Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SELECTIVE BACE1 INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2016/137788
Kind Code:
A1
Abstract:
The present invention provides a compound of Formula II: or a pharmaceutically acceptable salt thereof.

Inventors:
HEMBRE ERIK JAMES (US)
LOPEZ JOSE EDUARDO (US)
MCMAHON JENNIFER ANNE (US)
RICHARDS SIMON JAMES (US)
WINNEROSKI LEONARD LARRY JR (US)
WOODS TIMOTHY ANDREW (US)
Application Number:
PCT/US2016/018160
Publication Date:
September 01, 2016
Filing Date:
February 17, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
LILLY CO ELI (US)
International Classes:
C07D513/04; A61K31/542; A61P25/28
Domestic Patent References:
WO2012098461A12012-07-26
WO2012098213A12012-07-26
Foreign References:
EP2233474A12010-09-29
US8158620B22012-04-17
US8338407B22012-12-25
Other References:
DANIEL OEHLRICH ET AL: "The evolution of amidine-based brain penetrant BACE1 inhibitors", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 24, no. 9, 20 March 2014 (2014-03-20), AMSTERDAM, NL, pages 2033 - 2045, XP055233028, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2014.03.025
D. OEHLRICH, BIOORG. MED. CHEM. LETT., vol. 24, 2014, pages 2033 - 2045
L. ROCHIN ET AL., PROE. NATL. ACAD. SCI. USA, vol. 110, no. 26, 2013, pages 10658 - 10663
MORRIS ET AL., ARCH. NEUROL., vol. 58, 2001, pages 397 - 405
PETERSEN, ARCH. NEUROL., vol. 56, 1999, pages 303 - 308
"Remington: The Science and Practice of Pharmacy", 2006, LIPPINCOTT, WILLIAMS & WILKINS
PETER G.M. WUTS; THEODORA W. GREENE: "Greene's Protective Groups in Organic Synthesis", 2007, JOHN WILEY AND SONS, INC.
J. JACQUES: "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
BASTIN, R.J. ET AL., SALT SELECTION AND OPTIMIZATION PROCEDURES FOR PHARMACEUTICAL NEW CHEMICAL ENTITIES
JACS, vol. 121, 1999, pages 8649
NATIONAL FORMULARY, vol. 18, 1995, pages 1843 - 1844
VASSAR ET AL., SCIENCE, vol. 286, 1999, pages 735 - 742
YANG, J. NEUROEHEMISTRY, vol. 91, no. 6, 2004, pages 1249 - 59
YANG, J. NEUROCHEMISTRY, vol. 91, no. 6, 2004, pages 1249 - 59
VASSER ET AL., SCIENCE, vol. 286, 1999, pages 735 - 741
MAY ET AL., JOURNAL OF NEUROSCIENCE, vol. 31, 2011, pages 16507 - 16516
ESTERHAZY ET AL., CELL METABOLISM, vol. 14, 2011, pages 365 - 377
JOHNSON-WOOD ET AL., PROC. NATL. ACA . SCI. USA, vol. 94, 1997, pages 1550 - 1555
GAMES ET AL., NATURE, vol. 373, 1995, pages 523 - 527
Attorney, Agent or Firm:
LENTZ, Nelsen L. et al. (P.O. Box 6288Indianapolis, Indiana, US)
Download PDF:
Claims:
WE CLAIM:

1. A compound of the formula:

wherein R is methyl, ethyl, or cyclopropyl;

or a pharmaceutically acceptable salt thereof.

2. The compound or salt thereof according to claim 1 wherein R is methyl.

3. The compound or salt thereof according to either claim 1 or claim 2 wherein the compound is:

The compound or salt thereof according to any one of claims 1 -3 wherein compound is:

5. The compound accordin to any one of claims 1-4 which is:

6. The salt acc rding to any one of claims 1-4 which is:

7. The salt accordin to any one of claims 1-4 which is:

8. The compound accordin to any one of claims 1 -4 which is:

9. The compound or salt thereof according to any one of claims 1 -4 wherein the compound is N-[3-[(4aS,5S,7aS)-2-amino-5-(Ll-difluoroethyl)-4,4a,5,7- tetrahydrofuro [3 ,4-d] [ 1 ,3 ]thiazin-7a-yl]-4-fluoro-phenyl]-5 -( 1 ,2,4-triazol - 1 - yl)pyrazine-2-carboxamide.

10. The compound according to claim 5 which is -[3-[(4aS,5S,7aS)-2-amino-5- (1 ,1 -difluoroethy3)-4,4a,5,7-tetrahydrofuro[3,4-d] [1 ,3]thiazin-7a-yl]-4-fluoro- phenyl]-5-(l,2,4-triazol-l-yl)pyrazine-2-carboxamide.

11. The compound according to any one of claims 1-4 which is N-[3- [(4aS,5 S,7aS)-2-amino-5 -( 1 , 1 -difluoroethyl)-4,4a,5 ,7-tetrahydrofuro [3 A- d] [ 1 ,3] thiazin-7a-yl]-4-fluoro-phenyl] -5 -( 1 ,2,4-triazol- 1 -yl)pyrazine-2- carboxamide hydrate.

12. The salt according to any one of claims 1-4 which is N-[3-[(4aS,5S,7aS)-2- amino-5-( 1 , 1 -difluoroethyl)-4,4a,5,7-tetrahydrofuro[3,4-d] [ 1 ,3]thiazin-7a-yl]- 4-fluoro-phenyl]-5-(l,2,4-triazo]-l-yl)pyrazme-2-carboxamide 4- methylbenzenesulfonate.

13. The salt according to any one of claims 1-4 which is N-[3-[(4aS,5S,7aS)-2- amino-5-( 1 , 1 -difluoroethyl)-4,4a,5,7-tetrahydrofuro[3,4-d] [ 1 ,3]thiazin-7a-yl]- 4-fluoro-phenyl]-5-(l,2,4-triazol-l -y])pyrazme-2-carboxamide malonate.

14. 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 1-13, or a pharmaceutically acceptable salt thereof.

15. 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- 13, or a pharmaceutically acceptable salt thereof.

16. A compound or pharmaceuticaliy acceptable salt thereof according to any one of claims 1-13 for use in therapy.

17. A compound or pharmaceutically acceptable salt thereof according to any one of claims 1-13 for use in the treatment of Alzheimer's disease.

18. A compound or pharmaceutically acceptable salt thereof according to any one of claims 1-13 for use in treating the progression of mild cognitive impairment to Alzheimer's disease.

19. A pharmaceutical composition, comprising a compound or a pharmaceutically acceptable salt thereof according to any one of claims 1-13 with one or more pharmaceutically acceptable carriers, diluents, or excipients.

20. A process for preparing a pharmaceutical composition, comprising admixing a compound or a pharmaceutically acceptable salt thereof according to any one of claims 1-13 with one or more pharmaceutically acceptable carriers, diluents, or excipients.

Description:
SELECTIVE BACE1 INHIBITORS

The present invention relates to certain novel selective BACE1 inhibitors, 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 and other diseases and disorders involving amyloid β (Abeta) peptide, a neurotoxic and highly aggregatory peptide segment of the amyloid precursor protein (APP), 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 rather than halting, slowing, or reversing the disease, there is a significant unmet need in the treatment of Alzheimer's disease.

Alzheimer's disease is characterized by the generation, aggregation, and deposition of Abeta in the brain. Complete or partial inhibition of β-secretase (β-site amyloid precursor protein-cleaving enzyme; BACE) has been shown to have a significant effect on plaque-related and plaque-dependent pathologies in mouse models suggesting that even small reductions in Abeta peptide levels might result in a long-term significant reduction in plaque burden and synaptic deficits, thus providing significant therapeutic benefits, particularly in the treatment of Alzheimer's disease. In addition, two homologs of BACE have been identified which are referred to as BACEl and BACE2, and it is believed that BACEl is the most clinically important to development of Alzheimer's disease. BACEl is mainly expressed in the neurons while BACE2 has been shown to be expressed primarily in the periphery. (See D. Oehlrtch, Bioorg. Med. Chem. Lett., 24, 2033-2045 (2014)) In addition, BACE2 may be important to pigmentation as it has been identified as playing a role in the processing of pigment cell-specific melanocyte protein (See L. Rochin, ei al, Proc. Natl. Acad. Sci. USA, 110(26), 10658-10663 (2013)). BACE inhibitors with central nervous system (CNS) penetration, particularly inhibitors that are selective for BACEl over BACE2 are desired to provide treatments for Abeta peptide- mediated disorders, such as Alzheimer's disease.

United States Patent No. 8,158,620 discloses fused aminodihydrothiazine derivatives which possess BACEl inhibitory activity and are further disclosed as useful therapeutic agents for a neurodegenerative disease caused by Αβ peptide, such as Alzheimer's type dementia. In addition, United States Patent No. 8,338,407 discloses certain fused aminodihydrothiazine derivatives having BACEl inhibitory effect useful in treating certain neurodegenerative diseases, such as Alzheimer-type dementia.

The present invention provides certain novel compounds that are inhibitors of BACE, In addition, the present invention provides certain novel compounds that are selective inhibitors of BACEl over BACE2. Furthermore, the present invention provides certain novel compounds which penetrate the CNS. The present invention also provides certain novel compounds which have the potential for an improved side-effect profile, for example through selective inhibition of BACEl over BACE2.

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

Formula I

or a pharmaceutically acceptable salt thereof.

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

or a pharmaceutically acceptable salt thereof. The present invention further provides a compound of Formula II:

Formula II

wherein R is methyl, ethyl, or cyclopropyl;

or a pharmaceutically acceptable salt thereof.

In addition, the resent invention provides a compound of Formula Ha:

Formula Ila

or a pharmaceutically acceptable salt thereof.

The present invention also provides 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 Formulas I, la, II, or Ha, 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, comprising administering to a patient in need of such treatment an effective amount of a compound of Formulas I, la, II, or Ila, or a pharmaceutically acceptable salt thereof. The present invention also provides a method of inhibiting BACE in a patient, comprising administering to a patient in need of such treatment an effective amount of a compound of Formulas I, la, II, Ila, or a pharmaceutically acceptable salt thereof. The present invention also provides a method for inhibiting BACE -mediated cleavage of amyloid precursor protein, comprising admimstenng to a patient in need of such treatment an effective amount of a compound of Formulas L la, ii, or Ila, or a pharmaceutically acceptable salt thereof. The invention further provides a method for inhibiting production of Abeta peptide, comprising administering to a patient in need of such treatment an effective amount of a compound of Formulas 1, la, II, or Ila, or a pharmaceutically acceptable salt thereof.

Furthermore, this in vention provides a compound of Formulas I, la, II, or Ila, or a pharmaceutically acceptable salt thereof for use in therapy, in particular for the treatment of Alzheimer's disease or for the treatment of the progression of mild cognitive impairment to Alzheimer's disease. Even furthermore, this invention provides the use of a compound of Formulas I, la, II, or Ila, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of Alzheimer's disease.

The invention further provides a pharmaceutical composition, comprising a compound of Formulas I, la, II, or Ila, 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, la, II, or Ila, 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, la, II, and ila.

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. (Morris, et al. Arch. Neurol, 58, 397-405 (2001); Petersen, el al, Arch.

Neurol, 56, 303-308 (1999)). The term "treating 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.

The term "inhibition of production of Abeta peptide" is taken to mean decreasing of in vivo levels of Abeta peptide in a patient.

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 the attending diagnostician, as 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 by the attending diagnostician, 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 0.01 to about 20 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 (D.B. Troy, Editor, 21st Edition, Lippincott, Williams & Wilkins, 2006).

The compounds of Formulas I, la, II, or Ha, or pharmaceutically acceptable salts thereof are particularly useful in the treatment methods of the invention, but certain groups, substituents, and configurations are preferred. The following paragraphs describe such preferred groups, substituents, and configurations. It will be understood that these preferences are applicable both to the treatment methods and to the new compounds of the invention. Thus, the compound of Formulas I and II wherein the fused bicyclic ring is in the CIS configuration, or pharmaceutically acceptable salt thereof, is preferred. For example, one of ordinary skill in the art will appreciate that the compound of Formula la is in the CIS relative configuration for the centers labeled 1 and 2 as shown in Scheme A below. In addition, the compound of Formula la is comprised of a core that contains three chiral centers at the carbon atoms labeled 1, 2, and 3. it is understood by one of skill in the art, that the numbering system used in Scheme A may not correspond to the numbering system used for naming compounds herein, and as such, the numbering system used in Scheme A is for illustrative purposes. The preferred relative configuration for the three chiral centers of Formula la is shown in Scheme A:

Scheme A

Further compounds of the present invention are:

N-[3-[(4aS,7aS)-2-arnmo-5-(l,l-difluoroethyl)-4,4a,5 ^

d] [ 1 ,3]thiazin-7a-yl]-4-fluoro-phenyl]-5-( 1 ,2,4-triazol-l -yl)pyrazine-2-carboxamide;

N-[3-[(4aSR,5SR,7aSR)-2-amino-5-(l,l-difluoroethyl)-4,4a,5,7 - tetrahydrofuro [3 ,4-d] [ 1 ,3]thiazin-7a-yi] -4-fluoro-phenyl] -5-( 1 ,2,4-triazol- 1 -yl)pyrazine- 2-carboxamide, and pharmaceutically acceptable salts thereof.

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 especially preferred:

N~[3~[(4aS,5S,7aS)-2-amino~5~(l ,l-dif]uoroethyl)-4,4a,5,7-te1xahydro&ro[3,4- d] [ 1 ,3]thiazin-7a-yl]-4-fluoro-phenyl]-5-(l ,2,4-triazol- 1 -yl)pyrazine-2-carboxamide, and the pharmaceutically acceptable salts thereof; and

N- [3 - [(4aS,5 S,7aS)-2-amino-5 -( 1 , 1 -di fluoroethyl)-4,4a,5,7-tetrahydrofuro [3 ,4- d] [ 1 ,3]thiazin-7a-yl]-4-fluoro-phenyl]-5-(l ,2,4-triazol- 1 -yl)pyrazine-2-carboxamide hydrate.

In addition, N- [3 - [(4aS,5 S,7aS)-2-amino-5 -( 1 , 1 -di fluoroethyl)-4,4a,5 ,7- tetrahydrofuro [3 ,4-d] [ 1 ,3]thiazin-7a-yl] -4-fluoro-phenyl] -5-( 1 ,2,4-triazol-l -yl)pyrazine- 2-carboxamide is particularly preferred.

Furthermore, N-[3-[(4aS,5S,7aS)-2-amino-5-(l,l-difluoroethyl)-4,4a,5,7- tetrahydrofuro [3 ,4-d] [ 1 ,3]thiazin-7a-yl] -4-fluoro-pheny 1] -5-( 1 ,2,4-triazol- 1 -yl)pyrazine- 2-carboxamide malonate; and N-[3-[(4aS,5S,7aS)-2-amrao-5-(l,l-difluoroetiiy])-4,4a,5,7 elTahydrofuro[3,4- d] [ 1 ,3] hiazin-7a-yl]-4-fluoro-phenyl]-5-(l ,2,4-triazol- 1 -yl)pyrazine-2-carboxamide 4- methylbenzenesuifonate are especially preferred.

One of ordinar skill in the art will appreciate that compounds of the invention can exist in tautomeric forms, as depicted below in Scheme B. When any reference in this application to one of the specific tautomers of the compounds of the invention is given, it is understood to encompass both tautomeric forms and all mixtures thereof.

Scheme B

Additionally, certain intermediates described in the following preparations may contain one or more nitrogen or oxygen protecting groups. It is understood that protecting groups may be varied as appreciated by one of skill in the art depending on the particular- reaction conditions and the particular- transformations to be performed. The protection and deprotection conditions are well known to the skilled artisan and are described in the literature (See for example "Greene 's Protective Groups in Organic Synthesis", Fourth Edition, by Peter G.M, Wuts and Theodora W. Greene, John Wiley and Sons, Inc. 2007).

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, el ah, "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, such as a hydrochloride salt, can be formed, for example, by reaction of an appropriate free base of a compound of the invention, an appropriate pharmaceutically acceptable acid such as hydrochloric acid in a suitable solvent such as diethyl ether under standard conditions well known in the art. Additionally, the fonnation of such salts can occur simultaneously upon deproteetion of a nitrogen protecting group. The formation of such salts is well known and appreciated in the art. See, for example, Gould, P.L., "Salt selection for basic drags," 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 ah, "Pharmaceutical Salts," Journal of Pharmaceutical Sciences, 66: 1-19, (1977).

Certain abbreviations are defined as follows: "APP" refers to amyloid precursor protein; "BSA" refers to Bovine Serum Albumin; "CD!" refers to Ι,Γ- carbonyldiimidazole; "cDNA" refers to complementary deoxyribonucleic acid; "DCC" refers to 1,3-dicyclohexylcarbodiimide; "Deoxo-Fluor® " refers to bis(2- methoxyethyl)aminosulfur trifluoride; "DIG" refers to 1,3-diisopropylcarbodiimide; "DMAP" refers to 4-dimethylaminopyridine; "DMEM" refers to Dulbecco's Modified Eagle's Medium; "DMSO" refers to dimethyl sulfoxide; "EBSS" refers to Earle's

Balances Salt Solution; "EDCI" refers to 1 -(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride; "ELISA" refers to enzyme-linked immunosorbent assay; "F12" refers to Ham's F12 medium; "FBS" refers to Fetal Bovine Serum; "Fc" refers to fragment crystal lizable; "FLUOLEAD™" refers to 4-tert-butyl-2,6-dimethylphenylsu3fur trifluoride; "FRET" refers to fluorescence resonance energy transfer; "HATU" refers to (dimethylamino)-N,N-dime^

hexafluorophosphate; "HBTU" refers to (lH-benzotriazo3-l-y3oxy)(dimethylamino)-N,N- dimethylmethaniminium hexafluorophosphate; "HEK" refers to human embryonic kidney; "HF-pyridine" refers to hydrogen fluoride pyridine or Olah's reagent or

poly(pyridine fluoride); "HO At" refers to l-hydroxy-7-azabenzotriazole; "HOBT" refers to 1 -hydroxy lbenzotriazole hydrate; "IC50" refers to the concentration of an agent that produces 50% of the maximal inhibitory response possible for that agent; "IgG " refers to immunoglobulin-like domain Fc-gamma receptor; "MEM" refers to Minimum Essential Medium; "PBS" refers to phosphate buffered saline; "PDAPP" refers to platelet derived amyloid precursor protein; "Ph" refers to a phenyl group; "PyBOP" refers to

(benzolriazol-l-yl-oxytripyrrolidinophosphonivirn hexafluorophosphate); "PyBrOP" refers to bromo-tris-pyrrolidino phosphoniumhexafluorophosphate; "RFU" refers to relative fluorescence unit; "RT-PCR" refers to reverse transcription polymerase chain reaction; "SDS-PAGE" refers to sodium dodecy] sulfate polyacrylamide gel

electrophoresis; "SCX" refers to strong cation exchange; "SFC" refers to super critical chromatography;" T3P®" refers to propylphosphonic anhydride; "THF" refers to tetrahydrofuran; "TEMPO" refers to (2,2,6,6-tetramethyl-piperidin- 1 -yl)oxyl; "TMEM" refers to transmembrane protein; "trityl" refers to a group of the formula "(PhbO;

"XtalFluor-E® or DAST difluorosulfinium salt" refers to

(diethylamino)difluorosulfonium tetrafluoroborate or N,N-diethy 1-5,5- difluorosulfiliminium tetrafluoroborate; and "XtalFluor-M® or morpho-DAST

difluorosulfinium salt" refers to tetrafluoroborate or difluoro-4-mo holinylsulfonium tetrafluoroborate.

It is understood by one of ordinary skill in the art that the terms "tosylate",

"toluenesulfonic acid", "p-toluenesulfonic acid", and "4-methylbenzene sulfonic acid" refer to the compound of the following structure;

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 ait, 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. The following schemes, preparations, and examples further illustrate the invention.

In Scheme I, step A, trimethylsulfonium iodide is treated with an organic base such as n-butyllithium at a temperature of about -50 °C in a solvent such as THF. A protected oxymethyl oxirane, protected with a suitable protecting group, such as a trityl group, is then added to the basic solution at -10 °C and allowed to stir for about 2 hours to give the protected product of Scheme 1, Step A. "PG" is a protecting group developed for the amino group or oxygen group such as carbamates, amides, or ethers. Such protecting groups are well known and appreciated in the art. The protected product of Step A is reacted with an a-haloester such as tert-butoxy bromoacetate using tetra-N- butylammonium sulfate or other quaternary ammonium salt phase transfer catalysts in a solvent such as toluene and an aqueous inorganic base such as sodium hydroxide at about room temperature to give the compound of Scheme 1, Step B. S ch alkylations are well know in the art. Alternatively a base such as 60% sodium hydride in oil with solvents such as N,N-dimethylformamide or THF and a temperature range of 0 to 100 °C can be used to give the protected product of Step B. The fert-butoxy carbonyl acetate is converted to an oxime over a 2-step procedure. A reducing agent such as

isobutvlaluminum hydride in hexanes is added dropwise at a temperature of about -70 °C followed by the dropwise addition of an aqueous acid such as hydrochloric acid at a temperature of about -60 °C. The work-up is accomplished with an organic extraction to give the intermediate material. This material is dissolved in an organic solvent such as dichloromethane and treated with sodium acetate followed by hydroxylamine hydrochloride to give the oxime product of Step C. The oxime product of Scheme 1, Step C can be converted to the bicyclic 4,5-dihydroisoxazole product of Step D in a 3+2 cyclization by several methods such as using an aqueous solution of sodium hypochlorite or an alternative oxidant such as N-chlorosuccinimide and in a solvent such as tert-butyl methyl ether, toluene, dichloromethane, or xylene at a temperature of about 10-15 °C or with heating. The 2-fluoro, 5-bromo phenyl group ca be added to the dihydroisoxazole by generating the organometallic reagent. The organometallic reagent can be generated from 4-bromo- 1 -fluoro-2-iodo-benzene using halogen-metal exchange with reagents such as n-butyllithium or isopropylmagnesium chloride lithium chloride complex and dropwise addition at a temperature range of about -78 °C to 15 °C in a solvent such as THF. A Lewis acid such as boron trifluoride diethyl etherate is then added to give the product of Scheme 1, Step E. The resulting bicyclic tetrahydroisoxazole can be treated with zinc in acetic acid to form the ring opened product of Scheme 1, Step F. A alternate method to open the isoxazole ring uses Raney Nickel in a polar solvent such as ethanol under pressure with hydrogenation conditions. The product of Step F can then be reacted with benzoyl isothiocyanate in a solvent such as dichloromethane or THF at a temperature of about 5 °C to room temperature to give the thiourea compound of Step G. The thiazine ring can be formed using trifluoromethanesuifoiiic anhydride and an organic base such as pyridine in a solven t such as dichloromethane at a temperature of about -20 °C to give the product of Step H. The hydroxymethyl protecting group suc as a trityl group can be removed in Scheme 1, Step I using methods well known in the art such as formic acid at room temperature or with >-toluenesulfonic acid monohydrate in solvents such as dichloromethane and methanol to give the compound of Step Ϊ, The hydroxy methyl ca be oxidized to the carboxylic acid using oxidizing agents suc as 2-iodoxybenzoic acid (1BX) at temperatures of 0-22 °C in a solvent such as DMSO or addition of

(diacetoxyiodo)benzene portionwise or all at once in a solvent such as acetonitrile or acetonitrile and water with stirring at a temperature of about 5-25 °C to give the compound of Scheme 1, Step J. TEMPO can also be used as a catalyst in the oxidation. The Weinreb amide is prepared in Scheme 1, Step from the acid product of Step J with the addition of Ν,Ο-dimethylhydroxylamine hydrochloride, an organic base, such as txiethylamme, and a coupling reagent such as HATU. The mixture is stirred at room temperature to give the product of Step K. Other coupling agents that could be used include GDI, carbodiimides such as DCC, DIG, or EDO or other uronium or

phosphonium salts of non-nucleophilic anions, such as HBTU, PyBOP, and PyBrOP. The Weinreb amide is then converted to the ketone using an organometallic reagent such as a Grignard reagent or an organolithium reagent in Step L in a solvent such as THF. The appropriate Grignard reagent can be added as a solution in solvents such as ether or 2-methyltetrahydrofuran to the Wemreb amide at a temperature of about -78 °C to 0 °C to give the ketone of Step L. In Scheme 1, Step M, the R and ketone group of the compound of Step L can be converted to a difluoro-R group using Deoxo-Fluor® in a solvent such as dichloromethane at about -78 °C to room temperature. Another alternative procedure involves pre-mixing the fluorinating reagent such as Deoxo-Fluor® with boron trifluoride-diethyl etherate followed by the addition of the product of Scheme 1, Step L and iriethylamine trihydro fluoride to give the product of Scheme 1, Step M.

Alternatively, other fluorinating agents that may be used which are well known in the art are, diethyiaminosulfur trifluoride (also referred to as "DAST"), XtalFiuor-E® or Xta2F3uor-M® with an additive such as triethylamine trihydrofluoride or FLUOLEAD™ using an additive such as HF-pyridine. The 5-bromo of the phenyl is converted to the amine using (lR,2R)-N,N'-dimethyl-l,2-cyclohexanediarnine or trans- N,N'dimethylcyclohexane-l,2 -diamine in a solvent such as ethanol and adding sodium azide followed by sodium L-ascorbate and cupric sulfate. The reaction is heated to about 80-100 °C for several hours and then worked up with an extraction using a solvent such as ethyl acetate. The intermediate is then reduced under hydrogenation conditions using palladium on carbon such as 5-10% palladium in solvents such as methanol or ethanol and THF at a pressure of about 40-50 psi of hydrogen to give the aniline product of Scheme 1, Step N.

Scheme 2

Alternatively in Scheme 2, the protected product of Scheme 1, Step A, can be treated with 4-(2-chloroacetyl)morpholino and a base such as tetrabutyl ammonium hydrogen sulfate in a solvent such as toluene at a temperature of about 5 °C to give the product of Scheme 2, Step A. The morpholino group can then serve as a leaving group in Scheme 2, Step B. For example, the product of Scheme 2, Step A can be treated with the appropriate Grignard reagent which can be prepared in situ from isopropyl magnesium chloride lithium chloride complex and 4-bromo-l-fluoro-2-iodobenzene or if the appropriate Grignard reagent is available, the reagent can be added directly to the product of Scheme 2, Step A at a temperatixre of about 5 °C to give the product of Scheme 2, Step B. The carbonyi acetate can be converted to an oxime with hydroxy lamine hydrochloride and sodium acetate with heating to about 50 °C to give the product of Scheme 2, Step C. The oxime product of Scheme 2, Step C can then be converted to the product of Scheme

2, Step D (the same product as Scheme 1, Step E) using hydroquinone in a solvent such as toluene and heating to refl x. The amine product of Scheme 2, Step D can be acylated with acetyl chloride using an organic base such as DMAP and pyridine in a solvent such as dichloromethane at a temperature of about 0-5 °C to give the product of Scheme 2, Step E. The product of Scheme 2, Step E can then be converted to the product of Scheme

3, Step A as discussed below.

Scheme 3

In an alternate route, as described in Scheme 3, the isoxazole nitrogen of the compound of Scheme 1 , Step E, is protected with an acetyl group and the protecting group of the hydroxy methyl is removed in a two-step procedure. For example, the tetrahydroisoxazole is treated with an organic base such as DMAP and pyridine in a solvent such as dichloromethane and acetyl chloride is added. The temperature is maintained below about 10 °C and then allowed to stir at about room temperature. The reaction is diluted with water and extracted with a solvent such as dichloromethane. The organic extracts are washed with an aqueous acid such as I N hydrochloric acid and the aqueous extracted again with a solvent such as dichloromethane followed by an aqueous wash. The organic solvent can be partially removed and an acid such as formic acid or p- toluenesulfonic acid monohydrate in solvents such as dichloromethane and methanol can added to deprotect the hydroxy methyl. The mixture can be stirred at room temperature or heated to a temperature of about 40 °C until deprotection of the hydroxy is complete to give the compound of Scheme 3, Step A. The hydroxy methyl product of Scheme 3, Step A can be oxidized to the carboxylic acid product of Scheme 3, Step B in a manner analogous to the procedure described in Scheme 1, Step J, and the Weinreb amide can be further prepared in a manner analogous to the procedure described in Scheme 1 , Step using a coupling agent such as CDI in a portionwise addition or adding at once with a solvent such as dichloromethane, cooling to -20 °C and stirring for about 1 hour and adding ,0-dimethylhydroxylamine hydrochloride portionwise or all at once. An organic base such as triethylamine can also be used to promote the reaction. Further additions of CDI and Ν,Ο-dimethylhydroxylamine can be added until complete reaction is observed to give the Weinreb amide product of Scheme 3, Step C. The ketone of Scheme 3, Step D can be formed from the Weinreb amide in a manner analogous to the procedure described in Scheme 1 , Step L. The ketone of Step D can be converted to a difluoro-R group in a manner analogous to the procedure described in Scheme 1 , Step M to give the product of Scheme 3, Step E. The acetyl tetrahydroisoxazole can deprotected under acidic conditions well known in the art such as using hydrochloric acid and heating to about 100 °C to give the product of Scheme 3, Step F. The bicyclic tetrahydroisoxazole can be treated with zinc in acetic acid to form the ring opened product of Scheme 3, Step G in a manner analogous to the procedure described in Scheme I , Step F. The thiazine product of Scheme 3, Step H can be prepared in a one pot 2 step reaction using benzoyl isothiocyanate in a manner analogous to the procedure described in Scheme 1 , Step G. The mixture is evaporated to a residue and cyclohexane is added. The mixture is heated to about 60 °C and methyl fert-buty] ether is added to dissolve the residue. The solution is filtered and concentrated to dryness. The thiazine ring can then be formed in a manner analogous to the procedure described in Scheme 1 , Step H to give the product of Scheme 3, Step H. Scheme 4

In Scheme 4, Step A, the aniline product of Scheme 1, Step N can be coupled with a heteroaromatic carboxvlic acid utilizing coupling conditions well known in the art. One skilled in the art will recognize that there are a number of methods and reagents for amide formation resulting from the reaction of carboxylic acids and amines. For example, the reaction of an appropriate aniline with an appropriate acid in the presence of a coupling reagent and an amine base such as diisopropylethylamine or triethylamine, will give a compound of Scheme 4, Step A. Coupling reagents include carbodiimides such as DCC, DIG, EDCI, and aromatic oximes such as HOBt and HO At. Additionally, uronium or phosphonium salts of non-nucleophilic anions such as HBTU, HATU, PyBOP, and PyBrOP or a cyclic phosphoric anhydride such as T3P® can be used in place of the more traditional coupling reagents. Additives such as DMAP may be used to enhance the reaction. Alternatively, the aniline amine can be acylated using substituted benzoyl chlorides in the presence of a base such as triethylamine or pyridine. In Scheme 4, Step B, the protected thiazine amine can then be deprotected with an organic base such as pyridine and O-methylhydroxylamine hydrochloride in solvents such as THF and ethanoi and an organic base such as pyridine to provide the compound of Formula II.

Alternatively an inorganic base such as lithium hydroxide in methanol may be deprotect the thiazine to provide the compound of Formula II.

Scheme 5

Π

Alternatively, in Scheme 5, the bromide product of Scheme 1, Step M is converted to a protected aniline using trifluoroacetamide, copper iodide, a diamine such as trans, racemic-N,N'-dimethyl-l,2-cyclohexane diamine, an inorganic base such as potassium carbonate, and sodium iodide with heating to about 100-130 °C to give the protected aniline product of Scheme 5, Step A. The protected aniline and thiazine amine can then be deprotected stepwise. The trifluoroacetamide can be hydrolyzed using a base such as 7 N ammonia in methanol to give an aniline and protected thiazme, the same product of Scheme 1, Step N. The thiazine can then be deprotected under conditions well known in the art and described in Scheme 4, Step B using O-methylhydroxylamine hydrochloride in a solvent such as ethanol and THF with an organic base such as pyridine followed by heating to about 55 °C or stirring at room temperature followed by concentration and purification to give the product of Scheme 5, Step B. Alternatively, the order of deprotection could be reversed with the thiazine deprotected first and the aniline

deprotected last. In Scheme 5, Step C, the aniline product of Step B can then be reacted with the appropriate carboxylic acid or acid chloride as described in Scheme 4, Step A to give the products of Formula II,

A pharmaceutically acceptable salt of the compounds of the invention, such as a hydrochloride salt, can be formed, for example, by reaction of an appropriate free base of Formulas I, la, II, or Ha and an appropriate pharmaceutically acceptable acid such as hydrochloric acid, p-toluenesulfonic acid, or malonic acid in a suitable solvent such as diethyl ether under standard conditions well known in the art. Additionally, the formation of such salts can occur simultaneously upon deprotection of a nitrogen protecting group. 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,"

The following preparations and examples further illustrate the invention.

Preparation 1

(2S - 1 -Trityloxybut-3-en-2-ol

Scheme 1 , step A: Stir trimethylsulfonium iodide (193.5 g, 948.2 mmol) in THF (1264 mL) at ambient temperature for 75 minutes. Cool mixture to -50 °C and add n- butyllithium (2.5 mol/L in hexanes, 379 mL, 948.2 mmol) via cannula, over a period of 30 minutes. Allow the reaction to gradually warm to -30 °C and stir for 60 minutes. Add (2S)-2-trityloxymethyl oxirane (100 g, 316.1 mmol) portion wise, keeping the temperature below -10 °C. After the complete addition, allow the reaction mixture to warm to room temperature and stir for 2 hours. Pour the reaction into saturated ammonium chloride, separate the phases, and extract the aqueous phase with ethyl acetate. Combine the organic layers and dry over magnesium sulfate. Filter and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with methyl t-butyl ether: hexanes (10-15% gradient), to give the title compound (56.22 g, 54%). ES/MS m/z 353 (M+Na).

Alternate Preparation 1

(2S)- 1 -Trityloxybut-3-en-2-ol

Scheme 2, step A starting material: Add triphenylmethyl chloride (287 g, 947.1 mmol), DMAP (7.71 g, 63.1 mmol) and triethylamine (140 g, 1383.5 mmol) to a solution of (2S)-but-2-ene-l,2-diol (prepared as in JACS, 1999, 121, 8649) (64.5 g, 631 mmol) in dichloromethane (850 mL). Stir for 24 hours at 24 °C. Add 1 N aqueous citric acid (425 ml,). Separate the layers and concentrate the organic extract under reduced pressure to dryness. Add methanol (900 mL) and cool to 5 °C for 1 hour. Collect the solids by filtration and wash with 5 °C methanol (50 mL). Discard the solids and concentrate the mother liquor under reduced pressure to dryness. Add toluene (800 mL) and concentrate to a mass of 268 g to obtain the title compound (129 g, 67%) in a 48 wt% solution of toluene.

Preparation 2

1 -Morpholino-2-[(l S)-l -(trityloxymethyl)allyioxy]ethanone

Scheme 2, step A: Add tetrabutyl ammonium hydrogen sulfate (83.2 g, 245.0 mmol) and 4-(2-chloroacetyl)moφholine (638.50 g, 3902.7 mmol) to a solution of 1- trityloxybut-3-en-2-o2 ( 832.4 , 2519 mmol) in toluene (5800 mL) that is between 0 and 5 °C. Add sodium hydroxide (1008.0 g, 25202 mmol) in water (1041 mL). Stir for 19 hours between 0 and 5 °C. Add water (2500 mL) and toluene (2500 mL). Separate the layers and wash the organic extract with water (2 χ 3500 mL), Concentrate the organic extract under reduced pressure to dryness. Add toluene (2500 mL) to the residue and then add n-heptane (7500 mL) slowly. Stir for 16 hours. Collect the resulting solids by filtration and wash with n-heptane (1200 mL). Dry the solid under vacuum to obtain the title compound (1075.7 g, 98%),

Preparation 3

1 -(5-Bromo-2-fluoro-phenyl)-2-[(l S)-l -(trityloxymethyl)ailyloxy]ethanone

Scheme 2, step B: Add a 1.3 M solution of isopropyl magnesium chloride lithium chloride complex (3079 mL, 2000 mmol) in THF to a solution of 4-bromo-l-fluoro-2- iodobenze (673.2 g, 2237.5 mmol) in toluene (2500 mL) at a rate to maintain the reaction temperature below 5 °C. Stir for 1 hour. Add the resulting Grignard solution (5150 mL) to a solution of l-mo holmo-2-[(lS)-l-(tτitylo ymethyl)allyloxy]ethanone (500 g, 1093 mmol) in toluene (5000 mL) at a rate to maintain the reaction temperature below 5 °C. Stir for 3 hours maintaining the temperature below 5 °C. Add additional prepared Grignard solution (429 mL) and stir for 1 hour. Add a 1 N aqueous citric acid solution (5000 mL) at a rate to maintain the temperature below 5 °C. Separate the layers and wash the organic extract with water (5000 mL). Concentrate the solution under reduced pressure to dryness. Add methanol (2000 mL) to the residue and concentrate to give the title compound as a residue (793 g, 73.4% potency, 83%).

Preparation 4

1 -(5 -Bromo-2-fluoro-phenyl)-2- [( 1 S)- 1 -(trityloxymethyl)allyloxy]ethanone oxime

Scheme 2, step C: Add hydroxylamine hydrochloride (98.3 g) to l -(5-bromo-2- fluoro-phenyl)-2-[(l S)-l -(trityloxymethyl)allyloxy]ethanone (450 g, 707 mmol) and sodium acetate (174 g) in methanol (3800 mL). Heat the solution to 50 °C for 2 hours. Cool to 24 °C and concentrate. Add water (1000 mL) and toluene (1500 mL) to the residue. Separate the layers and extract the aqueous phase with toluene (500 mL).

Combine the organic extract and wash with water (2 χ 400 mL). Concentrate the solution under reduced pressure to give the title compound as a residue (567 g, 61.4% potency, 88%).

Preparation 5

tert-Butyl 2~[( 1 S)~ 1 -(trityloxymethyl)allyloxy]acetate

Scheme I , step B: Add (2S)-l -trityloxybut-3-en-2-ol (74.67 g, 226.0 mmol) to a solution of tetra-N-butyl ammonium sulfate (13.26 g, 22.6 mmol) in toluene (376 mL). Add sodium hydroxide (50% mass) in water (119 mL) followed by tert-butyl-2- bromoacetate (110.20 g, 565.0 mmol). Stir reaction mixture for 18 hours at ambient temperature. Pour into water, separate the phases, and extract the aqueous phase with ethyl acetate. Combine the organic layers and dry over magnesium sulfate. Filter the mixture and concentrate under reduced pressure to give the title compound (77.86 g, 77%). ES/MS m/z 467 (M+Na). Preparation 6

(lE)-2-[(lS)-l-(Trityloxymethyl)allyloxy]acetaldehyde

H

Scheme 1, step C: Cool a solution of teri-butyi 2-[(lS)-l

(trityloxymethyl)allyloxy]acetate (77.66 g, 174.7 mmol) in dichioromethane (582.2 mL) to -78 °C. Add a solution of diisobutylaluminum hydride in hexanes (1 mol L, 174.7 mL) dropwise over a period of 35 minutes and maintain the temperature below -70 °C. Stir at -78 °C for 5 hours. Add hydrochloric acid in water (2 mol/L, 192.1 mL) to the reaction mixture dropwise, keeping the temperature below -60 °C. Allow the reaction to gradually warm to ambient temperature and stir for 60 minutes. Separate the organic extract and wash with saturated sodium bicarbonate. Dry the solution over magnesium sulfate, filter, and concentrate under reduced pressure to give a residue. Dissolve the residue in dichioromethane. Add sodium acetate (28.66 g, 349.3 mmol), followed by

hydroxylamine hydrochloride (18.21 g, 262.0 mmol). Stir at ambient temperature for 18 hours. Pour into water, separate the phases, and extract the aqueous phase with dichioromethane. Combine the organic layers and dry over magnesium sulfate. Filter the mixture and concentrate under reduced pressure to give the title compound (68.38 g, 101%). ES/MS m/z 386 (M-H).

Preparation 7

(3aR,4S)-4-(Tri1yloxymethyl)-3,3a,4,6-te1iuhydrofuro[3,4-c]i soxazole

Scheme 1 , step D: Cool a solution of (lE)-2-[(lS)-l- (trityloxymethyl)allyloxy]acetaldehyde oxime (55.57 g, 143.4 mmol) in tert-butyl methyl ether (717 mL) to 5 °C. Add sodium hypochlorite (5% in water, 591 mL, 430.2 mmol) dropwise, keeping the temperature below 10 °C. Stir at 10 °C for 30 minutes. Allow the reaction to warm to 15 °C. Stir at 15 °C for 18 hours. Dilute the reaction mixture with ethyl acetate and wash with saturated sodium bicarbonate. Separated the phases, wash the organic phase with a 5% sodium hydrogen sulphite solution and brine. Dry the solution over magnesium sulfate, filter, and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with 50% methyl tert- butyl ether/dichloromethane: hexanes (20-27% gradient), to give the title compound (35.84 g, 65%). ES/MS m/z 408 (M-H a).

Preparation 8

(3aR,4S,6aR)-6a-(5-Bromo-2-fluoro-phenyl)-4-(trityloxymethyl )-3,3a,4,6- tetrahydrofuro[3,4-c]isoxazole

Scheme 1, step E: Cool a solution of 4-bromo-l-fluoro-2-iodo-benzene (86.94 g, 288.9 mmol) in THF (144.5 mL) and toluene (1445 mL) to -78 °C. Add n-butyllithium (2.5 M in hexanes, 120 mL, 288.9 mmol) dropwise, keeping the temperature below -70 °C. Stir for 30 minutes at -78 °C. Add boron trifluoride diethyl etherate (36.5 mL, 288.9 mmol) dropwise, keeping temperature below -70 °C. Stir the solution for 30 minutes at - 78 °C. Add a solution of (3aR,4S)-4-(trityloxymethyl)-3,3a,4,6-tetrahydrofuro[3,4- cjisoxazole (55.69 g, 144.5 mmol) in THF (482 mL) dropwise to the reaction, over a period of 30 minutes, keeping temperature below -65 °C. Stir at -78 °C for 90 minutes. Rapidly add saturated ammonium chloride, keeping temperature below -60 °C. Pour into brine, and extract the aqueous phase with ethyl acetate. Combine the organic extract and dry over magnesium sulfate. Filter and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with 10-15% diethyl ethenhexanes (0-70% gradient), to give the title compound (36,52 g, 45%). ES/MS m/e ( 79 Br/ 81 Br) 560/562 [M+H].

Alternate Preparation 8

Scheme 2, step D: Heat a solution of l-(5-bromo-2-fluoro-phenyl)-2-[(lS)-l- (trityloxymethyl)allyloxy]ethanone oxime (458 g, 502 mmol) and hydroquinone (56.3g 511 mmol) in toluene (4000 ml.) to reflux under nitiOgen for 27 hours. Cool the solution to 24 °C and add aqueous sodium carbonate (800 mL). Separate the layers and extract the aqueous phase with toluene (300 mL). Combine the organic extract and wash with water (2 x 500 mL). Concentrate the solution under reduced pressure to give a residue. Add isopropyl alcohol (1500 mL) and heat to reflux. Cool to 24 °C and collect the solids by filtration. Dry the solid under vacuum to obtain the title compound (212 g, 75%).

Preparation 9

l-[(3aR,4S,6aS)-6a-(5-Bromo-2-fluoro-phenyl)-4-(trityloxymet hyl)-3,3a,4,6- tetrahy dro f uro [ 3 ,4-c] i soxazol - 1 -y 3] ethanone

Scheme 2, step E: Add acetyl chloride (35.56 g, 503.9 mmol) to a solution of

(3aR,4S,6aR)-6a-(5-bromo-2-fluoi -phenyl)-4-(trityioxymethyl)-3,3a,4,6- tetrahydrofuro[3,4-c]isoxazole (235.3 g, 420 mmol), DMAP (5.13 g, 42.0 mmol), and pyridine (66.45 g, 840.1 mmol) in dichloromethane (720 mL) under nitrogen, maintaining internal temperature below 5 °C. Stir for 1 hour and then add water (300 mL) and 1 M sulfuric acid (300 mL). Stir the mixture for 10 minutes and allow the layers to separate. Collect the organic extract and wash with saturated sodium carbonate (500 mL) and water (500 mL), Dry the solution over magnesium sulfate. Filter and concentrate under reduced pressure to give l-[(3aR,4S J 6aS)-6a-(5-Bromo-2-fluoro-phenyl)-4- (trityloxymethyl)-3,3a,4,6 etTahydrofuro[3,4-c]isoxazol-l-y3]ethanone (235 g, 93%) as a grey solid.

Preparation 10

~ [(3aR,4S,6aS)~6a-(5 -Bromo-2-fj uorophenyl)-4~(hydroxymethyl)tetrahydro~ 1 H,3H- furo [3 ,4-c [1,2] oxazol- 1 -y 1] ethanone

Scheme 3, step A: In a 20 L jacketed reactor add acetyl chloride (290 mL, 4075 mmol) to a solution of (3aR,4S,6aR)-6a-(5-bromo-2-fluoro-phenyl)-4-(trityloxymethyl )- 3,3a,4,6-tetrahydrofuro[3,4-c]isoxazole (1996 g, 3384 mmol), DMAP (56.0 g, 458 mmol), pyridine (500 ml,, 6180 mmol) in dichloromethane (10 L) under nitrogen maintaining internal temperature below 10 °C. After complete addition (1 hour) warm to 20 °C and stir overnight. If reaction is incomplete, add acetyl chloride, DMAP, pyridine, and dichloromethane until complete reaction is observed. Cool the reaction mixture to 0 °C and slowly add water (5 L), stir the reaction mixture at 10 °C for 30 minutes and allow the layers to separate. Collect the organic extract and wash the aqueous with

dichloromethane (1 L). Wash the combined organic extracts with 1 N aqueous hydrochloric acid (2 x 4 L), extract the aqueous with dichloromethane (2 x 1 L). Wash the combined organic extracts with water (4 L) and remove the solvent under reduced pressure give total volume of approximately 5 L. Add 90% formic acid (1800 mL) and stand at ambient temperature for 3 days. Warm to 40 °C for 2 hours then remove the solvent under reduced pressure. Dilute the residue with methanol (4 L) and slowly add saturated aqueous sodium carbonate (3 L). Add solid sodium carbonate (375 g) to adjust the pH to 8-9. Stir at 45 °C for 1 hour then cool to ambient temperature. Remove the solids by filtration, washing with methanol (4 500 mL) then treat with 2 N aqueous sodium hydroxide (100 mL) and stand at ambient temperature for 1 hour. Remove the solids by filtration, washing with methanol (2 x 100 mL). Evaporate the solvent under reduced pressure and partition the residue between ethyl acetate (5 L) and water (2 L). Extract the aqueous with ethyl acetate (2 L) and wash the combined organic extracts with brine (2 x 1 L). Remove the solvent under reduced pressure, add methyl feri-butyl ether (2.5 L) and evaporate to dryness. Add methyl tert-butyl ether (4 L) and stir at 65 °C for 1 hour cool to ambient temperature and collect the solids by filtration, washing with methyl fert-butyl ether (3 x 500 mL). Dry under vacuum to a beige solid. Heat this solid in toluene (7.5 L) to 110 °C until fully dissolved, cool to 18 °C over 1 hour, and stir at this temperature for 1 hour. Warm to 40 °C and when precipitate forms, cool to 18 °C once more. Stir for 45 minutes then collect solids by filtration, washing with toluene (2 x 500 mL). Dry the solid under vacuum to obtain the title compound (443.1 g, 36%, 95% purity by LCMS). Evaporate the filtrate under vacuum to give a residue. Purify the residue by silica gel flash chromatography, eluting with 20% to 100% ethyl acetate in isohexane. Slurry the product containing fractions in methyl tert-butyl ether (2 L) at 60 °C for 30 minutes, cool to ambient temperature, and collect the solids by filtration, washing with methyl fert- butyl ether (2 x 200 mL). Dry the solids under vacuum to give the title compound as a beige crystalline solid (304 g, 24%, 88% purity by LCMS). Evaporate the filtrate under vacuum to a residue. Purify the residue by silica gel flash chromatography, eluting with 20% to 100% ethyl acetate in isohexane to give the title compound (57.8 g, 5%, 88% purity by LCMS). ES/MS: m/z ( 79 Br/ 8l Br) 360.0/362.0 [M+H].

Alternate Preparation 10

Scheme 3, step A: Add l-[(3aR,4S,6aS)-6a-(5-bromo-2-fluoro-phenyl)-4- (trityloxymethy3)-3,3a,4,6-tetTahydrofuro[3,4-c]isoxazol-l-y l]ethanone (69 g, 1 14.5 mmol) to a 15 °C solution ofp-toluenesulfonie acid monohydrate (2.2 g, 1 1.45 mmol), dichloromethane (280 mL) and methanol (700 mL). Stir for 18 hours and then remove the solvent under reduced pressure. Dilute the residue with dichloromethane (350 mL) and add 1 M aqueous sodium carbonate (140 mL) and water (140 mL). Separate the layers and evaporate the organic layer under reduced pressure. Add toluene (350 mL) to the residue and heat to reflux for 1 hour. Cool to 10-15 °C at a rate of 10 °C/hour.

Collect the solids by filtration and wash with toluene (70 mL). Dry the solid under vacuum to obtain the title compound (30 g, 65%) as a grey solid.

Preparation 11

(3aR,4S,6aS)-l -Acetyl-6a-(5-bromo-2-fluoro-phenyl)-3,3a,4,6 etTahydrofuro

c]isoxazole-4-carboxylic acid

Scheme 3, step B: Add water (2 L) to a suspension of l-[(4S,6aS)-6a-(5-bromo-2- fluoro-phenyl)-4-(hydroxymethyiy^

(804.9 g, 2177 mmoi), TEMPO (40.0 g, 251 mmol) in acetonitrile (4.5 L) in a 20 L jacketed reactor and cool to an internal temperature of 5 °C. Add (diacetoxyiodo)benzene (1693 g, 4993.43 mmol) portionwise over 30 minutes. Control the exotherm using reactor cooling and then hold at 20 °C until LCMS shows complete reaction. Slowly add a suspension of sodium bisulfite (70 g, 672.68 mmol) in water (300 mL) at ambient temperature, maintaining the internal temperature below 25 °C. Stir for 30 minutes and then cool to 5 °C. Add water (2 L), then slowly add 47 wt% aqueous sodium hydroxide (780 mL) over a period of 1 hour maintaining the internal temperature below 10 °C. Add ethyl acetate (2 L) and isohexane (5 L), stir vigorously and separate the layers. Extract the biphasic organic layers with water (1 L) and wash the combined aqueous with methyl tert-butyl ether (2.5L). Cool the aqueous extracts to 5 °C and slowly add 37%

hydrochloric acid (1.4 L) over 30 minutes maintaining the internal temperature around 5 °C. Add ethyl acetate (5 L), separate the layers and wash the organic with brine (3 χ 1 L). Extract the combined aqueous extracts with ethyl acetate (2.5 L), wash the combined organics with brine (1 L), then dr with sodium sulfate, and filter. Dilute the organics with heptane (2.5 L) and evaporate to dryness under reduced pressure. Add methyl tert- butyl ether (1.5 L) and heptane (1.5 L) and evaporate to dryness. Add heptane (2.5 L) and evaporate to dryness twice. Add heptane (500 mL) and methyl tert-butyl ether (500 mL) and stir at 40 °C for 30 minutes then collect the precipitate by filtration, washing with heptane/methyl tert-butyl ether (1 : 1, 1 L) then methyl fenf-butyl ether (3 χ 300 mL) and air dry to give the title compound as a beige crystalline solid (779 g, 91%). ES/MS: m/z ( 79 Br/ 81 Br) 374.0/376.0 [M+H].

[a] D 20 = -19.0 ° (C=1.004, chloroform).

Alternate Preparation 11

Scheme 3, step B: Add water (150 mL) and acetonitrile (150 mL.) to l-[(4S,6aS)- 6a~(5~brQmo-2~fiuoro-pheny3)-4-(hydroxym

1-yljjethanone (30 g, 73.3 mmol), TEMPO (1.14 g, 7.30 mmol) and (diacetoxyiodo) benzene (51 ,9 g, 161 mmol). Cool to 15 °C and stir for 2 hours. Slowly add sodium thiosulfate (21 g) and potassium carbonate (22 g) in water (150 mL) at ambient temperature. Stir for 1 hour and then add methyl teri-butyl ether (150 mL). Separate the layers and adjust the pH of the aqueous layer to 2-3 with concentrated sulfuric acid. Add ethyl acetate (150 mL) and separate the layers. Evaporate the organic layer to dryness under reduced pressure. Add n-heptane (90 mL) and heat to reflux for 1 hour. Cool to 15 °C and then collect the precipitate by filtration, washing with n-heptane (90 mL). Dry under vacuum to give the title compound as a white solid (27 g, 98%).

Preparation 12

(3aR,4S,6aS)-l-Acety2-6a-(5-bromo-2-fluoropheiiyl)-N-methoxy - -methyltetrahydro- 1 H,3H-furo[3,4-c] [ 1 ,2]oxazole-4-carboxamide

Scheme 3, step C: In a 10 L jacketed reactor, cool a solution of (3aR,4S,6aS)-l- ace1yl-6a-(5-bromo-2-fluoro-phenyl)-3,3a,4,6-te1rahydrofuro[ 3,4-c]isoxazole-4- carboxylic acid (771 g, 2019 mmol) in dichloromethane (7.0 L) to 0 °C under nitrogen and add GDI (400 g, 2421 mmol) portionwise over 40 minutes. Cool the reactor jacket to -20 °C and stir for 1 hour and then add ,0-dimethylhydroxylamine hydrochloride (260.0 g, 2612 mmol) portionwise over about 30 minutes. Stir at -20 °C for 1 hour, at 0 °C for 2 hours, and at 10 °C for 7 hours. Add GDI (175 g, 1058 mmo!) and stir at 10 °C overnight. Add further GDI (180 g, 1088 mmol) at 10 °C and stirr for 1 hour then add N,0-dimethylhydroxylamine hydrochloride (140 g, 1407 mmol) and continue stirring at 10 °C. If the reaction is incomplete, further charges of GDI followed by N,0~ dimethylhydroxylamine hydrochloride can be made until complete reaction is observed. Cool the reaction mixture to 5 °C and wash with 1 N aqueous hydrochloric acid (5 L) then 2 N aqueous hydrochloric acid (5 L). Extract the combined aqueous solution with dichloromethane (1 L), combine the organic extract and wash with water (2,5 L), 1 N aqueous sodium hydroxide (2.5 L), and water (2.5 L), dry over magnesium sulfate, filter, and evaporate under reduced pressure to give a residue. Add methyl tert-butyl ether (3 L) and evaporate under reduced pressure. Add further methyl iert -butyl ether (2 L) and stir at 50 °C for 1 hour, cool to 25 °C and stir for 30 minutes. Collect the resulting solids by filtration, wash with methyl tert-butyl ether (2 χ 500 mL) and dry under vacuum to give the title compound (760 g, 88%) as a white solid. ES/MS: m/z ( 79 Br/ 8i Br) 417.0/419.0 [M+H].

Alternate Preparation 12

Scheme 3, step C: Cool a solution of (3aR,4S,6aS)-l -acetyl-6a-(5-bromo-2- fluoro~phenyl)-3,3a,4,6-tetrahydrofuro[3,4-c]tsoxazole-4-car boxylic acid (27g, 70.7 mmol) in N,N-dimethylformamide (135 mL) to 0 °C under nitrogen and add CD! (14,9 g, 91.9 mmol). Stir for 1 hour and then add ,Ο-dimethylhydroxylamine hydrochloride (9.0 g, 92 mmol) and triethylamine (14.3 g, 141 mmol). Stir at 15 °C for 16 hours. Cool the reaction mixture to 0 °C and add 0.5 M aqueous sulfuric acid (675 ml,). Stir for 1 hour. Collect the resulting solids by filtration. Slurry the solids in methyl tert-butyl ether (90 mL) for 1 hour. Collect the solids by filtration, wash with methyl tert-butyl ether (30 mL). Dry under vacuum to give the title compound (23 g, 78%) as a solid.

Preparation 13

1 -[(3aR,4S,6aS)- 1 -Acetyl-6a-(5-bromo-2-fluoro-pheny )-3,3a,4,6-tetrahydrofiiro[3,4- c]isoxazol-4-yl]ethanone

Scheme 3, step D: In a 20 L jacketed reactor, cool a solution of (3aR,4S,6aS)-l- acetyl-6a-(5-bromo-2-fluorophenyl)-N-m

c][l,2]oxazole-4-carboxamide (654.0 g, 1536 mmol) in THF (10 L) to -60 °C and add a 3.2 M solution of methylmagnesium bromide in 2-metliyltetraliydrofuran (660 mL, 2110 mmol) dropwise, while maintaining the internal temperature below -40 °C. Stir the reaction mixture at -40 °C for 30 minutes then cool to -50 °C and add a solution of 1 N aqueous hydrochloric acid (2 L) in THF (2 L) maintaining the mtemal temperature below -38 °C. Increase the temperature to 10 °C and add ethyl acetate (5 L) and water (1 L), stir and allow internal temperature to reach 5 °C and separate the layers. Extract the aqueous layer with ethyl acetate (1 L) and combine the organic extracts. Wash the organic extracts with water (2 L) and extract the aqueous layer with ethyl acetate (1 L). Combine the organic extract and wash with brine (3 x 2 L) then dry over magnesium sulfate, filter, and evaporate under reduced pressure to a residue. Add cyclohexane (2.5 L), stir at 60 °C for 1 hour then at 20 °C for 30 minutes, and collect the solid by filtration, washing with cyclohexane (500 mL). Dry the solid under vacuum to obtain the title compound as a white solid (565 g, 99%). ES/MS: m/z ( 79 Br/ 8! Br) 372.0/374.0 [M-HH], [a] D 20 = -58.0 ° (C=1.000, chloroform). Alternate Preparation 13

Scheme 3, step D: Cool a solution of (3aR,4S,6aS)-l-acetyl-6a-(5-bromo-2- fluorophenyl)- -methoxy-N-methyltetrahydiO-lH,3H-furo[3,4-c][l,2]oxazole-4- carboxamide (4.0g, 9.59 mmol) in THF (60 mL) to -5 °C and add a 3.0 M solution of methylmagnesium bromide in 2-methyltetrahydrofuran (5.0 mL, 15 mmol) dropwise, while maintaining the internal temperature between -5 and 0 °C. Stir the reaction mixture between -5 and 0 °C for 60 minutes then add a solution of saturated ammonium chloride (20 mL). Add methyl fert-butyl ether (40 mL), allow the internal temperature to reach 5 °C and separate the layers. Evaporate the organic layer under reduced pressure to a residue. Add n-heptane (50 mL), stir, and collect the solid by filtration. Dry the solid under vacuum to obtain the title compound as a solid (3.0 g, 77%).

Preparation 14

l-[(3aR,4S,6aS)-6a-(5-Bromo-2-fluoiOphenyl)-4-(Ll-difluoi ethyl)tetrahydiO-lH,3H- furo [3 ,4-c ] 1 ,2] oxazol- 1 -yljethanone

Scheme 3, step E: Add l-[(3aR,4S,6aS)-l-acetyl-6a-(5-bromo-2-fluoro-phenyl)- 3,3a,4,6-tetrahydrofuro[3,4-c]isoxazol-4-yl]ethanone (5.08 g, 13.6 mmol) in a single portion to a stirred suspension of XtalFluor-M® (10.02 g, 39.18 mmol) in anhydrous dichloromethane (100 mL) at 0-5 °C. Stir the mixture for 10 minutes and add triethylamine trihydrofluoride (4.5 mL, 27 mmol) dropwise over 10 minutes. Stir the reaction mixture in the ice-bath for 8 hours then warm to ambient temperature and stir overnight. Add saturated aqueous sodium carbonate (100 mL) and stir for 1 hour.

Separate the layers and extract the aqueous with dichloromethane (2 χ 50 mL). Combine the organic extracts and wash with saturated aqueous sodium bicarbonate (100 mL), 2 N aqueous hydrochloric acid (2 x 100 mL), and brine (100 mL). Evaporate to dryness to a light brown solid and dissolve in methyl fert-butyl ether (300 mL) at 60 °C. Filter the hot solution and evaporate the filtrate to give a brown solid (5.3 g, 81%, 82% purity by LCMS) that is used without further purification. ES/MS: m/z ( 79 Br/ 81 Br) 393.8/395.8 [M+H].

Alternate Preparation 14

Scheme 3, step E: Add XtalFluor-M® (1.21 kg, 4.73 mol) in portions to solution of l-[(3aR,4S,6aS)-l-acetyl-6a-(5-bromo-2-fluoro-phenyl)-3,3a,4 ,6- tetrahydrofuro[3,4-c]isoxazol-4-yl]ethanone (565 g, 1.51 mol) in anhydrous dichloromethane (5 L) at -14 °C. Stir the mixture for 10 minutes and add txiethykmine trihydro fluoride (550 g, 3,34 mol) dropwise over 20 minutes. Stir the reaction mixture at -10 °C for approximately 10 hours then warm to ambient temperature and stir overnight. Add 50% aqueous sodium hydroxide (750 mL) slowly, maintaining the internal temperature below 10 °C, then add water (1.5 L) and saturated aqueous sodium hydrogen carbonate (1 L) and stir for 30 minutes. Separate the layers and extract the aqueous with dichloromethane (1 L). Combine the organic extracts and wash with brine (3 L), 2 N aqueous hydrochloric acid ( 5 L), and brine (3 L). Evaporate to give a residue and purify by silica gel chromatography eluting with 50-100% dichloromethane in iso-hexane then 10% methyl tert-butyl ether in dichloromethane to give the title compound as a white powder (467 g, 73%, 94% purity by LCMS). ES/MS: m/z ( 79 Br/ 8i Br) 393.8/395.8

[M+H],

Preparation 15

(3aR,4S,6aS)-6a-(5-Bromo-2-fluoro-phenyl)~4~(l,l-difluoroeth yl)-3,3a,4,6-tetrahydro- lH-furo 3,4-c]isoxazole

Scheme 3, step F: Add 37 wt% aqueous hydrochloric acid (1.3 L, 16 mol) to a solution of 1 - [(3aR,4S,6aS)-6a-(5 -bromo-2-fluorophenyl)-4-( 1 , 1 - difluoroethyl)tetTahydro-lH,3H-furo[3,4-c][l,2]oxazo3-l-yl]e thanone (570 g, 1.45 mol) in 1,4-dioxane (5 L) in a 10 L jacketed reactor and stir at 100 °C for approximately 3 hours or until LCMS shows complete reaction. Cool the reaction mixture to 10 °C, dilute with water (1 L) and add a mixture 50 wt% aqueous sodium hydroxide solution (800 mL) and water ( 1 L) slowly, maintaining the internal temperature below 20 °C. Add ethyl acetate (2.5 L) and stir vigorously, before separating the layers and washing the organic phase with brine (2 L), further brine (1 L), and water (1 L), Dry over magnesium sulfate, filter, and concentrate to dryness under reduced pressure to give a residue. Add cyclohexane (2.5 L) and evaporate to dryness then repeat to obtain the title compound as a brown oil (527 g, 89%, 86% purity by LCMS). ES/MS: m/z ( 79 Br/ 8! Br) 351.8/353.8 [M+H].

Preparation 16

[(2S,3R,4S)-4-Amino-4-(5-bromo-2-fluoropheny

3-yl]methanol

Scheme 3, step G: Add zinc powder (6.0 g, 92 mmol) to a solution of

(3aR,4S,6aS)-6a-(5-bromo-2-fluoro-phenyl)-4-( 1 , 1 -difluoroethyl)-3,3a,4,6-tetrahydro- lH-furo[3,4-c]isoxazole (5.06 g, 13.4 mmol) in acetic acid (100 mL) at ambient temperature and stir overnight. Dilute the mixture with ethyl acetate (200 mL) and water (300 mL) and stir vigorously while adding sodium carbonate (97 g, 915 mmol). Separate the layers and wash the organic layer with brine (2 x 200 mL), dry over magnesium sulfate, filter, and concentrate to give a residue. Purify the residue by silica gel chromatography eluting with 0% to 100% methyl fcrt-butyl ether in isohexane to give the title compound as a waxy solid (4.67 g, 89%, 90% purity by LCMS). ES/MS: m z

( 79 Br/ 81 Br) 354.0/356.0 [M+H].

Alternate Preparation 16

Scheme 3, step G: Add zinc powder (200 g, 3.06 mol) portionwise to a solution of (3aR,4S,6aS)-6a-(5-bromo-2-fluoro-phenyl)-4-(l,l-difluoroeth yl)-3,3a,4,6-tetrahydro- lH-furo[3,4-c]isoxazole (304 g, 75% purity, 647 mmol) in acetic acid (2 L) and water (2 L) at 20 °C then warm to 40 °C and stir overnight. Dilute the mixture water (2 L) and stir vigorously while adding sodium carbonate (4 kg, 43.4 mol) then adjust to pH 8-9 with further sodium carbonate. Add ethyl acetate (5 L) and water (2.5 L), stir for 30 minutes and filter through diatomaceous earth washing with 2: 1 acetonitrile/water. Separate the layers, extract the aqueous with ethyl acetate (2 χ 2.5 L) and wash the combined organic extracts with brine (2 x 2.5 L), dry over magnesium sulfate, filter, and concentrate to give a residue. Purify the residue by SFC, column: Chiralpak AD-H (5), 50 ¾ 250 mm; eluent: 12% ethanoi (0.2% diethylmethylamine in C(¾; flow rate: 340 g/minute at UV 220 nm to give the title compound as a white solid (197.7 g, 84%). [a] D 20 = -6.93 ° (0=0.678, chloroform). ES/MS: m/z ( 79 Br/ sl Br) 354.0/356.0 [M+H].

Preparation 17

[(2S,3R,4S)-4-Armno-4-(5-bromo-2-fluoro-phenyl)-2-^

yljmethanol

Scheme 1, step F: Add (3aR,4S,6aR)-6a-(5-bromo-2-fluoro-phenyl)-4-

(iri1yloxymethy2)-3,3a,4,6-tetrahydrofuro[3,4-c]isoxazo2e (31.30 g, 55.9 mmol) to acetic acid (186 mL) to give a suspension. Add zinc (25.6 g, 391 mmol) and stir the reaction mixture vigorously for 18 hours. Dilute the mixture with toluene and filter through diatomaceous earth. Concentrate the filtrate under reduced pressure. Solubilize the residue with ethyl acetate, wash with brine, and saturated sodium bicarbonate. Separate the phases, dry over magnesium sulfate, filter, and concentrate under reduced pressure to give the title compound (31.35 g, 99%). ES/MS m/e ( 79 Br/ 8i Br) 562/564 [M+H].

Preparation 18

~[[(3S,4R,5S)~3~(5-Bromo~2~fluoro-pheny])-4-(hydroxymethyl)- 5- (1xity4oxymethyl)te1iuhydrofuran-3-yl]carbam.othioyl]benzami de

Scheme 1, step G: Dissolve [(2S,3R,4S)-4-amino-4-(5-bromo-2-fluoro-phenyl)-2-

(tri1yloxyniethy2)terrahydrofuran-3-y2]methanol (31,35 g, 55.73 mmol) in

dichloromethane (557 mL) and cool to 5 °C. Add benzoyl isothiocyanate (9.74 mL, 72.45 mmol). After addition is complete, allow the reaction mixture to warm to room temperature and stir for 2 hours. Pour into saturated sodium bicarbonate, separate the phases, and extract the aqueous phase with dichloromethane. Combine the organic extract and dry over magnesium sulfate. Filter the solution and concentrate under reduced pressure to give the title compound (42.95 g, 106%). ES/MS m/e ( 79 Br/ 8i Br) 747/749 [M+Na].

Preparation 19

N-[(4aS > 5S,7aS)-7a-(5-Bromo-2-fluorophenyl)-5-(l,l-difluoroeth yl)-4a,5,7,7a- tetrahydro-4H-ftiro[3,4-d][ l,3]tmazin-2-yl]benzamide

Scheme 3, step H: Add benzoyl isothiocyanate (1.80 mL, 13.3 mmol,) to a solution of [(2S,3 R,4S)-4-amino-4-(5-bromo-2-fluorophenyl)-2-( 1,1- dif]uoroethyl)tetrahydrofuran-3-yl]methanol (4.67 g, 1 1.9 mmol) in dichloromethane (20 mL) at ambient temperature for 1 hour until LCMS shows reaction is complete.

Evaporate the reaction mixture to a residue under vacuum. Add cvclohexane (50 mL), warm to 60 °C and add methyl fert-butyl ether until precipitate is fully dissolved (100 mL). Filter the hot solution, cool to room temperature and slowly evaporate under reduced pressure until formation of a white precipitate. Remove the solvent under reduced pressure and dissolve the residue in anhydrous dichloromethane (30 mL), add pyridine (2.4 mL, 30 mmol), and cool the solution to -25 °C. Add

trifluoromethanesulfonic anhydride (2.2 mL 13 mmol) dropwise over 30 minutes and allow to warm 0 °C over 1 hour. Wash the reaction mixture with water (25 mL), 2 N aqueous hydrochloric acid (25 mL), water (25 mL), aqueous saturated sodium bicarbonate (25 mL), and water (25 mL), dry over magnesium sulfate, filter, and concentrated to dryness. Purify the residue by silica gel chromatography eluting with 5% methyl ten-butyl ether in dichloromethane to give the title compound as a light yellow foam (5.0 g, 76%, 90 % purity by LCMS). ES/MS: m z ( 79 Br/ 8l Br) 499.0/501.0 [M+H],

Alternate Preparation 19

Scheme 3, step H: Add benzoyl isothiocyanate (98 mL, 724,9 mmol,) to a solution of [(2S,3 R,4S)-4-amino-4-(5-bromo-2-fiuorophenyl)-2-( 1,1- difiuoroethyl)tetrahydrofuran-3-yl]methano3 (197.6 g, 546.7 mmol) in dichloromethane (1.2 L) at 30 °C for 1 hour. Add CD! (101 g, 610.4 mmol) and stir at ambient temperature for 3 hours. Further charges of GDI can be made to ensure complete consumption of the thiourea intermediate. Heat to 90 °C for 42 hours and cool the solution to ambient temperature. Dilute the reaction mixture with ethyl acetate (2 L) and add 2 N aqueous hydrochloric acid (2 L), stir, add brine (1 L) and separate the layers. Wash the organic layer with 2 N aqueous hydrochloric acid (0.5 L), brine (2 x 1 L) and aqueous saturated sodium bicarbonate (1 L). Dry over magnesium sulfate, filter, and concentrate to give a residue. Purify the residue by silica gel chromatography eluting with 0-100% ethyl acetate in iso-hexane to give the title compound as a light yellow solid (234 g, 83%). ES/MS: m z ( 79 Br/ 81 Br) 499.0/501.0 [M+H].

Preparation 20

N-[(4aS,5S,7aS)-7a-(5-Bi mo-2-fiuoro-pheny3)-5-(trityloxymethyl)-4,4a,

tetrahydrofuro[3,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 1, step H: Dissolve N-[[(3S,4R,5S)-3-(5-bromo-2-fluoro-phenyl)-4- (hydroxymethyl)-5-(tri1yloxymemyl)tetrahydrofuran-3-yl]carba mo

(42.95 g, 59.18 mmol) in dichloromethane (591 mL) and cool to -20 °C. Add pyridine (12.0 mL, 148.0 mmol), followed by trifluoromethanesulfonic anhydride (10.97 ml,, 65.10 mmol). Monitor the addition keeping the temperature below -20 °C. Stir the reaction mixture at -20 °C for 30 minutes. Allow the reaction mixture to warm to room temperature. Pour into saturated ammonium chloride, separate the phases, and extract the aqueous phase with dichloromethane. Combine the organic extract and dry over magnesium sulfate. Filter the solution and concentrate under reduced pressure to give the title compound (45.24 g, 108%). ES/MS m/e ( 79 Br/ 8i Br) 707/709 [M-i !j.

Preparation 21

N-[(4aS,5S,7aS)-7a-(5-Bromo-2-fluoro-phenyl)-5-(hydroxymethy l)-4,4a,

tetrahydrofu -d] [ 1 ,3]thiazin-2-yl]benzamide

H

Scheme 1, step I: Dissolve N-[(4aS,5S,7aS)-7a-(5-bromo-2-fluoro-phenyl)-5-

(trityloxymethyl)-4,4a,5 ,7-tetrahydrofuro[ 3 ,4-d] [ 1 ,3]thiazin-2-yi]benzamide (45.24, 63.93 mmol) in formic acid (160 mL) and stir at ambient temperature for 1 hour. Add water (29 mL) over a period of 5 minutes. Stir for 50 minutes. Concentrate the mixture under reduced pressure to a residue. Dissolve the residue in methanol (639 mL), add

triethylamine (26.7 mL, 191.8 mmol), and stir overnight at ambient temperature. Pour into brine, separate the phases, and extract the aqueous phase with chloroform. Combine the organic extract and dry over magnesium sulfate. Filter and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with acetone: hexanes (25-38% gradient), to give the title compound (16.04 g, 54%), ES/MS m/e ( 79 Br/ s ! Br) 465/467 [M+H] .

Preparation 22

(4aS,5S,7aS)-2-Benzamido-7a-(5-bromo-2-f!uoro~phenyl)~4,4a,5 ,7-tetrahydrofuro[3,4- d] L3]thiazine-5-carboxylic acid

Scheme 1, step J: Add N~[(4aS,5S,7aS)-7a~(5~bromo-2-fluoro-phenyl)-5- (hydroxymethyl)-4,4a,5,7-terrahydrofuro[3,4-d][l,3]thiazin-2 -yl]beiizamide (16.04 g, 34.47 mmol) to DMSO (172 mL), Add 2-iodoxybenzoic acid (35.56 g, 120.70 mmol) and stir at ambient temperature for 3 hours. Dilute the reaction mixture with chloroform (300 mL) and pour into saturated ammonium chloride (400 mL). Separate the organic phase and dry over magnesium sulfate. Filter the solution and concentrate under reduced pressure to give a residue. Dissolve the residue in ethyl acetate (400 mL) and wash with saturated ammonium chloride (2x250 mL). Separate the organic phase, dry over magnesium sulfate, filter, and concentrate under reduced pressure to give a residue. Dissolve the residue in a dichloromethane: methanol mixture and add diethyl ether until a solid precipitates. Collect the solid by filtration and dry under reduced pressure to give the title compound (5.78 g, 35%). ES/MS m/e ( 79 Br/ 8i Br) 479/481 [M+H].

Preparation 23

laS,5S,7aS)-2-Benzarmdo-7a-(5-bromo-2-fluoro-phenyl)-N-metho xy-N-methyl-

4,4a,5,7-tetrahydrofui [3,4-d][l,3]thiazine-5-carboxamide

Scheme 1, step K: Dissolve (4aS,5S,7aS)-2-benzamido-7a-(5-bromo-2-fluoro- phenyi)-4,4a,5 J 7 etrahydrofuro[3,4-d][L3]thiazine-5-carboxylic acid (5.78 g, 12,1 mmol) in dichloromethane (201 mL) and Ν,Ο-dimethylhydroxylamine hydrochloride (1.76 g, 18.1 mmol). Add triethylamine (5.29 mL, 36.2 mmol) followed by HATU (7.02 g, 18.1 mmol). Stir at ambient temperature for 3 days. Pour into saturated ammonium chloride, separate the phases, and extract the aqueous phase with ethyl acetate. Combine the organic extracts and dry over magnesium sulfate. Filter and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate; dichloromethane (0-50% gradient) to give the title compound (4.15 g, 66%). ES/MS m/e ( 79 Br/ 81 Br) 522/524 [M+H].

Preparation 24

N-[(4aS,5S,7aS)-5-Acetyl-7a-(5-bromo-2-fluoro-phenyl)-4,4a,5 ,7-tetrahydrofliro[3 J 4- d] 1 ,3]thiazin-2-yl]benzamide

Scheme 1 , step L: Add dropwise to a -78 °C solution of (4aS,5S,7aS)-2- benzarnido-7a-(5-bromo-2-fluoro-phenyl)-N-methoxy-N-methyl-4 ,4a,5,7- terrahydrofuro[3,4-d][l,3]thiazine-5-carboxamide (1.51 g, 2,89 mmol) in THF (57.8 mL) methylmagnesium bromide (3.0 mol/L in diethyl ether, 4.8 mL, 14.5 mmol). Stir the reaction at -78 °C for 5 minutes and allow to gradually warm to ambient temperature. Stir for 30 minutes. Quench the reaction with methanol (4 mL), dilute with saturated ammonium chloride, and extract with ethyl acetate. Combine the organic extract and dry over sodium sulfate. Filter and concentrate under reduced pressure to g ve a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate: hexanes (0- 100% gradient) to give the title compound (1.28 g, 93%). ES/MS m/e ( 79 Br/ 8 l Br) 477/479 [M+Na],

Preparation 24b

N-[(5S aS)-7a-(5-Bromo-2-fluorophenyl)-5-propanoyl-4a,5,7JA-tetrahy dro-4H- furo[3,4-d] [1 ,3]thiazin-2-yl]benzamide

Scheme 1, step L; Add (4aS,5S,7aS)-2-benzamido-7a-(5-bromo-2-fluoro- pheiiy3)-N-methoxy-N-methyl-4,4a,^^

(3.00 g, 5.74 mmol) to THF (1 15 mL) under nitrogen and cool to -78 °C. Add a solution of ethylmagnesium bromide (1.0 mol L) in THF (28.7 mL, 28.7 mmol) to the reaction dropwise, maintaining the temperature at -78 °C. After 1 hour, add methanol (10 mL) in one portion and then pour onto saturated ammonium chloride solution. Extract the mixture with ethyl acetate, dry the organic extracts over magnesium sulphate, filter, and concentrate in vacuo to give a residue. Purify the residue by silica gel chromatography, eluting with hexanes/ethyl acetate (100-40% gradient) to give the title compound (2,844 g, 5.788 mmol, 100%). ES/MS m/z ( 79 Br/ 81 Br) 491/493/513/515 (M+H/ a).

Preparation 24c

N-[(5S,7aS)-7a-(5-Bromo-2-fluoi phenyi)-5-(cyc2opropylcarbonyl)-4a,5,7,7a-tetrahydiO- 4H-furo [3 ,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 1 , step L: Add dropwise to a -78 °C solution of (4aS,5S,7aS)-2- benzarnido-7a-(5-bromo-2-fluoro-phenyl)-N-methoxy-N-methyl-4 ,4a,5,7- tefrahydrofuro[3,4-d][l,3]thiazine-5-carboxainide (1.51 g, 2,89 mmol) in THF (51.2 mL) cyclopropylmagnesium bromide (1.0 mol/L in 2-methyltetrahydroturan, 14 mL, 14.4 mmol). Stir the reaction at -78 °C for 5 minutes and allow to gradually warm to ambient temperature. Stir for 30 minutes. Quench the reaction with methanol (4 mL), dilute with saturated ammonium chloride, and extract with ethyl acetate. Combine the organic extract and dry over sodium sulfate. Filter and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate: hexanes (0-100% gradient) to give the title compound (1.299 g, 89%). ES/MS m/z ( 79 Br/ 81 Br) 503/505 [M+Ffj.

Preparation 25

N-[(4aS,5S,7aS)-7a-(5-Bromo-2-fluoi -phenyl)-5-(l,l-difluoroethyi)-4,4a,5,7- tetrahydrofuro 3,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 1 , step M: Add together dichloromethane (34 mL), Deoxo-Fluor® (1.52 mL, 6.88 mmol), and boron trifluoride diethyl etherate (0.89 mL, 6.88 mmol). Stir at ambient temperature for 2 hours. Add -[(4aS,5S,7aS)-5-acetyl-7a-(5-bromo-2-fluoro- phenyl)-4,4a,5,7-tetrahydrofuro[3,4-d][l,3]thiazm-2-yl]benza mtde (0.821 g, 1.72 mmol) in one portion, followed by triethylamine tiihydrofluoride (1.13 mL, 6.88 mmol). Stir at ambient temperature for 18 hours. Pour into saturated ammonium chloride, separate the phases, and extract the aqueous phase with ethyl acetate. Combine the organic extract and dry over magnesium sulfate. Filter and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with dichloromethane: hexanes (80-100 % gradient), to give the title compound (0.552 g, 64%). ES MS m/e

( 79 Br/ 8! Br) 499/501 [M+H]. Preparation 25b

-[(4aS,5S,7aS)-7a-(5-Bromo-2-fluorophenyl)-5-(l,l-difluoropr opyl)-4a,5,7,7a- tetrahydro-4H-furo[3,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 1 , step M: Add N-[(5S,7aS)-7a-(5-bromo-2-fluorophenyl)-5-propanoyl- 4a,5,7,7A-tetrahydro-4I-i-furo[3,4-d][l,3]thiazm-2-yl]benzam ide (0.486 g, 0.989 mmol) to dichloromethane (19.8 mL) under nitrogen at -78 °C, followed by Deoxo-Fluor® (1.75 g, 1.75 mL, 3.96 mmol). Stir for 30 minutes at -78 °C and then warm to room temperature overnight. Pour the reaction onto saturated sodium bicarbonate solution and extract with dichloromethane. Separate organics using a hydrophobic frit and concentrate in vacuo to give a residue. Purify the residue by silica gel chromatography, elutmg with hexanes/ethyl acetate (100-50% gradient) to give the crude title compound with an impurity (0.230 g, 0.448 mmol, 45%) that is used without further purification. ES/MS ni/z 513/515 (M+H).

Preparation 25c

N-[(4aS,5S,7aS)-7a-(5-Bromo-2-fluoro-phenyl)-5-[cyclopropyl( difluoro)methyl]-

4,4a,5,7-tetrahydrofuro[3,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 1, step M: Add Deoxo-Fluor® (1.9 mL, 6.68 mmol) to an ambient solution of N-[(5S,7aS)-7a-(5-bromo-2-fluorophenyl)-5-(cyclopropy]carbon yl)-4a,5,7,7a- tetrahydro-4H-furo[3,4-d][l,3]thiazin-2-yl]benzamide (0.812 g, 1.61 mmol) in dichloromethane (24 mL). Stir at ambient temperature for 18 hours. Pour into saturated sodium bicarbonate and extract the aqueous phase with ethyl acetate (3x). Combine the organic extracts and dry over sodium sulfate. Filter and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate; hexanes (5-100 % gradient), to give the title compound (41 mg, 5%).

ES/MS mix ( 79 Br/ 81 Br) 525/527 [M+H].

Preparation 26

N-[(5S,7aS)-5-(l,l-Difluoroethyl)-7a-{2-fluoro-5-[(trifluoro a(x1yl)arnino]phenyl}- 4a,5,7,7a-tetrahydi -4H-furo[3,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 4, step A: Dissolve N-[(4aS,5S,7aS)-7a-(5-bromo-2-fluorophenyl)-5- (l,l-difluoroethyl)-4a,5,7,7a etrahydro-4H-furo[3,4-d][13]thiazin~2 » yl]benzamide (234 g, 454.6 mmol) in 1,4-dioxane (2 L) and add 4 A molecular sieves (37 g), 2,2,2- trifluoroacetamide (91 g, 780.9 mmol), finely ground potassium carbonate (114 g, 824.9 mmol), sodium iodide (117 g, 780.6 mmol), copper (I) iodide (17.5 g, 91.9 mmol) and racemic trans-N,N' -dimethyl- 1,2-cyclohexane diamine (20 g, 140.6 mmol) under a stream of nitrogen. Purge the vessel with 3 vacuum nitrogen switches and heat to 123 °C for 18 hours. Cool to ambient temperature and filter the solution through diatomaceous earth, and wash with ethyl acetate. Add saturated aqueous ammonium chloride (2 L) and vigorously stir for 45 minutes. Separate the layers and wash the organic layer with saturated aqueous ammonium chloride (3 x 1 L), brine (300 mL), dry over magnesium sulfate, filter, and evaporate to give a residue. Purify the residue by silica gel chromatography eluting with 0-100% ethyl acetate in iso-hexane to give the title compound as a light yellow solid (297.9 g, 95%, 81% purity). ES/MS: m/z 532.0 [M+Hj. Preparation 27

N-[(4aS,5S,7aS)-7a-(5-Arnino-2-fluoro-phenyl)-5-(l,l-difluor oethyl)-4,4a,5,7- terraliydrofuro 3,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 1, step N: Combine N-[(4aS,5S,7aS)-7a-(5-bromo-2-fluoro-phenyl)-5-

(1,1 -difluoroethyl)-4,4a,5,7-tetrahydrofuro [3 ,4-d] [1 ,3] thiazin-2-yl]benzamide (0.372 g, 0.74 mmol) and (lR,2R)-N, '-dimethyl~l,2-cyclohexanediamtne (0.037 mL, 0.22 mmol) in ethanol (30 ml). Add sodium azide (0.194 g, 2.98 mmol), followed by sodium L- ascorbate (0.66 M solution, 0.50 ml, 0.33 mmol). Purge the top of the flask with nitrogen and add cupric sulfate (0.33 M solution, 0.68 ml, 0.22 mmol). Heat the reaction mixture to 80 °C and stir for 5 hours. Cool the reaction and add cold water. Extract the mixture with ethyl acetate. Combine the organic extract and dry over sodium sulfate. Filter and concentrate under reduced pressure to give a residue. Combine the residue with palladium (10 mass% on carbon, 0.35 g, 0.16 mmol) in ethanol (50 ml) and THF (10 ml). Purge the mixture with nitrogen and with hydrogen. Stir at ambient temperature under 50 psi of hydrogen for 1 hour. Filter off the catalyst and wash with ethyl acetate.

Concentrate the solution under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate: dichloromethane (0-20% gradient), to give the title compound (0.2184 g, 67%). ES/MS m/z 436 (M+H),

Alternate Preparation 27

Scheme 4, step B: Add 7 N ammonia in methanol (600 mL, 4.2 mol) to a stirred suspension of -[(5S,7aS)-5-(l,l-difluoroethy2)-7a-{2-fluoro-5- [(trifluoroacetyl)amino]phenyl} -4a,5,7,7a-tetrahydro-4H-furo[3,4-d] [1 ,3]thiazin-2- yljbenzamide (250 g, 80% purity, 376.3 mmol) in methanol (200 mL) at room temperature and stir at ambient temperature for 18 hours. Evaporate to dryness to give the title compound as a brown gum (190 g, 375.2 mmol, 86% purity). ES/MS: m/z 436.0 [M+H].

Preparation 27b

S,5 S,7aS)-7a-(5- Amino-2-fluorophenyl)-5-(l , 1 ~difluoropropyl)-4a,5 ,7,7a-tetrahydro- 4H-fur -d] [ 1 ,3]thiazm-2-amine

Scheme 1, step N: Add together N-[(4aS,5S,7aS)-7a-(5-bromo-2-fluorophenyl)-5- (1,1 -difluoropropyl)-4a,5,7,7a-tetrahydro-4H-ruro [3 ,4-d] [ 1 ,3]thiazin-2-yl]benzamide (0.567 g, 1.104 mmol), 1,4-dioxane (4.802 mL) and ethanol (1 1.04 mL), followed by sodium azide (0.2154 g, 3.313 mmol), trans -Ν,Ν'-dimethy Icy clohexane- 1 ,2-diamine (0.04760 g, 0.0528 mL, 0.3313 mmol), sodium L-ascorbate 0.66 M (0.74 g, 0.74 mL, 0.4859 mmol) and water (0.1699 mL). Finally add cupric sulphate 0.33 M (0.74 g, 0.74 mL, 0.2430 mmol) and heat to 100 °C. Stir overnight at 90 °C. Add further cupric sulfate 0.33 M (0.74 g, 0.74 mL, 0.2430 mmol), sodium L-ascorbate 0.66 M (0.74 g, 0.74 mL, 0.4859 mmol), sodium azide (0.2154 g, 3.313 mmol) and trans-Ν,Ν'- dimethy Icy clohexane- 1 ,2-diamine (0.04760 g, 0.0528 mL, 0.3313 mmol). Heat the reaction at 100 °C for 1 hour, and then cool to room temperature. Pour the reaction onto brine and extract the product with chloroform. Filter mixture through diatomaceous earth and separate the organics, dry over magnesium sulphate, filter, and concentrate in vacuo. Transfer the material to a Parr flask with palladium (5 mass%) in Lindlar's catalyst (0.113 g, 0.0533 mmol), and add methanol (55.22 mL) under nitrogen. Hydrogenate under 276 kPa hydrogen pressure with vigorous shaking for 3 hours. Add further palladium (5 mass%) in Lindlar's catalyst (0.1 13 g, 0.0533 mmol) and hydrogenate under 345 kPa hydrogen pressure for a further 4 hours. Filter the reaction through diatomaceous earth, and wash with chloroform. Concentrate the filtrate in vacuo and then add methanol (55.22 mL) followed by lithium hydroxide hydrate (0.4634 g, 0.182 mL, 11.04 mmol). 8

Heat the reaction mixture to 70 °C for 3 hours and then cool to room temperature. Pour the reaction onto brine and extract with chloroform. Dilute the organics with methanol, and pour onto an SCX-2 cartridge. Flush the cartridge with one column volume of methanol and discard. Then flush the SCX-2 cartridge with one column volume of 7 M methanolic ammonia and concentrate in vacuo to give the title compound (0.290 g, 0.840 mmol, 76%). ES/MS m/z 346 (M+H).

Preparation 27c

N-[(4aS,5S,7aS)-7a-(5-Arnmo-2-fluoro-phenyl)-5-[cyclopropyl( difluoro)methyl]

4,4a,5,7-tetrahydrofuro[3,4-d] [ 1 ,3]thiazin-2-yl]benzamide

Scheme 1 , step N: Combine N-[(4aS,5S,7aS)-7a-(5-bromo-2-fluoro-pheny])-5- [cyclopropyl(difluoro)methyl] -4,4a,5 ,7-tetrahydrofuro[3 ,4-d] [ 1 ,3]thiazin-2-yl]benzamide (31 mg, 0.059 mmol) and (lR,2R)-N J N'-dimethyl-l,2-cyclohexanediamine (0.0049 mL, 0.030 mmol) in ethanol (3 ml). Add sodium azide (31 mg, 0.47 mmol), followed by sodium L-ascorbate (0.66 M solution, 0.089ml, 0.059 mmol). Purge the top of the flask with nitrogen and add cupric sulfate (0.33 M solution, 0.18 ml, 0.059 mmol). Heat the reaction mixture to 80 °C and stir for 3 hours. Cool the reaction and add cold water. Extract the mixture with ethyl acetate. Combine the organic extract and dry over sodium sulfate. Filter and concentrate under reduced pressure to give a residue. Combine the residue with palladium (10 mass% on carbon, 30 mg, 0.014 mmol) in ethanol (20 ml) and THF (5 ml). Purge the mixture with nitrogen and with hydrogen. Stir at ambient temperature under hydrogen at 40 psi for 4 hours. Filter off the catalyst and wash with ethyl acetate. Concentrate the solution under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate: hexanes (0-100% gradient), to give the title compound (21 mg, 77%). ES/MS m/z 462 (M+H). Preparation 28

aS)-7a-(5-Ammo-2-fluorophenyl)-5-(l,l-difluoroethyl)-4a,5,7, 7a-tetrahyd]

4H-furo [3 ,4-d] [ 1 ,3]thiazm-2-ainine

Scheme 4, step B: Dissolve N-[(4aS,5S,7aS)-7a-(5-ammo-2-fluoro-phenyl)-5-

(1,1 -difluoroet yl)-4,4a,5,7-tetrahydroruro [3 ,4-d] [ 1 ,3]thiazin~2~yl]benzamide (216,4 g, 88% purity, 435.9 mmol) in pyridine (400 mL), ethanol (100 mL) and THF (300 mL). Add O-methylhydroxylamine hydrochloride (190 g, 2275.0 mmol) and stir at ambient temperature for 18 hours. Dilute with 2-methyltetrahydrofuran (1 L) and wash with water (2 x 300 mL). isolate the organic layer and add 35% aqueous ammonium hydroxide (100 mL) to the aqueous. Extract with 2-methyltetrahydrofuran (300 mL) then saturate with sodium chloride and extract with 2-methyltetrahydrofuran (2 x 300 mL). Combine the organic extracts, wash with brine (300 mL) and evaporate to a residue. Dissolve in methanol (200 mL), add 7 N ammonia in methanol (100 mL, 700 mmol) and stir at room temperature for 18 hours. Further ammonia can be added if any trifluoracetamide impurity remains. Remove the solvent under reduced pressure and dissolve the residue in aqueous 2 N aqueous hydrochloric acid (1.5 L). Extract with dichloromethane (6 * 500 mL), combine the organic layers and remove the solvent under reduced pressure to a total volume of about 1 L. Wash with 2 N aqueous hydrochloric acid (300 mL) and combine all aqueous washings. Add 2-methyltetrahydrofuran (1 L) and stir vigorously while adjusting the pH to basic with sodium bicarbonate until no gas evolution is observed. Separate the layers and extract the aqueous with 2-methyltetrahydrofuran (2 χ 500 mL). Dry the combined organic extracts with magnesium sulfate, filter, and evaporate to give a brown solid. Purify the residue by silica gel chromatography eluting with 0-100% dichloromethane in THF . Evaporate the product containing fractions with ethyl acetate/heptane to give the title compound as a fine beige powder (106 g, 70%, 95% purity). ES/MS: m/z 332.0 [M+H], [a] D 20 = +42.11 ° (C= 0.532, chloroform). Preparation 29

5 -(1 H- 1 ,2,4-Triazol- 1 -yl)pyrazine-2-carboxylic acid

Stir a mixture of methyl 5-chloropyrazine-2-carboxylate (124 g, 718.55 mmol), lH-l ,2,4-triazole (198.5 g, 2874.2 mmol) and potassium carbonate (297.92 g, 2155.6 mmol) m N,N-dimethylformamide (1 L) at 100 °C for 15 hours. Cool to ambient temperature and pour into water (2 L). Adjust the pH of the solution to 2-3 using concentrated aqueous hydrochloric acid (about 500 mL) and stir for 30 minutes. Collect the resulting solid by filtration and wash with water. Add water (500 mL) and ethanol (500 mL), heat to 50-60 °C for 4 hours, and cool to ambient temperature. Collect the solids by filtration and dry under vacuum at 40 °C to give the title compound as a white solid. ES/MS: m/z 190.0 (.VI -I I). Preparation 30

N-[3-[(4aS,5S,7aS)-2-Benzamido-5-(l,l -difluo

d] [ 1 ,3]thiazin-7a-yl -4-fluoro-phenyl]-5- 1 ,2,4-triazol- 1 ~yl)pyrazme~2-earboxamide

Scheme 3, step A: Add together N-[(4as,5s,7as)-7a-(5-amino-2-fluoro-phenyl)-5- (l,l-difluoroethy )~4,4a,5,7-tetrahydrofiiro[3,4~d][l ,3]thiazin-2-yl]benzamide (0.139 g, 0.32 mmol), 5-(lH-l,2,4-triazol-l-yl)pyrazrne-2-carboxylic acid (0.0852 g, 0.45 mmol), and HOAt (0.0575 g, 0.41 mmol) in dichloromethane (4 ml): dimethylformamide (1 mL). Add N,N-diisopropylethylamine (0.11 mL, 0.63 mmol) to the solution followed by EDCI (0.079 g, 0.41 mmol) in one portion. Stir the reaction mixture at ambient temperature for 18 hours. Dilute the solution with ethyl acetate, wash with water and brine, and separate the phases. Extract with ethyl acetate. Combine the organic extract and dry over magnesium sulfate. Filter the solution and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate: dichloromethane (0-30% gradient), to give the title compound (0.1140 g, 59%). ES/MS m z 609 (M+H).

Preparation 30a

N-[3-[(4aS,5S,7aS)-2-Benzamido-5-[cye!opropy

telxahydroffliO [3 ,4-d] [ 1 ,3]thiazin-7a-y¾^

2-carboxamide

Scheme 3, step A: Add together N-[(4aS,5S,7aS)-7a-(5-amino-2-fluoro-phenyl)-

5-[cyclopropyl(difluoro)methyl]-4,4a,5,7 e1rahydrofuro[3,4-d][L3]thiazm-2- yljbenzamide (21 mg, 0.045 mmol), 5-(lH-l,2,4-triazol-l-yl)pyrazine-2-carboxylic acid (10 mg, 0.054 mmol), and HOBT (10 mg, 0.059 mmol) in dichloromethane (2.5 ml): dimethylformamide (0.5 mL). Add N,N-diisopropylethylarnine (0.016 mL, 0.091 mmol) to the solution followed by EDC1 (11 mg, 0.059 mmol) in one portion. Stir the reaction mixture at ambient temperature for 18 hours. Dilute the solution with ethyl acetate, water, and 1 N NaOH (0.5 mL) and extract with ethyl acetate (3*). Combine the organic extracts and dry over sodium sulfate. Filter the solution and concentrate under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with ethyl acetate: dichloromethane (0-100% gradient), to give the title compound (20 mg, 69%). ES/MS m/z 635 (M+H). Example 1

N-[3-[(4aS,5S,7aS)-2-Amino-5-iU-dM^

d] [ 1 ,3] thiazin-7a-yl]-4-fluoro- henyl]-5-( 1 2,4-triazol- 1 -yl)pyrazine-2-carboxamide

Scheme 3, step B: Heat a mixture of N-[3-[(4aS,5S,7aS)-2-benzamido-5-(l,l- difSuoroethyl)-4,4a,5,7-tetrahydrofuro[3,4-d][l ,3]thiazm-7a-yl]-4-fluoro-phenyl]-5- (l,2,4-triazol-l-yl)pyrazme-2-carboxamide (0.1148 g, 0.189 mmol), O- methylhydroxyiamine hydrochloride (0.1575 g, 1.886 mmol), and pyridine (0.15 mi, 1.886 mmol) in THF (2 mL) and ethanoi (2 mL) at 45 °C for 5 hours. Cool the reaction mixture to ambient temperature and stir for 2 days. Concentrate the solution under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with 7 N NH 3 in methanol: diehloromethane (0-3% gradient), to give the title compound (0.086 g, 90%). ES/MS m 505 (M+H).

Alternative Preparation Example 1

Scheme 4 Step D: Stir (4aS,5S,7aS)-7a-(5-amino-2-fluorophenyl)-5-(l,l- difluoroethyl)-4a,5,7,7a-te1xahydro-4H-fuiO[3,4-d][l ,3]thiazin-2~amine (96.5 g, 291 mmol) in ethyl acetate (1 L) under a nitrogen atmosphere at 50 °C and add 5-(lH-l,2,4- triazol-l-yl)pyrazine-2-carboxylic acid (84 g, 439.45 mmol) slowly to the wann solution. Stir for 10 minutes and add T3P® (1.67 M in ethyl acetate, 350 mL, 585 mmol) and stir at 50 °C for 17 hours. Cool to ambient temperature, dilute with diehloromethane (1 L) and stir while quenching with a solution of sodium carbonate in water (128 g, 1.21 mol in 1 L). Dilute with d ehloromethane (1 L) and water (2 L) and stir vigorously for 1 hour. Filter through diatomaceous earth and wash with diehloromethane (3 ¾ 500 mL), methanol (500 mL), water (500 mL), aqueous saturated sodium bicarbonate (500 mL), and 1 : 1 methanol:dichloromethane (6 χ 500 mL). Separate the layers and extract the aqueous with dichloromethane (3 χ 1 L). Combine all organic phases and evaporate to give a residue. Sonicate the residue in dichloromethane (1 L) for 15 minutes and collect the solids by filtration washing with dichloromethane (5 x 200 niL). Add saturated aqueous sodium hydrogen carbonate until pH 8 is obtained and stir vigorously with dichloromethane (1 L) and methanol (500 ml,). Remove the solids by filtration and extract the filtrate with dichloromethane (2 χ 500 mL). Dissolve the solids with

dichloromethane:methanol (1 : 1, 500 mL) and combine this solution with the other organic phases. Remove the solvent under reduced pressure adding dichloromethane to maintain a solution and then once a final volume of about 300 ml, is obtained, purify the solution by silica gel chromatography, eluting with 5% of 0,3 M ammonia/methanol in dichloromethane to give a light brown solid. Dissolve the solid in hot ethanol (2.5 L), filter while hot, and cool to ambient temperature over 1 hour. Collect the solids by filtration and wash with ethanol (2 x 250 mL) and dry under vacuum. Evaporate the filtrate to dryness and further purify by silica gel chromatography eluting first with 65% ethyl acetate in 50: 1 iso-hexane/7 N ammonia in methanol then 50: 1 ethyl acetate/7 N ammonia in methanol. If required, further purification can be completed by SFC, column: Chiralpak AD-H (5 μ), 50 x 250 mm; eluent: 35% isopropanol (0.2%

diethylmethylamine) in CO?,; flow rate: 300 g/minute at IJV 220 nm. After evaporation and vacuum drying, slurry the material in ethanol (1.5 L) and stir with gentle wanning between (36 and 45 °C) for 20 minutes. Collect the solid by filtration washing with ethanol (100 mL). Further material can be recovered from the filtrate; evaporate to dryness, reflux in ethanol, remove the solids by hot filtration and then cool the filtrate to ambient temperature. Collect solids by filtration, washing with ethanol and combine with the material obtained from the above filtration. Dry the combined solids under vacuum at 40 °C to give the title compound as a white solid (103.3 g, 68%, containing 2.5 wt% ethanol). ES/MS m/z 505.0 (M+H), [a] D 20 - +149.4 ° (C= 1, chloroform).

Example 1A

-[3-[(4aS,5S,7aS)-2-amino-5-(l,l-difl.uoroethyl)-4,4a,5,7-te trahydrofuro[3,4- d][l,3]thiazin-7a-yl]-4-fluoro-pheiiyl]^^ 4- methy lbenzenesul fonate

Dissolve N-[3-[(4aS,5S,7aS)-2-amino-5-(l,l-difluoroethyl)-4,4a,5,7- tetrahydrofuro [3 ,4-d] [ 1 ,3]thiazin-7a-yl] -4-fiuoro-pheiiyl] -5-( 1 ,2,4-triazoi- 1 -yl)pyrazine- 2-carboxamide (600 mg, 1.189 mmol) in acetone (9 mL)and water (1 niL). Heat the resulting suspension to 60 °C. Add ?-toluenesulfonic acid monohydrate (420 mg, 2.208 mmol) dissolved in acetone (1 niL). Stir the mixture overnight at 60 °C. Cool the mixture to room temperature, filter the solids by vacuum and wash with acetone (1 mL) and air dry overnight to give the title compound (743 mg, 73%). X-Ray Powder Diffraction (XRD)

The XRD 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 0.009° in 2Θ, a scan rate of 0.5 seconds/step, with 0.6 mm divergence, 5.28 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. The crystal form diffraction patterns are collected at ambient temperature and relative humidity, it is well known in the crystallography art that, for any given crystal forrn, 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 United States Pharmacopeia #23, National Formulary #18, pages 1843-1844, 1995. 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 eiystal 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, were adjusted based on NIST 675 standard peaks at 8.853 and 26.774 degrees 2-theta.

A prepared sample of crystalline N-[3-[(4aS,5S,7aS)-2-amino-5-(l,l- difluoroe1hyl)-4,4a,5,7-telTahydrofuro[3,4-d][l,3]thiazm-7a- yl]-4-f]uoro-ph

(l,2,4-triazol-l-yl)pyrazme-2-carboxamide 4-methylbenzenesulfonate is characterized by an XRD pattern using Cu a radiation as having diffraction peaks (2-theta values) as described in the Table below. Specifically, the pattern contains a peak at 17.3° in combination with one or more of the peaks selected from the group consisting of 14.8, 12.7, and 4.9; with a tolerance for the diffraction angles of 0.2 degrees. Table 1: X-ray powder diffraction peaks of Example 1A.

Example IB

N-[3-[(4aS,5S,7aS)~2~armno-5-(i,! -difiuoroe ^

d] [1 ,3]thiazin-7a-yl]-4-fluoro-phenyl]-5-(l ,2,4-triazol-l -yl)pyrazine-2-carboxamide malonate

Add together N-[3-[(4aS,5S,7aS)-2-amino-5-(l, l-difluoroethyl)-4,4a,5,7- tetrahydrofuro [3 ,4-d] [ 1 ,3]thiazin-7a-yl] -4-fluoro-phenyl] -5-( 1 ,2,4-triazoi- 1 -yl)pyrazine- 2-carboxamide (201 mg, 0.398 mmol) and malonic acid (104 mg, 0.999 mmol)in 95% ethanol- water (15 mL). Stir the mixture 65 °C until a solution a clear solution is obtained. A thick white solid precipitates after a few minutes. Stir the suspension for 1 hour at 55 °C and then cool to room temperature with stirring. Filter the solids under vacuum and air dry for 2 days to give the title compound (477 mg, 80%).

A prepared sample of crystalline N-[3-[(4aS,5S,7aS)-2-amino-5-(l,l- difluoi ethy2)-4,4a,5,7-tetrahydromro[3,4-d][l J 3]thiazm-7a-yl]-4-fluoi -phenyi]-5-

(l,2,4-triazol-l -y])pyrazme-2-carboxamide malonate is characterized by an XRD pattern using CuKa radiation as having diffraction peaks (2-theta values) as described in Table 2 below. Specifically, the pattern contains a peak at 22.7 in combination with one or more of the peaks selected from the group consisting of 16.8, 17.2, and 24.0; with a tolerance for the diffraction angles of 0.2 degrees.

Table 2: X-ray powder diffraction peaks of Example IB.

Angle Relative Intensity

Peak

(°2-Theta +/- 0.2°) (% of most intense peak)

1 5.5 39

2 10.3 44

3 11.8 55

4 15.3 39

5 16.8 62

6 17.2 57

7 18.3 41

8 22.4 60

9 22.7 100

10 24.0 53 Example 1C

N-[3-[(4aS,5S,7aS)-2-arrdno-5-(l,l-difluoroe1hyl)-4,4a,5,7-t etrahy(kofuro[3,4- d] [1 ,3]thiazin-7a-yl]-4-fluoro-phenyl]-5-(l ,2,4-triazol-l -yl)pyrazme-2-carboxamide

h drate

Suspend N- [3 ~[(4aS,5 S,7aS)-2-amino-5 - ( 1 , 1 -difluoroethyl)~4,4a,5 ,7- tetrahydrofuro [3 ,4-d] [ 1 ,3]thiazin-7a~yl] -4-f! uoro-phenyl] -5-( 1 ,2,4-triazol-l -yl)pyrazine- 2-carboxamide (116 mg, 0.23mmol ) in 1 : 1 THF: water (2 mL) at 70 °C. Stir the solution for at least 2 days, filter the solid, and dry under a nitrogen stream to give the title compound,

A prepared sample of N-[3-[(4aS,5S,7aS)-2-amino-5-(l ,l~difiuoroethyl)-4,4a,5,7- tetrahydrofm [3,4-d] [1 ,3]thiazm^

2-carboxamide hydrateis characterized by an XRD pattern using CuKa radiation as having diffraction peaks (2-theta values) as described in Table 3 below. Specifically, the pattern contains a peak at 13.0 in combination with one or more of the peaks selected from the group consisting of 7.8, 10.5, 1 1.0, 14.9, 19.7, 21.3, and 26.9 with a tolerance for the diffraction angles of 0.2 degrees.

Table 3. X-ray powder diffraction peaks of Example 1C.

Example 2

N-[3-[(4aS,5S,7aS)~2-Amino~5~(l,l-difluoropropyl)-4,4a,5,7-t etrahydrofiiro[3,4- d][l,3]t azin-7a-y2]-4-fluoro-pheny2]-5-(l,2,4-triazol-l-yl)pyrazine- 2-carboxaniide

Scheme 3, steps A and B: Add together 5-(lH-l,2,4-triazol-l-yl)pyrazine-2- carboxylic acid (0.116 g, 0.608 mmol), acetonitrile (4.05 mL), dimethyl formamide

(0.00314 mL), and then oxalyl chloride (0.154 g, 0.105 mL, 1.22 mmol) dropwtse under nitrogen. Stir the reaction for 30 minutes and then concentrate in vacuo. Dissolve the residue in acetonitrile (4.05 mL,) and add dropwise to a mixture of (4aS,5S,7aS)-7a-(5- amino-2-fluorophenyl)-5-(l , 1 -difluoropropyl)-4a,5,7,7a-tetrahydro-4H-furo[3,4- d][L3]thiazin-2 -amine (0.140 g, 0.405 mmol) in ethanol (4.05 mL) and water (1.35 mL) with stirring. Upon complete addition, dilute the reaction with chloroform and wash with saturated sodium bicarbonate solution. Dilute the organics with methanol and add to an SCX-2 cartridge. Flush the SCX-2 cartridge with one column volume of methanol and discard, and then flush the SCX-2 cartridge with one column volume of 7 M methanolic ammonia. Concentrate the methanolic ammonia flush in vacuo and purify by silica gel chromatography, eluting with dichioromethane/methanol (100-85% gradient) to give a residue. Further purify by achiral SFC (Supercritical Fluid Chromatography) (Column: benzenesulfonamide (BzS) (5 μ), Princeton Chromatography, 21.2 x 250 mm; eluent: 22% methanol (1% 2 M ammonia in methanol) in CC¾ flow rate; 70 mL/rninute at UV 250 nm; back pressure; 100 bar; temperature: 40 °C). Solubilize the residue in chloroform and wash with brine. Pass the organics through a hydrophobic frit and concentrate in vacuo to give the title compound (0.0658 g, 0.127 mmol, 31 %). ES/MS m/z 519 (M+H). Example 3

N- [3 - [(4aS,5 S,7aS)-2-Amino-5 - [eyclopropyl(difluoro)methyl] -4,4a,5 ,7- te1xahydro&ro[3,4-d][l,3]thiazin-7a-yl]-4-fluoro-pheny2] -5-(l,2,4-triazol-l-yl)p^^

2-carboxamide

Scheme 3, step B: Heat a mixture of -[3-[(4aS,5S,7aS)-2~benzamido-5-

[cyclopropyl(difluoro)methyl]-4,4a,5,7-te1xahydrofuro[3,4 -d][l,3]thiazin-7a-yl]-4-fluoro- phenyi]-5-(l,2,4-triazo2-l-yi)pyrazine-2-carboxamide (20 mg, 0.0315 mmol), O- methyihydroxylamme hydrochloride (26 mg, 0.315 mmol), and pyridine (0.026 ml, 0.315 mmol) in ethanol (3 ml,) at 55 °C for 18 hours. Cool the reaction mixture to ambient temperature and concentrate the solution under reduced pressure to give a residue. Purify the residue by silica gel chromatography, eluting with 7 N N¾ in methanol: dichloromethane (0.5-10% gradient), to give the title compound (15 mg, 90%). ES/MS m/z 531 (M+H).

In vitro Assay Procedures:

To assess selectivity of BACE1 over BACE2, the test compound is evaluated in

FRET-based enzymatic assays using specific substrates for BACE1 and BACE2 as described below. For in vitro enzymatic and cellular assays, the test compound is prepared in DMSO to make up a 10 mM stock solution. The stock solution is serially diluted in DMSO to obtain a ten-point dilution eun'e with final compound concentrations ranging from 10 μΜ to 0.05 nM in a 96-well round-bottom plate before conducting the in vitro enzymatic and whole cell assays.

In vitro protease inhibition assays:

Expression of AwBACEl :Fc and AwBACE2:Fc.

Human BACE1 (accession number: AF 190725) and human BACE2 (accession number: AF 204944) are cloned from total brain cDNA by RT-PCR. The nucleotide sequences corresponding to amino acid sequences #1 to 460 are inserted into the cDNA encoding human TgGi (Fc) polypeptide (Vassar et al, Science, 286, 735-742 (1999)). This fusion protein of BACE1(1 -460) or BACE2( 1-460) and human Fc, named

AwBACEl :FC and AwBACE2:Fc respectively, are constructed into the pJB02 vector.

Human BACEl(l-460):Fc (AwBACEl :Fc) and human BACE2(l-460):Fc (AwBACE2:Fc) are transiently expressed in HEK293 cells. 250 ,ug cDNA of each construct are mixed with Fugene 6 and added to 1 liter HEK293 cells. Four days after the transfection, conditioned media are harvested for purification. AwBACEl :Fc and AwBACE2:Fc are purified by Protein A chromatography as described below. The enzymes are stored at - 80 °C in small aiiquots. (See Yang, et. al, J. Neurochemistry, 91(6) 1249-59 (2004),

Purification of AwBACEl :Fc and AwBACE2:Fc.

Conditioned media of HEK293 cell transiently transfected wit AwBACEl :Fc or AwBACE2:Fc cDNA are collected. Cell debris is removed by filtering the conditioned media through 0.22 μηι sterile filter. 5 ml Protein A-agarose (bed volume) is added to 4 liter conditioned media. This mixture is gently stirred overnight at 4 °C. The Protein A- agarose resin is collected and packed into a low-pressure chromatography column. The column is washed with 20x bed volumes of PBS at a flow rate 20 ml per hour. Bound AwBACEl :Fc or A«BACE2:Fc protein is eluted with 50 mM acetic acid, pH 3.6, at flow rate 20 ml per hour. 1 ml fractions of eluent are neutralized immediately with 0.5 ml 200 mM ammonium acetate, pH 6.5. The purity of final product is assessed by

electrophoresis in 4-20% Tris-Glycine SDS-PAGE. The enzyme is stored at -80 °C in small aiiquots.

BACEl FRET Assay

Serial dilutions of the test compound are prepared as described above. The compound is further diluted 20 x in KH2PO4 buffer. Ten uL of each dilution is added to each well on row A to H of a corresponding low protein binding black plate containing the reaction mixture (25 uL of 50 mM H 2 PG 4 , pH 4.6, 1 mM TRITON® X-100, 1 mg mL BSA, and 15 μΜ of FRET substrate based upon the sequence of APP) (See Yang, et. ah, J. Neurochemistry, 91(6) 1249-59 (2004)). The content is mixed well on a plate shaker for 10 minutes. Fifteen uL of two hundred pM human BACEl (l-460):Fc (See Vasser, et ah, Science, 286, 735-741 (1999)) in the KH 2 PO 4 buffer is added to the plate containing substrate and the test compound to initiate the reaction. The RFU of the mixture at time 0 is recorded at excitation wavelength 355 nm and emission wavelength 460 nm, after brief mixing on a plate shaker. The reaction plate is covered with aluminum foil and kept in a dark humidified oven at room temperatui'e for 16 to 24 hours. The RFU at the end of incubation is recorded with the same excitation and emission settings used at time 0. The difference of the RFU at time 0 and the end of incubation is representative of the activity of BACEl under the compound treatment. RFU differences are plotted versus inhibitor concentration and a curve is fitted with a four-parameter logistic equation to obtain the IC 50 value. (May, et ah, Journal ofNeuroscience, 31. 16507-16516 (2011)).

The compound of Example 1 herein is tested essentially as described above and exhibits an iC 50 for BACEl of 1.19 nM ± 0.48, n= (Mean ± SEM; SEM = standard error of the mean). This data demonstrates that the compound of Example 1 inhibits purified recombinant BACEl enzyme activity in vitro. BACE2 TMEM27 FRET Assay

Transmembrane protein 27 (TMEM27) (Accession Number NM_020665), also known as Collectrin) is a recently described substrate for BACE2, but not BACE1 (Esterhazy, et al, Cell Metabolism, 14, 365-377 (201 1 )). To evaluate the test compound for inhibition of BACE2 enzymatic activity, a FRET peptide (dabcyl-QTLEFLKIPS- Luc Y) based upon the amino acid sequence of human TMEM27 is used as a substrate (Esterhazy, et al, Cell Metabolism, 14, 365-377 (2011)). Serial dilutions of the test compound are prepared as described above. The compound is further diluted 20x in ΚΉ 2 ΡΟ 4 buffer. Ten uL of each dilution is added to each well on row A to H of a corresponding low protein binding black plate containing the reaction mixture (25 iL of 50 mM KH2PO4, pH 4.6, 1 mM TRITON® X-100, 1 mg/mL BSA, and 5 μΜ of TMEM FRET substrate). Fifteen uL of twenty μΜ human B ACE2 (l-460);Fe (See Vasser, et ah, Science, 286, 735-741 (1999)) in KH 2 PO4 buffer is then added to the plate containing substrate and the test compound to initiate the reaction. The conten t is mixed well on a plate shaker for 10 minutes. The RFU of the mixture at time 0 is recorded at excitation wavelength 430 ran and emission wavelengt 535 rail The reaction plate is covered with aluminum foil and kept in a dark humidified oven at room temperature for 16 to 24 hours. The RFU at the end of incubation is recorded with the same excitation and emission settings used at time 0. The difference of the RFU at time 0 and the end of incubation is representative of the activity of BACE2 under the compound treatment. RFU differences are plotted versus inhibitor concentration and a curve is fitted with a four-parameter logistic equation to obtain the IC50 value. (May, et ah, Journal ofNeuroscience, 31, 1650746516 (2011)).

The compound of Example 1 herein is tested essentially as described above and exhibits a BACE2 IC 50 of 479 nM ± 202, n=4(Mean ± SEM; SEM === standard error of the mean). The ratio of BACE1 (FRET IC50 enzyme assay) to BACE2 (TMEM27 FRET IC50 assay) is about 400-fold, indicating functional selectivity for inhibiting the BACE1 enzyme. The data set forth above demonstrates that the compound of Example 1 is selective for BACE1 over BACE2. SH-SY5YAPP695Wt Whole Cell Assay

The routine whole cell assay for the measurement of inhibition of BACE1 activity utilizes the human neuroblastoma cell line SH-SY5Y (ATCC Accession No. CRL2266) stably expressing a human APP695Wt cDN A. Cells are routinely used up to passage number 6 and then discarded.

SH-SY5YAPP695Wt cells are plated in 96 well tissue culture plates at 5.0* 10 4 cells/well in 200 \iL culture media (50% MEM/EBSS and Ham's F12, 1. x each sodium pyruvate, non-essential amino acids and Na bicarbonate containing 10% FBS). The following day, media is removed from the cells, fresh media added then incubated at 37 °C for 24 hours in the presence/absence of test compound at the desired concentration range.

At the end of the incubation, conditioned media are analyzed for evidence of beta- secretase activity by analysis of Abeta peptides 1-40 and 1 -42 by specific sandwich ELISAs. To measure these specific isoforms of Abeta, monoclonal 2G3 is used as a capture antibody for Abeta 1-40 and monoclonal 21 F 12 as a capture antibody for Abeta 1-42. Both Abeta 1-40 and Abeta 1 -42 ELISAs use biotinylated 3D6 as the reporting antibody (for description of antibodies, see John son- Wood, et ah, Proc. Natl. Acad. Sci. USA 94, 1550-1555 (1997)). The concentration of Abeta released in the conditioned media following the compound treatment corresponds to the activity of BACE1 under such conditions. The 10-point inhibition curve is plotted and fitted with the four- parameter logistic equation to obtain the IC50 values for the Abeta-lowering effect. The compound of Example 1 is tested essentially as described above and exhibits the following activity for Abeta-lowering as shown in table 4,

Table 4

(Mean ± SEM; SEM = standard error of the mean) In vivo Inhibition of Beta-Secretase

Several animal models, including mouse, guinea pig, dog, and monkey, may be used to screen for inhibition of beta-secretase activity in vivo following compound treatment. Animals used in this invention can be wild type, transgenic, or gene Imockout animals. For example, the PDAPP mouse model, prepared as described in Games et ah, Nature 373, 523-527 (1995), and other non-transgenic or gene knockout animals are useful to analyze in vivo inhibition of Abeta and sAPPbeta production in the presence of inhibitory compounds. Generally, 2 month old PDAPP mice, gene knockout mice or non- transgenic animals are administered compound formulated in vehicles, such as com oil, beta-cyclodextran, phosphate buffers, PHARMASOLVE®, or other suitable vehicles via oral, subcutaneous, intra- venous, feeding, or other route of administration. One to twenty-four hours following the administration of compound, animals are sacrificed, and brains are removed for analysis of Abeta 1-x. "Abeta 1-x" as used herein refers to the sum of Abeta species that begin with residue 1 and end with a C-terrninus greater than residue 28. This detects the majority of Abeta species and is often called "total Abeta". Total Abeta peptides (Abeta 1-x) levels are measured by a sandwich ELISA, using monoclonal 266 as a capture antibody and biotinylated 3D6 as reporting antibody. (See May, et al., Journal ofNeuroscience, 31, 16507-16516 (2011)).

For acute studies, compound or appropriate vehicle is administered and animals are sacrificed at about 3 hours after dosing. Brain tissue, is obtained from selected animals and analyzed for the presence of Abeta 1-x. After chronic dosing brain tissues of older APP transgenic animals may also be analyzed for the amount of beta-amyloid plaques following compound treatment.

Animals (PDAPP or other APP transgenic or non-transgenic mice) administered an inhibitor}' compound may demonstrate the reduction of Abeta in brain tissues, as compared with vehicle-treated controls or time zero controls. For example, a 3, 10, and 30 mg/kg oral dose of Example 1, to young female PDAPP mice reduced Abeta 1-x peptide levels in brain hippocampus by 23% (non-significant), 43% (p<0.05), and 58% (p<0.01), respectively. In brain cortical tissue, doses of 3, 10, and 30 mg/kg of Example 1 reduced Abeta 1-x levels by 43%, 59%, and 73% (all values p<0.01) compared to vehicle-treated mice three hours after dosing. Given the activity of the Example 1 , against the BACEl enzyme in vitro, these Abeta- lowering effects are consistent with BACE inhibition in vivo, and further demonstrate CNS penetration of Example 1.

These studies show that compound of the present invention inhibits BACEl and is, therefore, useful in reducing Abeta levels.