitrile (590 mL) at room temperature. The reaction mixture is stirred in an ice-water bath and sodium tert-butoxide (54.3 g, 548 mmol) is added in portions over 10 minutes, maintaining an internal temperature below 10 °C. The reaction mixture is stirred in an ice-water bath at an internal temperature of 5 °C for 9 hours, then is warmed slowly to room temperature and is stirred overnight. The reaction mixture is cooled in an ice- water bath and saturated aqueous ammonium chloride solution (200 mL) is added over 5 minutes, maintaining an internal temperature below 10 °C during the addition. The mixture is then diluted with water (100 mL) and warmed to room temperature. The mixture is extracted with methyl tert-butyl ether (2 x 300 mL) and the combined organics are washed with brine (300 mL). The combined organics are dried over sodium sulfate, filtered and concentrated to give a residue. The residue is passed quickly through a pad of silica gel (300 g) eluting with methyl tert-butyl ether (1 L) and the filtrate is

concentrated to give the title compound (46.5 g, 109%). MS m/z 334.0 (M+Na). Alternative synthesis of tert-butyl (2S.4S)-2-methyl-4-r(5-methyl-1.2.4-oxadiazol-3- vPmethoxylpiperidine-l-carboxylate.

To a solution of tert-butyl (2S,4S)-4-hydroxy-2-methyl-piperidine-l-carboxylate (0.25g, 1.16 mmol) and 3 -(chloromethyl)-5 -methyl- 1,2,4-oxadiazole (0.308g, 2.32 mmol) in Ν,Ν-dimethylformamide (3mL) under nitrogen at 0 °C is added portionwise sodium tert-butoxide (0.35 g, 3.5mmol) over 5 min. The reaction mixture is stirred at rt for 10 min then at 40 °C for 12h. The reaction mixture is cooled to room temperature then quenched with water (lOmL). The layers are separated and the aqueous phase is extracted with methyl tert-butyl ether (2x l0mL). The combined organic extracts are washed with an aqueous solution of lithium chloride (5%), dried over magnesium sulfate, filtered and concentrated under reduced pressure to afford the title compound (0.49 g, 0.7 mmol, 81% yield, 60% purity) as a brown oil. MS m/z 334.0 (M+Na). Preparation 5

Synthesis of 5-methyl-3-[[(2S,4S)-2-methyl-4-piperidyl]oxymethyl]-l,2,4-o xadiazole hydrochloride.

A flask containing tert-butyl (2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazol-3- yl)methoxy]piperidine-l-carboxylate (4.03 g, 12.9 mmol) is submerged in an ice-water bath. To this flask is added a 4 M solution of hydrochloric acid in 1,4-dioxane (25.9 mL, 104 mmol) dropwise over 5 minutes with stirring, maintaining an internal temperature below 20 °C during the addition. The reaction mixture is stirred at room temperature for lhour, then is concentrated to give the title compound (3.56 g, 92% yield based on 83% purity measured by l U NMR. MS m/z 212.0 (M+H).

Alternative synthesis of 5-methyl-3-[[(2S,4S)-2-methyl-4-piperidylloxymethyll-L2,4- oxadiazole hydrochloride.

Add methanol (50 mL) to tert-butyl (2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazol-

3-yl)methoxy]piperidine-l-carboxylate (12.9 g, 0.041 mol). The mixture is cooled to 0 °C. A 4M solution of hydrochloric acid in methanol (80 mL) is added dropwise to the cooled mixture, maintaining an internal temperature below 20 °C. The reaction mixture is then stirred at room temperature for 18 hours. The mixture is then concentrated to remove solvent. Acetone (10 mL) is added and the mixture is stirred for 20 min.

Tetrahydrofuran (40 mL) is added and the mixture is stirred for 3 hours. The solid is collected by filtration under nitrogen and the filtered solid cake is rinsed with

tetrahydrofuran. The filtered solid is then dried under vacuum at 45 °C for 2 hours to give the title compound as a 90% purity. Recrystallization using acetone can increase purity of title compound to 95%. Preparation 6

Synthesis of 5-methyl-3-[[(2S,4S)-2-methyl-4-piperidyl]oxymethyl]-l,2,4-o xadiazole.

To a solution of tert-butyl (2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazol-3- yl)methoxy]piperidine-l-carboxylate (0.49g, 1.6 mmol) in dichloromethane (lOmL) under nitrogen is added trifluoroacetic acid (1.8 mL, 23 mmol). The mixture is stirred at room temperature for 3h. The mixture is concentrated under reduced pressure to afford a yellow oil. The residue is dissolved in methanol (5mL) and poured onto a cation exchange cartridge, eluted with methanol (2x lOmL) then a 2 M ammonia solution in methanol (lOmL). The filtrate is concentrated under reduced pressure to give title compound (0.3g, 1.4 mmol, 91%). MS m/z 212.0 (M+H).

Example 1

Synthesis of N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazo l-3- yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide.

N-(4-Fluoro-5-formyl-thiazol-2-yl)acetamide (28.3 g, 150 mmol) is added to 5- methyl-3-[[(2S,4S)-2-methyl-4-piperidyl]oxymethyl]-l,2,4-oxa diazole hydrochloride (48.7 g, 185 mmol, 94% purity) in ethyl acetate (707 mL) at room temperature. The reaction mixture is stirred at room temperature and N,N-diisopropylethylamine (34.1 mL, 195 mmol) is added dropwise over 1 minute, then sodium triacetoxyborohydride (98.5 g, 451 mmol) is added in one portion. The reaction mixture is stirred in a 31 °C heating block overnight with an internal temperature of 30 °C, then is cooled in an ice-water bath to an internal temperature of 5 °C. To the mixture is added 2 M aqueous hydrochloric acid solution (226 mL) over 15 minutes, maintaining an internal temperature below 10 °C. To the mixture is added water (250 mL) and the mixture is stirred at room

temperature for 5 minutes. The layers are separated and the organic layer is extracted with a mixture of 2 M aqueous hydrochloric acid solution (28 mL) in water (50 mL). The first aqueous layer is stirred in an ice-water bath and 50% aqueous sodium hydroxide solution (25.7 mL) is added dropwise over 10 minutes, maintaining an internal temperature below 10 °C. The mixture is diluted with saturated aqueous sodium bicarbonate solution (100 mL), then is stirred at room temperature for 10 minutes and then is extracted with ethyl acetate (3 x 400 mL). The combined organics are dried over sodium sulfate, filtered and concentrated to give a residue. The second aqueous layer from the extraction with aqueous hydrochloric acid is diluted with

2-methyltetrahydrofuran (200 mL) and the mixture is passed through a short pad of diatomaceous earth. The filtrate is transferred to a separating funnel and the layers are separated. The aqueous layer is stirred in an ice-water bath and 50% aqueous sodium hydroxide solution (3.15 mL) is added dropwise over 5 minutes, maintaining an internal temperature below 10 °C. The mixture is diluted with saturated aqueous sodium bicarbonate solution (10 mL), then is stirred at room temperature for 5 minutes and then is extracted with ethyl acetate (3 χ 40 mL) and 10% isopropanol in ethyl acetate (100 mL). The combined organics are dried over sodium sulfate, filtered and concentrated to give a residue, which is combined with the residue from the first part of the workup. The combined residue is passed through a pad of silica gel (350 g) eluting with ethyl acetate (3.5 L) and the filtrate is concentrated to give a residue (45.8 g).

The residue (47.5 g of combined lots, 123.9 mmol) is purified by flash

chromatography, eluting with 50-100%) ethyl acetate in heptane. The product-containing fractions are concentrated to residue, which is suspended in a 1 : 1 mixture of methyl-tert- butyl ether and heptane (448 mL). The mixture is stirred in a 46 °C heating block for 30 minutes at an internal temperature of 45 °C, then is cooled to room temperature over 2 hours with stirring. The mixture is filtered, washing the solid with a 1 : 1 mixture of methyl-tert-butyl ether and heptane (30 mL). The filtered solid is dried under vacuum at 40 °C overnight to give the title compound (28.5 g). MS m/z 384.0 (M+H);

[a] D 20 = +33.4 ° (C=0.26, methanol).

Alternative synthesis of N-r4-fluoro-5-rr(2S.4SV2-methyl-4-r(5-methyl-1.2.4-oxadiazol - 3-yl)methoxy1-l-piperidyl1methyl1thiazol-2-yl1acetamide.

To a solution of N-(4-fluoro-5-formyl-thiazol-2-yl)acetamide (0.05g, 0.28 mmol) and 5-methyl-3-[[(2S,4S)-2-methyl-4-piperidyl]oxymethyl]-l,2,4-o xadiazole (0.04g,

0.19mmol) in dichloromethane (10 mL) under nitrogen are added

Ν,Ν-diisopropyl ethyl amine (0.1 mL, 0.57 mmol) and sodium triacetoxyborohydride (0.12g, 0.57 mmol). The reaction mixture is stirred at room temperature for 12h. The reaction mixture is poured into a saturated aqueous solution of sodium bicarbonate (lOmL). The layers are separated and the aqueous phase is extracted with

dichloromethane (2* lOmL). The combined organic extracts are dried over magnesium sulfate, filtered and concentrated under reduced pressure to afford an orange oil.

The residue is taken up in methanol (to a total volume of 9.8 ml), filtered and purified by prep-HPLC (Phenomenex Gemini-NX 10 Micron 50* 150mm C-18) (CH3CN & Water with 10 mM ammonium bicarbonate adjusted to pH 9 with ammonium hydroxide, 15 % to 100% CH3CN over lOmin at 1 lOml/min) (1 injection) (271/204 nm) to give the title compound (0.02g, 0.05 mmol, 28%). MS m/z 384.2 (M+H).

Example 1A

Crystalline N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazo l-3- yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide.

Suspend crude N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl-l,2,4-oxadiazo l-3- yl)methoxy]-l-piperidyl]methyl]thiazol-2-yl]acetamide (29.9g) in 448 mL of 50% methyl tert butyl ether in heptane at 46 °C for 30 minutes. Stir the mixture and cool to 19 °C over two hours before filtering following with a wash of 30 mL of 50% methyl tert butyl ether in heptane to provide the title compound (28.5g, 95% yield). X-Rav Powder Diffraction (XRPD) of Example 1 A

The XRPD patterns of crystalline solids are obtained on a Bruker D4 Endeavor X-ray powder diffractometer, equipped with a CuKa source (λ = 1.54060 A) and a Vantec detector, operating at 35 kV and 50 mA. The sample is scanned between 4 and 40° in 2Θ, with a step size of 0.0087° in 2Θ and a scan rate of 0.5 seconds/step, and with 0.6 mm divergence, 5.28mm fixed anti-scatter, and 9.5 mm detector slits. The dry powder is packed on a quartz sample holder and a smooth surface is obtained using a glass slide. It is well known in the crystallography art that, for any given crystal form, the relative intensities of the diffraction peaks may vary due to preferred orientation resulting from factors such as crystal morphology and habit. Where the effects of preferred orientation are present, peak intensities are altered, but the characteristic peak positions of the polymorph are unchanged, (see, e.g. The U. S. Pharmacopeia 38 - National Formulary 35 Chapter 941 Characterization of crystalline and partially crystalline solids by X-ray powder diffraction (XRPD) Official May 1, 2015). Furthermore, it is also well known in the crystallography art that for any given crystal form the angular peak positions may vary slightly. For example, peak positions can shift due to a variation in the temperature or humidity at which a sample is analyzed, sample displacement, or the presence or absence of an internal standard. In the present case, a peak position variability of ± 0.2 in 2Θ will take into account these potential variations without hindering the unequivocal identification of the indicated crystal form. Confirmation of a crystal form may be made based on any unique combination of distinguishing peaks (in units of ° 2Θ), typically the more prominent peaks. The crystal form diffraction patterns, collected at ambient temperature and relative humidity, aree adjusted based on NIST 675 standard peaks at 8.85 and 26.77 degrees 2-theta.

A prepared sample of crystalline N-[4-fluoro-5-[[(2S,4S)-2-methyl-4-[(5-methyl- l,2,4-oxadiazol-3-yl)methoxy]-l-piperidyl]methyl]thiazol-2-y l]acetamide is characterized by an XRPD pattern using CuKa radiation as having diffraction peaks (2-theta values) as described in Table 1 below. Specifically the pattern contains a peak at 12.1° in combination with one or more peaks selected from the group consisting of 15.3°, 21.6°, 22.2°, 22.7°, 23.5°, 24.3°, and 26.8° with a tolerance for the diffraction angles of 0.2 degrees. Table 1 : X-ray powder diffraction peaks of crystalline N-[4-fluoro-5-[[(2S,4S)-2-methyl- 4-[(5-methyl-l,2,4-oxadiazol-3-yl)methoxy]-l-piperidyl]methy l]thiazol-2-yl]acetamide,

Example 1A.

In vitro human OGA enzyme assay

Generation of OGA proteins

The nucleotide sequence encoding full-length human O-GlcNAc-β-Ν- acetylglucosaminidase ( M_012215) is inserted into pFastBacl (Invitrogen) vector with an N-terminal poly-histidine (HIS) tag. Baculovirus generation is carried out according to the Bac-to-Bac Baculovirus Expression system (Invitrogen) protocol. Sf9 cells are infected at 1.5 x 10 6 cells/mL using 10 mL of PI virus per Liter of culture and incubated at 28 ° C for 48hrs. Cells are spun down, rinsed with PBS and the pellets stored at -80 ° C. The above OGA protein (His-OGA) is purified as follows: 4 L of cells are lysed in 200 mL of buffer containing 50 mM Tris, pH 8.0, 300 mM NaCl, 10% glycerol, 10 mM Imidazol, 1 mM Dithiothreitol (DTT), 0.1% Triton™ X-100, 4 tablets of protease inhibitors (complete EDTA-Free, Roche) for 45 min at 4 ° C. This cell lysate is then spun for 40 min at 16500 rpm at 4 ° C, and supernatant incubated with 6 mL of Ni-NTA resin (nickel-nitrilotriacetic acid) for 2 hours at 4 ° C.

Resin is then packed onto column and washed with 50 mM Tris, pH 8.0, 300 mM NaCl, 10% glycerol, 10 mM Imidazole, 0.1% Triton™ X-100, 1 mM DTT, followed by 50 mM Tris, pH 8.0, 150 mM NaCl, 10 mM Imidazol, 10% glycerol, 1 mM DTT. The proteins are eluted with 50 mM Tris, pH 8.0, 150 mM NaCl, 300 mM Imidazole, 10% glycerol, 1 mM DTT. Pooled His-OGA containing fractions are concentrated to 6 ml and loaded onto Superdex75 (16/60). The protein is eluted with 50 mM Tris, pH 8.0, 150 mM NaCl, 10%) glycerol, 2 mM DTT. Fractions containing His-OGA are pooled and protein concentration measured with BCA (Bradford Colorimetric Assay).

OGA enzyme assay

The OGA enzyme catalyses the removal of O-GlcNAc from nucleocytoplasmic proteins. To measure this activity Fluorescein di-N-acetyl-P-N-acetyl-D-glucosaminide (FD-GlcNAc, Kim, Eun Ju; Kang, Dae Ook; Love, Dona C; Hanover, John A.

Carbohydrate Research (2006), 341(8), 971-982) is used as a substrate at a final concentration of 10 μΜ (in the 96 well assay format) or 6.7 μΜ (in the 384 well assay format). This fluorogenic substrate becomes fluorescent upon cleavage by OGA, so that the enzyme activity can be measured by the increase in fluorescence detected at 535 nm (excitation at 485nm).

The assay buffer is prepared to give a final concentration of 50 mM

H2NaP03-HNa 2 P0 3 , 0.01% bovine serum albumin and 0.01% Triton™ X-100 in water, at pH 7. The final enzyme concentration is 3 nM (in the 96 well assay format) or 3.24 nM (in the 384 well assay format). Both assay formats yield essentially equivalent results.

Compounds to be tested are diluted in pure dimethyl sulfoxide (DMSO) using ten point concentration response curves. Maximal compound concentration in the reaction mixture is 30 μΜ. Compounds at the appropriate concentration are pre-incubated with OGA enzyme for 30 minutes before the reaction is started by the addition of substrate. Reactions are allowed to proceed for 60 minutes at room temperature. Then, without stopping the reaction, fluorescence is read. IC50 values are calculated by plotting the normalized data vs. log of the compound and fitting the data using a four parameter logistic equation.

The compound of Example 1 was tested essentially as described above and exhibited an IC50 of 2.36 nM ± 0.786 (n=8). This data demonstrates that the compound of Example 1 inhibits OGA enzyme activity in vitro. Whole cell assay for measuring the inhibition of OGA enzyme activity

Cell Plating:

Utilizing standard conditions known in the art, TRex-293 cells modified for inducible expression of the P301 S-1N4R form of the microtubule associated protein tau are generated and maintained in growth media, consisting of DMEM High Glucose (Sigma# D5796), supplemented with 10 % Tetracyclin-free Fetal Bovine Serum (FBS, Sigma F2442), 20 mM HEPES, 5 μg/mL Blasticidin (Life Technologies# Al 1139-03) and 200 μg/mL Zeocin (Life Technologies# R250-01). For the experiments, cells are plated in growth media at 10,000-14,000 cells per well in a Corning Biocoat (356663) 384 well plate coated with poly-D-Lysine, and incubated 20-24 h in a cell incubator at 37°C/5% C0 2 . Experiments are performed without inducing Tau expression.

Compound treatment:

Compounds to be tested are serially diluted 1/3 in pure DMSO using ten point concentration response curves and further diluted in growth media. 20-24 h after plating, cells are treated with test compound in growth media; maximal compound concentration is 15 μΜ (0.15% DMSO). The maximum inhibition is defined by replicate measurements of 15 uM Thiamet G and the minimum inhibition is defined by replicate measurements of 0.15% DMSO treatment. The cells are returned to the incubator at 37°C/5% C0 2 for 20- 24 hours. Compounds are tested in duplicates within each plate.

Immunostainmg:

After 20-24 hours of compound treatment, the media is removed from the assay plate and 25 μΐ ^ of 3.7 % Formaldehyde solution (Sigma # F1635) in DPBS (Sigma

#D8537) are added to each well and incubated for 30 minutes. The cells are then washed once with DPBS and then permeabilized with 0.1% Triton™ X-100 (Sigma# T9284). After 30 minutes, cells are washed twice with DPBS and then blocking solution(l% BSA/DPBS/0.1% Triton™ X-100) is added to each well and incubated for 60 minutes. The blocking solution is removed and a 0.40-0.33 μg/mL solution of O-GlcNAc Protein antibody (RL2 clone, Thermo, MAI 072) in blocking solution is added to the cells and allowed to sit overnight at 2-8 °C. The next day, the cells are washed twice with DPBS and the secondary antibody, Alexa Fluor 488 goat anti-mouse IgG (Life Technologies # Al 1001) at 2 ug/mL in DPBS is added to each well and allowed to sit at room

temperature for 90 min. The secondary antibody is removed, cells washed twice with DPBS and a solution of DAPI (Sigma #D9564) and RNase (Sigma, R6513) in DPBS at a concentration of 1 and 50 ug/mL, respectively, is added to each well. The plate is sealed, incubated for one hour and analyzed on an Acumen eX3 hci (TTP Labtech). A 11 the incubations and washing steps described above are done at room temperature, except for the primary antibody. Analysis and Results:

The plates are analyzed on an Acumen eX3 instrument using a 488 and 405 nm excitation lasers and two emission filters FL2 (500-530 nm) and FL1 (420-490 nm). The FL2 filter is the signal corresponding to the O-GlcNAc Protein antibody (RL2 clone) and the FL1 filter is the signal corresponding to the cell nuclei (DAPI). The ratio Total FL2/Total FL1 (Total fluorescence of each well without object or population selection) is used for data analysis. The data is normalized to a maximum inhibition as referenced by a 15 μΜ treatment of Thiamet G and a minimum inhibition as achieved by a 0.15% DMSO treatment. The data is fitted with a non-linear curve fitting application (4- parameters logistic equation) and IC50 values are calculated and reported.

The compound of Example 1 was tested essentially as described above and exhibited an IC50 of 21.9 nM ± 7.3 (n=5). This data demonstrates that the compound of Example 1 inhibits OGA enzyme activity in a cellular assay.