![]() compound, use of a compound, pharmaceutical composition, and pharmaceutical product
专利摘要:
COMPOUND, USE OF A COMPOUND, PHARMACEUTICAL COMPOSITION, AND PHARMACEUTICAL PRODUCT The present invention relates to a fused aminodihydrothiazine derivative of formula (I): wherein X is hydrogen or fluorine; A is CH or N; Y is methyl, ethyl, monofluoromethyl, trifluoromethyl, difluoroethyl, methoxy, ethoxy, methoxymethyl or -C < 154 > N and its pharmaceutically acceptable salts; whose compound has an inhibitory effect on the production of Aß or an inhibitor of BACE1 and is useful as a prophylactic or therapeutic agent against a neurodegenerative disease caused by Aß and typified by dementia of the Alzheimer type. 公开号:BR112013018602A2 申请号:R112013018602-0 申请日:2012-01-20 公开日:2020-09-01 发明作者:Adrian Hall;Christopher Neil Farthing;Jose Luis Castro Pineiro 申请人:Eisai R&D Management Co., Ltd.; IPC主号:
专利说明:
DECLARATION Regarding the Priority Document The Applicant declares, for the purposes of article 16, § 2 of Law 9.279 of 05/14/96 and, in accordance with item 3.1 of Normative Instruction 17/2013 or, if applicable, in accordance with with Article 25 of Resolution 77/2013, or even with Article 8 of Normative Instruction 13/2013, that the identifying data of the priority document are as follows: Applicant EISAI LIMITED, European Knowledge Center, Mosquito Way, Hatfield, Herts AL 9SN, United Kingdom. Deposit Date January 21, 2011. Deposit country UNITED KINGDOM. Order Number 1101l40.0 Title FUSED AMINODIHYDROTHIAZINE DERIVATIVES. Furthermore, the Applicant declares that the information above is true. Rio de Janeiro, T [iqAAns) m p / p Kasznar Leonardos Intellectual Property P119190 HGPassos Tatiana Almeida Silvelra Property Agent Mustriai MaWcda n ° 1677103 "COMPOUND, USE OF A COMPOUND, PHARMACEUTICAL COMPOSITION, AND PHARMACEUTICAL PRODUCT" The present invention concerns a derivative of fused aminodidothrotiazine and its pharmacological use. More particularly, this The invention relates to a fused aminodi-idrotiazine derivative having an inhibitory effect on the production of amyloid- [3 'protein (hereinafter referred to as Aj3) or an inhibiting effect of amyloid-B precursor protein cleavage enzyme. beta 1 (hereinafter referred to as BACEI or beta-secretase) and is effective for the treatment of a neurodegenerative disease caused by the Aj3 protein, in particular, Alzheimer's-type dementia, Down syndrome or the like and a pharmaceutical composition comprising the derivative of fused aminodi-idrotiazine as an active ingredient. Akheimer's disease is a disease characterized by degeneration and loss of neurons as well as the formation of senile plaques and neurofibrillary tangles. Currently, only the symptoms of Ahheimer's disease are treated using a symptom-enhancing agent typified by an acetylcholinesterase inhibitor and a drug remedy} to inhibit disease progression has not yet been developed. It is necessary to develop a method to control the cause of the onset of the pathology in order to create a fundamental remedy against Alzheimer's disease. It is assumed that Af3-proteins as breakdown products of amyloid precursor proteins (hereinafter referred to as APP) are highly involved in the degeneration and loss of neurons and onset of dementia symptoms. The Af3-proteins have, as main components, Af340 consisting of 40 amino acids and Af342 with two amino acids added at the C-terminus. A {340 and AB42 are known to be highly prone to aggregation and are the main components of senile plaques. In addition, Aj340 and AÍJ are known to be increased by mutations in APP and presenilin genes that are observed in familial Alzheimer's disease. consequently, a compound that reduces the production of AB40 and A) 342 is expected to be a disease progression inhibitor or prophylactic agent against Alzheimer's disease. : A | 3 is produced by APP cleavage by beta-secretase '5 (BACEI) and subsequently by gamma-secretase. For this reason, attempts have been made to create gamma-secretase and beta-secretase inhibitors in order to inhibit Af3 production. The published international patent application WO2011 / 005738 (Eli Lilly and Company) describes compounds of the formula (A) and their use as 10 BACE inhibitors: "O '"' xSí "'' ^ '(R3) p where R', Rj R3, X, m, nep are defined in this. Aminodi-idrotiazine compounds of the formula (B) have already been disclosed in published International patent application WO2009 / 091016 (Eisai R&D Management Co., Ltd.): 1a) —L- (B):) jj) R '( B) R4 'R3 15 where ring A represents a C6-14 aryl group or the like; L represents NR'CO- [where R 'represents a hydrogen atom or the like] or similar; ring B represents a C 6 -14 aryl group or the like; X represents a C1-3 alkylene group or the like; Y represents a simple or similar link; Z represents a C1-3 alkylene group or the like; RJ and R2 independently represent a hydrogen atom or semelbante; and R3, R4, R5 and R6 independently represent a hydrogen atom, a halogen atom or the like. 5 The additional fendid aminodihydrazide compounds of formula (C) have been disclosed in published patent application WO20l0 / 038686 (Eisai R&D Management Co., Ltd.): (A) —L — jb) R5 R6l R1,) ' U-, 2 (C) Y), À R4 'R3 wherein ring A represents a C6J4 aryl group or the like; L represents —NR'CO- [where R "represents a hydrogen atom or the like] or similar; ring B represents a C6-14 aryl group or the like; X represents a Cl-3 alkylene group or the like; Y represents a single or semi-bonding; Z represents an oxygen atom or similar; R1 and R2 each independently represents a hydrogen atom or similar; and R3, RJ, R5 and R6 each independently represents a hydrogen atom, a halogen atom or similar. The present invention represents a selection of the genus of compounds disclosed in WO2009 / 091016. An object of the present invention is to provide additional compounds having an inhibiting effect on Afil production or an inhibiting effect on BACEI and are useful as a pronlatic or therapeutic agent against a neurodegenerative disease caused by Af3 and typified by dementia of the Ahheimer type, whose compounds are derived of fimed aminodi-idrotiazine. In this way, the present invention provides the compound of ) formula (1) · 4. XNH oQjÍ ')!): "'" '~ F, Ê' where X is hydrogen or fluorine; AéCHouN; 5 Y is methyl, ethyl, && monofluoromethyl, difluoromethyl, trifluoromethyl, difluoroethyl, methoxy, ethoxy, methoxymethyl or -C "N; and their pharmaceutically acceptable salts. In an embodiment of the present invention, X is hydrogen. In another embodiment of the present invention, A is N. In another embodiment of the present invention, Y is methyl, monofluoromethyl, diRuoromethyl, trifluoromethyl or methoxy. A favored group of compounds of the present invention is the compound of formula (Ia) and its pharmaceutically acceptable salts: OyjÍ '' ,, l)) 'NH2 "" F, È' where Y C as previously defined. Preferably, Y is methyl, monofluoromethyl, difhioromethyl, trifluoromethyl, difluoroethyl, methoxy, ethoxy or methoxymethyl. In one embodiment, the present invention provides a Z is a compound of formula (Ia) in which Y is methoxy or monofluoromethyl. Another favored group of compounds of the present invention is the compound of the formula (lb) and its pharmaceutically acceptable salts: ojÍ:, l) 'NH, "" F, Ê i: where Y is as previously defined. Preferably, Y is methyl, monofluoromethyl, difluoromethyl or methoxy. 10 Another favored group of compounds of the present invention is the compound of the formula (Ic) and its pharmaceutically acceptable salts: o ~ Á, | '1'NH2 "" ,, È À where Y is as previously defined. Preferably, Y is methyl, ethyl, trifluoromethyl, methoxy or -C * N. Preferred compounds of the present invention are: N- (3 - ((4aS, 5S, 7aS) -2-mino-5- (tnnuoromethyl) -4a, 5,7,7a- tetrahydro-4HAlro [3,4-d ] [1,3] thiazin-7a-yl) -4-nuorophenyl) -5-methoxypyrazine-2- carboxamide: o, u ° ") N) 'NH, FÂ' ÈF N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (Oduoromethyl) -4a, 5,7,7a- tetrahydro-4H-mro [3,4-d] [1,3] thiazine -7a-i]) - 4-nuorophenyl) -5-cyanopicol jt7 "" 'Ílj)' NH2 jF ': N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a- tetrahydro-4H-hro [3,4-d] [1,3] ti = in-7a-yl) -4-nuorophenyl) -5- (difluoromethyl) pyrazine-2-carboxamide: f) II: 1) H2 'Qf' N- (3 - (((4aS, 5S, 7aS) -2-amino -5- (trifluoromethyl) -4a, 5,7,7a- tetrai & o-4H-hro [3,4-d] [1,3] thiazin-7a-yl) -4-nuorofem]) - 5- (trifluoromethyl) picolinamide: Ç' F O ~ N ^ | J <; . Ú) "" FÁ 'È'. > N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tetrahydro-4H-hro [3,4-d] [1,3] thiuin -7a-yl) -4-nuoropheni!) - 5-methyl] pyrazine-2-carboxamide: o) U ', l)' NH2 '/' F ' N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (triMoromethyl) -4a, 5,7,7a- teÜhydro-4H-Nro [3,4-d] [1,3] tiazin-7a-yl) -4-nuorophenyl) -5-methylpicolinamide: o> 7 F!> j) HNH, O ', S FÂ' ÈF. > N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tetrahydro-4HA] ro [3,4-d] ijl, 3] thiazin- 7a-yl) -4-fluorophenyl) -5-ethi1picolinamide: ojj ', JJNH,' ÇF ' N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tehydro-4H-hrol3,4-d] [1,3] thiazin- 7a-yl) -4-nuoropheni]) - 5- (nuoromethyl) pyrazine-2-carboxamide: Og) ' jjNH, FÂ 'ÈF. N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (tMuoromethyl) -4a, 5,7,7a tehydro-4H-hro [3,4-d] [1,3] thiam-7a-iI) -4-nuorophenyl) -5-methoxypicolinamide: oj "°" ) N) 'NH, FAN' is F. : N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (trinuoromethi]) - 4a, 5,7,7a- tetrahydro-4HAüro [3,4-d] [1,3] thiazinja -il) -4Auorophenyl) -5-ethoxypyrazine-2-carboxamide: oyjC {om F CN) HNH2 The 'rS FÃ 'ÈF. - N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tetrahydro-4H-hro [3,4-d] [1,3 ] thiazin-7a-yl) -4-nuorophenyl) -5- (1,1-difluoroethyl) pyrazine-2-carboxamide: F , F ojj f i>: N) HNH, O 'S' ¢ F '. N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tehydro-4H-Wro [3,4-d] [1,3] tiazin- 7a-yl) -4-nuoropheni]) - 5- (triAuoromethyl) pyrazine-2-carboxamide: F , F o) ü ^ ' FC NH ° jj "'' ÇF '> N- (3 - ((4aS, 5S, 7aS) -2- = ino-5- (tnnuoromethyl) -4a, 5,7,7a- tetrahydro-4H-hro [3 , 4-d] [1,3] thiazin-7a-i |) -4-nuorofeml) -5- (methoxymethyl) pyrazine-2-carboxamide: °) ü ^ ° "F CN) HNH, O ', S' ÇF ' N- {3 - [(4aS, 5S, 7aS) -2-amino-5- (tnnuoromethyl) -4a, 5-di-i & o- 4H-fiiro [3,4 d1 [1,3] tiazin-7a (7H ) -yl] -4-nuorophenyl} -5 - [(2H3) methyloxy] pyrazine-2-carboxamide: 10 o, u2iH ': J))' nh2 'Gf' N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (tnnuoromethyl) -4a, 5,7,7a tehydro-4H-Nro {3,4-d] [1,3] thiazin -7a-i!) - 4,5-difluorophenyl) -5- (dif1uoromethyl) pyrazine-2-carboxamide: F OyXj '')) ',,, FÀ' Èf .7 N- (3 - (((4aS, 5S, 7aS) -2-amino-5- (trièuoromethyl) -4a, 5,7,7a- tetrahydro-4HAi ] ro [3,4-d] [l3] thiazin-7a-yl) -4,5-difluorophenyl) -5-methoxypyrazine-2-carboxamide: o% u ° "":, I)) ,, FÀ ' ÈF 7 N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (triAororomethyl) -4a, 5,7,7a- teUhydro-4H-Hro [3,4-d] [l, 3] thiazin-7a-yl) -4,5-dinuorophenyl) -5-methylpyrazMo-2-carboxamide: ç3t "')) NH," t f- {' ff. N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tetrahydro-4H-hro [3,4-d] [1,3] thiazin- 7a-yl) -4,5-dinuorophenyl) -5- (nuoromethyl) -pyrazine-2-carboxamide: F) 7 and its pharmaceutically acceptable salts. In one embodiment, the present invention provides a compound that is N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4H -füro [3,4-d] [1,3] thiazin-7a-i |) -4-nuorophenyl) -5-methoxypyrazine-2-carboxamide or a pharmaceutically acceptable salt thereof. In another embodiment, the present invention provides a compound that is N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) - 4a, 5,7,7a- tetrahydro-4HAüro [3,4-d] [1,3] thiazin-7a-yl) -4-fluorophenyl) -5- (fluoromethyl) pyrazine-2-carboxamide, or a pharmaceutically acceptable salt thereof. Specific compounds within the scope of this invention include those named in the Examples below and their pharmaceutically acceptable salts. As used herein, the term "difluoroethyl" refers to an alkyl group having two carbon atoms and replaced by two fluorine atoms. Examples of the group are CH, -CF, -, CH, F-CHF- and CHF, -CH, -. In the present invention, the group is preferably CH3-CF2-. The compound of formula (I) is not limited to a specific isomer and includes all possible isomers (such as a ketoenol isomer, an imine-enamine isomer and a rotamer) and mixtures thereof. For example, the compound of formula (I) includes the following tautomers: ojjÍ "oj; í 'xnh x), Nh)) j"' ")) Í:" The compounds of the present invention contain three chiral centers licalized in the tetrahydrofúro-thiazinyl ring within formula (J). The stereochemical configuration at each of these chiral centers is preferably S, that is, these are stereoisomers (4aS, 5S, 7aS). For the avoidance of doubt, the stereoisomers (4aS, 5S, 7aS) of the present invention can be present as a mixture with one or more of the other possible stereoisomers, for example, in a racemic mixture. In one embodiment, the present invention provides a compound of formula (I) that is stereochemically pure at the chiral centers' (4aS, 5S, 7aS). In the context of the present specification, the term stereochemically pure indicates a compound having 80 ° /) or greater by the weight of the stereoisomer (4aS, 5S, 7aS) and 20 ° 4 or less by weight of the other stereoisomers. In an additional embodiment, the compound of formula (J) is 90 "/, or greater in weight of the stereoisomer (4aS, 5S, 7aS) and 10 ° /) or less in weight of the other stereoisomers. In yet another form In addition, the compound of formula (I) has 95 ° /, or greater by weight of the stereoisomer (4aS, 5S, 7aS) and 5 ° /) or less by weight of the other stereoisomers. , the compound of formula (I) is 97 ° /) or greater by weight of the stereoisomer (4aS, 5S, 7aS) and .3 ° / 0 or less by weight of the other stereoisomers. In this specification, although the crystalline polymorphs of Compound may be present, the compound is simially not "5 Limit to it and may be present as a simple crystalline form or a mixture of simple crystalline forms. The compound can be an anhydride or a hydrate. Any of these forms is included in the claims of this specification. The present invention also includes the isotopically labeled compounds 10, which are identical to the compounds of the form (I)> except one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number observed in natural state. Examples of isotopes that can be incooperated in compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine, phosphorus, chlorine, technetium and iodine, such as 2H, 3H, 11, 14C,! 3N, 15O , l8F, 32p, 99mTC, l23j and 13lj. The compounds of the present invention and the pharmaceutically acceptable derivatives (for example, salts) of said compounds which contain the isotopes already mentioned and / or other isotopes of other atoms 20 are within the scope of the present invention. The isotopically labeled compounds of the present invention, for example, those in which radioactive isotopes, such as 3H and / or 14C are incorporated, are drug-dependent and / or substrate tissue distribution assays. 3H and 14C are considered useful due to their ease of preparation and detectability. The 25 'lç, l'O and l8F isotopes are considered useful in PET (positron emission tomography) and the 99 "TC, 123I and l3j isotopes are considered useful in SPECT - - (computed tomography emission tomography) , all useful in brain imaging. Substitution with the heavier isotopes such as 2H can produce certain therapeutic advantages that result from increased metabolic stability, for example, increased in vivo half life or reduced dosage requirements and are therefore considered useful in some circumstances The isotopically labeled compounds of formula (I) of this invention can generally be prepared by carrying out. "5 of the procedures disclosed in the Schemes and / or in the Examples below, by substituting an easily available isotopically labeled reagent for a non-isotopically labeled reagent. The aminodi-idrotiazine derivative of formula (I) according to the present invention can be a pharmaceutically acceptable salt! 10 Pharmaceutically acceptable salts include those described by Berge, Bighley and Monkhouse, J. Pharm. Sci., 1977, 766, 1-19. Specific examples of pharmaceutically acceptable salts include salts of organic acids (such as such as sulfates, nitrates, perchlorates, phosphates, carbonates, bicarbonates, fluorides, chlorides, bromides and iodidides), organic carboxylates (such as acetates, oxalates, maleates, tartrates, fumarates, citrates, malonates and lactates), organic sulfonates ( such as methanesulfonates, trifluoromethanesulfonates, ethanesulfonates, benzenesulfonates, toluenesulfonates and camphorsulfonates), amino acid salts (such as as aspartates and glutamates), quaternary amine salts, 20 alkali metal salts (such as sodium salts and potassium salts) and alkaline earth metal salts (such as magnesium salts and calcium salts). The compound of formula (I) according to the present invention can be converted to a pharmaceutically acceptable salt by a conventional method "when necessary. The salt can be prepared by a method in 25 that the methods typically used in the field of organic synthetic chemistry and the others are appropriately combined. Specific examples of the method include neutralization titration of a free solution of the compound of the present invention with an acidic solution. The final aminodi-idrotiazine derivative of formula (I) or pharmaceutically acceptable salt according to The present invention can be a solvate thereof Examples of a solvate include a hydrate The compound of the formula (I) according to the present invention can be converted to a solvate by subjecting the compound to a known 5-forming reaction per se when necessary The present invention further provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in therapy. The fused aminodi-idrotiazine derivative or pharmaceutically acceptable salt thereof or solvate thereof according to the present invention has an excellent AB production inhibitory effect or BACEI inhibitory effect and is useful as a prophylactic or therapeutic agent against a neurodegenerative disease caused by Ajj and typified by Alzheimer's type dementia. The compounds of the invention reduce both Aj340 and A | 342. In addition, the compounds of the present invention may have a BACE 2 inhibitory effect. In this way, in another aspect, the present invention provides a compound of formula (I) as defined above, or a pharmaceutically acceptable salt thereof, for inhibit the production of amyloid-B protein. In another aspect, the present invention provides a compound of the formula (I) as defined above, or a pharmaceutically acceptable salt thereof, to inhibit beta 1 amiioid precursor protein cleavage enzyme (BACE 1) ). In another aspect, the present invention provides a compound of formula (I) as defined above, or a pharmaceutically acceptable saj thereof, for the treatment of a neurodegenerative disease. Examples of neurodegenerative diseases include Alzheimer's dementia (AD), Down syndrome, cerebrovascular amyloid angiopathy (CAA), mild cognitive impairment (MCI), memory loss, pre-senile dementia, senile dementia, hereditary cerebral hemorrhage with amyloidosis and other degenerative dementias such as mixed degenerative vascular dementias, such as mixed degenerative vascular dementias, dementia associated with basal cortical degeneration, '5 dementia associated with Parkinson's disease (PD) and dementia associated with diffuse Lewy body type AD. In one embodiment, the neurodegenerative disease is Ahheimer-type (AD) dementia. In another aspect, the invention provides for the use of a compound of formula (I) as defined above, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment or prevention of a neurodegenerative disease, such as dementia of the type AlZheimer (AD), Down syndrome, cerebrovascular amyloid angiopathy (CAA), mild cognitive deterioration (MCI), memory loss, pre-senile dementia, senile dementia, hereditary cerebral hemorrhage with amyloidosis and i5 other degenerative dementias such as dementia dementias vascular origin of mixed degenerative, such as dementia of vascular origin of mixed degenerative, dementia associated with basal cortical degeneration, dementia associated with Parkinson's disease (PD) and dementia associated with diffuse type AD Lewy body. In a form of rehabilitation, neurodegenerative disease is Alzheimer's-type dementia (AD). In another aspect, the invention provides a method of inhibiting the production of amyloid-B protein and / or treating or preventing neurodegenerative disease, such as Alzheimer's dementia (AD), P Down, cerebrovascular amyloid angiopathy (CAA), mild cognitive impairment (MCI), memory loss, pre-senile dementia, senile dementia, hereditary cerebral hemorrhage with amyloidosis and other degenerative dementias such as mixed degenerative vascular dementias, such as mixed degenerative vascular dementias, dementia associated with basal cortical degeneration, dementia associated with Parkinson's disease (PD) and dementia associated with diffuse Lewy body type AD, which involves administration to a human patient in need. a therapeutically or profiquely effective amount of> a compound of formula (I) or a pharmaceutically acceptable salt thereof. Examples of neurodegenerative diseases include those listed above. In one embodiment, the neurodegenerative disease is Akheimer-type (AD) dementia. "Effective amount" means an amount sufficient to cause a benefit to the patient or at least to cause a change in the patient's condition. Additional conditions that can be treated by the compounds of the present invention include type 2 diabetes, Creutzfield-jakob disease (CJD), peripheral nerve damage, peripheral neuropathy, progressive supranuclear palsy, stroke, amyotrophic lateral sclerosis (ALS), autoimmune diseases, inflammation, arterial thrombosis, anxiety disorders, psychotic disorders, epilepsy, attacks, convulsions, tension disorders, vascular amyloidosis, pain, Gerstmann-Straeussler-Scheinker syndrome, scrapie, encephalopathy, cerebellar spinal ataxia , Wilson's disease, Graves' disease, Huntington's disease, Whipple's disease, Kostmann's disease, glaucoma, hereditary cerebral hemorrhage with amyloidosis, cerebral hemorrhage with amyloidosis, vascular amyloidosis, cerebral inflammation, Eagile X syndrome, stroke, syndrome Tourette, inclusion body myositis, tension disorders, depression, bipolar disorder and obsessive compulsive disorder. In one aspect, the present invention further provides a compound of formula (I) as defined above, or a pharmaceutically acceptable salt thereof, for the treatment of type 2 diabetes. In another aspect, the present invention further provides the use of a compound of formula (I) as defined above, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicinal product for J '" 18 the treatment or prevention of type 2 diabetes. Yet, in another aspect, the present invention further provides a method of inhibiting the production of B-alnyloid protein and / or treating or preventing type 2 diabetes that involves administration to a "5 human patient in need of a therapeutically or prophylactically effective amount of a compound of the formula (I) or a pharmaceutically acceptable salt thereof. Another aspect of the invention provides a pharmaceutical composition comprising a compound of formula (I) as defined 10 above, or a pharmaceutically acceptable salt thereof, as the active ingredient in association with a pharmaceutically acceptable carrier. the composition can be in any suitable form, depending on the intended method of administration. For example, this can be in the form of a tablet, capsule or liquid for oral administration or a solution or suspension for parenteral administration. The fused aminodi-idrotiazine derivative or pharmaceutically acceptable salt thereof according to the present invention can be formulated by a conventional method. Preferable examples of the dosage form include tablets, coated tablets, such as 20-film tablets and sugar-coated tablets, soft granules, granules, powders, capsules, syrups, lozenges, inhalants, suppositories, injections, ointments, eye drops, drops nasal drops, ear drops, poultices and lotions. These solid preparations, such as tablets, capsules, granules and powders can generally contain from 0.01 to 100 ° /) by weight and preferably from 0.1 to 100 ° /) by weight of the aminodi-idrotiazine derivative molten or pharmaceutically acceptable salt thereof according to the present invention as an active ingredient. The active ingredient is stimulated by the combination of ingredients commonly used as materials for a pharmaceutical preparation and the addition of an excipient, a disintegrant, a binder, a lubricant, a dye and a concealer typically used and add a stabilizer, an emulsifier, an absorption inducer, a surfactant, a pH adjuster, a preservative and an antioxidant when needed, for example 5 example, using a conventional method. Examples of such ingredients include animal and vegetable oils, such as soybean oil, beef tallow and synthetic glyceride; hydrocarbons, such as liquid paraffin, squalane and solid paraffin; Ester oils, such as octyldodecyl myristate and isopropyl myristate; higher alcohols, such as cetostearyl alcohol and 10 behenyl alcohol; a silicone resin; silicone oil; surfactants, such as polyoxyethylene fatty acid ester, " sorbitan, glycerol fatty acid ester, polyoxyethylene sorbitan fatty acid ester, polyoxyethylene Hydrogenated castor oil and a polyoxyethylene-polyoxypropylene block copolymer; water-soluble polymers, such as 15 such as hydroxyethylcellulose, polyacrylic acid, a carboxyvinyl polymer, polyethylene glycol, polyvinylpyrrolidone and methylcellulose; lower alcohols, such as ethanol and isopropanol; polyhydric alcohols, such as glycerol, propylene glycol, dipropylene glycol and sorbitol; sugars, such as glucose and sucrose; inorganic powders, such as silicic anhydride, magnesium aluminum silicate and 20 aluminum silicate and purified water. Examples of the excipient used include lactose, piss starch, sucrose, glucose, mannitol, sorbitol, crystalline cellulose and silicon dioxide. Examples of the binder used include polyvinyl alcohol, polyvinyl ether, methyl cellulose, ethyl cellulose, gum arabic, tragacanth, gelatin, shellac, hydroxypropylmethylcellulose, 25 hydroxypropylcellulose, polyvinylpyrrolidone, a block copolymer of polypropylene glycol-polyoxyethylene "and meglumine. Examples of disintegrants include starch, agar, gelatin powder, crystalline cellulose, calcium carbonate, sodium bicarbonate, calcium citrate, dextrin, pectin and /) carboxymethylcellulose calcium. Examples of the lubricants used include magnesium stearate, talc, polyethylene glycol, silica and hydrogenated vegetable oil. Examples of the dye used include those allowed to be added to pharmaceutical products. Examples of the correctives used include cocoa powder, menthol, empasm, peppermint oil, bomeol and powder. '5 cinnamon. Obviously, the ingredients are not limited to the above additive ingredients. For example, an oral preparation is prepared by the addition of the fused aminodi-idrotiazine derivative or pharmaceutically acceptable salt thereof according to the present invention as an active ingredient, an excipient and, when necessary, a binder, a disintegrant, a fertilizer, a dye, a concealer and others and then forming the mixture into powder, fine granules, granules, tablets, coated tablets, capsules or the like by a conventional method. obviously, tablets or granules can be properly coated, for example, coated with sugar 15 when needed. For example, a syrup or preparation for injection is prepared by adding a pH adjuster, a solubilizer, an ionizing agent and others and a solubilizing agent, a stabilizer and others when needed by a conventional method. The injection can be a 20 previously prepared solution or can be powder alone or powder containing a suitable additive, which is dissolved before use. The injection can usually contain from 0.01 to 100 ° / by weight and preferably from 0.1 to 100 ° by weight of the active ingredient. In addition, a liquid preparation for oral administration such as a suspension or syrup may contain 25 usually from 0.01 to 100 ° /) by weight and preferably from 0.1 to 100 ° /) by weight of the active ingredient. For example, an extreme preparation can be prepared by any conventional method without specific limitations. As a base material, any one of several materials usually used for a pharmaceutical, quasi-drug, cosmetic or the like can be used. Examples of the base material include materials, such as C such as animal and vegetable oils, mineral oils, "ester oils, waxes, higher alcohols, fatty acids, silicone oils, surfactants, phospholipids, 5 alcohols, polyhydric alcohols such as water-soluble polymers, clay minerals and purified water, a pH adjuster, an antioxidant, a chelator, a preservative and fungicide, a dye, a flavoring agent or the like can be added when needed. In addition, ingredients such as an ingredient having a differentiating inducing effect, a blood flow enhancer, a bactericide, an antiphlogistic, a cell activator, vitamin, amino acid, a humectant and a keratolytic agent can be combined when necessary The dose of the aminodi-idrotiazine derivative or the pharmaceutically acceptable salt thereof according to the present invention varies according to the degree of symptoms, age, sex, body weight, mode of administration, type of salt and specific type of disease, for example. In addition, the active ingredient is orally administered to an adult from 30 µg to 10 g, preferably 100 µg to 5 g and more preferably 100 µg alg per day or is administered to an adult by injection around 20 30 µg a 1 g, preferably 100 µg to 500 mg, more preferably, 100 µg to 300 mg per day, in one or several doses, respectively. The compounds of formula (I) can be used in combination with other therapeutic agents, for example, drugs claimed to be useful as a disease modifier or symptomatic treatments of a neurodegenerative disease, such as Alzheimer's disease. Thus, in another aspect, the present invention provides a pharmaceutical product which comprises, in combination, a first active ingredient which is a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one additional useful active ingredient. in the treatment of a neurodegenerative disease. In an embodiment of the invention, the neurodegenerative disease is Alzheimer's-type (AD) dementia. Suitable examples of such additional active ingredients can be symptomatic agents, for example, those known as 5 by Inodinc.in cholinergic transmission! as M1 and M3 muscarinic receptor agonists or allosteric modulators, M2 muscarinic antagonists, M4 agonists or positive allosteric modulators (PAMs), acetylcholinesterase inhibitors (such as tetrahydroaminoacridine, donepezil hydrochloride and rivastigmine), nicotinic receptor agonists (10) such as Ct7 agonists or allosteric modulators or a4j32 agonists or allosteric modulators), PPAR agonists (such as PPARy agonists), 5-HT4 receptor agonists or partial agonists, H3 histamine agonists, 5-HT6 receptor antagonists or 5HT1a receptor ligands and NMDA receptor antagonists or modulators, 15 5-HT2a antagonists, 5-HT7 antagonists, D1 or PAMS agonists, D4 or PAMS agonists, D5 or PAMS agonists, GABA- reverse agonists A5 or negative allosteric modulators (NAMs), GABA-A a2 / 3 agonists or PAMS, mGluR2 modulators (PAMs or NAMs), mGluR3 PAMS, mGluR5 PAMs, inhibitors of PDE 1, PDE 2 inhibitors, 20 PDE 4 inhibitors, PDE 5 inhibitors, PDE 9 inhibitors, PDE 10 inhibitors, GlyTl inhibitors, DAAO inhibitors, ASCl inhibitors, AMPA modulators, SIRTI activators or inhibitors , AT4 antagonists, GalRI antagonists, GalR3 ligands, adenosine A1 antagonists, A2a adenosine antagonists, a2A antagonists or agonists, 25 selective or non-selective norepinefPine reabsorption inhibitors (SNRIs) or "disease-modifying agents" such as gamma secretase inhibitors or modulators, alpha secretase activators or modulators, amyloid aggregation inhibitors, amyloid antibodies, tau aggregation inhibitors or inhibitors of tau / kinase phosphorylation, tau dephosphorylation / phosphatase activators, inhibitors of mitogen activated protein kinase kinase 4 (MKK4 / MEK4 / MAP2K4), inhibitors of c-jun N-terminal kinase (JNK), casein kinase inhibitors, MK2 inhibitors (protein activated by r protein kinase mitogen activated by protein kinase 2), "5 inhibitors of MARK (microtubule affinity regulating kinase), inhibitors of CDK5 (cyclin-dependent kinase 5), inhibitors of GSK-3 (glycogen synthase kinase-3) and inhibitors of tau-tubulin kinase-l (TTBKI). Additional examples of such other therapeutic agents can be calcium channel blockers, HMG-CoA inhibitors (3-hydroxy-3-methyl-10 giutaryl-CoA) reductase (statins) to lipid-reducing agents, imitations of NGF (factors nerve development), antioxidants, ") GPR3 ligands, plasmin activators, neprilisin activators (NEP), IDE (insulin-degrading enzyme) activators, melatonin MTI and / or MT2 agonists, TLX / NR2E1 ligands (X-tailless receptor), GluRl ligands, 15 RAGE antagonists (receptor for advanced glycation end products, EGFR inhibitors (epidermal development factor receptor), FPRL-I ligands (l-formyl peptide-like receptor) , GABA antagonists and MICAL inhibitors cell that interacts with casL), for example oxoreductase, CBl antagonists / inverse agonists, 20 non-steroidal anti-inflammatory drugs (NSAIDs), anti-inflammatory agents (for example, agents that can be used to treat neuroinflammation by intensifying or reducing neuroinflammation), amyioid precursor protein (APP) ligands, anti-amyloid vaccines and / or antibodies, agents that promote or enhance amyloid efkixo and / or release, histone deacetylase inhibitors (HDAC), EP2 antagonists, ll-beta HSDI (hydroxysteroid dehydrogenase) inhibitors, liver X receptor (LXR) agonists or PAMs, imitations of lipoprotein receptor (LRP) -related protein and / or ligands and / or enhancers and / or inhibitors, inhibitors of butyryl cholinesterase, cinurinic acid antagonists and / or cinurenine aminotransferease (KAT) inhibitors, FF orphanage antagonists / nociceptin (NOP) / l agonist-like receptor (ORLI), trans ligands carrier of excitatory amino acid (EAAT) (activators or inhibitors) and inhibitors of plasminogen activator-1 inhibitor (PAl-1), niacin and / or GPR109 agonists or PAMs in combination with cholesterol-lowering agents and / or HMGCoA reductase inhibitors (statins), dimebolin or similar agents, antihistamines, metal binding / chelating agents, antibiotics, hormone secretagogues development agents, cholesterol-lowering agents, vitamin E, cholesterol absorption inhibitors, promoters of cholesterol efflux and / or insulin activators and super-regulating agents. In one embodiment, the present invention provides a pharmaceutical product that comprises, in combination, a first active ingredient that is a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one other active ingredient selected from: - 0 Cholinesterase inhibitors, for example, donepezil, galantamine, rivastigamine, tetrahydroaminoacridine and their pharmaceutically acceptable salts, 0 5-HT6 antagonists, for example, SB-742457 and their pharmaceutically acceptable salts, 0 HMGCoA reductase inhibitors, for example, lovastatin, rosuvastatin, atorvastatin, simvastatin, fluvastatin, pitavastatin, pravastatin and their pharmaceutically acceptable salts. The individual components of such combinations can be administered sequentially or simultaneously in separate or combined pharmaceutical formulations. Accordingly, the pharmaceutical product can, for example, be a pharmaceutical composition comprising the first and other active ingredients in admixture. and, Alternatively, the pharmaceutical product may, for example, comprise the first and other active ingredients in separate pharmaceutical preparations suitable for simultaneous, sequential or separate administration to a patient in need thereof. ) The combinations mentioned above can be conveniently presented for use in the form of a pharmaceutical formulation, and in this way pharmaceutical formulations comprising a combination as defined above together with a pharmaceutically acceptable carrier or excipient comprise another aspect of the invention. When a compound of formula (I) or a pharmaceutically acceptable salt thereof is used in combination with a second active therapeutic agent, the dose of each compound may differ from that when the compound is used alone. Appropriate doses will be easily estimated by those skilled in the art. In this way, a further aspect of the invention provides a method of preparing a pharmaceutical composition, which involves mixing at least one compound of formula (I) as defined above or a pharmaceutically acceptable salt thereof, with one or more adjuvants, diluents or carriers pharmaceutically acceptable and / or with one or more other therapeutically or prophylactically active agents - In one embodiment, the present invention provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, one or more others agents for the treatment of Ahheimer's disease, such as an M1 and M3 muscarinic 25 receptor agonist or allosteric modulator, an M2 muscarinic antagonist, an acetylcholinesterase inhibitor, a nicotinic receptor agonist or allosteric modulator, a PPAR agonist 5-HT4 receptor agonist or partial agonist, a histamine H3 agonist, a 5-HT6 receptor agonist, a receptor ligand 5HTlA ptor, an antagonist or - "" "~ 26 NMDA receptor modulator, a 5-nT2A antagonist, a 5-HT7 antagonist, a DI agonist or positive allosteric modulator (PAM), a D4 or PAM agonist, an inverse GABA-A ct5 agonist or & ' negative allosteric modulator (NAM), a GABA-A a2 / 3 agonist or '5 PAM, a mGluR2 modulator (PAM or NAM), a mGlulü PAM, a mGluR5 PAM, a PDE 1 inhibitor, a PDE 2 inhibitor, a PDE 4 inhibitor, a PDE 5 inhibitor, a PDE inhibitor 9, a PDE 10 inhibitor, a GlyTl inhibitor, a DAAO inhibitor, an ASCl inhibitor, an AMPA modulator, an SIRTI activator or inhibitor, an AT4 10 antagonist, a GalRl antagonist, a GaiR3 ligand , an adenosine A1 antagonist, an A2a adenosine antagonist, an a2A antagonist or agonist, a selective or non-selective norepinehine reabsorption inhibitor (SNRI), a secretase gamma inhibitor or modulator, an alpha secretase activator or modulator, an amyloid aggregation inhibitor, a 15 amyloid anti-type, an inhibitor of tau aggregation, an inhibitor of phosphorylation of tau, an inhibitor of MK2 (protein kinase 2 activated by itogen-activated protein kinase), a MARK inhibitor (microtubule affinity regulating kinase), inhibitors CDK5 (cyclin-dependent kinase 5), GSK-3 inhibitors (glycogen synthase kinase-3) and 20 inhibitors of tau-tubulin kinase-1 (TTBKI). Additional examples of such other therapeutic agents can be calcium channel blockers, HMG-CoA inhibitors (3-hydroxy-3-methyl-glutaryl-CoA) reductase (statins) to lipid-lowering agents, imitations of NGF (nerve development factors), antioxidants, GPR3 ligands, plasmin activators, neprilysin activators (NEP), IDE activators (insulin-degrading enzyme), melatonin MTI and / or MT2 agonists, TLX / NR2E1 ligands (X-tailless receptor), GluRl ligands, RAGE antagonists (receptor for RAGE advanced glycation end products, EGFR (epidermal development factor receptor) inhibitors, FPRL-I ligands (formyl peptide-like receptor-1), GABA antagonists and MICAL inhibitors (mojécula that interacts with casL), for example oxoreductase, CB 1 antagonists / inverse agonists, in association with a pharmaceutically acceptable carrier. In a further embodiment "5 the present invention provides a combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, together with a therapeutic agent as described above for sequential or simultaneous administration in separate pharmaceutical formulations or combined. In another aspect, the invention provides a method of inhibiting the production of B-nanoid protein and / or treating or preventing neurodegenerative disease, such as Akheimer-type dementia (AD), Down syndrome, cerebrovascular amyloid angiopathy (CAA ), mild cognitive impairment (MCI), memory loss, pre-senile dementia, senile dementia, 15 hereditary cerebral hemorrhage with amyloidosis and other degenerative dementias such as. dementias of mixed degenerative vascular origin, such as dementias of mixed degenerative vascular origin, dementia associated with basal cortical degeneration, dementia associated with Parkinson's disease (PD) and dementia associated with · Lewy 20 body type AD, the method involving administration to a human patient suffering from the condition, a therapeutically or prophylactically effective amount of the pharmaceutical composition described above or a compound of formula (I) as defined above, or a pharmaceutically acceptable salt thereof. "Effective amount" means an amount sufficient to cause a benefit to the patient or at least to cause a change in the patient's condition. Alzheimer's disease (AD) is pathologically characterized by the presence of neurofibrillary tangles (NFTs) and plaques, consisting of amyloid peptides (Aj3) peptides of varying lengths, for example 42 amino acids (A | 342) and 40 amino acids (Aj340). In addition to these pathological markers, cerebral atrophy is also evident. Plaque formation is believed to occur due to the aggregation of Afl peptides. AB peptides are formed in the brain by sequential cleavage of '5 amyloid precursor protein (APP) by f3-secretase (BACE-I) and Y-secretase. Therefore, potential AD drugs aimed at inhibiting amyloid formation by inhibiting BACE-I or y-secretase, must be able to achieve adequate exposure in the brain in order to exert an effect on AD. Although BACE-I represents an alternate target for 10 to interrupt or decrease the production of amyloid peptides, several groups have found it challenging to identify BACE-I inhibitors that can penetrate the central nervous system (CNS) and thereby inhibit the enzyme in place of action. The brain is protected by several barriers including the 15 blood brain barrier (BBB) and transporters (Hitchcock and Pennington, J Med Chem 2006, 29, 7559; Ueno, Curr. Med. Chem. 2007, i4, 1199; Gloor et al. , Brain Res. Rev. 2001, 36, 258). Several efkíxo transporters have been characterized preventing the compounds from entering the brain. One of the best characterized and most prominent in preventing the penetration of xenobiotic 20 CNS is P-glycoprotein (Pgp) (Kusuhara and Sugiyama, Drug Discovery Today, 2001, 6, 150; Mahar Doan et al., J. Pharm. Expt 2002, 303, 1029; Lin, Drugs of Today 2004, 40, 5; Lin & Yamazaki, 'Clin Pharmacokinet. 2003, 42, 59; Schirikel, Adv. Drug Deliv. Rev. 1999, 36, 179). Pgp efflux has been shown to be important for BACE-I 25 inhibitors (Hussain et al., J. Neurochem. 2007, 100, 802). In this way, the control of Pgp efflux is important. Those skilled in the art will estimate that there are several ways to measure or predict CNS penetration in vitro or in vivo. The potential for CNS penetration can be estimated in vitro by determining whether a compound can be subjected to Pgp efflux, that is, by conducting an in vitro Pgp assay. Those skilled in the art will estimate that several cell lines can be used and that these cell lines may or may not affect test results. One such test is' 5 described below (Cyprotex UK). The following MDR-I MDCK assay was used to estimate the flow of Pgp. The assay was conducted at Cyprotex Discovery Ltd. 15 Beeçh Lane, Maccles / íe / d, Cheshire, UK, SK / O 2DR Permeability of MDRI-MDCK (Bi-directional; pH 10 7.4 / pH 7.4) protocojo MDCK cells are an epithelial cell line of canine renal origin. These cells can be transfected to stably express the active P-glycoprotein (MDRI-MDCK) and are ideal for studying drug efflux. The test compound was added to the apical or basolateral iateral of a confluent monolayer of MDRI-MDCK cells and permeability was measured by monitoring the appearance of the test compound on the opposite side of the membrane using LC-MS / MS. For this, an apparent permeability / age coefficient / age 20 (Pa ,,) and e / 7uro ratio were measured / calculated. objective To measure the permeability of the test compound in the apical to basolateral (A-B) and basolateral to apical (B-A) direction using MDRI-MDCK cells. A permeability ratio B-A and A-B was calculated (efflux ratio 25) to show whether the compound undergoes the efflux of P-glycoprotein. The compounds were supplied as a 200 µL solution of 10 niM of test compound in DMSO. Experimental Procedure MDRI-MDCK cells obtained from NIH (Rockvijle, MD, USA) are used. Following the culture until the confluence, the monolayers were prepared by rinsing both apical and basolateral surfaces twice with buffer at pH 7.4 at 37 ° C. The Z cells " were then incubated with pH 7.4 buffer in compartments both '5 apical and basolateral for 40 minutes to stabilize the physiological parameters. The pH 7.4 buffer was then removed from the apical companion and replaced with the test compound dosing solutions. Solutions were prepared by diluting 10 niM of test compound in 10 DMSO with buffer to give a final test compound concentration of 10 µM (final DMSO concentration adjusted to 1 ° / 0). The yellow fluorescent integrity marker Lucifer was also included in the dosing solution. The apical inserts were then placed in 'company' plates containing Eesco buffer at pH 7.4. The patterns 15 analytics were made from dosing solutions. For basolateral to apical experiments (B-A) the experiment was started by replacing the plug in the inserts, then placing them on the company plates containing dosing solutions. Incubations were performed in an atmosphere of 5 ° /) CO2 with a relative unit of 95 20 ° / 0 to 37 ° C for 60 minutes. After the incubation period, the company plate was removed and the apical and basolateral samples were diluted for analysis by LC-MS / MS. The permeability of the test compound was estimated in duplicate. On each plate, compounds with known penneability characteristics 25 were performed as controls. Control test compounds were quantified by analyzing the LC-MS / MS cassette using a 5-point calibration with appropriate sample dissolution. Generic Cyprotex analytical conditions were used. The initial concentration (C0) was determined from the dosage solution m and the experimental recovery calculated from Cq and compartment concentrations both apical and basolateral. The integrity of the monolayers throughout the experiment was verified by monitoring the Lucifer yellow coloration using the '5 fluorometric analysis. Lucifer yellow permeation is low if the monolayers are not damaged. If a P, pp value of Lucifer yellow is above the QC limits in an individual test compound reservoir, then an n = 1 result has been reported. If the P, pp values of · Lucifer yellow were above the QC limits in both replica reservoirs 10 for a test compound, the compound was tested again. If on repeat, permeation and high Lucifer yellow was observed in both reservoirs then the inherent toxicity or fluorescence of the test compound was assumed. No experiments add! was done in this example. 15 Data Analysis The permeability coefficient for each compound (Papp) was calculated from the following equation: Pa ,, = (dQ dt) (Co x A) When dQ / dt is the rate of drug transfer through cells, Cq is the concentration of donor compartment in zero period and 20 A is the area of the cell monolayer. C0 was obtained from the analysis of the dosage solution at the beginning of the experiment. In addition, an efflux ratio (ER) was calculated from the average of data A-B and B-A. This is derived from: ER = ((Papp (B - A)) "((Papp (A - B)) Two control compounds were evaluated alongside the 25 test compounds, propranolol (highly permeable) and prazosin (a substrate for P-glycoprotein.) Surprisingly, the compounds of the present invention were found to have a lower Pgp efflux ratio than the compounds exemplified in WO2009 / 091016 indicating that they have the potential to show greater CNS penetration. Selected G's are shown in Table 1 below. "5 Table 1: MDR-I MZ Test Data) CkPgp Example P% ER Comparative Example 1 26.2 Comparative Example 2 16.6 Comparative Example 3 24.0 Comparative Example 4 20.7 2 1.7 3 1.4 4 1.0 1 0.7 Comparative example 5 4.7 Comparative example 6 4.5 10 0.6 5 1.5 6 1.0 7 0.8 8 1.7 9 1.2 ll 1.0 I] 2 I 0.8 13 1.7 15 1.6 16 l, l 17 1.4 18 1.1 Note: Example 14 is intentionally not included. Comparative Examples 1 to 6 are covered by the published International patent application WO2009 / 091016; Comparative Examples 1 to 4 are specifically described in WO2009 / 091016 as Examples 32, 35, 54 and 73 respectively. . 10 Comparative Examples 5 and 6 are N {3 - ((4aS, 5S, 7aS) -2-amino-5- (fluoromethyl) -4a, 5,7,7a- tetrahydro-4HAlro [3,4-d] [ 1.3] thiazin-7a-ii) -4-fluorophenyl) -5-ethoxypicolinamide and N- (3 - ((4aS, 5R, 7aS) -2-amino-5-methyl-4a, 5,7,7a- tetrahydro-4H-filter [3,4A] [1,3] thiazin-7a-yl) -4Aorophenyl) -5-ethoxypicolinamide. 15 respectively. The data demonstrate that the compounds of the present invention and specific examples 1 to 13 and 15 to 18 have less Pgp efflux and therefore CNS penetration potentially greater than the representative examples of WO2009 / 0910l6 using the previously mentioned recognized method. estimated CNS penetration. For example, Comparative Examples 1 to 6 have higher Pgp efflux ratios than the compounds of the present invention. In addition, Comparative Examples 5 and '5 6 have higher Pgp efflux ratios than a close analog, Example 10 of the present invention, which clearly demonstrates the effect that the trifluoromethane group on the tetrahydrofuran ring has on Pgp efflux. , that is, the trifluoromethyl group reduces the efflux of Pgp. Those skilled in the art will estimate that the Pgp 10O in vivo assay described above is a predictive assay for CNS penetration in vivo. in this way it is also desirable for the decreased pgp-mediated efflux to translate to the in vivo situation. those skilled in the art will estimate that there are many ways to estimate the CNS penetration of compounds in vivo. For example, one can quantify compound concentrations in blood or plasma and brain and calculate a ratio of brain: blood (Br: Bl) or brain: plasma (Br: Pl). This method has been used historically and has been widely accepted as a method of determining CNS penetration (Summerfield et al., J Phannacol. Expt. The R. 2007, 322, 205). Those skilled in the art will estimate that this type of test can be conducted 20 in a fixed state, a simple time point, or can be performed by quoting the area under the curve (AUC) ratios. All methods are equally valid, but each may have certain warnings that will be appreciated by those skilled in the art. Recent literature has been published to suggest that it is important to consider the three concentrations in vivo and that when 25 efflux occurs from the brain, the plasma-free concentration must be the same or equivalent to the free brain concentration (Kalvass and Maurer, Biopharmaceutics & Drug Disposition 2002, 23, 327; Mauer et al, Drug Metab. Disposition 2005, 33, i75; Trainor Expert Opin. Drug Discov. 2007, 2, 5 1). In this way, a compound that can freely penetrate the CNS and is not subject to active efflux, for example, by Pgp or another transporter, must demonstrate a free brain: free plasma (BrE: PÍ &) or an unbound brain: plasma not on (Br,: Plj of approximately 1: 1. Those skilled in the art will estimate that free or unbound concentrations "5 may be due to the multiplication of total brain or total plasma concentration by the Non-Bonded Hation in brain tissue or plasma, which can be measured by the assay described below. Those skilled in the art will estimate that the unbound Eation can change with the experimental actors, for example, concentration or temperature, etc. Those skilled in technique 10 will be able to estimate this and select the most appropriate setting of conditions. Those skilled in the technique will also estimate that no matter how much the conditions are the same for each compound evaluated, then the assay will give consistent data for the range of compounds tested, thereby minimizing any discrepancies. It has also been proposed that the 15 drug concentrations in the cerebrospinal fluid (CSF) are · equivalent to the concentrations brain cells for compounds that are not actively effected from r of the brain (He et al., Xenobiotica 2009, 39, 687). Thus, another method of determining CNS penetration should be to estimate CSF: free plasma (CSF: P | r) or CSF: unbound plasma (CSF: PJ,). If the free drug in plasma is able to permeate the CNS and is not inflow or outflow then the CSF: Plf, or CSF: P! U should be approximately 1: l. Those skilled in the art will estimate that the tissues associated with the objectives associated with determining CSF drug concentrations and extracting CSF, for example CSF 25 can be contaminated with blood depending on the withdrawal method, also CSF concentrations can be accurate lower, depending on the dose used. Thus, it was observed that a GlaxoSmithKline BACE inhibitor (GSKl88909), BACE-Í IC50 5 nM, having exposure to low CNS, was ineffective in reducing Afj40 production in the brains of human TASTPM mice (which overexpress both human and AppsweK595N / M596j mice). PS 1m] 46v) in acute administration (Hussain et a!., J. Neurochem. 2007, 100, 802 - 809). Following an oral dose of 250 mg / kg at 5 brain co-concentration of GSKl88909 in TASTPM mice it was 0.62 µM. When a Pgp inhibitor (GFl20918) was dosed 5 hours before oral administration of GSK188909, the cerebral concentration of GSK188909 was observed to be 5.43 UM following an oral dose of 250 mg / kg, that is, the co-administration of a Pgp inhibitor caused an increase of almost 9 times iO in the penetration of CNS, presenting efflux of Pgp is an important mechanism preventing BACE inhibitors from penetrating the CNS. In addition, in the absence of a Pgp inhibitor, an oral dose of 250 mg / kg of GSK188909 has no effect on brain AÜ40 levels in TASTPM mice, since when a Pgp inhibitor was co-administered (5 hours before of the administration of GSK188909) a 68 ° reduction in the levels of Af340 in the brain with respect to mice treated with the vehicle was observed. Another document reported a similar effect with three Bristol-Myers Squibb BACEJ inhibitors (Meredith et al., J. Pharm. Expt. Ther. 2008, 326, 502-513). The three compounds reported were observed to be Pgp substrates in vitro. When dosed to mice, the three compounds showed low CNS penetration and did not decrease amyloid levels in the brain, but were able to decrease plasma amyloid levels. When the same three compounds were added to the Pgp knockout (KO) mice, the level of penetration into the CNS increased and the compounds were able to decrease amijoid levels in the brain. Schering-Plow researchers have also published documents (Iserloh et al., Bioorg. Med. Chem. Lett. 2008, 18, 418) to show that BACE-I inhibitors in their series (for example, 11 from the reference mentioned above) , are subject to Pgp efflux, as a result whose compound has been observed to have a low Br: Pl (<0.1) in the rat. '5 The literature cited above emphasizes the difficulties in identifying BACE-I inhibitors that are not subjected to Pgp efflux. Such inhibitors must be highly desirable and many research groups have tried to discover such as to no avail. In this way, BACE-I inhibitors that are not Pgp substrates and can therefore penetrate 10 easily in the lower amyloid CNS in the brain should be desirable. More recently, Wyeth researchers have reported extensive work to overcome Pgp efflux in a series of cyclic acylguanidine BACE-I inhibitors (Malamas et al, Bioorg. Med. Chem. Lett. 2010, 20, 6597). Compotes were found to be substrates of Pgp 15 Hacos and with Br: Pl approaching l: í. however, two main examples of reduced Pgp efflux (84 and 89 from the aforementioned reference) do not decrease A | 340 in the brain of Tg2576 mice 8 hours after an oral dose of 30 mg / kg. The loss of efficacy was attributed to the fact that the compounds showed ajta brain tissue binding. In this way, it is important to discover BACE-I inhibitors that are not Pgp substrates but still have reasonable & unbound action! in brain tissue and are able to dominate amyloid in the brain. It has also been shown that BACE inhibitors that are not Pgp substrates in vitro can penetrate CNS (eg, 25 Merck TC-1) and can decrease AB40 levels in the brain of APP-YAC mice and monkeys (Sankaranarayanan et al., J. Pharmacol. Expt. The R. 2009, 328, 13 1-140). In this way, in vitro pgp assays showed TC-1 should not be a substrate for Pgp and then TC-1 was dosed to APP-YAC mice (100 mg / kg i.p.) was able to penetrate N, modestly the CNS as shown by brain concentrations and the brain: plasma ratio and its capacity resulted in moderate decrease in cerebral amyloid. Weather Conc. Plasma Conc. Cerebral Br: Pl Reduction in AB40! (µM)) (µM) I cerebral (%) I 2h! 25! 1.6 I 0.06 I 26 I 4h | 13 l 1.8 I 0.14 I 29 The brain and plasma concentration of TC-1 following dose 5 of 100 mg / kg and corresponding effects on brain Ab40 levels in APP-YAC mice. In separate experiments it was shown that TC-1 can penetrate the CSF of monkeys when co-administered with a CYP3A4 inhibitor (ritonavir). In these experiments, the mean plasma concentration of TC-1 was observed to be 2.7 µM, while the CSF concentration was observed to be 0.025 µM. However, when TC-1 is -99 ° /) bound to plasma proteins, the concentration of free plasma was calculated to be -0.027 nNI. It was observed that the CSF Afl40 levels showed a decrease of 42 ° /, compared to a control group treated with vehicle. In this way, a BACE inhibitor that can freely penetrate the CNS should be expected to be able to decrease the amyloid levels in the CNS. It should be beneficial not to be co-dosed with a CYP3A4 inhibitor. The compounds of the present invention have been shown to decrease Aj3 production in cellular assays that correlate with their ability to decrease Aj3 production in animals. In this way, the compounds of the present invention will be useful in decreasing the production of N in humans and, in this way, they will be useful in the treatment of neurodegenerative diseases such as Alzheimer's disease. CNS penetration in vivo in rats Male Sprague Dawley rats were "purchased from Charles River UK Ltd. (Margate, UK) and housed in accordance with the instructions of the UK Home Office. The drugs were manufactured in the appropriate concentrations in methyl cellulose at 0.5 ° /) The animals were dosed orally (2 mL / kg) by gavage in doses summarized in Tables 2 to 4 below. At the post dosing time points, specified in Tables 2 to 4 below, the animals were administered by an i.p. of "5 sodium pentobarbitone (approximately 330 mg / kg for terminal anesthesia). Using a guillotine, the animals were decapitated and stem blood collected in 15 mj Falcon tubes containing 100 IU of heparin. The blood was swirled followed by centrifugation at 6000 rpm, 4 ° C for 5 minutes Plasma was collected by 10 DMPK and ELISA assays and stored at -80 ° C until use.The brains were dissected and divided along the midline, weighed and stored at -80 ° C until further use. Mé, t, o, for Plasma, Brain and CSF 15 Preparation of Acetonitrile Working Solutions The test compound was prepared as a 1 mg solution free from the base solution / mL in DMSO, swirled and sonified for 5 minutes. The 1 mg / niL DMSO solution was diluted to 10 and 30 µg / mL acetonitrile stocks by adding 10 µL to 990 µL of 20 acetonitrile and 30 µL to 970 µL of acetonitrile, respectively. The 10 and 30 µg / niL acetonitrile stocks were then serially diluted 1: 9 (V / V) (100 µL stock in 900 µL acetonitrile) to give the following solutions: 0.003, 0.01, 0.03, 0.1, 0.3,!, 3, 10 and 30 µg / ml of acetonitrile. Preparation of Standards, Blanks and Plasma Samples 25 Control male Sprague Dawley mouse plasma and study plasma samples were stored at -80 ° C until the day of analysis using these were thawed at room temperature. The control plasma was centrifuged (2,000 g per 10 minutes) and divided (90 µL) into eppendorf tubes for the preparation of standards and blank samples. The study samples were previously divided (100 µL) in eppendorf tubes immediately following the plasma collection. An aliquot of (10 µL) from the appropriate acetonitrile stock was added to the control plasma (to give a final volume of 100 µL) "5 to give the required calibration standards that range from 1 to 3000 ng / niL. Double blanks and blank samples were prepared by adding 10 µL of acetonitrile to 90 µL of white plasma. Preparation of Standards, Blanks and Brain Samples from a male Sprague Dawley control mouse and the 10 study brain samples were weighed after collection and stored at - 80 "C until the day of analysis when they were thawed at room temperature. The brains, once thawed, were diluted with water (4 nL per gram of tissue) and homogenized using a mechanical homogenizer. An aliquot (100 µL) of each study sample was made in 15 Micronics tubes ready for analysis and sufficient aliquots (90 µL) of homogenate of control brain prepared for the preparation of standards and blanks. An aliquot (10 µL) of the appropriate acetonitrile stocks was added to the control brain homogenate (to give a final 20 volume of 100 µL) to give the required calibration standards covering the range of 1.5 to 5000 ng / g. Double blanks and blank samples were prepared by adding 10 µL of acetonitrile to 90 µL of blank brain homogenate. Extraction of Plasma and Samples, Patterns and Brain Blanks Each sample, standard and white of plasma homogenate and 25 brain (100 µL) was extracted with an aliquot of (300 µL) acetonitrile (containing 0.1 ° formic acid / ) and 100 ng / ml of an appropriate internal standard). The double blanks were extracted with an aliquot (300 µL) of acetonitrile containing formic acid at 0.1 ° 4). All samples, standards and blanks were then swirled mixed and centrifuged (2000 g for 15 minutes). An aliquot (50 µL) of the resulting supernatant was then used in a plate of 96 2 ml deep reservoirs and diluted with acetonitrile: water (50:50 v / v) (150 µL) ready for analysis by an LC- Specific MS / MS. 5 Preparation of Samples, Standards and Blanks of CSF Maje Sprague Dawley control mice and study CSF samples were stored at -80 ° C until the day of analysis when they were thawed at room temperature. One aliquot (50 µL) of each study sample was used in Micronics tubes ready for analysis and 10 sufficient aliquots (45 µL) of CSF control prepared from standards and blanks. An aliquot (5 µL) of the appropriate acetonitrile stocks was added to the control CSF (to give a final volume of 50 µL) to give the required calibration standards that range from 1 to 1000 ng / niL. 15 Double white and blank samples were prepared by adding 5 µL of acetonitrile to 45 µL of blank CSF. CSF Sample, Standard and White Extraction Each CSF standard and white sample (50 µL) was extracted with an aliquot (150 µL) of acetonitrile (containing 0.1 ° /) of formic acid 20 and 100 ng / mL of an appropriate internal standard). The double blanks were extracted with an aliquot (150 µL) of acetonitrile containing 0.1 ° / 0 formic acid. All samples were then subjected to mixed swirling and an aliquot (50 µL) of each was further diluted in 150 µL of acetonitrile: water (50/50 v / v) in a block of 96 deep 25 mL reservoirs ready for use. LC-MSMS analysis. All samples were then analyzed using a Waters Acquity UPLC connected to a Waters Xevo TQ mass spectrometer. LC conditions: P Column: Acquity UPLC BEFI Cl8, 1.7 um, 2.1 x 50 mm, maintained at 40 ° C Mobile Phase: A = 95 ° /) water: 5 ° /) McOH containing 0.01 M ammonium acetate B = 5 ° /) water: 95 ° /) MeOlil containing ammonium acetate '5 0.01 M Gradient: Time (minute) B (° / 0) 0 I' 1.2 I 95 1.5 I 95 1.7 'I 5 2.0 i 5 Flow rate: 0.6 ml / minute; injection volume 5 µL; autosampler temperature 6 ° C LC Mxo was diverted to waste the first 0.3 10 min of each injection MS / MS transitions were automatically optimized by Waters QuanOptimise software Amyloid Detection DEA / NaCl extraction of AB peptides from brain of 15 rabbit: 100 ml of 0.2 ° 4 esh diethyl amine (DEA) in 50 niM NaCl (pH 10) was recently prepared and 1 ml / 25 mg of brain tissue was added to each hemisphere (i.e. 40 x brain volume). The brains were immediately homogenized using a Politron PT 1200 for 20 1.5 minutes and the samples left to incubate on ice for one hour after homogenization. 3 ml of the homogenate was transferred to a polyalomer tube (Beckman # 362333) and rotated at 133,000 x g (55,000 µm / min) for 45 minutes at 4 ° C. The supernatant was then neutralized at pH 8 to 8.3 by the addition of 1/10 volume 0.5 M Tris / HCl, pH 6.8. Samples 25 can be used & stored or frozen on dry ice and stored at -80 ° C until required for Human / Rat (40) ELISA (Wako Kit) analysis The Wako Af340 ELISA Kit (Code No. 294-62501) uses the 4 monoclonal antibody! BNT77, elevated against the Af3 epitope (jL28) and the monoclonal antibody! BA27, which specifically detects the C-terminal portion of AB40. The kit used for the quantitative determination of human or rat Af3 (1-40) and also N | terminally truncated A | 340 (Aj3 (x-40)) species in biological matrices, such as tissue culture medium, homogenated from tissue, CSF and plasma. For analysis, plasma and brain samples are diluted 1: 1 10 with the standard diluent contained in the kit and CSF samples are diluted 1: 8 with the standard diluent contained in the kit. The test is performed according to the manufacturer's instructions and the samples are analyzed in duplicate. The data are analyzed using Microsofí Excel 2003 and statistical analysis is performed using Genstat 9 ° Edition. Thus, when Comparative Example 4 was administered at a dose of 10 mg / kg p.o. and plasma, brain and CSF samples were collected 2, 4, 6 and 8 hours post-dosing following the following concentrations were measured (Table 2): Table 2: Data for Comparative Example 4 Time (h) {Pl] (' ÍM) '[Plj (nh4) [B'] ('ÍM)' [B ',,] (nM) [CSF] (nM) Br ,,,: Pl ,,, Br,: Pl, CSF: Pl, 2 1257 440 971 65 104 0.8 0.15 0.24 4 1162 4 () 7 874 59 88 0.7 0.14 0.22 6 834 292 570 38 63 0.7 0.13 0.22 8 484 169 368 25 26 0.8 0.15 0.15 L Calculated by multiplying [Pl] by Pl Fu. 2 Calculated by multiplying [Br] by Br Fu. 20 From the study above, Comparative Example 4 showed a reduction of 59 ° /) and 64 ° /) of A | 340 in the brain in 4 and 6 hours respectively and a reduction of 76 ° /) and 70 ° /) of AB40 at CSF at 4 and 6 hours respectively. Certain compounds of the present invention have been estimated in vivo in the rat to corroborate the levels of penetration into CNS; these data are presented in the tables below. Surprisingly, it has been observed that the compounds of the present invention show increased CNS penetration in the rat with respect to the compounds of WO2009 / 091016 by any of the recognized methods already mentioned for determining CNS penetration. In this way, the compounds of the present invention may have improved profiles in which they target the site of action, the brain, and therefore may show improved efficacy or efficacy at lower concentrations or doses or peripherally reduced mediated side effects by of preferred CNS division or a combination of any 10 or all of these aspects. Thus, when Example 8 of the present invention was administered in a dose of 10 mg / kg p.o. and plasma, brain and CSF samples were collected 2, 4, 6 and 8 hours after dosing the following measured concentrations (Table 3): 15 Table 3: Data for Example 8 Time (h) [lj ('IM)' { Pl,] (nM) [B ') (' ÍM) '[B'J_ (' ÍM) [CSF] (nM) Br ,,,: Pl ,,, Br,: Pl ,, CSF: Pl, 2 1189 124 3961 87 65 3.3 0.7 0.5 4 657 68 2582 57 39 3.9 0.8 0.6 6 229 24 845 i9 12 3.7 0.8 0.5 8 186 19 709 16 'nq . 3.8 0.8 'n.d. 1 Caused by the multiplication of [Pl] by Pl Fu 2 Calculated by multiplying [Br] by Br Fu. 3. not quantifiable. Concentrations close to or below the lower limit of quantification and may not be precisely quantified. 4. Not determined. From the study above, Example 8 showed a reduction of 68 ° 4 and 72 ° /) of Afl40 in the brain in 4 and 6 hours respectively and a reduction of 82 ° /) and 74 ° 4 of AB40 in CSF in 4 and 6 hours respectively. In this way, the compounds of the present invention show decreased Pgp efflux from previous disclosures while demonstrating efficacy in the CNS. Effectiveness is thus achieved with lower circulating plasma concentrations. When Example 1 of the present invention was administered at a dose of 10 mg / kg p.o. and plasma, brain and CSF samples were collected 2, 4, 6 and 8 hours post-dose, the following concentrations were measured (Table 4): è Table 4: Data for Example 1 ' Ã Time (h) {Pl] ('ÍM)' [Pl ,,] (nhí) [B'j (nM) '[B',] (nM) [csFj (nM) Br ,,,: Pl ,, , Br ,,: P1, CSF: Pl ,, 2 462 23 2338 30 23 5.1 1.3 1.0 4 298 15 1550 20 18 5.2 1.3 1.2 6 401 20 1492 19 10 3.7 1.0 0.5 8 228 ll 1194 16 18 5.2 1.5 1.6 I Calculated by multiplying [jpl] by Pl Fu. 2 Calculated by multiplying [Br] by Br Fu. From the study above, Example] showed a reduction of 64 ° /) and 70 ° /) of Aj340 in the brain in 4 and 6 hours respectively and a reduction of 80 ° /) and 85 ° '6 of AB40 in CSF in 4 and 6 hours respectively. In this way, the compounds of the present invention exhibited decreased Pgp efflux over previous inventions while demonstrating the effectiveness of CNS. Effectiveness is thus achieved with lower circulating plasma concentrations. Method for Determination of Plasma Protein Binding (PPB) and Brain Tissue Binding (BTB) Preparation of Compound The compounds were dissolved in DMSO to give a 1 mg 15 free base / mL solution, after the further dissolution of 100 µg / niL in acetonitrile (100 µL of 1 mg / mL in 900 µL of acetonitrile). Matrix Preparation On the morning of dialysis, plasma and male Sprague Dawley rat brain, previously stored at -80 ° C, were thawed at 20 ° C at room temperature. The plasma was reached for pH and, if necessary, adjusted to pH 7.4 with 1M HCl. The plasma was then centrifuged (2000 g per 10 min) and the brains diluted with 2 ml of phosphate buffered saline solution (pH 7.4) per gram of tissue and homogenized using a mechanical homogenizer. An aliquot (10 µL) of the 25 µg / niL acetonitrile compound solution was then added to 1 niL of plasma and brain homogenate and subjected to mixed swirling to give a final compound concentration of 1 µg / mL in the matrix. Preparation of RED Plate The Rapid Balance Dialysis (RED) plate (Thermo Scientific) was prepared according to the manufacturer's instructions, that is, the '5 base plate was embedded in 20 ° /) (V / V) of ethanol for 10 minutes and then rinsed twice with deionized water before being allowed to dry. The base plate was then filled with the appropriate number of disposable inserts (n = 3 per compound) (Thermo Scientific) and matrix containing 1 µg / mL of compound added in the matrix chamber of the inserts (200 µL) and 10 an aliquot (350 µL) of PBS added to the buffer chamber. The plate was then covered with an adhesive and incubated in air at 37 ° C for 6 h with shaking at 130 rjpm. Sampling Following the 6 h incubation, the seal was removed and a 15 aliquot (50 µL) taken from PBS chambers and dispensed in Micronics tubes. Also, an ajiquette (50 µL) was removed from the matrix chambers and placed in separate Micronics tubes. Plasma and brain were then compared in the matrix with 50 µL of drug-free PBS and PBS samples with 50 µL of the drug-free matrix - 20 corresponding, to give equal compositions and tonal volumes (100 µL). Sample analysis The samples were mixed vortex and an aliquot (300 µL) of acetonitrile containing 0.1 ° /) formic acid and 100 ng / mL of an appropriate internal standard added. The samples were then mixed and 25 centrifuged (2000 g for 15 min) and an aliquot of the supernatant (100 µL) removed in a 96-well plate and diluted with an equal volume of water ready for analysis by LC-MS / MS. The following data was obtained by the following compounds in the above test (Table 5). Table 5: Rat PPB compound (° / 0) Rat BTB rat k plasma (%) Rat brain fu) Comparative Example 4 I 65.0 l 0.350 I 93.3 I 0.067. : Example 1 I 95.1 I 0.049) 98.7 I 0.013 I Example 8 I 89.6 | 0.104 I 97.8 I 0.022 The data represent an average of n = 3 replicates of the unbound fraction. From the data presented in the above, it will be apparent to the person skilled in the art that the compounds of Examples 1 and 8. achieve a similar reduction in the Af340 brain that of Comparative Example 5, but with a lower plasma concentration and free plasma concentration. This is advantageous and indicates that the compounds of the invention will have better or similar efficacy at lower concentrations than the compounds of WO2009 / 091016 and consequently will be less similar to cause the peripherally mediated side effects, ok! 10 such as cardiovascular effects, phospholipidosis, liver toxicity, renal toxicity and gastrointestinal toxicity. Evaluation of the effects on the QTc interval in guinea pigs Male Dunkin-Hartley guinea pigs were weighed 15 and anesthetized using 4 ° /) isoflurane in carbogen. Anesthesia was maintained at 1.5 ° / isoflurane and the animals were kept under anesthesia for the duration of the study. Xylazine at 2mg / kg i.m. was administered to the rear limb as a bradycardic agent to enable the detection of QTC prolongation by sofiware. 20 The carotid artery and jugular vein were canuated with lines containing heparinized saline and a 3-lead ECG connected and monitored using the LabCbm Pro software. The animals were allowed to stabilize for 30 minutes after the completion of the surgical procedure, before the initiation of an infusion vehicle iv (5 ° /) of DMSO / 90 ° '6 of MilliQ / 5 25 ° 4 of O, IN of HCl) from time zero (infusion rate = 0.2 ml / kg / min) . In 10 mins, a peripheral blood sample was collected by PK analysis (150 ul; all coIection syringes were heparinized). In 12 mins, drug infusion was started @ 2.0 mg / kg / 10 min i.v .. The dosage was increased to 6.0 mg / kg / 10 min, then 20 mg / kg / 10 i.v., with a period. 10-minute infusion and two minutes of blood sampling at every "5 dosage. After the final dosage, a blood sample was taken and a second infusion vehicle started. Eight minutes later a terminal blood sample was collected by plasma analysis PK and animal killed by a list method 1. QTc changes (Bazett's) were analyzed using the LabChart Pro 10 software. QTc was not changed until the highest tested dosage / concentration, which corresponds to a 9503 unbound piasma concentration nM for example 8 and 296 nNl for example 1. Evaluation of the effects on the QTc interval in beagle dogs Male beagle dogs were weighed and injected with K 15 sodium thiopental for induction of anesthesia. Anesthesia was maintained with a mixture of 1-J, 5 ° /) isoflurane and oxygen and the animals kept under artificial respiration and anesthesia using isoflurane for the duration of the study. The carotid artery and saphenous vein were cannulated with lines containing heparinized saline and a LII ECG connected and monitored 20 using the polygraph system. The animals were allowed to stabilize for 30 minutes before infusing the compound solution and the infusion through the cannula was started from time zero with I mg / kg / 10 min. The dosage was increased to 3 mg / kg / 10 min, then 10 mg / kg / 10 min. An arterial blood sample was collected after each dosage by PK analysis. 25 QTc was not changed until the highest concentration / dosage tested, which corresponds to an unbound plasma concentration of 4128 nMporexample 8 and 1329 nMporexample 1. Next, the methods for preparing the compound of formula (I) or a pharmaceutically salt acceptable from this according to ) The present invention will be described. A. Preparation method A generates !: I Fj ° ^ (') °; ° r A (") ° J" "°" A (-) ° QL-A, «': ,,, '": jj' 7j ': J ,,,, ", ~ j t '^ c, E ,, ,,, ": jj, O 'O ,,,,,, Jí: NH2.: 7 F'è' -i: 'F" j H i F F F OH A- (5) A- (6) A- (7) U'jjNrhj ^,)) ÍÚ "j Ijjhz H Fè OH f f A- (8) F F A- (9) A- (1O) X i, i NO, X i l NO2 X i, | NH, A- (x) F ^ r A- (xi) fAY A- (xii) FA ~, ": j> f" '': í7sÀ '"°'":, Ár '°' A- (11) A- (12) F A- (13) o¶A ::] r "oÁy" X (<1 NH 'X i I NH A- (xiii) F ^} A- (xiv) FA ~ l.' Y q rN ~ S NHBOC "F,, C" Y '"r" "S A- (15) F | H F A- (14) F F, H In the formula, X, Y and A are as defined above. General preparation method A is a method for preparing a compound A- (l5) which corresponds to compound (I) according to the present invention from compound A- (l) as a crude material through the steps multiple of step A- (i) to step A- (xiv). Compound A- (1) is commercially available. Step A- (i): This step and a step to obtain a compound A- (2) by opening the epoxide A- (l) with a sulfide ilide to generate an intermediate allyl alkoxide which is then alkylated to give the compound A- (2). That person skilled in the art will appreciate that this transformation can be "5 conducted in a pot or as two individual reactions. That person skilled in the art will appreciate the benefits and discounts of a compared pot reaction to conduct two separate reactions and choose the best method for your requirements accordingly. Specifically, the A- (1) epoxide can be opened by the anion 10 of thmethylsulfonium iodide and resulting loss of dimethylsulfide to give the corresponding allyl alkoxide. Trimethylsulfonium iodide can be deprotonated with a suitable base, for example, butyl lithium. The amount used in the reaction is not particularly limited as it does not interfere with the reaction. Examples of suitable solvents include THF. That person skilled in the art will appreciate that the word solvent in this example is used to indicate the liquid in which the reaction is carried out and that the reagents cannot be dissolved. Preferably the reaction should be conducted below room temperature, preferably -30 to 20 ° C. In addition, the reaction can be heated at room temperature to the filtration reaction. A reaction time is not particularly limited and is usually 5 minutes to 24 hours, preferably 1 to 6 hours. Those skilled in the art will appreciate that the alkoxide generated from this reaction can be reacted with an alkylating agent directly, such as tert-butyl bromoacetate and that this reaction can proceed with or without additional solvents. If additional solvents are required for the filtration reaction, then solvents such as DMF or NMP are suitable. The reaction temperature is not particularly limited. Suitable reaction temperatures include room temperature at 80 ° C, preferably room temperature. A reaction time is not particularly limited and is usually 5 minutes to 1 week, preferably i - 48 hours. That person skilled in the art will appreciate the alkoxide. intermediate must be extinguished, isolated and purified then subjected to the '5 independent alkylation conditions. This reaction can be carried out under the same conditions as those usually used in the O-alkylation reaction of an alcohol compound (such as the conditions described in · Tetrahedron Lett. 46 (2005) 45, 7751-7755). In this reaction, compound A- (2) can be obtained by adding a base such as sodium hydride in a solution of the intermediate alcohol in THF to prepare an alkoxide and then reacting the alkoxide with tert-butyl bromoacetate, for example. example. The solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved therein to a certain extent. Examples of the solvent include solvents such as THF, DMF and dimethyl sulfoxide. The reaction can be carried out by causing 1 to 3 equivalents of an appropriate base to act in the presence of such a solvent. Examples of the base used include sodium hydride, potassium hydride and t-butoxypotassium. A reaction time is not particularly limited and is usually 0.5 to 72 hours and preferably 0.5 to 12 hours. The reaction temperature is usually -20 ° C to 50 ° C. A more preferred result such as improved yield can already be achieved by adding a salt such as tetrabutylammonium iodide to this reaction. Step A- (ii): 25 This step is a two-step sequential reaction to obtain compound A- (3) from compound A- (2) by deprotection of the ester group then forming a Weinreb amide. Specifically, the tert-butyl ester of compound A- (2) can be deprotected under the same conditions as those generally used in the deprotection of a tert-butyl ester compound (such as the conditions described in a document such as TW Greene and PGM Wuts, "Protective Groups in Organic Chemistry, Third Edition", John Wiley ± & Sons (1999), pp. 404-408). In this reaction, compound A- (2) can be reacted '5 with an appropriate acid in a suitable solvent, such as formic acid, such as solvent and acid, for example. The solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved therein to a certain extent. A time . reaction time is not particularly limited and is usually 0.5 to 72 hours and 10 preferably 0.5 to 24 hours. The reaction temperature is usually a freezing temperature of 60 ° C. The intermediate acid can then be transformed to the Weinreb amide (Tetrahedron Lett. 1981, 22, 3815) by the reaction of N, O-dimethylhydroxylamine hydrochloride under the conditions of standard amide formation, ie 15 by condensation of the intermediate acid with hydrochloride of N, O-dimethylhydroxylamine using a condensing agent. Alternatively, this step is a step to obtain a compound A- (3) by condensing the intermediate acid with N, O-dimethylhydroxylamine hydrochloride by the acylation reaction. 20 The condensation reaction of the intermediate acid - with N, O-dimethylhydroxylamine hydrochloride using a condensing agent can be carried out under the same conditions as those usually used and described in the following documents. Examples of the known method include that in Rosowsky et al .; J. Med. Chem., 34 (1), 227-234 (1991), 25 Brzostwska et al .; Heterocycles, 32 (10), 1968-1972 (1991) and Romero et al .; J. Med. Chem., 37 (7), 998-1014 (1994). The N, O-dimethylhydroxylamine hydrochloride can be a free form or a saj. The solvent in this reaction is not particularly limited as it does not inhibit the reaction. Examples of the solvent include tetrahydrofuran, 1,4-dioxane, ethyl acetate, methyl acetate, dichloromethane, chloroform, N, N-dimethylformamide, toluene, acetonitrile and 2 [xylene]. Examples of the condensing agent include CDT (N) N'-carbonyldi- '5 imidazole), Bop (1H-1,2,3-benzotriazol-1-yloxy (tri (dimethylanine)) phosphonium hexafluorophosphate), WSC (hydrochloride hydrochloride) l-ethyl-3- (3-dimethylaminopropyl) carbodiimide), DCC (N, N-dicyclohexylcarbodiimide), diethylphosphoryl cyanide, PyBOP (benzotriazolj-yloxytris (pyrrolidine) hexafluorophosphate phosphonium) and EDC · l-hydrochloride ethyl-3- (3-10 dimethylaminopropyl) carbodiimide). Suitable conditions include an acid-activating agent, such as N, N'-carbonyl diimidazo !. An ample excess equivalent of N, O-dimethylhydroxylamine is used with respect to the intermediate acid. A large excess equivalent of an organic base such as ethylamine can be added where needed. 15 A reaction time is not particularly limited and is usually 0.5 to 72 hours and preferably 0.5 to 24 hours. The reaction temperature varies according to the raw material used, the solvent and others and is not particularly limited. The temperature, from chilled to the reflux temperature of the solvent is acceptable, from chilled to room temperature is preferable. Step A- (iii): This step is a step to obtain a compound A- (4) by reacting an organometallic reagent (arylithium reagent or a Grignard reagent) with compound A- (3) as described in Tetrahedron Lett . 25 1981,22,3815. The reaction in this step can be carried out under the same conditions as described in Tetrahedron Lett. 1981, "22, 3815, for example. Arylithium reagent (including heterocyclic) or Grignard reagent (including heterocyclic) can be prepared by a method known to a person skilled in the art. Specifically, the corresponding phenyl lithium reagent or phenyl magnesium reagent (Grignard) can be prepared by exchanging metal halogen between a compound of. 'aryl halide and organometallic reagent A commercially available ta! like "5 'is an alkylithium reagent such as n-, sec- or tert-butyllithium or a Grignard reagent such as isopropylmagnesium bromide, or metallic magnesium, for example. The solvent used in this step varies according to a starting material and the reagent used and is not particularly limited as it does not inhibit the reaction, allows the starting material to be dissolved in it to a certain extent and is always inert during the reaction. Preferred Examples of the solvent include organic solvents such as diethyl ether, tetrahydrofuran, 1,4-dioxane, 1,2-dimethoxyethane, benzene and toluene and mixed solvents thereof. A reaction time is not particularly limited and is usually 0.1 to 48 hours and preferably 0.1 to 12 hours. The reaction temperature varies according to a starting material, the reagent used and others and is preferably kept low, for example, at -78 to - 60 ° C. Step A- (iv): This step is a step to obtain a compound A- (5) by the oxidation of compound A- (4). The reaction in this step can be carried out under the same conditions as those usually used in the oxidation reaction of a carbonyl compound such as the conditions described in Org. Lett. 9 (2007) 25 5, 753-756, Tetrahedron: Asymmetry 5 (1994) 6, 1018-1028 and Tetrahedron 54 (1998) 22, 5868-5882. Specifically, compound A- (5) can be obtained by reacting compound A- (4) with hydroxylamine or a hydroxylamine salt (such as hydroxylamine hydrochloride or hydroxylamine sulfate) in the presence of a base or in the absence of a base , for example. The solvent used in this reaction is not particularly limited as it does not inhibit the reaction. Preferred examples of the solvent include Z organic solvents such as ethanol, methanol, tetrahydrofuran, 1,4-dioxane, 5 l, 2-dimethoxyethane and dichloromethane and mixtures of these solvents and water. Examples of the base used include sodium acetate, pyridine, sodium hydroxide, cesium hydroxide, barium hydroxide and 2,6-lutidine. A reaction time is not particularly limited and is usually 5 minutes to 24 hours and preferably 5 minutes to 12 hours. The reaction temperature is usually - 10 20 ° C at the solvent reflux temperature and most preferably 0 ° C at solvent reflux temperature. Step A- (v): This step is a step to obtain a compound A- (6) by an intramolecular thermal cycloaddition of alcenij oxime A- (5). 15 The reaction is carried out in the presence of an additive, for example, hydroquinone. The solvent used in this reaction is not particularly limited as it does not inhibit the reaction. Suitable reaction solvents include high-boiling solvents such as xylenes. The reaction temperature 20 is not particularly limited and is usually 80 to 200 ° C or solvent reflux temperature. A reaction time is not particularly limited and is usually 0.5 to 48 hours and preferably 0.5 to 24 hours. Step A- (vi): This step is a step to obtain a compound A- (7) to subject compound A- (6) by the reductive cleavage reaction of the deN-O bond. The reductive cleavage reaction of the N-O bond can be carried out under the conditions using acetic acid-zinc, a metallic catalyst such as hydrogen-platinum oxide, or lithium aluminum hydride, for example. The reaction using zinc such as acetic acid-zinc can be carried out under the same conditions as described in J. Org. Chem. 2003,. 68, 207-1215 and Org. Lett. 7 (2005) 25, 5741-5742, for example. "5 Examples of the acid used include acetic acid, formic acid and hydrochloric acid. The solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved in it to a certain extent. only include methanol, ethanol, 1,4-dioxane, THF and water. The above acid can also be used 10 as a solvent. The reaction temperature is usually -20 ° C to the reflux temperature of solvent and preferably temperature to solvent reflux temperature A reaction time is not particularly limited and is usually 5 minutes to 48 hours and preferably 5 minutes to 24 hours. 15 The reaction using a metal catalyst such as hydrogen-platinum oxide can be carried out under the same conditions. conditions as described in Tetrahedron: Asymmetry 5 (1994) 6, 1018-1028 and Tetrahedron, Vol. 53, No. 16, pp 5752-5746, 1997, for example: Compound A- (7) can be obtained by hydrogenating the compound A- (6) using platinum oxide 20 as a catalyst in a solvent such as methanol, for example. The reaction using lithium aluminum hydride can be carried out under the same conditions as described in Bull. Chern. Soc. Jpn., 66, 2730-2737 (1993), for example. Compound A- (7) can be obtained by reducing compound A- (6) using aluminum hydroxide in a solvent such as 25 ether, for example. Step A- (vii): This step is a step to obtain a compound A- (8) from compound A- (7). The thiourea derivative A- (8) can be obtained from compound A- (7) by a method known to a person skilled in the art. The compound A- (8) can be obtained in this step by the reaction of the compound A- (7) with benzoyl isothiocyanate in a single solvent! such as 4 'dichloromethane or toluene. This reaction can be carried out under the same "5 conditions as described in J. Med. Chem. 1990, 33, 2393-2407, for example. The solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved in it to a certain extent. Examples of the solvent include dichloromethane, chloroform, toluene, 1,4-dioxane and THF. The reaction temperature is usually -20 ° C the reflux temperature of solvent and preferably freezing temperature to solvent reflux temperature A reaction time is not particularly limited and is usually 5 minutes to 48 hours and preferably 5 minutes to 24 hours. Step A- (viii): 15 This step is a method of obtaining a compound A- (9) by cyclizing the compound A- (8). In this reaction, compound A- (8) can be cyclized under various conditions to obtain compound A- (9) by activating the alcohol of compound A- (8). For example, compound A- (9) can be obtained in this reaction by heating compound A- (8) in a solvent such as methanol in the presence of a ta! as concentrated hydrochloric acid, for example. The solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved in it to a certain extent. Examples of the solvent include solvents such as methanol, ethanol, 1-propanol and water, mixed solvents thereof, and acids used as a solvent. The reaction can be carried out by using an equivaiente in a large excess of an appropriate acid to act in the presence or absence of such a solvent. Examples of the acid used include concentrated hydrochloric acid, hydrobromic acid, sulfuric acid, triAuoroacetic acid, methanesulfonic acid, trifluoromethanesulfonic acid and mixtures thereof. A reaction time is not particularly limited and is usually 0.5 to 72 hours and preferably 0.5 to 24 hours. The reaction temperature is usually chilled to reflux temperature of solvent. Alternatively, compound A- (9) can be obtained by reacting compound A- (8) with trinuoromethanesulfonic anhydride in a solvent such as dichloromethane in the presence of a base such as pyridine. The solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved therein to a certain extent. That person skilled in the art will appreciate that a solvent is not always required and the reaction can also be conducted in the absence of a solvent, for example, when the base is pyridine. Examples of the solvent include solvents such as dichloromethane, chloroform, 1,2-15 dichloroethane, THF, 1,2-dimethoxyethane and toluene and mixed solvents thereof. The reaction can be carried out using 1 to 20 equivalents of an appropriate base in such a solvent. Examples of the base used include pyridine, 2,6-lutidine, sodium carbonate, potassium carbonate and mixtures thereof. A reaction time is not particularly limited and is usually 0.5 to 24 hours 20 and preferably 0.5 to 12 hours. The reaction temperature is usually -78 ° C at room temperature. Step A- (ix): This step is a method of obtaining compound A- (10) by deprotecting the protection group of compound A- (9). Compound A- (10) 25 can be obtained under deprotection conditions known to a person skilled in the art. When the protecting group is a benzoyl group, compound A- (1O) can be obtained in this reaction by heating compound A- (9) in a solvent such as methanol in the presence of a base such as DBU, for example. This reaction can be carried out under the same conditions as described in Synth. Commun. 32 (2), 265-272 (2002), for example. The solvent used in the reaction is not particularly limited as it . does not inhibit the reaction and allows the starting material to be dissolved in it in "5 a certain extent. Examples of the solvent include solvents such as methanol, ethanol and 1-propanol. The reaction can be carried out using 1 to 20 equivalents of an appropriate base in such a solvent. Examples of the base used include DBU. A reaction time is not particularly limited and is usually 0.5 to 2 hours and preferably 0.5 to 12 hours. The reaction temperature is usually room temperature at solvent reflux temperature. Alternatively, compound A- (10) can be obtained in this reaction by heating compound A- (9) with an inorganic base such as potassium carbonate, for example. The solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved in it to a certain extent. Examples of the solvent include solvents such as methanol, ethane! and l-propanol. The reaction can be carried out using 1 to 20 equivalents of an appropriate base in such a solvent and preferably a light excess is used. Examples of the base used include potassium carbonate. A reaction time is not particularly limited and is usually 0.5 to 24 hours and preferably 0.5 to 12 hours. The reaction temperature is usually room temperature at solvent reflux temperature and preferably 50 to 100 ° C. That person skilled in the art will appreciate that the selected solvent will limit the reaction temperature to its reflux temperature. Examples of suitable solvents include refluxing methanol. Step A- (x): This step is a step to obtain compound A- (l1) by the nitration reaction of compound A- (1O). In this nitration reaction, the compound A- (l1) can be obtained from compound A- (l0) by a method known to a person skilled in the art. Examples of the nitration agent used Y in the reaction include potassium nitrate / concentrated sulfuric acid, fumigation of nitric acid / concentrated sulfuric acid and fumigation of "5 nitric acid / acetic anhydride. Suitable solvents for the reaction include trifluoroacetic acid. The reaction temperature is not particularly limited and it is usually -20 ° C at room temperature and preferable reaction temperatures include 0 to 10 ° C. Step A- (xi): 10 This step and one step to obtain a compound A- (l2) by t-butoxycarbonylation of the amino group of the compound A- (l1) .The reaction can be carried out under the same conditions as those commonly used in t-butoxycarbonylation of an amino compound such as the conditions described in a document such as TW Greene and 15 PGM Wuts, "Protective Groups in Organic Chemistry, Third Edition ", John Wiley & Sons (1999), P. 518-525. Compound A- (12) can be obtained by reacting compound A- (1l) with di-tert-butyl dicarbonate using a solvent such as tetrahydrofuran, for example plo. Alternative solvents include acetonitrile and DMF. That person skilled in the art will appreciate it. 20 that a base can also be added to a reaction mixture, although it is not essential. Suitable examples of a base include, but are not limited to, triethylamine and diisopropylethylamine. The reaction temperature is not particularly limited and is usually at room temperature at reflux and preferably room temperature at 60 ° C. Step A- (xii): This step is a step to obtain a compound A- (13) from compound A- (12). The compound A- (l3) is synthesized by reducing the nitro compound A- (l2) by a synthetic method known to a person skilled in the art. Examples of the method include reduction by catalytic hydrogenation using a noble metal catalyst such as Raney nickel, palladium, ruthenium, rhodium or platinum. Other reducing agents include tin chloride, for example. Examples of the solvent include alcoholic solvents as "5 such as methanol, ethanol and l-propanol, preferably ethanol. A reaction time is not particularly limited and is usually 0.5 to 24 hours and preferably 0.5 to 18 hours. The reaction temperature is usually room temperature. Alternative reduction reaction conditions include reaction with iron with an additive such as ammonium chloride or acid 10 hydrochloric, in an alcoholic solvent such as ethanol, at an appropriate reaction temperature, for example, 65 ° C. Step A- (xiii): This is a step to obtain a compound A- (14) from compound A- (13) by condensing compound A- (13) with a carboxylic acid and a condensing agent. The condensation reaction can be carried out under the same conditions as those usually used and described in the following documents. Examples of the known method include that in Rosowsky et al .; J. Med. Chem., 34 (1), 227-234 (1991), Brzostwska et al .; Heterocycles, 32 (10), 1968-1972 (1991) and Romero et al .; J. 20 Med. Chem., 37 (7), 998-1014 (1994). Compound A- (13) can be a free form or a saj. The solvent in this reaction is not particularly limited as it does not inhibit the reaction. Examples of the solvent include tetrahydrofuran, 1,4-dioxane, ethyl acetate, methyl acetate, dichloromethane, chloroform, N, N-dimethylformamide, toluene, acetonitrile and xylene. Examples of the condensing agent include CD1 (N, N'-carbonyldiimidazole), Bop. (1H-1, 2,3-benzotriazol-1-yloxy (tri (dimethylamino)) phosphonium hexafluorophosphate), WSC (l-ethyl-3- (3-dimethylaminopropyl) carbodiimide hydrochloride), DCC (N, N -dicyclohexylcarbodi-Mida), 6i diethylphosphoryl cyanide, PyBOP (benzotriazol-1-yloxytri (pyrrolidine) phosphonium hexafluorophosphate) and EDC · HC1 (1-ethyl-3- (3-dimethylaminopropyl) carbodiimide hydrochloride). Suitable conditions include an acid-activating agent, such as N, N'-carbonyl diimidazole. An equivalent '' 5 in a large excess of the acid can be used with respect to compound A- (13). An equivalent in a large excess of an organic base such as triethylamine or N, N-diisopropylethylaminy 'can be added where required. A reaction time is not particularly limited and is usually 0.5 to 72 hours and preferably 0.5 to 24 hours. The reaction temperature varies according to the raw material used, the solvent and others and is not particularly limited. Cold temperature at solvent reflux temperature is acceptable, cold at room temperature is preferable. Alternatively, compound A- (14) can be obtained by converting the desired carboxylic acid to the corresponding acid chloride and then reacted with the acid chloride with compound A- (13). Acid chloride can be synthesized by means known to a person skilled in the art. For example, the desired carboxylic acid can convert to the corresponding acid chloride by reaction with thionyl chloride in the presence or absence of a solvent, for example, dichloromethane, N, N'-dimethylimidazoline-2-one, NMP or DMF. One to two equivalents or a large excess of thionyl chloride can be used with the desired carboxylic acid. The reaction temperature is -30 ° C at reflux and preferably -10 ° C at room temperature. Acid chloride can also be formed by treating the acid with oxalyl chloride in a solvent such as dichloromethane in the presence of DMF. The reaction temperature is -30 ° C at room temperature and preferably -10 ° C at room temperature Alternatively, compound A- (14) can be obtained by converting the desired carboxylic acid to a mixed acid anhydride and then reacted by the acid anhydride mixed with compound A- (J3). The mixed acid anhydride can be synthesized by means known to a person skilled in the art. The synthesis is carried out by reacting the desired "5 carboxylic acid with a chloroformioate such as ethyl chloroformioate in the presence of a base such as trietijamine, for example. One to two equivalents of the chlorofonnioate and the base are used with respect to the desired carboxylic acid. The reaction temperature is -30 ° C at room temperature and preferably -20 ° C at room temperature 10 The condensation step of the acid anhydride mixed with compound 1- (13) is carried out by the reaction of the acid anhydride mixed with the compound 1- (13) in a solvent such as dichloromethane, tetrahydrofuran or N, N-dimethylphonamide, for example An equivalent in a large excess of the desired carboxylic acid is used with respect to compound A- (13). 15 A reaction time is not particularly limited and is usually 0.5 to 48 hours and preferably 0.5 to 12 hours. The reaction temperature is -20 ° C to 50 ° C and preferably -20 ° C at room temperature. Alternatively, compound A- (i4) can be obtained by converting the desired carboxylic acid into an activated ester and then reacting the active ester with the A- compound (13). The step of obtaining the active ester is carried out by reacting the desired carboxylic acid with a reagent for the active synthesis of the ester in a solvent such as 1,4-dioxane, tetrahydrofuran or N, N-dimethylformamide in the presence of a condensing agent such as DCC, for example. Examples of the active ester synthesis reagent include 25 N-hydroxysuccinimide. One to 1.5 equivalents of the active ester synthesis reagent and the condensing agent are used with respect to compound A- (13). A particularly limited non-C reaction time and is usually 0.5 to 48 hours and preferably 0.5 to 24 hours. The reaction temperature is -20 ° C to 50 ° C and preferably - 20 ° C at room temperature. The stage of condensation of the active ester with compound A- (13). it is carried out by reacting the active ester with compound A- (13) in a solvent such as dichloromethane, tetrahydroxy or N, N-dimethylformamide, for example. A large excess equivalent of the active ester is used with respect to compound A- (13). A reaction time is not particularly limited and is usually 0.5 to 48 hours and preferably 0.5 to 24 hours. The reaction temperature is -20 ° C to 50 ° C and preferably -20 ° C at room temperature. 10 Step A- (xiv): This step is a step to obtain compound A- (15) by deprotection of the t-butoxycarbonyl group of compound A- (14) - The reaction can be carried out under the same conditions as those generally used in the deprotection reaction of a t-15 butoxycarbonyl group such as the conditions described in a document such as TW Greene and PGM Wuts, "Protective Groups in Organic Chemistry, Third Edition", Jobn Wiley & Sons (1999), P. 518 -525. Compound A- (i5) can be obtained by reacting compound 1- (14) with a strong acid, for example, trifluoroacetic acid in the presence or absence of a solvent. Suitable solvents include dichloromethane. Alternative acids include hydrochloric acid in suitable solvents, such as dichloromethane or dioxane, for example. The reaction temperature is usually cooled to 80 ° C, preferably at room temperature. A particularly limited non-C 25 reaction time is usually 5 minutes to 48 hours and preferably 5 minutes to 12 hours. B. General preparation method B: In the formula, X, Y and A are as defined above. General preparation method B is an alternative method for the preparation of a compound A- (15) corresponding to the compound (I) according to the present invention from compound A- (l1) as a crude material through the steps multiple of step B- (i) to step B- (ii). Compound A- (l1) can be prepared as described in "5 General preparation method A or the examples. O Ü" ~ ^ X 'x / I no, xi I nh2 Í n fAV b () FA "db (" ) xY ^ rNH f: j! 7 "'' ÇJÀ" '':) JNH, 'é' f, h '1h) s a- (15) a- (11) f a- (16) ff Step B- (i): This step is a step to obtain a compound A- (16) from compound A- (1l). Compound A- (16) is synthesized by reducing the compound 10 nitro A- (1l) by a method of synthesis known to a person skilled in the art. Examples of the method include reduction by catalytic hydrogenation using a noble metal catalyst such as Raney nickel, palladium, ruthenium, rhodium or platinum. Other reducing agents include tin chloride, for example. Examples of the solvent include alcoholic solvents such as 15 such as methanol, ethanol and l-propanol, preferably ethanol. A reaction time is not particularly limited and is usually 0.5 to 24 hours and preferably 0.5 to 18 hours. The reaction temperature is usually room temperature. Alternative reduction reaction conditions include reaction with iron with an additive such as ammonium chloride or acid 20 hydrochloric, in an alcoholic solvent such as ethanol, at an appropriate reaction temperature, for example, 65 ° C. Step B- (ii): This is a step to obtain a compound A- (15) from compound A- (16) by condensing compound A- (13) with a carboxylic acid and a condensing agent. The condensation reaction can be carried out under the same conditions as those usually used and described in the following documents. Examples of the known method include that in Rosowsky et al .; J. Med. Chein., 34 (1), 227-234 (1991), 5 Brzostwska et al .; Heterocycles, 32 (10), 1968-1972 (1991) and Romero et al .; J. Med. Chem., 37 (7), 998-1014 (1994). Compound A- (15) can be obtained by converting the desired carboxylic acid to the corresponding acid chloride and then reacted with the acid chloride with compound A- (16). Acid chloride can be synthesized iO by means known to a person skilled in the art. For example, the desired carboxylic acid can convert to the corresponding acid chloride by reaction with thionyl chloride in the presence or absence of a solvent, for example, dichloromethane, N, N'-dimethylimidazoline-2-one, NMP or DMF. One to two equivalents or a large excess of thionyl chloride can be 15 used with respect to the desired carboxylic acid. That person skilled in the art will appreciate that the choice of reaction conditions used can affect the reaction result, for example, the conditions can affect whether the acid chloride reacts with the portions of aniline or isothiourea. That person skilled in the art will appreciate that reaction of thionyl chloride with a carboxylic acid results in the concomitant formation of 1 equivalent of hydrochloric acid in addition to the formation of the desired acid chloride. That person skilled in the art will appreciate that current conditions do not use a method of removing formed hydrochloric acid in this way. The hydrochloric acid formed in this reaction may or may not affect the selectivity of the reaction which may or may not result in a beneficial result. A reaction time is not particularly limited and is usually 0.5 to 48 hours and preferably 0.5 to 12 hours. The reaction temperature is -30 ° C at reflux and preferably -110 ° C at room temperature. Acid chloride can also be formed by treating the acid with oxalyl chloride in a solvent such as dichloromethane in the presence of DMF. The reaction temperature is -30 ° C at room temperature and preferably -10 ° C at room temperature. C. General preparation method C: "5 General preparation method C is an alternative method for the preparation of a compound A- (2) which is a synthetic intermediate of compound (I) according to the present invention from the compound A- (17) as a crude material through step C- (i) Compound A- (17) is commercially available. O "" . ) 7 ° YA (17) 'lj, 10 Step i: This step is a step to obtain a compound A- (2a) of A- (! 7) by adding a trifluoromethyl anion to compound A- (l7) to generate an intermediate or intermediate allyl alkoxide of trimethylsilyl ether which is then alkylated to give compound A- (2a). That person skilled in the art will appreciate that this transformation can be conducted in a pot or as two individual reactions. That person skilled in the art will appreciate the benefits and discounts of a compared pot reaction to conduct two separate reactions and choose the best method for their requirements accordingly. 20 Specifically, acrolein A- (17) can react with a trifluoromethyl anion that can be generated by the action of fluoride reagents such as (triMoromethyl) trimethylsilane to generate the corresponding allyl alkoxide or timetylsilyl ether. The solvent used in the reaction is not '-. particularly limited as it does not interfere with the reaction. Examples of suitable solvents include THF. Acceptable temperature ranges for the reaction include -10 ° C at reflux of the solvent, 67 preferably below room temperature. That person skilled in the art will appreciate that certain chemical reactions can be exothermic and that control measurements should be put in place of the control of these exotherms. That person skilled in the art will also appreciate that the "5 reaction exotherm can be controlled by allowing the solvent to reflux. Suitable precursors for generating triuoromethyl anion include, but are not limited to, (tnnuoromethyl) trimethylsilane (Rupert's reagent, Chem Rev '1997, 97, 757) and suitable fluoride sources include, but are not limited to, tetrabutylammonium fluoride (TBAF), tetrabutylammonium difluorotriphenylsilicate (TBAT) and cesium fluoride. Although the initial reaction temperature may be below room temperature it is acceptable to allow the reaction temperature to emerge the reflux of the solvent during the course of the reaction to the filtration reaction A reaction time is not particularly limited and is usually 5 minutes to 24 hours, preferably 1 to 6 hours. Those skilled in the art will appreciate that the alkoxide generated from this reaction can be reacted with an alkylating agent directly, such as tert-butyl bromoacetate and that this reaction can proceed with or without additional solvents. If additional solvents are required for the filtration reaction, then solvents such as DMF or NMP 20 are suitable. The reaction temperature is not particularly limited. Suitable reaction temperatures include room temperature at 80 ° C, preferably room temperature. A reaction time is not particularly limited and is usually 5 minutes to 1 week, preferably 1 to 48 hours. Alternatively, the alkylation reaction can be conducted under the conditions of phase transfer, for example, by the addition of an aqueous base, for example, aqueous sodium hydroxide. That person skilled in the art will appreciate when these conditions are applied when the use of a phase transfer catalyst is required. Suitable phase transfer catalysts include, but are not limited to, tetrabutylammonium hydrogen sulfate. That person skilled in the art will appreciate that the intermediate alkoxide must be extinguished, isolated and purified then subjected to independent alkylation conditions. This reaction can be carried out under the "5 same conditions as those usually used in the O-alkylation reaction of an alcohol compound (such as the conditions described in Tetrahedron Lett. 46 (2005) 45, 7751-7755). In this reaction, compound A- (2a) can be obtained by adding a base such as sodium hydride in a solution of the intermediate alcohol in THF to prepare an alkoxide and then reacting the alkoxide with tert-butyl bromoacetate, for example. solvent used in the reaction is not particularly limited as it does not inhibit the reaction and allows the starting material to be dissolved in it to a certain extent. Examples of the solvent include solvents such as THF, DMF and dimethyl sulfoxide. performed by causing] 3 15 equivalents of a suitable base to act in the presence of such a solvent. Examples of the base used include sodium hydride, potassium hydride and t-butoxypotassium. A reaction time is not particularly limited and is used 0.5 to 72 hours and preferably 0.5 to 12 hours. The reaction temperature is usually -20 ° C to 50 ° C. A more preferred result such as an improved yield can be achieved by adding a salt such as tetrabutijammonium iodide to this reaction. That person skilled in the art will appreciate that this reaction generates a new chiral center in compound A- (2a) and that compound A- (2a) is the same 25 as compound A- (2) except that compound A- (2) is enantiomerically pure considering compound A- (2a) to be racemic. That person skilled in the art will appreciate the enantiomerically pure compounds A- (2) and the - racemic compound A- (2a) are indistinguishable by analytical techniques such as NMR and liquid chromatography, however these are distinguishable by chiral HPLC. That person skilled in the art will appreciate the most appropriate methods for obtaining the. enantiomer desired from compound A- (2a) or a more advanced synthetic intermediate or final compound. Appropriate methods and appropriate stages of enantiomeric purification include those as detailed in the examples. Still in one aspect, the present invention provides a process for preparing a compound of formula (T), which comprises reacting a compound of formula A - (! 6), where X is defined in formula (I), 10 with a compound of the formula (IT) in which A and Y are as defined in the formula (I), or a C1-6 alkyl ester, acid anhydride or acid halide thereof, to produce a compound of the formula (I) and optionally convince the compound in an additional compound of formula (I) or formulation of a pharmaceutically acceptable salt thereof. 'y "N-N'2 Hojj' X) À: j 'Jz"' ° "". I'J)) ,, f, G H A- (16) f, G H 15 The reaction of A- (16) and (II) can be convenient. conducted in a solvent (such as tetrahydrofuran, 1,4-dioxane, ethyl acetate, methyl acetate, dichloromethane, chloroform, N, N-dimethijformamide, toluene, acetonitrile or xylene) at a temperature in the range of -30 ° C to 100 ° C. In an embodiment of the invention, compound (II) can conveniently take the form of an acid halide (for example, chloride) as it can be prepared by reacting the acid with a suitable reagent (for example, - thionyl chloride) It will be appreciated by one skilled in the art that in the process of the present invention certain functional groups such as groups of hydroxyl, carboxy or amino in the starting reagents may need to be protected by the protecting groups. In this way the preparation of the compounds of formula (I) may additionally involve the incorporation and removal of one or more protecting groups. The protection and deprotection of. functional groups is for example, described in T. W. Greene and P. G. M. "5 Wuts," Protective Groups in Organic Chemistry, Third Edition ", John Wiley & Sons (1999). The present invention will be described more specifically below with reference to Examples, Preparation Examples and Test Examples. However, the present invention does not is abbreviated to this. The abbreviations used in the Examples are conventional abbreviations known to a person skilled in the art. Some abbreviations are below: LCMS, LC / MS & LC-MS (liquid chromatography / mass spectrometry); MS (mass spectrum) ); MDAP (mass-directed self-purification); NMR (nuclear magnetic resonance); s, d, t, dd, m, br (singlet, doublet, triplet, doublet of doublets, multiplet, broad); Ph, Me, Et , Pr, Bu, Bn (phenyl, methyl, ethyl, propyl, butyl, benzyl); THF (tetrahydrofuran); DCM (dichloromethane); DMF ÇN, N-dimethylformamide); h, hr, hrs (hours); EDC & EDAC (N-3- (dimethylaminopropyl) -N'ethylcarbodiimide hydrochloride); DMAP (4-N, N-20 dimethylaminopyridine); DMSO (dimethyl sulfoxide); UV (ultraviolet); RT & rt (room temperature); Rt (retention time); min & mins (minutes); EtOAc (ethyl acetate); Et2O (diethyl ether); McCN (acetonitrile); EtOH (ethanol); MeOH (methanol); PhCH3 & P11Me (toluene); tlc (thin layer chromatography); TFA (trifluoroactic acid); NaOH (sodium hydroxide); HCl 25 (hydrochloric acid); NMP (N-methylpyrrolidinone or 1-methyl-2-pyrrolidinone); HPLC (high performance liquid chromatography); TBAF (tetrabutylammonium fluoride); BuLi (n-butyl lithium); PyBOP: benzotriazol-yloxytris (phrolidino) phosphonium hexafluorophosphate; Pd2dba3: useful (dibenzylidenoacetone) dipaladium; Pd (t-Bu3P) 2: bis (tri-t- butylphosphine) palladium; TFA: trifluoroacetic acid; pTLC: preparative thin layer chromatography; HRMS (high resolution mass spectrometry); Tr or Trt (trityl or triphenylmethyl). r 1 H NMR spectrum, a Bruker "5 AM series spectrometer was recorded operating at a 400 MHz (related) Eequence. Chemical changes in the proton nuclear magnetic resonance spectrum are recorded in Ô units (ppm) relative to tetramethylsilane and constants of connection (J) are recorded in Hertz (Hz) .The patterns are indicated as s: singlet, d: doublet, t; triplet, br; broad. "Room temperature" in the following Examples and Preparation Examples typically refers to about 10 ° C to about 35 ° C. "° /)" indicates ° /) by weight, unless otherwise specified. The chemical names were generated from the chemical structures using ChemBioDraw Ultra 11.0 and 12.0. 15 Description of the figures: Figure 1 is a typical chromatogram of an HPLC chiral isolation of compound 1- (20). Preparation example 1 Synthesis of ((4aS, 5S, 7aS) -7a- (5-amino-2-fluorophenyl) -5-20 (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4HAúro [3,4- d] [1,3] thiazin-2-yl) tert-butyl carbamate 1- (13) 1- (2) Synthesis of {K2S) JJJ-trinuorobut-3-en-2-i]] oxy} acetate tert-butyl To a suspension of trimethylsulphonium iodide (1 10 g) in 25 THF (500 ML) at -30 ° C was added lithium hexamethyldisilazide (530 ml, IN in THF) in portions for 45 mins. After stirring at -20 ° C for 20 mins, (S) -2-trifluoromethyloxy (37.97 g) was added at the same temperature for 15 mins and the mixture was allowed to warm up in RT and stirred for 3 hours. The paste was then added in portions in an ice-cold solution of tert-butyl bromoacetate (105.68 g) in NMP (200 niL). The resulting mixture was allowed to warm up in RT and stirred for 2 days, before dilution. with EtOAc (1 L). The organic layer was washed with sodium bicarbonate (sat., Aq., 4 x 400 ml), dried over MgSO4 and evaporated. The residue was purified by column chromatography on silica gel (5 ° /) of EtOAc in hexanes) to obtain the title compound (70.1 g) which was used in the purification without the subsequent step. H-NMR (400 MHZ, CDCl) δ (ppm): 1.30 (s, 9 H) 3.83 -3.96 (m, 2 H) 4.14 -4.2! (M, IH) 5.34 -5.48 (m, 2 H) 5.56 - 5.71 (m, IH) 'r' d) j '_, jjl "fj: jJ F" is 1- (4) 1- (1) F 1- (2) F 1- (3) i; '-) O) is Fjj,, ÁJ fj O, ""! _ "" F'E' - {FF '' F7ylH 1- (5) 1- ( 6), :) ÍZ C _ jjj _,: ^ iyNH2 F, 1 HH Fè H OH FF FF 1- (8) 1- (9) 1- (10) / | NO2 / l NO2 i I NH, 'fjjjÍ "' 'F:' ^ jj" "° ''> jY" ° ' F I H F I H FF 1- (11) 1- (12) F 1- (13) 10 1- (3) Synthesis of (S) -N-methoxy-N-methyl [-2 - ((1, 1,] - tnnuorobut- 3-en-2-yl) Oxy) acetamide {[(2S)-1,1,1-trifluorobut-3-en-2-yl] Oxy} tert-Butyl acetate (70.1 g, crude) was dissolved in icy formic acid (200 mL). The mixture was allowed to warm to RT and stir overnight. The reaction mixture was then concentrated under reduced pressure, toluene (200 ml) was added, the mixture concentrated, before the second addition of toluene (200 ml) and concentration in an Oil. The residue was dissolved in DCM (600 niL), cooled in an ice bath and N, N'-carbonyl diimidazole (35 g) was _ 5 added in portions over 20 mins. After stirring for 45 mins, N, O-dimethyl hydroxylamine hydrochloride (22 g) was added and the mixture was · Reaction was allowed to warm to RT and stir overnight. Saturated NaHCO3 (500 niL) and brine (250 ml) were then added and the mixture extracted with EtOAC (3 x 750 ml). The combined organic portions were dried over MgSO4 and evaporated. The residue was purified by column chromatography on silica gel (1 ° /) to 30 ° /) of EtOAc in hexanes) to obtain the title compound (25.17 g). 'H-NMR (400 MHZ, CDCl) Õ (ppm) 3.21 (s, 3 H), 3.71 (m, 3 H), 4.36 - 4.51 (m, 3 H), 5.54 - 5.69 (m, 2 H), 5.84 (ddd, j = 17 , 7, 10.4, 7.3 Hz, 1 H) 15 1- (4) Synthesis of (S) J {2 Auorophenyl) -2 - ((1,1, l-l-trifluorobut-3- en-2-yl ) oxy) ethanone A solution of n-butyllithium in hexane (2.50 M; 90 niL) was added to the drops for 25 mins in a solution of 2-bromofluorobenzene (40.35 g) in THF (250 niL) under an atmosphere N2 at - 20 78 ° C. The reaction solution was allowed to warm to -60 ° C and stirred for 60 min. (S) -N-methoxy-N-methyl-2 - ((1,1,1-trinuorobut-3-en-2-yl) oxy) acetamide (40 g) in THF (25 niL) was added to the drops in a reaction solution, and after stirring at -60 ° C for 2 h, NH, aqueous CI (100 mL) was added to a reaction solution, followed by heating in RT. Brine (200 ml) was added to a reaction solution and the mixture was extracted with EtOAc (3 x 400 ml). The combined organic portions were dried over MgSO4, evaporated and the residue was purified by silica gel column chromatography (1 ° /) to 10 ° 4 EtOAc in hexanes) to obtain the title compound (33.59 g). 'H-NMR (400 MHz, CDCl): lij (ppm): 4.40 (pentet, J 6.3 Hz, 1 H) (3aR, 4S, 6aS) -6a- (2-nuorophenyl) -4- (trinuoromethyl) hexahydron [3,4-c] isoxazole (28.76 g) was dissolved in acetic acid (200 niL) and the solution was ground at 0 ° C. Zinc (50 g) was added and the reaction mixture was allowed to warm up and stir at RT for 16 h. The reaction mixture was then "5 diluted with EtOAc (500 ml) and filtered through celite, washing with an additional 500 ml of EtOAc. The combined organic portions were evaporated, dissolved in chloroform (200 ml), and Ammonia (28 ° / ) aq., 250 ml) was added slowly, the layers were separated and the aqueous portion was still extracted with chloroform (2 x 250 ml). The combined organic extracts 10 were dried over anhydrous MgSO4 and evaporated to produce the titer (3 1.12 g) that was used in the subsequent step without further purification. H-NMR (400 MHz, CDCl) δ (ppm): 2.93 (ddd, j = 7.7, 4.9, 2.5 Hz, 1 H), 3.84 (dd, j = 12 , 4, 4.8 HZ, 1 H), 4.05 (dd, J = 9.2, 3.2 Hz, 1 H), 4.17 (dd, J =] 2,4,2,3 Hz, 1Hj), 43l (d, j = 93Hz, 1Fn, 4.72 (quin, j = 7.3Hz, IH), 7.13 15 (ddd, j = 13,1,8,8,1,3 Hz, IH), 7.22 (td, j = 7.6, 1.3 Hz, 1H), 7.31-7.40 (mjH), 7.5l (td, j = 8.0.1.6Hz, 1H) 1- (8) Synthesis of N - ((((3S, 4R, 5S) -3- (2-nuorophenyl) -4- (hydroxyl) -5- (hinuoromethyl) tetraidromrmo-3-yl) cMbamothioyl) bemamide Isoticianate benzoyl (19.0 mL) was added to a solution containing ((2S, 3R, 4S) -4-mino-4- (2-Quorophenyl) -2- (WiRuoromethyl) tetrahydrofuran-3-yl) methanol (28 , 72 g) in DCM (150 ml) and the mixture was stirred at RT for 8 h. Sodium bicarbonate (sat., Aq., 200 ml) was then added, the mixture extracted with EtOAc (3 x 300 ml), dried over MgSO4 and concentrated under reduced pressure The residue was purified by column chromatography on silica gel (5 ° /) at 30 ° 4 EtOAc in hexanes) to obtain the title compound (37.07 g). 'H-NMR (400 MHz, CDCJ,) Ô (ppm): 3.22 (dd, J = 8, lA5HL1H), 331 (td, j = 8,0,3,0HL1H), 3,94-4, 07 (m, lH), 4.3l -4.46 (m, 1 H), 4.53 (d, j = 9.9 Hz, IH), 4.83 (d, j = 9.9 HZ, IH), 6.97 - 7.14 (m, 1 H), 7.22 (td, J = 7.7, 1.3 HZ, 1 H), 7.31 - 7.45 (m, 1 H ), 7.49 - 7.61 (m, 2 H), 7.61-7.70 (m, IH), 7.75 (td, j = 8.1, 1.5 Hz, IH), 7.79-7.93 (m, 2H), 8.90 (s, 1H), 11.85 (s, 1H) 1- (9) Synthesis of N {(4aS, 5S, 7aS) -7a- (2-fluorophenyl) -5- (trinuoromethyl) -4a, 5,7,7a-tetrahydro-4H-Wo [3,4-d] [1,3] tiwin-2-yl) benzamide "5 N - ((((3 S, 4R, 5S) -3- (2-Fluorophenyl) -4- (hydroxyl) -5- (triyl fluoromethyl) tetrahydro-amine-3-yl) carbamothioyl) benzamide (3 l, lg) was dissolved in pyridine (150 ml) and the mixture sieved at -20 ° C. Trifluoromethanesulfonic anhydride (14.0 inlj) was added to the drops for 30 minutes and the reaction was allowed to warm to 0 ° C. After stirring for 2 h, the The reaction was quenched by the addition of ammonium chloride (sat., Aq., 400 niL) and extracted with EtOAc (3 x 500 mL). The combined organic extracts were dried over MgSO4, concentrated in vacuo and purified by column chromatography on silica gel (2 ° 4) at 30 ° /) of EtO (c / hex) to obtain the title compound (18.50 g ). 'H NMR (400 MHz, CDCl3) 6 ppm 2.86 (dd, J = 13.9, 3.5 15 Hz, 1 FI), 3.25 (d, J = 13.6 Hz, 1 H), 3.61 (br, s ,, 1 H), 4.00 -4.10 (m, 1 H) , 4.66 (d, J = 8.8 Hz, 1 H), 4.78 - 4.87 (m, 1 H), 7.12 - 7.60 (m, 6 H), 7.68 - 7.73 (m, 1 H), 7.99 - 8.16 (br, s ,, 2 H), 8.62 - 8.66 (m, 1 H) 1- (10) Synthesis of N - ((4aS, 5S, 7aS) -7a- (2-fluorophenyl) -5- (triMoromethyl) -4aSj, 7a-tetrahydro-4H-nlro [3,4-d] [13] thiazin-2-yl) benzamide 20 N - ((4aS, 5 S, 7aS) Àa {2-Fluorophenyl) -5- (tMuoromethyl) - 4a5,7,7a-tetrahydro-4HAúro [3,4-d] [1,3] thiazin-2- il) benzamide (21.5 g) was dissolved in methanol (160 ml), 1,8-diazabicyclo [5.4.0] undec-7-ene (16.29 g) was added and the solution was heated to reflux (heating block temperature of 80 ° C). After 16 h, the reaction mixture was concentrated 25 under reduced pressure and the residue purified by column chromatography on silica gel (10 ° /) at 60 ° / 6 EtOAc in hexanes) to yield the title compound (13.82 g). 'H NMR (400 MHZ, CDCl3) 6 ppm 2.85 (dd, J = 13.6, 3.8 Hz, 1 H), 3.14 (dd, J = 13.5, 3.2 HZ, 1 H), 3.33 - 3.45 (m, 1 H), 3.92 (dd, J = 8.1, 2.0 Hz, 1 H), 4.49 (br, s ,, 2 H), 4.63 - 4.76 (m, 2 H), 7.08 (ddd, J = 12, 6, 8.1, 1.0 Hz, 1 H), 7.13 - 7.22 (m, 1 H), 7.25 -7.36 (m, 1 H), 7.44 (td, J = 8 , 0, 1,9Hz, IH) 1- (ll) Synthesis of (4aS, 5S, 7aS) -7a- (2-nuoro-5-nitrophenyl) -5- (trinuoromethyl) -4a, 5,7,7a- tetrahydro-4H-hro [3,4-dl [1,3] thi-in-2-amine "5 N - ((4aS, 5 S, 7aS) -7a {2-Fluorophenyl) -5- (trifluoromethyl) - 4a, 5,7,7a-tetrahydro-4H-hro [3,4-d] [1, 3] thiazin-2-yl) benzamide (5.15 g) was dissolved in TFA (75 ml) and the solution was poured at 0 ° C. Sulhiric acid (conc., 20 niL) was added, followed by fumigation of nitric acid (2 mlj) in drops for 20 mins. After stirring at 0 ° C for 90 mins, the mixture of The reaction was poured into ice (200 g) and basified at pFI 12 with 6N NaOlil (aq.). After allowing the ice to melt, the mixture was extracted with EtOAc (3 x 500 ml) and the combined organic portions dried over MgSO4 and evaporated to produce the title compound (22.1 g, purity approximately 71 ° 4) which was used in purification without the subsequent step. È NMR (400 MHz, 15 CDCl,) ç; ppm 2.89 (d, J = 3.8 Hz, 1 H), 3.09 (br, s ,, 1 H), 3.28 - 3.54 (m, 1 H), 3.80 - 4 , 03 (m, 1 H), 4.50 - 4.70 (m, 3 H), 4.71 - 4.86 (m,] H), 7.21 - 7.30 (m, 1 H) , 8.18 -8.28 (m, i H), 8.45 (dd, J = 6.8.2.8 Hz, IH) 1- (12) Synthesis of ((4aS, 5S, 7aS) - 7a- (241uoro-5-nitrophenyl) -5- (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4H-nlro [3,4-d] [1,3] thiazin-2-ij) carbamate 20 tert-butyl (4aS, 5 S, 7aS) -7a {2-Fluoro4-nitrophenyl) -5- (trifluoromethyl) - 4a, 5,7,7a-tetrahydro-4H-Wo [3,4-d] [ 1.3] tiwin-2-mine (20.6 g, brut) was dissolved in THF (300 mL), di-tert-butyl dicarbonate (12 g) was added in portions over 20 mins and the reaction mixture was heated to 25 60 ° C. Still the portions of di-tert-butyl dicarbonate (10 g) were added until the starting material was consumed by TLC. The reaction mixture was drained and sodium bicarbonate (sat., Aq., 200 ml) and brine (200 ml) were added. The mixture was then extracted with EtOAc (3 x 500 mL) and the combined organic portions were dried over MgSO4 and evaporated. The residue was purified by column chromatography on silica gel (10 ° /) to 25 ° /) of EtOAc in hexanes) to yield the title compound (16.62 g). 'H NMR (400 MHz, CDCl) Õ ppm 1.55 (s, 9 H), 2.73 - 2.84 (m, IH), 2.92 - 3.05 (m, 1 H), 3.43 - 3.55 (m, 1 H), 3.81 - 3.94 (m, 1 H), 4, 57 "5 (d, J = 8.3 Hz, 1 H), 4.73 - 4.83 (m, 1 H), 7.19 - 7.39 (m, 2 H), 8.20 - 8, 29 (m, 1H), 8.32 (d, j = 6.8Hz, 1H) 1- (13) Synthesis of ((4aS, 5S, 7aS) -7a- (5-amino-2Aorophenyl) -5- ( trinuoromethyl) -4a, 5,7,7a-tetrahydro-4H-Wo [3,4-d] [1,3] thiazin-2-yl) tert-butyl carbamate 10 ((4aS, 5S, 7aS) -7a {2Auoro-5-nitrophenyl) -5- (trifluoromethyl) - 4a, 5,7,7a-tetrai & o-4H-mro [3,4-d] [1,3, tiwin-2-yl) tert-Butylbamate (! 6.61 g) was dissolved in ethanol (250 ml) and tin dihydrate chloride (25.0 g) was added. After stirring at RT for 18 h, the solution was poured into NaOifl (2N aq., 300 niL) and ceiite® (-50 g) was added. The mixture The resulting 15 was filtered through more celite® and extracted with EtOAC (2 x 500 mL). The combined organic portions were dried over MgSO4 and evaporated to yield the title compound (15.52 g). This material must be used crude but a portion was purified by column chromatography on silica gel (20 ° /) to 50 ° /) of EtOAc in hexanes) to produce pure material 20 (recovery of 79 ° 4). 1 H NMR (400 MHz, CDCl 3 ppm 1.53 (s, 9 H), 2.77 (d, J = 14.4 Hz, 1 H), 3.09 (br, s ,, 1 H), 3.46 (br, s ,, 1 H), 3.62 (br, s ,, 2 H), 3.87 (br, s ,, 1H), 4.6l (d, j = 8.6 HZ , 1H), 4.71 (br, s ,, IH), 6.61 (br, S ,, 2 H), 6.85 6.95 (m, IH) Preparation example 2 25 Synthesis of ((4aS , 5S, 7aS) -7a- (5-mino-2-duorophenyl) -5- (hinuoromethyl) -4a, 5,7,7a-tetrahydro-4H-hro [3,4-d] [l ', 3] thiazin-2-yl) carb = tert-butyl act 2- (10) ) j '"°". : jj. jjjj: í f 1- (3) 'i ff 2- (1): f "2- (2)' fi I" ff AJ], ^ UA, 1] h ¢ nnf ^ l "° b" G ^> °: í! "" 'O, r'jç i- j)' 7 ('F "j' 6h '> rú>' ó ff 2- (4) ff oh 2- (3) 2- (5) _ ,) J'3_, JNH2 -, j)) ff 'È f 2- (6) 2- (7) 2- (8): J, J no2::;, j mh2'> ÁY "°" "ÀJ '"°" f 2- (9) f 2- (10) 2- (1) Synthesis of (S) -1- (2,3-dinuorophenyl) -2 - ((1,1,1-trifluorobut-3 -en-2-yl) oxy) ethanone A solution of n-butyllithium in hexane (2.50 M, 13.5 mL) was added to the drops for 20 mins in a solution containing 1-bromo-2,3-5 difluorobenzene (6.50 g) in Et2O (50 inlj) under an N2 atmosphere at -78 ° C. The reaction solution was allowed to stir for 60 mins. (S) -N-methoxy-N-methyl-2-. ((l, lj-tMuorobut-3-en-2-ii) oxy) acetamide) (5.20 g) in Et2O (10 niL) was then added to the drops in a reaction solution, and after stirring at -78 ° C for 1 h, aqueous yci (50 mjj) was added to a reaction solution, followed by heating at RT. NaHCO3 (sat. Aq., 100 ml) was added to a reaction solution and the mixture was extracted with EtOAc (3 x 100 niL). The combined organic portions were dried over MgSO4, evaporated and the residue was purified by column chromatography on silica gel (1 °) to 0 ° /) of EtOAc in hexanes) to obtain the title compound (3.91 g) . 1H NMR (400 MHz, CDCl) δ ppm: 4.33 - 4.43 (m, 1 H), 4.80 - 4.84 (m, 2 H), 5.55 - 5.67 (m, 2 H), 5.76 - 5.94 (m, 1 H), 7.18 - 7.28 (m, 1 H), 7.37 - 7.47 (m, 1 H ), 7.70 (ddt, j = 7.9, 6.0, 1.7 Hz, 1 H) 2- (2) Synthesis of (S) -1- (2,3-dinuoropheni]) - 2- ((1,1, 1, 1-triorobut-3-en-2-yl) oxy) ethanone oxime "5 (S) -1- (2,3-diAuorophenyl) -2 - ((1, 1, 1-trinuorobut- 3-en-2-yl) oxy) ethanone (3.91 g) was dissolved in anhydrous methanol (40 ml) and hydroxylamine hydrochloride (1.25 g) and sodium acetate (1.68 g) were added. The reaction mixture was heated to 50 ° C for 90 min, then dried in RT, concentrated in vacuo and the residue purified by chromatography on 10 silica gel (2 ° / 0 to 20 ° /) of EtOAc in hexanes) to produce the title compound as a mixture of geometric isomers (4.10 g) .1H-NMR (400 MHz, CDCl) δ (ppm): 4.04 - 4.26 (m 1 H), 4.43 - 4, 55 (m, 0.4 H) 4.80 - 4.89 (m, 1.6 H) 5.39 - 5.55 (m, 2 H) 5.64-5.80 (m, 1 H) 7.05 - 7.30 (m, 3 H), 7.76 (br s, 0.2H), 8.30 (brs, 0.8H). J5 2- (3) Synthesis of (4S) -6a- (2,3-difluorophenyl) -4- (triBuoromethyl) hexahydrofúro [3,4-c] isoxazole (S) -l- (2,3-dinuorophenyl) - 2 - ((1,1,1-trinuorobut-3-en-2-yl) oxy) ethanone oxime (4.10 g) was dissolved in xylenes (40 ml) and hydroquinone (380 mg) was added. The reaction mixture was heated to reflux 20 (heating the block temperature 140 ° C) for 20 h, then chilled and evaporated. The residue was purified by cojuna chromatography on silica gel (1 ° /) to 25 ° /) of EtOAC in hexanes) to obtain the title compound (3.16 g). 'H NMR (400 MHz, CDCl) Õ ppm 3.77 (br, s ,, IH), 3.99 - 4.16 (m, 1 H), 4,! 6- 4.22 (m, 1H ), 4.22 -4.44 (m, 2 H), 4.51 (d, j = 9.9 Hz, IH), 5.44 25 (s, 1 H), 7.07 - 7.24 ( m, 2 H), 7.38 (br, s ,, IH) 2- (4) Synthesis of ((2S, 3R, 4S) -4-amino-4- (2,3-difluorophenii) -2- ( trif1uoromethyl) tetrai & oWrano-3-yl) methanol (4S) -6a- (2,3-dinuorophenyl) -4- (trinuoromethyl) hexahydro [3,4-cjisoxazole (3.16 g) was dissolved in acetic acid (20 ml) and the mixture of 8i reaction started at 0 ° C. Zinc (5.0 g) was added and the reaction was allowed to warm up and stir at RT for 20 h. The reaction mixture was then diluted with EtOAc (50 mL) and filtered through celite®, washing with an additional 100 niL of EtOAc. The combined organic portions were evaporated, dissolved "5. in CHCl3 (20 ml / ml), and Ammonia (28 ° / aq., 25 ml / ml) was added slowly. The layers were separated and the aqueous portion was further extracted with CHCl3 (2 x 25 mL). The combined organic extracts were dried over anhydrous MgSO4 and evaporated to produce the title compound (3.12 g) which was used in the subsequent step without further purification. 'H NMR (400 MHZ, CDC! 3) 10 6 ppm 2.93 (ddd, j = 7.8, 5.1, 2.8 Hz, 1 H), 3.85 (dd, J = 12.4, 5.1 Hz, 1 H), 4.03 (dd, J = 9.1, 2.8 Hz, 1 H), 4.14 (dd, J = 12.3, 2.7 Hz, 1 H), 4.35 (d, J = 9.1 Hz, 1 H), 4.68 (quin, J = 7.3 Hz, 1 H), 7.09 - 7.25 (m, 2 H), 7.25 - 7.34 (m, 1 H) 2- (5) Synthesis of N - ((((3 S, 4R, 5S) -3- (2,3-dinuorophenyl) -4- i5 (hydroxyl) -5- (trinuoromethyl) tetrahydro-3-yl) carb = otioyl) benzamide Benzoyl isoticyanate (2.0 mlj) was added to a solution containing ((2S, 3R, 4S) -4-amino-4- (2,3-dinuorophenyl) -2- (trinuoromethyl) tetrahydroWrano-3-yl) methanol (3.12 g) in DCM (20 ml) and the mixture was stirred at RT for 18 h. Sodium bicarbonate (sat., Aq., 50 niL) was then added, 20 the mixture extracted with EtOAc (3 x 75 niL), dried over MgSO4 and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (5 ° /) to 40% EtOAc in hexanes) to obtain the title compound (4.18 g). 'H NMR (400 MHZ, CDCl) δ ppm 3.12 (dd, J = 7.6, 4.3 Hz, 1 H), 3.18 - 3.29 (m, 1 H), 4.03 (ddd, j = 12.3, 7.2, 4.5 Hz, 1 H), 4.35 - 25 4.49 (m, IH), 4.59 (d, J = 9.9 Hz, 1 H), 4.81 (d, j = 9.6 Hz, 1 H), 7.07-7, 23 - (m, 2H), 7.49 (t, j = 7.2HL1H), 7.56 (t, j = 7.7Hz, 2H), 7.67 (t, j = 7.5 W, lH) , 7.88 (d, j = 7, lW, 2H), 8.92 (s, lW, 11.89 (s, 1H 2- (6) Synthesis of N {(4aS, 5SjaS) -7a- (2,3-difluorophenyl) -5- (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4H-fúro [3,4-d] [i, 3] thiazin-2-yl) benzamide N {(((3S, 4R, 5S) -3 {2,3-Difluorophenyl) -4- (hydroxyl) -5- (trifluoromethyl) tetrai & ohrano-3-yl) carbamothioyl) benzamide (2.99 g) was dissolved in pyridine (14 ml) and the mixture is stirred at -20 ° C. The trifluoromethanesulfonic anhydride (1.55 ml) was added to the drops over 10 min and the reaction mixture was allowed to warm to 110 ° C. After stirring for 2 h, an additional portion of trifluoromethanesulfonic anhydride (1.0 mL) was added to the drops for 10 min, the reaction was added for an additional 2 hours and was then quenched by the addition of ammonium chloride (sat., Aq. , 50 ml-) and extracted with EtOAc (3 x 100 ml). The combined organic extracts 10 were dried over MgSO4, concentrated in vacuo and purified by column chromatography on silica gel (5 ° / 0 to 20 ° 4) EtOAc / hex) to obtain the title compound (1.20 g) . 'H NMR (400 MHz, CDCl3) Õ ppm 2.86 (d, J = 10.6 Hz, 1 H), 3.20 (br, s ,, 1 H), 3.55 (br, s ,, 1 H), 4.04 (br, s ,, IH ), 4.65 (d, J = 8.8 Hz, 1 H), 4.81 (br, s ,, i H), 7.06 - 7.24 (m, 3 H), 7.40 - 7.64 15 (m, 3 H), 7.82 - 8.21 (m, 2 H) 2- (7) Synthesis of (4aS, 5S, 7aS) -7a- (2,3-difluorophenyl) -5 - (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4H-njro [3,4-d] [1,3] thiazin-2-amine N - (((4aS, 5S, 7aS) -7a- (2 , 3-DiQuoropheni]) - 5- (trinuoromethyl) - 4a5j, 7a-tetrahydro-4HAijro [3,4-d] [i, 3] thiazin-2-yl) benzamide (2.00 g) was 20 dissolved in methanol (250 niL), 1,8-diazabicyclo [5.4.0] undec-7-ene (1.53 g) was added and the solution was heated to reflux (heating the block temperature 80 ° C). After 3 h, the reaction mixture was concentrated under reduced pressure, diluted with water (100 nL) and extracted with EtOAc (3 x 100 ml). The combined organic portions were dried over MgSO4, 25 evaporated and the residue purified by column chromatography on silica gel (0 ° /) at 50 ° 4 EtOAc in hexanes) to yield the title compound (1.42 g). 'H NMR (400 MHz, CDCl) 15 ppm 2.86 (dd, J = 13.6, 3.8 Hz, 1 H), 3.15 (dd, J = 13.8, 3.2 Hz, 1 H), 3.27 - 3.42 (m, 1 H), 3.93 (dd, J = 8.2, 1.9 Hz, 1 H), 4.39 - 4.78 (m, 4 H), 6.96 - 7.25 (m, 3 H) 2- (8) Synthesis of (4aS, 5S, 7aS) -7a- (2,3-dinoro-5-nitrophenyl) -5- (trifluoromethyl]) - 4a, 5,7,7a-tetrahydro-4H-n] ro [3,4-d] [1,3] thiazin-2-amine (4aS, 5S, 7aS) -7a- (2,3-Dinuorophenyl) -5- (trinuoromethyl) - 4a, 5,7,7a- tetrahydro-4HAlro [3,4-d] [1,3] thiazin-2-amine (1.42 g) was dissolved "5 in TFA (6 ml) and the solution was cooled at 0 ° C. Sulfuric acid ( conc., i mjj) was added, followed by fumigation of nitric acid (0.30 ML) in drops for 20 mins After stirring at 0 ° C for 1 h, the reaction mixture was poured into ice (50 g) and basified at pH 12 with 2 N NaOH (aq.) After allowing the ice to melt, the mixture was extracted with EtOAc (3 x 75 nil) and the combined organic portions dried over MgSO4 and evaporated to produce the title compound (1.91 g, purity approximately 80 ° 4) which was used in the purification without the subsequent step. 'H NMR (400 MHz, CDCl3) δ ppm 2.88 (dd, J = 13.8, 3.9 HZ, 1 H), 3.11 (dd, J = 13.6, 2.8 FIz, 1 H), 3.37 (dt, J = 7.4, 3.5 Hz, 1 H), 3.93 ( d, J = 7.8 Hz, 1 H), 4.53 - 4.83 (m, 4 H), 8.09 15 (ddd, j = 9.0, 6.3, 2.9 Hz, 1 H), 8.27 (dt, J = 5.2, 2, 6 FLz, 1 H) 2- (9) Synthesis of ((4aS, 5S, 7aS) -7a- (2,3-dinuoro-5-nitrophenyl) -5- (triAuoromethyl) -4a, 5,7,7a- teuahydro-4H-hro [3,4-d] [1,3] thiazin-2-yl) tert-butyl carbamate (4aS, 5 S, 7aS) -7a- (2,3-Dinuoro-5-nitrophenyl) -5- (mnuoromethyl) - 20 4a, 5,7,7a-tetrahydro-4H-hro [3,4-d] [1,3] tiwin-2-amine (1.91 g, crude) was dissolved in THF (10 ml), di-tert-butyl dicarbonate (1.3 g) was added in portions over 20 mins and the reaction mixture was heated to 65 ° C. After 3 h, the reaction mixture was drained and sodium bicarbonate (sat., Aq., 50 niL) was added. The mixture was then extracted with EtOAc (3 x 750 25 ml) and the combined organic portions were dried over MgSO4 and evaporated. The residue was purified by gel column chromatography. silica (0 ° /) to 20 ° /) of EtOAc in hexanes) to produce the title compound (1.43g crude, in a mixture with the bis-boc version). 2- (10) Synthesis of ((4aS, 5S, 7aS) -7a- (5-amino-2,3- dinuorophenyl) -5- (trinuoromethyl) -4a, 5,7,7a-teKaidro-4H-hro [3,4-d] [1,3, thiazm- 2-yl) tert-butyl carbamate The crude mixture obtained in preparation example 2- (9) containing ((4aS, 5S, 7aS) -7a- (2,3-dinoro-5-nitrophenyl) -5- (trinuoromethyl) - "5 4a, 5,7,7a-tetrahydro- 4n-hro [3,4-d] [1,3] ti = in-2-yl) tert -Butylbamate) (1.43 g) was dissolved in ethanol (25 ml) and dihydrochloride chloride tin (2.50 g) was added in. After stirring for 18 h, the solution was poured into NaOlil (2N aq., 100 mL) and extracted with EtOAc (3 x 100 niL). The combined organic portions were dried over MgSO4, evaporated and purified 10 by column chromatography on silica gel (0 ° /) to 30 ° /) of EtOAc in hexanes) to first produce the bis-boc product (730 mg) and secondly the title compound (300 mg H NMR (400 MHz, CDCIJ 15 ppm 1.53 (s, 9H), 2.76 (dd, j = 13.9.3.8 Hz, 1 H), 3.08 (d, j = 13.6 HZ, IH), 3.36 -3.45 (m, 1 H), 3.71 (br, s ,, 2 H), 3.90 (d, J = 8.3 Hz, 1 H ), 4.57 (d, J = 15 8.6 Hz, 1 IF), 4.68 -4.79 (m, 1 H), 6.30 - 6.37 (m, 1 H), 6.42 - 6.49 (m, 1 H) Preparation example 3: Synthesis of 5-ethoxypyrazine-2-carboxylic acid ( 3- (2)) ,,: j "· c,;) ° í ·, 0: J ° Õ Õ 3- (1) 3- (2) Synthesis of 5-ethoxypyrazine-2-carboxy! - (1) A stirred solution of 20 methyl 5-chloropyrazine-2-carboxylate (0.50 g) in ethanol (10 ML) was dissolved at 0 ° C and sodium ethoxide (21 ° /, w / w of the solution in ethanol, 1 ml) was added over 10 mins. After allowing to warm up in RT and stirred for 2 h, water (100 ml) was added and the mixture extracted with EtOAc (2 x 150 ml). The combined organic portions were dried over MgSO4 and evaporated to produce the title compound. 25 (0.65 g, purity approximately 85 ° /). 'H NMR (400 MHz, CDCl3) 6 ppm 1.45 (t, j = 7, lW, 3H), 1.46 (t, j = 7, lW, 3H), 4.48 (q, j = 7 , lW, 2H), 4.49 (q, j = 7, lHz, 2Hi), 8.28 (d, j = 1.3Hz, lH), 8.88 (d, j = l, 3Hz, iH) Synthesis of 5-ethoxypyrazine acid -2-carboxylic acid 3- (2) 5-ethoxypyrazine-2-carbonlate (0.65 g, approximately 85 ° /} purity) was dissolved in dioxane (3 ml) and water "5 (3 mL) was added, followed by lithium hydroxide monohydrate (255 mg, in portions over 10 mins). After stirring at RT for 24 h, Et2O (25 mlj) and NaHCO3 (sat., Aq., 25 mlj) have been added. The layers were separated and the organic layer was extracted with NaOH (1 N, aq., 25 ml). The combined aqueous portions were acidified with 6N HCl at pH 2 and the mixture extracted with EtOAc (3 x 40 niL). The combined EtOAc extracts were dried over MgSO4 and evaporated to produce the title compound as a yellowish-white powder. 'H NMR (400 MHz, CDCl3) Ô pprn 1.47 (t, J = 7.1W, 3H), 4.53 (q, j = 7.1 &, 2H), 8.16 (d, j = 1.2, 2H, lH), 8.98 (d, j = 1.2Hz, IH) 15 Example of preparation 4: Synthesis of 5-ethoxypyrazine-2-carboxylic acid (4- (3)) OOF , FF,, F H, N :: r "-—>; jA -, 0: ^ - HO 7" d Ô Ó Õ 4- (1) 4- (2) 4- (3) Methyl 5-acetylpyrazine-2-carboxylate 4- (1) 5-Acetylpyrazine-2-carboxamide (3.275 g) was dissolved in methanolic HCl (1.25 N, 150 ml) and the reaction mixture was heated to reflux and stirred during the night. After drying, sodium bicarbonate was added and the mixture was extracted with EtOAc. The EtOAc layer was dried over MgSO4 and evaporated to yield the title compound (3.79 g, approximately 90 ° /) of purity). 'H NMR (400 MHz, CDC] 3) Õ ppm 2.78 (s, 3H), 4.10 (s, 3H), 9.33 (d, j = 1.5w, 1H), 9.36 ( d, j = 1.5m, 1H) 25 methyl 5- (1,1-difluoroethyl) pyrazine-2-carboxylate 4- (2) 5-acetylpyrazine-2-carboxylate methyl (300 mg, "T 86 approximately 90 ° /) of purity) was dissolved in DCM (15 mL) and dissolved at 0 ° C under nitrogen. Bis (2-methoxyethyl) aminoenxoEe trifluoride (0.61 mlj) was added to the drops and the reaction mixture allowed to warm in RT and stir> " during the night. Sodium bicarbonate (sat., Aq.) Was added "5 carefully and the mixture extracted with DCM. The organic portions were dried over MgSO4, evaporated and purified by silica gel chromatography (35 ° 4 EtOAC in hexane) to yield the title compound (155 mg) as a white solid, 'H NMR (400 MHz, CDC] 3) Ô ppm 2.00 (t, J = 18.8Hz, 3H), 4.01 (s, 3Hj) N, 98 (d, j = 1sI1z, 1H), 9.24 (d, j = l, 5Hz, 10 IH) methyl 5- (1,1-Difluoroethyl) pyrazine-2-carboxylic acid 4- (3) 5 {1,1,1-difluoroethyl) pyrazine-2-carboxylate (0.65 g, approximately 85 ° / ) of purity) was dissolved in dioxane (2 nL) and water (2 nil) was added, followed by Jithium hydroxide monohydrate (54 mg, 15 in portions). After stirring at RT for 90 mins, the mixture was concentrated to 2 ml and Et2O (20 niL) added. The mixture was then extracted with NaOll (1 N, aq., 20 niL) and the aqueous portions acidified with 6N HCI to pH 2. A: the aqueous portion was then extracted with EtOAc, dried over MgSO4 and evaporated to yield the title compound as a white solid (119 mg). lH 20 NMR (400 MHZ, CDCl) 15 ppm 2.11 (t, J = 18.8 Ilz, 3 H), 9.01 (d, J = 1.3 HL1Hj), 9.47 (d, j = 1.3Hz, 1Hj) . Preparation example 5: Synthesis of 5- (fluoromethyl) pyrazine-2-carboxylic acid (5- (3)) ,,,, jj) j ,,,, jí "° - jO:> c HO tj ^ 'o o o o 5- (1) 5- (2) 5- (3) Sodium 5- (hydroxyl) pyrazine-2-carboxylate 5- (1) 25 In a solution of methyl 5-formylpyrazine-2-carboxylate (2.47 g) in THF (20 ml) was added sodium borohydride (170 mg) in portions for 10 mins. After stirring for 1 h, methanol (10 ml) was added. The reaction mixture was added for an additional 20 minutes and then HCl (1 N, aq., 20 niL) and brine (20 mL) were added. The "" mixture was extracted with EtOAc (3 x 40 mL) and the organic portions "5 combined dried over MgSO4 and evaporated to yield the 'title compound (1.31 g).' H NMR (400 MHz, CDCl) 15 lpm ppm 4.07 (s, 3 H), 4.98 (br, s ,, 2 H), 8.80 (s, 1 H), 9.27 (s, 1 H) 5- (fluoromethyl) pyrazine-2-carboxylate 5- (2) In a solution of 5- (hydroxyl) pyrazine- 10-methyl 2-carboxylate (0.64 g) in THF (20 ml), triethylamine (2.30 g) was added and the solution was poured at 0 ° C. Triethylamine trihydrofluoride (1.22 g) was then added followed by nonafluorobutanesulfonyl fluoride (2.28 g) to the drops for 5 mins. After heating at RT and stirring for 2 h, NaHCO3 (sat., Aq., 100 niL) was added and the mixture was extracted with EtOAc (2 x 50 15 ml). The combined organic portions were dried over MgSO4, evaporated and purified by chromatography on silica gel (5 ° /) to 50 ° /) EtOAc in hexane) to produce the title compound (94 mg). 1H NMR (400 MHz, CDCl) 15 ppm 4.07 (s, 3 H), 5.67 (d, J = 46.5 HZ, 2 H), 8.89 (s, 1 H), 9.28 ( s, 1 H) 20 Methyl 5- (Fluoromethyl) pyrazine-2-carboxylic acid 5- (3) 5- (fluoromethyl) pyrazine-2-carboxylate (94 mg) was dissolved in dioxane (1 nil) and water (1 niL) was added, followed by lithium hydroxide monohydrate (60 mg). After stirring at RT for 18 h, Et2O (20 ml) was added and the mixture was then extracted with NaOH (I N, 25 aq., 2 x 20 ml). The aqueous portions were acidified with 6N HCl to pH 1, extracted with EtOAc (2 x 40 ml), the combined organic portions dried over MgSO4 and evaporated to produce the title compound as a white solid (71 mg). 'H NMR (400 MHz, CDCl) δ ppm 5.70 (d, J = 46.2 Hz, 2 H), 8.85 (s, 1H), 9.40 (s, 1H) Preparation example 6: Synthesis of 5-difjuoromethylpyrazine-2-carboxylic acid (6- (5) H °)) y "yj; j" "y Jn "6- (1) 6- (2) FF ho ~, j :: jL - y jr "- y Ij" 'O 643) 6- (4) 6- (5) Synthesis of t-butyl 5-methylpyrazine-2-carboxylate 6- (1) A complex boron trifluoride diethyl ether (91.7 µL) was added to the drops to a suspension of tert-butyl 2-methylpyrazine-5-carboxylic acid (I g) and 2,2,2-trichloroacetimidate (4.75 g ) in THF (20 mL) under drying on ice. The reaction solution was heated at RT, followed by stirring for 2 h. The saturated NaCl solution and EtOAc were added to a reaction solution and the organic layer was separated. The organic layer was dried over anhydrous MgSO4 and the insoluble substance was separated by filtration. The filtrate was concentrated and purified by silica gel column chromatography to obtain the title compound (1.4 g). 'H-NMR (CDCj3) δ (ppm): 1.65 (s, 9 H), 2.65 (s, 3 H), 8.57 (d, J = 1.2 Hz, 1 H), 9 , 10 (d, J = 1.6 FIZ, IH). Synthesis of t-butyl 5 {(E) -2-dimethylamino vinyl) -pyrazine-2-carboxylate 6- (2) A mixture of t-butyl 5-methylpyrazine-2-carboxylate (1.35 g), DMF (25 niL) and N, N-dimethylformaniide dimethylacetal (25 mL) was stirred at 130 ° C for 5 h. "The reaction solution was streaked in RT and diluted with EtOAc. The mixture was washed with saturated NaCl solution The organic layer was dried over anhydrous MgSO4 and the insoluble substance was filtered off.The filtrate was concentrated and the residue was purified by column chromatography on silica gel to obtain the title compound (648 mg). -RMN (CDCl,) Ô (jppm): 1.63 (s, 9 H), 3.00 (s, 6 H), 5.16 (d, J = 12.8 HznH), 7.72 (d, j = 12Mlz, 1H), 8.16 (d, j = 1.2Hz, 1H), 8.8l (d, j = 1.6 IJz, IH). 5 Synthesis of t-butyl 5-formylpyrazine-2-carboxylate 6- (3) Sodium periodate (1.67 g) was added in a solution of 5 - ((E) -2-dimethylamino-vinyl) -pyrazine -2-t-butyl carboxylate (645 mg) in 50 ° / TW of water (26 ml) and the mixture was stirred at RT for 4 h. The saturated NaHCO3 solution and EtOAc were added to a reaction solution and the organic layer was separated. The organic layer was washed with the saturated NaCl solution and dried over anhydrous MgSO4. The insoluble substance was separated by filtration and the filtrate was concentrated. The residue was purified by silica gel column chromatography to obtain the title compound (249 mg). 'H-NMR (CDCl,) δ (jppm): 1.68 (s, 9 H), 9.25 (d, J = 1.2 Hz, 1 H), 15 9.36 (d, j = 1 , 6HZ, IH), 10.2 (S, IH). Synthesis of t-butyl 5-difluoromethylpyrazine-2-carboxylate 6- (4) [Bis (2-methoxyethyl) amino] sulfur & e (662 µL) trifluoride was added to the drops in a solution of 5-formylpyrazine-2-carboxylate t-20 butyl (249 mg) in CH2C! 2 (12 ml) under an N atmosphere, under cooling on ice. The reaction solution was added over 2 h while gradually taken up in RT. The saturated NaHCO3 solution and EtOAc were added to a reaction solution and the organic layer was separated. The organic layer was washed with saturated NaCl solution and dried over anhydrous MgSO4. The insoluble substance was separated by filtration and the filtrate was concentrated. The residue was purified by column chromatography on silica gel to obtain the title compound (175 mg). 1 H-NMR (CDCl 6 '(ppm): 1.67 (s, 9 H), 6.75 (t, J = 54.4 Hz, 1 H), 9.02 (d, J = 0.8 Hz , 1 H), 9.25 (d, j = 0.8 HZ, IH). Synthesis of 5-difluoromethylpyrazine-2-carboxylic acid 6- (5) tMuoroac-ethical acid (1 ml-) was added in a solution of t-butyl 5-difluoromethylpyrazine-2-carboxylate (175 mg) in dichloromethane (1 ml-) and the mixture was stirred at RT for 5 h. Ether and 5 N of '5 NaOlil were added to a reaction solution. The aqueous layer was separated and made acidic with 5 N hydrochloric acid. EtOAc was added in an aqueous layer and the organic layer was separated. The organic layer was dried over anhydrous MgSO4 and the insoluble substance was filtered off. The filtrate was concentrated to obtain the title compound (100 mg). 1 H-1 H NMR (CDCl 1) δ (ppm): 6.80 (t, J = 54.4 Hz, I H), 9.02 (s, 1 H), 9.47 (s, IH). Preparation example 7: Synthesis of 5-cyanopyridine-2-carboxylic acid (7- (2)) "cn jCN io] 1A, N-1]" jOyjj "'°" f "" N " o o 7- (1) 7- (2) Synthesis of methyl 5-cyanopyridine-2-carboxylate 7 (1) A mixture of methyl 5-bromopyridine-2-carboxylate (2,8 15 g) and copper cyanide (3.6 g) in NMP (30 ml) was heated with stirring at 170 ° C for 1.5 h. Water was added to a RT reaction solution and the substance was insoluble! has been removed by filtration. The filtrate was extracted with EtOAC. The extract was washed with the saturated NaCl solution and then dried over anhydrous MgSO4. The drying agent was removed by filtration and the Mrado was concentrated under reduced pressure. The resulting crude product was purified by column chromatography on silica gel (EtOAc-heptane system) to obtain the title compound (920 mg). 1H-NMR (400 MHz, CDCl,) iij (ppm): 4.06 (s, 3 H), 8.16 (dd, J = 2.0, 8.0 Hz, 1 H), 8.27 (d, J = 8.0 Hz, 1H), 9.0l (d, j = 2, OHz, IH). 25 Synthesis of 5-cyanopyridine-2-carboxylic acid 7- (2) A solution of the compound of Preparation Example 13- (1) (920 mg) and a 5 N solution of NaOH (2.26 ml) in ethanol (30 ml) was stirred at RT for 10 min. 5 N of hydrochloric acid (5.2 mL) was added to a RT reaction solution, followed by extraction with EtOAc. The extract was dried over anhydrous MgSO4. The drying agent was removed by filtration and the filtrate was concentrated under reduced pressure to obtain the title compound (800 mg). 'H-NMR (400 MHz, DMSOd ,;) Õ (ppm): 8.18 (d, j = 8, OHz, IH), 8.5l (dd, j = 2.0, 8, OHz, 1H) , 9, J2-9,18 (m, IH). Preparation example 8: Synthesis of 5- (methoxyl) pyrazine-2-carboxylic acid (8- (2)) is') "° · 10 ,,:> °" '· ho ,,:> ° "' 10- KÁ "NA ooo 5- (1) 8- (1) 8- (2) Synthesis of methyl 5- (methoxyl) pyrazine-2-carboxylate 8-10 (1) The compound obtained in preparation example 5- (1 ) (279 mg) was dissolved in DMF and the solution was dissolved at O ° C. Sodium hydride (60% in Mineral oil, 70 mg) was added, followed by iodomethane (250 15 mg). After 2 days, water (25 ml) was added and the solution extracted with EtOAc (100 ml). The aqueous layer was saturated with NaCl and further extracted with EtOAc (2 x 50 ml). The combined organic portions were dried over MgSO4, evaporated and purified by chromatography on silica gel (30 ° /) at 50 ° /, EtOAc in hexanes) to produce the title compound (55 mg, approximately 65 ° /) ). 1 H NMR (400 MHz, CDCl 3) B ppm 3.55 20 (s, 3 H), 4.05 (s, 3 H), 4.72 (s, 2 H), 8.84 (d, J = 1.0 Hz, 1 H), 9.25 (d, J = 1.0 HZ, 1 H) Synthesis of 5- (methoxyl) pyrazine-2-carboxylic acid 8- (2) '5- (methoxyl) pyrazine Methyl -2-carboxylate, 8- (1), (55 mg, 25 crude) was dissolved in], 4-dioxane (1 niL) and water (1 niL) was added followed by lithium hydroxide monohydrate (50 mg ). After stirring at RT for 1 h, water (20 ml) was added and the mixture was extracted with ether (20 ml). The aqueous portion was acidified to pH 2 and extracted with EtOAc (2 x 25 ml). The combined EtOAc layers were dried over MgSO4 and evaporated to yield the title compound (19 mg). 'H NMR (400 MHz, CDCl3) 6' 5 ppm 3.58 (s, 3 H), 4.77 (s, 2 H), 8.80 (br, s ,,] H), 9.38 ( br, s ,, 1 H) Preparation example 9: 5-Methoxypyrazine-2-carboxylic acid 1. mcoh, h, so, rNmc | 2. NaOH 2. NaOMe NUOMe ho}, j 'hoj, joo 5-Chloropyrazine-2-carboxylic acid (5.0 g, 0.032 mol) was loaded in a round flute equipped with a thermo-connection, 10 suspended stirrers and condenser in reflux. Methanol (37.5 ml, 0.926 mol) was loaded followed by concentrated sulfhyllic acid (0.2 ml, 0.004 mol). The 3-mouthed dung was equipped with a heating mantle and then the reaction mixture was heated to ca. 65.0 ° C (internal T). The continuous reaction mixture to stir at ca. 65.0 ° C (internal T) per ca. 4 h. The reaction mixture 15 cooled to ca. 25.8 ° C (internal T). Methanol (12 niL, 0.31 mol) was loaded and the slurry continued to stir at ca. 22.3 ° C (internal temperature) per ca. Then 15 min at ca. 1.0 ° C (internal temperature) under an atmosphere of nitrogen 25 ° /) of sodium methoxide in methanol (1: 3, sodium methoxide: methanol, 7.7 mL) was charged to the jar while the temperature remains below 30 ° C (inner T 20). The reaction mixture was adjusted to 20.4 "C (internal T). After 30 min., Sodium hydroxide (2.0 g, 0.04 mol) and water (37.5 ml, 2.08 mol) were combined to form a solution and then the solution was loaded into a reaction mixture Water (50.0 mL, 2.78 mo!) was loaded and then the reaction mixture was heated to 40.0 ° C (internal temperature) for about 60 mins The heating mantle was removed and then the reaction mixture was heated to about 25.4 ° C (internal temperature) 38 ° /) aqueous HCl solution (38:62, hydrogen chloride : water, 4.0 niL) was added at a rate (ca. 5 min.) such that the temperature remained below 30.0 ° C (internal temperature). The thick paste was stirred for 1 h at ca 2l, 4 ° C (internal T) and then filtered through a sintered fin. The solids were rinsed with water (10.0 mL, 0.555 mol) and 5 dried under vacuum overnight to produce 5-methoxypyrazine-2-carboxylic acid (3.59 g). 'H NMR (500 MHz, DMSO) 15 13.24 (IH, br S), 8.79 (IH, d, J' - 1.2 HZ), 8.37 (IH, d, J - 1.2 Hz), 3.98 (s, 3H); "C NMR (125 MHz, DMSO) 6 165.36, 161.88, 143.88, 136.82, 135.55, 54.69. General procedure for the binding of anilines prepared in the 10 preparation examples 1- (13) and 2- (10) with aryl carboxylic acids: Preparation of N- (3 - ((4aS, 5S, 7aS) -2-mino- 5- (tnnuoromethi |) -4a, 5,7,7a- tetrahydro-4HAlro [3,4-d] [l3] thiazin-7a-yl) -4-nuorophenyl) -5-methoxypyrazine-2-carboxamide (Example 1 ) ° YiC "Ç'NH, FÂ- 'Èf ((4aS, 5S, 7aS) -7a- (5-amino-2-nuorophenyl) -5- (trinuoromethyl) - 15 4a, 5,7,7a-tetrahydro-4H @ ro [3,4-d] [i , 3] thiazin-2-yl) tert-butyl carbamate (Preparation example 1, 56 mg) was dissolved in DCM (2 ml) and 5-methoxypyrazine-2-carboxylic acid (40 mg), N, N-di -isopropylethylamine (80 mg) and hexafluorophosphate (1FPbenzotriazol-yloxy) tripinolidinj-yl) phosphonium (135 mg) were added. The reaction mixture was stirred at RT for 18 h and 20 sodium bicarbonate (sat., Aq., 25 niL) was added. The mixture was extracted with EtOAc (2 x 40 niL), the combined organic portions were dried over MgSO4, evaporated and purified by silica gel chromatography (EtOAc gradient / hexanes) to produce the amide (40 mg) as a solid White. The amide was dissolved in DCM (2 niL) and TFA (1 ml) was added. After stirring at RT for 2 h, the reaction mixture was evaporated and sodium bicarbonate (sat., Aq., 25 ml) was added. The mixture was extracted with EtOAc (2 x 25 ml) and the combined organic portions dried over MgSO4 and evaporated to produce the title compound as a white solid (34 mg). 1 H NMR (400 MHz, CDCl) 6 ppm 2.78 - 2.87 (m, 1 H), 3.11 - 3.21 (m, 1 H), 3.38 - 3.48 (m, 1 H), 3.82 - 4.06 (m, 4 H), 4.48 - 4.72 (m, 2 H), 6.96 - 7.10 (m, 1 H), 7.52 ( d, j = 4.8 Hz, IH), 7.87 (d, J = 8.8 HZ, 1 H), 8.09 (s, 1 H), 8.95 (s, 1 H), 9 , 46 (br, s ,,! H) Example 2: N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (üif1uoromethyl) - 10 4a, 5,7,7a-tetrahydro- 4H-Wo [3,4-d] li, 3] tiwin-7a-i |) -4-nuorophenyl) -5- cyanopicolinamide JLJ j) N) 'NH,' (F 'Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2-nuorophenyl) -5-trinuoromethyl-4aS, 7,7a-tetrahydro-4HAiIro [3 , 4-d] tert-butyl [1,3] thiazin-2-yl] carbamate and 5-cyanopyrazine-2-carboxylic acid according to a general procedure.1 H NMR (400 MHZ, CDCl) jj; ppm 2.82 (dd, J = 13.6, 3.5 Hz, 1H), 3.14-3.21 (m, 1 H), 3.37-3.45 (m, 1 H), 3 , 91 (d, j = 9.1 Hz, 1 H), 4.50 - 4.71 (m, 2 H), 7.08 (dd, J = 11.9, 8.8 Hz, 1 H) , 7.46 - 7.57 (m,! H), 7.92 (dt, J = 8.8, 3.4 Hz,] H), 8.17 (dd, J = 8.3, 2, 0 Hz, 1 H), 8.34 (d, J = 8.1 Hz, IH), 8.81-8.92 (m, IH) 20 Example 3: N- (3 - ((4aS, 5 S , 7aS) -2-amino-5- (trifluoromethyl) - 4a, 5,7,7a-tetrahydro-4H-hro [3,4-d] [1,3] tiwin-7a-yl) -4-Auorofeml) -5- (difluoromethyl) pyrazine-2-carboxamide F ojj ') N)' NH, 'ÇF' Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2-fluorophenyl) -5- Mnuoromethyl-4a, 5,7,7a-tetrahydro-4H -hro [3,4-d] [1,3] tiwin-2-yl] tert-butyl carbamate and 5-difluoromethyl-piraúna-2-carboxylic acid according to a general procedure. 'H NMR (400 MHZ, CDCl) δ ppm 2.80 (dd, J = 13.8, 3.7W, 1H), 3.1l (dd, j = l3.6,2.8W, 1H), 3.31 - 3.41 (m, lH), 3.86 (d, j = 8.3W, 1H), 4.57 (d, j = 8.3k, lH), 4.64 (dt, j = l4.7.7, lW, 1H), 4.75 (brs, 2H), 6.69 (t, j = 56.3m, 1W, 7.07 (dd, j = 11.6.8.8w , 1W, 7.57 (dd, j = 7.1, 2.8Hz, lH), 7.87 (dt, j = 8.5.3.6m, 1H), 8.85 (s, lH), 9.45 (s, 1 H), 9.58 (br, s ,, 1 H) Example 4: N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) - 4a, 5j, 7a-tetrahydro-4HAilro [3,4-d] [1,3] thiazin-7a-i]) - 4-nuorophenií) -5- (trifluoromethi1) picolinamide F,, jO tj 'ÇF' Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2Aorophenyl) -5-trif1uoromethi] Aa, 5,7,7a-tetrahydro-4H-nlro [3,4 -dj [1,3] thiazin-2-yl] tert-butyl carbamate and 5-trifluoromethyl-pyridine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHz, CDCl) δ ppm 2.88 (dd, J = 13.6, 3.8 Hz, IH), 3.20 (dd, j = 13.6, 3.0 Hz, IH ), 3.37 -3.53 (m, 1 H), 3.94 (dd, j = 8.3.2.3Hzmg, 4.40-4.89 (m, 4H), 7j4 (dd, j = ll, 9.8.8Hz, 1H), 7.66 (dd, J = 6.8, 2.8 Hz, 1 H), 7.98 (ddd, J = 8.8, 4.0, 3.0 Hz, 1 H), 8.20 (dd, J = 8.2, 1.6 Hz, 1H), 8.45 (d, j = 83HLnlj), 8.90 (s,! H), 9.95 (s, lH) Example 5: N- (3- ( (4aS, 5 S, 7aS) -2-amino-5- (trinuorometij) - 4a, 5,7,7a-tetrahydro-4H-hro [3,4-d1 [1,3] tiwin-7a-yl) - 4-nuorofeml) -5-methylpyrazine-2-carboxamide om: 3 ": Í))" "FÁ 'Èf Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2-fluorophenyl) -5- tnRuoromethyl-4a, 5,7,7a-teWahydro-4H-hro [3,4-d] [1,3] ti = in-2-yl] tert-butyl cbamate and 5-methyl-pyrazine-2- acid carboxylic acid according to a general procedure. 'H NMR (400 MHZ, CDCl) δ ppm 2.72 (s, 3 H), 2.88 (dd, J = 13.6, 3.8 Hz, 1 H) , 3.21 (dd, J = 13.6, 3.0 Hz, 1 H), 3.42 -3.49 (m, 1 H), 3.94 (d, J = 8.3 Hz, 1 H), 4.39 - 4.80 (m, 4 H), 7.14 (dd, J = 11.9, 8.8 Hz, 1 H), 7.62 (dd, J = 7.1, 2.8 Hz, l H), 7.93 - 7.99 (m, l H), 8.46 (d, J = 1.0 Hz, 1H), 9.39 (d, j = 13 Hz, lH ), 9.65 (s, 1H) Example 6: N- (3 - ((4aS, 5S, 7aS) -2-mino-5- (trinuoromethyl) - 4aS, 7,7a-tetrahydro-4HAi1ro [3,4 -d] [1,3] thiazin-7a-yl) -4-f1orophenyl) -5- methylpicolinamide oj -Ç tj FÂ ' Synthesized EF of [(4aS, 5 S, 7aS) -7a- (5-amino-2-fluorophenyl) -5-trifluoromethyl-4a, 5,7,7a-tetrahydro-4H-nlro [3,4-d] [ 1.3] tert-butyl thiazin-2-yl] carbamate and 5-methyl-pyridine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHz, CDCl3) δ ppm 2.37 (s, 3 H), 2.78 (dd, J = 13.6.3.8 Hz, 1 H), 3.12 (dd, J = 13.6, 2.8 FIz, 1 H), 3.28 - 3.40 (m, 1 H), 3.87 (d, J = 8.1 Hz, 1 H), 4.28 - 5, 02 (m, 4 H), 7.02 (dd, J = 11.9, 8.8 Hz, 1m, 7.55 (dd, j = 7.1.2.8w, 1m, 7.63 (dd , j = 8,0,1,4w, lH), 7,88 "5 (ddd, j = 8,8,4,0,2,9W, lW, 8,10 (d, j = 8,1Hz, 1H), 8,31-8,40 (m, 1W, 9, 90 (s, IH) Example 7: N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (trifjuoromethyl) - 4a, 5,7,7a-tetrahydro-4H-Wof3,4-d ] [1,3] ti & .in-7a-yl) -4-Ruorophenyl) -5-ethylpicolinamide %THERE 10 Synthesized from,)) J "'[(4aS, 5S, 7aS) -7a- (5-amino-24uorophenyl) -5-trinuoromethyl-4aS, 7,7a-tetrahydro-4H-nlro [3,4-d] [1,3] thiazin-2-yl] tert-butyl carbamate and 5-ethyl-pyridine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHZ, CDCl3 ppm 1.33 (t, J = 7.6 Hz, 3 H), 2.78 (q, J = 7.6 Hz, 2 H), 2.88 (dd, J = 13.5, 3.7 Hz, 1 H), 3 , 22 (dd, J = 15 13,6,2,8 Hz, 1H), 3,42-3,48 (m, 1H), 3,96 (d, j = 7,3 Hz, IH), 4 , 44-4.95 (m, 4H), 7j2 (dd, j =] 1,6,8,8HZ, lH), 7: 62 (dd, j = 6,9,2,7Hz, lH), 7 , 74 (dd, j = 8.1, 1.8 JJz, IH), 7.98 (dt, j = 8.7, 3.5 Hz, 1 H), 8.21 (d, j = 8, 1 Hz, 1 H), 8.44 - 8.48 (m, 1 H), 10.00 (s, 1 H) Example 8: N- (3 - ((4aS, 5S, 7aS) -2-amino -5- (trifluoromethyl) - 20 4a, 5,7,7a-tetrahydro-4H-hro [3,4-dj [1,3] thiam-7a-i |) -4-nuorophenyl) -5- (Ruoromethyl) pyrazine-2-carboxamide I ' oj3 ^ ' j)) 2 'if Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2-fluorophenyl) -5-trifluoromethyl-4a, 5,7,7a-tetrahydro-4H-n] ro [3 , 4-d] [], 3] thiazin-2-yl] tert-butyl carbamate and 5Aororomethyl-pyrazine-2-carboxylic acid according to a general procedure. Details of a current preparation are as follows: ((4aS, 5 S, 7aS) -7a- (5-amino-2-nuorophenyl) -5- (trinuoromethyl) -4a, 5,7,7a- tetrai & o-4H-hro [3,4-d] [1,3] úwin-2-yl) tert-Butyl carbate (500 mg) was dissolved in DCM (10 niL) and 5-fluoromethyl-pyrazine-2-carboxylic acid (223 mg), N, N-diisopropylethylamine (521 mg) and hexafluorophosphate (1H-benzotriazol-1-yloxy) tripirrolidin-1-yl) phosphonium (750 mg) were added. The reaction mixture was stirred at RT for 1 h and sodium bicarbonate (sat., Aq., 50 niL) was added. The mixture was extracted with EtOAc (2 x 75 inl), the combined organic portions were dried over MgSO4, evaporated and purified by gel chromatography. silica (0 ° 4 to 30 ° /) EtOAc gradient / hexanes) to produce the amide (613 mg) as a white solid. The amide was dissolved in DCM (2 ml) and TFA (1 ml) was added. After stirring at RT for 2 h, the reaction mixture was evaporated and sodium bicarbonate (sat., Aq., 25 niL) was added. The mixture was extracted with EtOAc (3 x 25 ml) and the combined organic portions dried over MgSO4 and evaporated to yield the title compound as a white solid 'H NMR (400 MHz, CDCl) 6 ppm 2.89 (dd, J = 13.8, 3.7 Hz,] H), 3.21 (dd, j = 13.9.2.5 Hz, IH), 3.40 -3.51 (m, 1 H), 3 , 96 (d, j = 7.3 Hz, 1H), 4.42-4.85 (m, 4H), 5.69 (d, j = 46.5W, 2H), 7.15 (dd, j = ll, 9, 8.8Hz, 1W, 7.64 (dd, j = 7.1.2.8W, 1H), 7.94-8.00 (m, lW, 8.77 (s, l H ), 9.47 (s, 1 H), 9.68 (s, 1 H) , Example 9: N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) - 4aS, 7,7a-tetrahydro-4HAilro [3,4-d] [1,3] thiazine -7a-yl) -4-nuorophenyl) -5- methoxypicolinamide OJ: I l O " F ij; NH c1: "'' jF 'Synthesized from [(4aS, 5S, 7aS) -7a {5-amino-2-fluorophenyl) -5- 5 trifluoromethyl-4a, 5,7,7a-tetrahydro-4Hjilro [3,4 -d] [1,3] thiujn-2-yl] tert-butyl carbamate and 5-methoxypyridine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHz, CDC] 3) 6 ppm 2, 88 (dd, J = 13.8, 3.7HzmgÁ, 23 (dd, j = 13À2.4HL1H), 3.46 (dj = 7.3Hz, 1H), 3.89- 4.02 (m, 4 H ), 4.54 - 5.00 (m, 4 H), 7.1 l (dd, j = 11.9, 8.8 Hz, 1 H), 7.36 (dd, 10 j = 8.8, 2.8W, 1H), 7.6l (dd, j = 7, l, 2.8W, lW, 7.97 (dt, j = 8.5.3.6W, IH), 8.23 - 8, 30 (m, 2 H), 9.86 (s, 1 H) Example 10: N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) - 4a, 5,7,7a -tetrahydro-4H-füro [3,4-d] [1,3] thiazin-7a-yl) -4-fluorophenyl) -5-ethoxypyrazine-2-carboxamide ojj; oj ,,,),) ',,, FÂ' Èf '15 Synthesized from [(4aS, 5 S, 7aS) -7a- (5-amino-2-fluorophenyl) -5- trinuoromethyl-4a, 5,7,7a-tetrahydro-4HAlro [3,4-d] [1,3] thiazin-2-yl] tert-butyl carbamate and 5-ethoxypyrimidine-2-carboxylic acid according to a general procedure. 'H RNIN (400 MHz, CDCl3) Ô ppm 1.47 (t, J = 7.1 Hz, 3 H), 2.87 (dd, J = 13.6, 3.5 FIz,] H), 3.20 (d, J = 13.6 Hz, 1 H), 3.40 - 3.50 (m , 1H), 3.94 (d, j = 7.8w, 1H), 4.46-4.95 (m, 6m, 7.1] (dd, j = l1.5, 9, OHz, 1H) , 7.54-7.63 (m, 1H), 7.90-8.0l (m, IH), 8.13 (S, 1H), 9.0O (S, 1H), 9.52 (br , s ,, lH) Example 11: N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (tnnuoromethyl) - 4a, 5,7,7a-tetrahydro-4HAijro [3,4-d ] [1,3] thiazin-7a-yl) -4-fluorophenyl) -5- (1,1-difluoroethyl) pyrazine-2-carboxamide f , fo) K "f |) N) HN h, the 's 'Qf' Synthesized from [(4aS, 5S, 7aS) -7a {5-amino-2-fluorophenyl) -5-trinuoromethyl-4a, 5,7,7a-teWai & o-4H-hro [3,4-d] [1,3 ] tert-butyl thiazin-2-ylcarbamate and 5- (1,1-Difluoroethyl) pyrazine-2-carboxylic acid according to a general procedure. 'H NMR (600 MHZ, CDCl3) Õ ppm 2.03 (t, J = 18.8 Hz, 3 H), 2.82 (d, J = 12.0 Hz, 1 H), 3.14 (d, J = 12.4 Hz, 1 H), 3.35 - 3.46 (m, 1 H), 3.77 - 4.07 (m, 1 H), 4.20 - 4.93 (m, 4 H), 7.08 (dd, J = 11.7, 9.0 Hz, 1H ), 7.56 (d, j = 4.5W, lH), 7.80-7.93 (m, lH), 8.87 (s, 1H), 9.43 (s, j H), 9.59 (br, s ,, 1 H) Example 12: N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (tMuoromethyl) - 4aÁ7,7a-tetrahydro-4HmIro [3,4-d1 [1,3] thiazin-7a-yl) -4-fluorophenyl) -5- (trifluoromethyl) pyrazine-2-carboxamide F , F oM7m U Ç'NHZ · ÇF 'Synthesized from [(4aS, 5 S, 7aS) -7a- (5-anIino-2-nuoropheni!) - 5-trinuoromethyl-4a, 5,7,7a-tetrahydro-4H-bore [3,4- dj [1,3] ti & in-2-yl] tert -butyl and 5-trinuoromethylpyrazine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHz, CDClj Ô ppm 2.80 (dd, J = 13.6, 3.8w, 1H), 3, ll (dd, j =] 3,8,2,9w, 1n), 3 , 30-3.44 (m, lW, 3.87 (d, j = 8.3 Hz, IH), 4.25 -5.14 (m, 4 H), 7.07 (dd, j = 1 !, 9, 8.8 Hz, IH), 7.57 (dd, J = 6.8,2.8W, 1W, 7.86 (dt, j = 8,4,3,6R, lH), 8 , 89 (s, 1W, 9.53 (s, 2H) Example 13: N- (3 - ((4aS, 5S, 7aS) -2-mino-5- (tri8uoromethyl) - 10 4aS, 7,7a-tetrai -4HAlro [3,4-d] [1,3] thiazin-7a-yl) -4-fluorophenyl) -5- (methoxyl) pyrazine-2-carboxamide Ç3 ^ ° "Ü)" "'(F' Synthesized from [(4aS, 5 S, 7aS) -7a- (5-amino-2Aorophenyl) -5-trinuoromethyl-4a, 5,7,7a-tetrai & o-4H- hro [3,4-d] [1,3] tiwin-2-yl] tert -butyl and 5- (methoxyl) pinmidMa-2-carbonyl acid chromatography according to a general procedure. 'H NMR (400 MHz, CDCl,) Ô ppm 2.79 (dd, J = 13.6, 3.8Hz, 1H), 3.11 (dd, j = 13.6.2.8N, 1H), 3.32-3.39 (m, lW, 3.49 (s, 3 m, 3.84 (d, j = 8.3w, 1m, 4.34-4.73 (m, 6w, 7.05 (dd, j = ll, 6.8.8w, 1W, 7.55 (dd, j = 6.8.2.8W, lH), 7.88 (dt, j = 8.4.3.6W, lH), 8.63 ( s, 1 H), 934 (d, j = 1.0 Hz, 1H), 9.60 (s, 1H) Example 14: N- {3 - [(4aS, 5S, 7aS) -2-amino-5- (tnnuoromethyl) - 4a, 5-dihydro-4HAIro [3,4d] [1,3] thiazin-7a ( 7H) -yl] -4-fluorophenyl} -5 - [('H,) methylOxy] pyrazine-2-carboxamide 14- (2) 5 - [(2H3) methyloxy] pyrazine-2-carboxylic acid synthesis MeO, C Á'1 ^ ' N "D, CO, C jíi'j" ° "' N 'HO, C jíi'j "°"' N Part (I): 5 - [(2H3) methyloxy] (2H3) methyl pyrazine-2-carboxylate Recently cut sodium metal (160 mg) was added in portions over 10 mins to ('H3) methan (2Hj) ol ( 5 ml) and the solution was added until the sodium was dissolved. This solution was then added to the methyl 5-chloropyrazine 2 carboxylate (1.02 g) in (2H3) methan (2H) ol (5 naL) and the solution was allowed to stir at RT for 1 hr. The solution was then concentrated under reduced pressure to a volume of about 2 ml and water (50 ml) was added. The mixture was extracted with EtOAc (2 x 50 niL), the combined organic portions were dried over MgSO4 and evaporated to produce the title compound (745 mg). 'H NMR (400 MHz, CDCl) at ppm 8.30 (d, J = 1.3Hz, 1H), 8.9l (d, j = 1.3Hz, 1H) Part (II): acid 5- [ (2H3) methyloxy] pyrazine-2-carboxylic A 'A stirred solution of 5 [(2H3) methyl] Oxy] pyrazine 2 (2H3) methyl carboxylate in 1,4-dioxane (5 mL) was added water (5 mL) followed by lithium hydroxide monohydrate (300 mg). After stirring for 1 hr, the reaction mixture was concentrated under reduced pressure to about 5 ml and extracted with diethyl ether (25 niL). The organic layer was extracted with 1N NaOlil (aq., 10 niL) and the combined aqueous portions were acidified to pH 2 with 6N hydrochloric acid. After stripping in a refrigerator, the mixture was filtered to yield the title compound as a light brown powder (660 mg). 'H NMR (400 MHz, CDCl) 6 ppm 8,21 (d, J = 1,3 Hz, 1 H), 9,0 l (d, j = 1, 3 Hz, 1W, l0, l2 © rs ,, lH) Part (III): N- {3 - [(4aS, 5S, 7aS) -2-amino-5- (tnnuoromethi!) - 4a, 5-dihydro-4HAúro [3.4 d] [13] tiazin- 7a (7Hj) -yl] -4-nuorophenyl} -5- [('H,) methyloxy] pyrazine 2 carboxamide o, ug:) N) NH, '' ÇF '((4aS, 5S, 7aS) -7a- (5-amino-2-nuorophenyl) -5- (Onuoromethyl) - 5 4a, 5,7,7a-tetrahydro-4H-hro [3,4- dj [1,3] tiwin-2-yl) tert-Butyl carbamate (100 mg) was dissolved in DCM (2 ml) and 5 - [('H,) methyloxy] pyrazine-2-carboxylic acid (55 mg) , N, N-diisopropylethylamine (112 mg) and (1H-benzotriazol-yloxy) tripirrolidinj-yl) phosphonium hexafluorophosphate (180 mg) were added. The reaction mixture was stirred at RT for 18 h and 10 sodium bicarbonate (sat., Aq., 25 ml) was added. The mixture was extracted with EtOAC (2 x 40 mL), the combined organic portions were dried over MgSO4, evaporated and purified by chromatography on silica gel (2 ° /) at 25 ° /) of EtOAc in hexanes) to produce the amide (127 mg) as a white solid. The amide was dissolved in DCM (2 ml) and TFA (1 ml) was added. After 15 RT stirring for 2 h, the reaction mixture was evaporated and sodium bicarbonate (sat., Aq., 25 niL) was added. The mixture was extracted with EtOAc (2 x 40 ml) and the combined organic portions dried over MgSO4 and evaporated to produce the title compound as a white solid (104 mg). jÍJ NMR (400 MHZ, CDCl,) Õ ppm 2.87 (dd, J = 13.6, 3.8 Hz, IH), 3.21 (dd, J = 20] 3,6,2,8w, 1H , 3.39-3.53 (m, lm, 3.95 (d, j = 8.3w, 1H), 4.65 (d, j = 8.3W, 1W, 4.72 (quin, j = 7.2W, 1W, 4.87 (brs, 2W, 7, l2 (dd, j = 1l, 9, 8.8W, 1W, 7.60 (dd, j = 6,9,2,7W, lH) , 7,95 (dt, j = 8,5,3,6W, lH), 8J6 (d, j = 1,0HzjH), 9,02 (d, j = 1,0Hz, 1H), 9,52 ( s, 1H) Example 15: N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (tMuoromethyl) - L}. "T 4a, 5,7,7a-tetrahydro-4H-Nro [3,4-d] [1,3] thiazin-7a-yl) -4,5-dinuorophenyl) -5- (diMorometii) pyrazine-2- carboxamide F oÇj '')) ',,, FÂ' Èf Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2,3-diylorophenyl) -5- trifluoromethi] -4a, 5,7,7a -tetrahydroH-n] ro [3A-d] [1,3] thiazin-2-yl-1-tert-butylcarbamate and 5-dinuoromethyl-pyrazine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHz, CDCI) Õ ppm 2.90 (dd, J = 13.8, 3.7 Hz, IH), 3.19 (dd, J = 13.8.2.7 Hz,! H), 3.31 - 3.49 (m, 1 H), 3.95 (d, j = 7.6 Hz, IH), 4.44-5.15 (m, 4H), 6.81 (t , j = 55.8 Hz, 4H), 7.22 -7.35 (m,] H), 8.08 (ddd, j = l1,2,6,8,2,7R, 1H), 8, 94 (s, lm, 9.53 (s, 1H), 9.67 10 (s, 1 H) Example 16: N- (3 - ((4aS, 5S, 7aS) -2-steel-5- (trinuoromethyl) - 4a, 5,7,7a-teKaidro-4H-hro [3,4-d] [1,3, thiazin-7a-yl) -4,5-dinuorophenyl) -5-methoxypyrazine-2-carboxamide The "1 {OMe)) ',,,'" y Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2,3-difluorophenyl) -5-trifIuorometij-4aS, 7,7a-tetrahydro -4H-füro [3,4-d] [1,3] thiazin-2-i1] tert-butyl carbamate and 5-methoxypyrazine-2-carboxylic acid according to a general procedure. 'NMR (400 MHz, CDCl, + MeOlj) δ ppm 2.83 (dd, J = 13.9, 3.8 Hz, 1 H), 3.14 (dd, J = 13.9, 3.0 Hz, i H), 3.29-3.39 (m, 1 H), 3.87 (dj = 8.3HLlH), 4.04 (s, 3H), 4.60 (d, j = 8.3Hz, 1Fn, 4.67 (quin, j = 6.3w, 1H), 7 , ll-7,2l (m, 1H), 8.03 (ddd, j = 11,6,6,9,2,7m, lH), 8.15 (d, j = 1,3W, lH), 8.96 (d, j = j, 3W, 1H) Example 17: N {3 {(4aS, 5S, 7aS) -2-amino-5- (tMuoromethyl) - 4a, 5,7,7a-tetrahydro-4H -hro [3,4-d] [1,3] ti & in-7a-yl) -4,5-dinuorophenyl) -5-methylpyrazine-2-carboxamide om23 "' The "'Qf' Synthesized from [(4aS, 5 S, 7aS) -7a {5-amino-2,3-difluorophenyl) -5-trinuoromethyl-4a, 5,7,7a-tetrahydro-4H-hro [3, 4-d] [1,3] tiwin-2-yl] carb = tert-butyl act and 5-methylpyrazine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHz, CDCl, + MeOD) (3 ppm 2.68 (s, 3 H), 2.84 (dd, J = 13.6, 3.8 Hz, 1 Il), 3.15 (dd, J = 13.9, 3.0 Hz , IH), 3.30 - 3.42 (m, lH), 3.88 (d, j = 10.4W, 1H), 4.6l (d, j = 8.6W, 1W, 4.68 ( quin, J = 7.2Hz, 1H), 7.13-7.25 (m, lH), 8.05 (ddd, j = 1i, 6.6.8.2.8Hz, 1H), 8.46 (s, 1H), 9.30 (d, j = j, 3Hz,! H) Example 18: N- (3 - ((4aS, 5S, 7aS) -2-mino-5- (tnnuoromethyl) - 4a, 5,7,7a-tetrai & o-4H-hro [3,4-d] [1,3] ti = in-7a-yl) -4,5-dinuorophenyl) -5- (nuoromethyl) -pyrazin-2-carboxamide 1) '(F' Synthesized from [(4aS, 5S, 7aS) -7a- (5-amino-2,3-diüuorophenyl) -5- trifluoromethyl-4a, 5,7,7a-tehaidro-4H-fúro [3,4-d] [1,3] thiazin-2-yl] tert-butyl carbamate and 5- (fluoromethyl) pyrazine-2-carboxylic acid according to a general procedure. 'H NMR (400 MHZ, CDCl) i5 ppm 2.89 (dd, J = 13.6, 3.8 Hz, 1 H), 3.19 (dd, J = 13.6, 3.0 Hz, 1 H), 3.43 (dd, J = 7.5, 3.4 llz, 1 H), 5 3.86 - 3.99 (m, 1 H), 4.39 - 4.67 (m, 3 H), 4.74 (quin, J = 7.1 Hz, 1 H), 5.76 (d, j = 45.5 Hz, 2H), 8.10 (ddd, j = 11.4, 6 , 8.2.8 Hz, IH), 8.77 (s, 1H), 9.46 (s, 1 H), 9.69 (s, 1 H) The alternate preparations of the compounds of Examples 1 and 8 are described in this below. For these alternate preparations, 1H NMR and '3C NMR spectrum were recorded on a Varian 400 MHz or 500 MHz instrument with sofWare vRMN 6.1C. Alternative preparation of N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (tMuoromethi]) - 4a5j, 7a-tetrahydro-4H-nIro [3,4-d] [1,3] thiazine -7a-yl) -4-fluorophenyl) -5-methoxypyrazine-2-carboxamide (Example 1) 1- (14) Synthesis of 2- (1,1,1-trifluorobut-3-en-2-yloxy) acetate tert-Butyl OHC) ,, M, "jji </ NaOH aq., Iy U 1 < Cfjms y f CF, cat. TBAF 3C Bu, N HSO4 toluene 0 ° C A reaction vessel was loaded with toluene (3.2 L), THF (0.60 L) and Acrolein (0.40 L, 5.985 mol) at room temperature under nitrogen. (Trifluoromethyl) trimethylsilane (1.003 kg, 7.059 mol) was added at 17 ° C. The reaction mixture was heated at 2.5 ° C and TBAF (0.01 M in THF, 0.400 L, 0.004 mol) was added over 2 h. During the addition of TBAF, the temperature of the reaction mixture increases to 65 ° C. The reaction mixture was spun at 0 ° C, and After 2h, tetra-n-butylammonium hydrogen sulfate (0.171 kg, 0.503 mol) was added, followed by tert-butyl bromoacetate (0.987 kg, 5.064 mol). Sodium hydroxide (50 ° /, weight in water, 4.2 kg, 52.6 mol) was added over 2h while maintaining a temperature under 10 ° C. After 2h at 0-5 ° C, water (2.9 L) and tert-butyl methyl ether (6.0 L) were added to a reaction mixture. The aqueous phase was extracted once more with tert-butyl methyl ether (6.0 L). The organic phases were combined and washed with 14% aqueous NaC3 (3 x 1.6 L). The organics were concentrated in vacuo to produce the title compound as an oil (1,150 kg, 94.5 ° / 0) which was used in the subsequent stage without further purification. 'H NMR (500 MHz, CDC !,) Õ ppm 5.86 - 5.74 (m, 1 H), 5.59 (d, J = 17.5m, lW, 5.56 (d, j = 10 , 9w, 1W, 4.37-4.30 (m, lH), 4.1l (d, j = 16.5 HZ, 1 H), 4.06 (d, J = 16.4 Hz, 1 H ), 1.40 (s, 9H); "C NMR (125 MHZ, CDCl,) D ppm 168.51, 128.49 (d, J = 1.7 Hz), 123.86, 123.71 (q, J = 281.8 Hz), 82.22, 78.67 (q , J = 31.5 Hz), 66.60, 28.02. 1- (15) Synthesis of Nmethoxy-N-methyl-2- (1,1 l-tMuorobut-3-en-2-yloxy) acetamide ) Uoj <HCOOH ('V)) ° jjijA' CF3 MeONHMe-HCl "CF, CDl, DCM Stage 2 To a reactor containing 2- (1,1,1-trifluorobut-3-en-2-yloxy) tert-butyl acetate (1,150 kg, 4,788 mol) was added formic acid (6.2 kg) at room temperature. The reaction mixture was heated to 55-60 ° C for 4-5 h. The formic acid was evaporated in vacuo (T = 40 to 45 ° C) and challenged with toluene (2 x 3.0 L). To the residue, CH2Cl2 (2.0 L) was added and further concentrated in vacuo. To the resulting residue was added CH2C12 (4.6 L) and the solution was spun at 0 ° C, followed by N, N-carbonyldiimidazole (1.05 kg, 6.49 mol) in five portions. The mixture was added for 30 mins and N, O-dimethyl & oxylamine hydrochloride (0.67 kg, 6.72 mol) was added in portions while maintaining the temperature below 10 ° C. The reaction mixture was heated at rt and stirred for 14 h. The reaction mixture was heated at 3.2 ° C and imidazole (100.7 g, 1.48 mol) was loaded in two portions. The reaction mixture was heated in rt and water (1.4 kg) was added, followed by tert-butyl methyl ether (14.0 L). The organic phase was washed with 2.0 N of aqueous HCl (1.0 L and 0.7 L), followed by NáHCO, sat. aq. (1.2 L) and sat. NaCl. aq. (1.20 L). The organics were concentrated to produce the title compound as an Oil (0.95 kg, 87.2 ° /). 'H NMR (500 MHz, cDclj) Ô ppm 5 5.85 -5.76 (m, IH), 5.62 (d, J = 17.2 Hz, 1 H), 5.56 (d, J = 10.4 Hz, IH), 4.49 -4.34 (m, 3 H), 3.68 (S, 3 H), 3.67 (s, IH), 3.18 (s, 3 H), 3.08 (s, IH); "C NMR (126 MHz, cdcl,) Ô ppm 169.9 *,! 63.4 *, 128.61,! 23.87 (d, J = 282.0 Hz), 123.82, 78.54 (q, J = 31.3 Hz), 66.12, 6, 52, 60.56, 36.20, 32.24. Note: this compound is a 3: 1 mixture of the amide bonding rotamers. 10 * chemical changes of carbonyl estimated 'indirectly through iH_ 13ç HMBC (heteronuclear multiple bond correlation). HRMS Calculated by C, H ,, F, NO, [M + H] "228.0848; observed 228.0836. 1- (16) Synthesis of 1 {2-Fluorophenyl) -2- (1,1,1- trif1uorobut-3-en-2-yloxy) ethanone ) ojji> 'j,): °) CF3 nBuLi, THF Stage 3 In a solution of 1-bromo-2-fluorobenzene (0.967 kg, 5.527 mol) in THF (6.2 L) at -75 ° C, n-butyllithium (2.50 M in hexane, 2.09 L, 5 , 22 mol) while keeping the temperature below -65 ° C (ca. 100 min.). After 15 mins, a solution of N-methoxyN-methyl-2- (1,1,1-trifluorobut-3-en-2-yloxy) acetamide (0.949 kg, 4.178 mol) in THF (1.6 L) was added while keeping the temperature below -65 ° C (ca. 70 min.). After 2.5 h at - 78 ° C, the reaction was quenched by the addition of NH4Cl sat. aq. (3.0 L) and tert-butyl methyl ether (9.0 L). The reaction mixture was heated to rt, the aqueous phase was extracted again with tert-butyl methyl ether (2.5 L). The organic phases were combined, washed with sat. NaCl. aq. (2 x 0.3 L) and concentrated in vacuo to produce the title compound as an oil (1.007 kg, 80.0 ° 4). 'H NMR (500 MHz, CDCl) çj ppm 7.96 (td, J = 7.6, 1.8 Hz, 1 H), 7.62 - 7.54 + (m, 1 H), 7, 33 - 7.25 (m, 1 H), 7.20 - 7.12 (m, 1 H), 5.86 (ddd, j = 17.5, 10.4, 7.3 Hz, IH), 5.60 (dd, j = 20.5, 13.8 Hz, 2 H), 4.91 - 4.76 (m, 2 H), 4.39 (dq, 5 J = 12.8, 6, 4 Hz, IH); "C NMR (125 MHz, CDCl,) δ ppm 193.55, 162.14 (d, Jc, = 254.1 Hz), 135.36 (d, Jc, = 9.2 Hz), 130.62 ( d, Jcf = 3.2 Hz), 128.49, 124.85 (d, Jcf = 3.3 Hz), 123.89, 122.93, 122.72 (d, Jc, = 24.5 Hz) , 116.50 (d, Jc, = 23.7 Hz), 78.97 (q, Jcf = 31.4 Hz), 74.56 (d, Jc, = 12.4 Hz). HRMS Calculated by C ,, H, 0F4O, [M + H] "263.0695; 10 observed 263.0709. 1- (17) Synthesis of 1 {2-Fluorophenij) -2- (1,1,1-triMorobut-3-en-2-yloxy) ethanone oxime f NH, OH HCl, ho-n f NaOAc) o /! MeOH. , C ^ yO ", | CF3 C / Stage 4 CF3 C, To a reactor was added hydroxylamine hydrochloride (0.34 kg, 4.95 niol), sodium acetate (0.47 kg, 5.70 mol) and MeOH (2.68 L). To this suspension was added a solution of 1 (2Auorophenyl) -2- (1,1-trifluorobut-3-en-2-yloxy) ethanone (0.998 kg, 3.806 mo!) In McOH (1.8 L) and the reaction mixture was heated to 40 to 50 ° C- On completion (ca- 2 h) the reaction mixture was spun in RT and filtered through Celite (0.5 Wt) and rinsed with EtOAc (3.0 L) The filtrate was concentrated in vacuo and to the resulting residue 20 was added tert-butyl methyl ether (6.3 L), water (0.94 L) and sat. NaHCO3. aq. (2.5 L). The organic phase was washed once with water (1.6 L) and sat. aq. (0.1 .L). The organic phase was concentrated in vacuo to yield the title compound as an oil (1.03 kg, 95.0 ° /,), 1 H NMR (500 MHz, CDCl,) tj ppm 7.49 - 7.35 ( m, 2 Il), 7.24 - 7.06 (m, 2 H), 5.78 - 5.65 (m, l 25 H), 5.54 - 5.40 (m, 2 Hj), 4 , 89 - 4.81 (m, 1 H), 4.53 (d, J = 12.6 HZ, 1 H), 4.47 (d, j = 12.6 Hz, 0.5 H), 4 , 27 -4.18 (m,] H), 4.13 -4.05 (m, 0.5H). 11 The HRMS Calculated by C ,, H ,, F, NO2 [M + H] "278.0804; observed 278.0780. Note: 1- (2-F! Uorophenyl) -2- (1,!, J-MAuorobut-3-en-2-yloxy) ethanone oxime exists as a balance of structural isomers, which takes into account the values integrals smaller than the total number. 1- (18) Synthesis of (3aR *, 4S *, 6aS *) - 6a- (2-fluorophenyl) -4- (triàuoromethyl) hexahydro [3,4-c] isoxazol '° " ' Fjh hydroquinone ·) o "-" "" ") / -jN cf, C /" o, J,; O xylenes 130 ° C F3cr A In a solution of 1 {2Auorophenyl) -2- (1, l, l-trifluorobut-3- en -2-yloxy) etmone oxime (1.085 kg, 3.328 mol) in xylenes (6.9 L) hydroquinone (86.2 g, 0.8 mol) was added in rt. The solution was heated to 1228 ° C (internal temperature ) for J8 h. The solution was tested in RT and hexanes (7.0 L) was added, followed by 4.0 M aq. HCl (2.4 L). The reaction mixture was added for 1 h and filtered. water (2.0 L), tert-butyl methyl ether (7.0 L) and 25 ° 4 by weight of aqueous NaOlyl (0.4 L) were added, the aqueous layer was extracted once with tert-methyl ether -butyl (7.0 L), the organics were combined, washed with 27 ° 4) aqueous NaCl (2.0 L) and concentrated under vacuum in a black oil (512.0 g, 56 ° /). NMR (500 MHz, CDCl) 6 ppm 7.64 - 7.52 (m, 1 H), 7.39 - 7.31 (m, 1 H), 7.19 (td, J = 7.7, 1.2W, 1H), 7.1l (dd d, j = 11,9,8,2,1,0W, lH), 4,54 (d, j = l0, lHz, IH), 4,34-4,23 (m, IH), 4,26 -4.17 (m, 1 H), 4.16 (d, J = 10.2 FIz, IH), 4.10 (d, J = 8.5 Hz, 1 H), 3.71 (d, J "= 20.2 Hz, IH); "C NMR (125 MHZ, CDC !,) Õ ppm 160.59 (d, Jcf = 247.0 Hz), 130.50 (d, Jcf = 8.7 Hz), 128.72, 124.69 (d , Jc, = 3.3 Hz), 124.45 (q, Jcf = 281.8 Hz), 124.43 (d, Jcf = 11.9 Hz), 116.66 (d, Jcf = 22.7 Hz ), 83.70 (q, Jcf = 32.1 FIZ), 78.17 (d, Jc, = 3.1 HZ), 77.63, 54.53. III HRMS Calculated by C ,, H ,, F, NO2 [M + H] "278.0804; observed 278.0802. 1- (19) Synthesis of) ((2S *, 3R *, 4S *) - 4-amino-4- (2Aorophenyl) - 2 (trifluoromethyl) tetrahydro-3-yl) methanol Á. ,,,,,,,, J) lí ,, O 'O THF, water "O, - ^ W, F, Ê Êi F, i racemic 5 Zinc-o (389.2 g, 5.95 mol) was placed in a reaction vessel and water was added (893 niL), acetic acid (135 niL, 2.38 mol) was added while maintaining a temperature below 10 ° C. After 15 min, 6a- (2-nuorophenyl) ) -4- (winuoromethyl) hexahydroMo [3,4-c] isoxazole (550.0 g, 1.98 mol) was added as a solution in THF (665 mL). The 10 reaction mixture was added over 16 h in rt Methylene chloride (1.89 L) was added, followed by 28 ° / aq. of NjH4oH aq. (552 mL) while the temperature was kept below 30 ° C. The mixture was added for 30 min and then filtered over Celite (80 g) rinsing with methylene chloride (378 niL). The aqueous layer was extracted with methylene chloride (i, 89 L). The organics were combined, washed with sat. NaCl (1.0 L) and concentrated in vacuo to produce an Oil (502 g, 90.6 ° 4) The crude residue was used in the next step without further purification - HRMS Calculated p or C ,, H ,, F, NO2 [M + H] "280.0961; observed 280.0972. 1- (20) Synthesis of ((2SÀR, 4S) -4 ^ nino-4- (2-fluorophenyl) -2- (trifluoromethyl) tetrahydro-aluminum-3-yl) methanol (2S, 3S) -2,3-bis ( benzoyloxy) succinate ) nhz D-DB'A,) ,,, Hojj '- · PI ,, EtOH, ag "r" "| F3C OH F3C OH In a solution of 4-amino-4- (2-fluorophenyl) -2- (trifluoromethyl) tetrahydro-3-yl) methanol (0.502 kg, 1.798 mol) in ethanol (4.865 L), dibenzoyl-D-tartaric acid ( 0.642 kg, 1.788 mol). The resulting suspension was heated to 67 ° C. Water (94.0 ml, 5.2 mol) was added over 15 min while maintaining the temperature> 66 ° C. The resulting sojering was ground at 45 ° C while precipitation occurred. The paste was reheated to 60 ° C and then stirred at room temperature at 5 ° C / hour. The slurry was filtered and the solid was rinsed with spherical and pre-mixed solution of ethanol (950 ml) and water (20 ml). The solid was dried to a constant 10 ° weight under vacuum (370 g, 97.6 ° / e). 'H NMR (500 MHZ, CD, OD) δ ppm 8.13 (d, J = 7.2 IIz, 4 H), 7.66 - 7.58 (m, 3 H), 7.54 - 7, 45 (m, 5 H), 7.36 - 7.20 (m, 2 H), 5.92 (s, 2 H), 4.79 - 4.66 (m, 1 H), 4.40 - 4.28 (m, IH), 4.04 (dd, j Á 12.1, 3.4 IIZ, 1 H), 3.92 (dd, J = 12.1, 5.4 Hz, 1 H) , 3.30 - 3.24 (m, IH); "C NMR (125 MHz, DMSO) D ppm 169.61, 165.81, 160.23 (d, J = lS 246.1 Hz), 133.00, 131.34 (d, J = 9.1 Hz ), 129.65, 129.55, 128.08, 127.97 (d, J = 3.5 Hz), 124.95 (d, J = 3.3 Hz), 116.56 (d, J = 23.5 Hz), 77.48 (q, Jcf = 31.0 Hz), 76.33, 73.20, 65.61 (d, j = 3.1 Hz), 57.11 HRMS Calculated by C ,, H ,, F, NO, [M + H] "280.0961; observed 280.0967 (for amino alcohol). The absolute stereochemistry of the title compound was indicated by comparison with a starting sample prepared from enantioenriched by (S) -2- (Üifluoromethyl) oxirane. Chiral HPLC parameters: Equipment, Reagents and mobile phase: Equipment: "T 113 HPLC column: Cbiralcel OD, 4.6 x 250 mm,] 0 µm, Daicel Chemical Industries, Ltd., catalog No. 14025. Solvent release system: Agilent llOO HPLC, low pressure mixture, low pressure mixture with degasser in im or equivalent Autosampler: autosampler A $ ilent 1100, 0.1 to 100 µL of bandZ or equivalent Detector: variable wavelength detector A *] ent i 100 or equivalent Crygraphic software: Sofware Agilent ChemStation version A.09.03 or higher by HPLC, Waters Empower 2 Build 2154 or equivaknte volumetric glass: Class A. Volumetric pipette: CIasse A. pipette Volume calibrated Eppendorf, or equivalent. Scale: Analytical balance & capable of weighing ± 0.1 mg . Reagents: Heptane: HPLC grade. Baker (catalog No. 9177-03) or equivalent. 2-Propanol: Grade HPLC, Baker (catalog and No. 9095-03) or equivalent. Triethylamine: Z 99 ° / ,, SiQma-Aldrich (catalog No. T0886) Oll equivalent. Mobile phase: Add 70 niL of 2-propanol and 930 mL of heptane (measured separately with 100 mL and 1000-ML graduated cylinders) and 1.0 niL of 5 triethylamine (measured with a vojumetric glass pipette) in an Easco and mix appropriate. Degas in-line during use. Dilution solution: 2-Propanol HPLC parameters: HPLC column: Chiralcel OD, 4.6 x 250 mm. 10 µm, Daicel Chemical Tndustries. Ltd., catalog no. 14025. Temperature: 35 "C Flow rate *: 0.8 ml / min Gradient: NA Injection volume: 5 µL Detection: 262 nm UV Data acquisition time: 30 min Total performance time: 30 min Maximum column pressure: 35 Bar Needle wash: 2-propanol! * "Flow rate can be adjusted ± 0.2 ml / min to maintain specified retention times. Retention times for analytes and impurities: Compound peak Retention time (Relative retention time, RRT) | 20.6min ± 10% fJJ) H, (RRT 1.00) F, C OH 19.2 min (RRT 0.93) Jm °; jj; "'F, C" ÓH (Enantiomer 10 A typical chromatogram of an E- chiral HPLC isolation. compound 1- (20) is shown in figure 1. 1- (21) Synthesis of N - ((3S, 4R, 5g-3- (2-nuoropheni]) - 4- (hydroxyl) -5- (trinuoromethyl) -tetrahydro-3-ylcarbamothioyl) benz = outward F ') ,,, Hojçj 'cj' ".) J- ,, O> J · tt, j EtOAc, 1N NaOH rb] S FAITH OH F, CH OH To the chiral salt ((2S, 3R, 43-4- = inno- 4- (2-nuorophenyl) -2- 5 (trinuoromethyl) tetrahydronlrano-3-yl) methanol (2S, 3S) -2,3-bis (benzoyloxy) succinate (0.361 kg, 0.556 mo!) EtOAc (1 , 08 L) and the suspension was sieved at -3 ° C. 1.0 N of NaOlil aq. (1.30 L) was added over 20 mins while maintaining T <5 ° C. After 5 mins, benzoyl isoticyanate ( 80.0 mL, 594 mmol) was added over 8 mins while maintaining T <5 ° C. After] h, EtOAc (722 ml) was loaded. The aqueous layer was removed and the organics were washed with sat. aq. (361 mL) and sat. NaCl. aq. (361 ml). The organics were filtered through celite (90 g) and rinsed with EtOAc (360 ml). The organics were concentrated in vacuo to produce a residue that was redissolved in CH2C | 2 (1.1 L) and concentrated to produce the title compound as yellow foam (261 g, 99 ° /) yield taking into account the residual solvents ) that was used in the following step. Yh NMR (500 MHz, DMSO) δ ppm 12.04 (s, 2 H), 11.20 (s, 2 H), 7.95 (d, J = 7.4 Hz, 2 H), 7.69 - 7.60 (m,] H), 7.56 - 7.42 (m, 2 H), 7.37 - 7.28 (m, 1 H), 7.24 - 7.12 (m, 2H ), 5.59 (t, J = 4.5 Hz, 1 H), 5.03 (d, J = 9.7 Hz, 1 H), 4.92 (d, J = 9.7 Hz, 1 H), 4.75 - 4.63 (m, 1 H), 3.92 - 3.74 (m, 2 H), 2.77 - 2.66 (m, IH); "C NMR (125 MHZ, DMSO) Õ ppm 179.98, 167.85, 159.75 (d, Jcf = 245.0 Hz), 133.44, 132.58, 129.88, 129.81, 129 , 04, 128.85, 126.31 (d, Jcf = 9.8 Hz), 124.36, 116.83 (d, Jc, = 23.4 Hz), 76.11 (q, Jc, = 31 , 0 Hz), 74.37 (d, Jc, = 6.1 Hz), 68.77 (d, Jcf = 3.4 Hz), 57.03, 52.23 HRMS Calculated by C20Hl8F4N2O3S [M + H ] "441.0896; observed 441.0818. 1- (22) Synthesis of N - ((4aS, 5SJaS) "7a {2Auorophenyl) -5- (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4HAüro [3,4-d] [1, 3] tiazin-2-yl) benzamide "J- ,, ,,,,,,,,,,,,. ) Nj- ,, O) s CH, Cl, F, Ô 'ÔH F, Ê A A solution of N - ((3S, 4R, 5S [) - 3- (2Auorophenyl) -4-5 (hydroxyl) -5 - (tnnuoromethyl) -tetraidromrano-3-i] carbmothioyl) well = flow (258.3 g, 583.8 mmol) in CH, Cl2 (1.55 L) was cooled to -19.4 ° C. Pyridine (1118 niL, 1.46 mol) was added while maintaining the temperature at -20 ° C and then the reaction mixture was cooled to -24 ° C. In another nitrogen-purged container, CH2Cl2 (258 mL) was added followed by 10 trifluoronethanesulfonic anhydride (108.0 niL, 642.2 mmol). The resulting solution was added to a reaction mixture over 30 min, while maintaining the temperature <-19.7 ° C. Upon complete addition, the reaction mixture was added for 30 min at -20 ° C to -15 ° C and then heated at -1l ° C for 20 min. Saturated NH4Cl aq. (646 niL) and water (390 ml) was added. The mixture was warmed to room temperature and the aqueous layer was reinovided. The organics were washed with saturated aq. premixed (646 mlj) and water (390 niL). The aqueous layers were combined and extracted once with CH2Cl2 (520 niL). The organics were combined and concentrated in vacuo to produce a light orange foam (250 g, 100 ° / 0). The residue was used in the next stage without purification. 'H NMR (500 MHz, CDC] 3) δ ppm 8.03 (d, j = 6.7Hz, 2H), 7.52 (t, j = 7, OHz, 1H), 7.48-7.31 ( m, 4H), 7.20 (t, j = 7.4 Hz, IH), 7.12 (dd, J = 12.0, 8.4 Hz, 1H), 4.82-4.73 (m, l H), 4.60 (d, j = 8.9N, 1H), 4.03 (d, j = 8.3W, lH), 3.57 (d, j = 2.7Hz, l H), 3.20 (d, J = 13.6 Hz, 1 H), 2.81 (dd, J = 13.8, 2.5 Hz, IH); "CRMN (125 MHz, CDCl,) t: ppm 171.50, 159.57 (d, Jcf = 247.2 Hz), 134.62, 132.49, il6 130.65 (d, Jc, J = 8.8 Hz), 129.77, 128.51, 128.45, 125.14 (q, Jc, = 281.8 Hz), 124.97 (d, Jc , = 3.0 Hz), 124.66 (d, Jc, = 10.3 Hz), 117.05 (d, Jc, = 23.5 Hz), 66.81 (d, Jc, = 5.2 Hz), 38.90, 23.20. HRMS Calculated by C20HiS4N, O2S [M + H] "425.0947; 5 observed425.0945. 1- (23) Synthesis of (4aS, 5S, 7aS) -7a- (2-nuorophenyl) -5- (trifluoromethyl) -4aS, 7,7a-tetrahydro-4H-nIro [3,4-d] [1,3] thiazin-2-amine , I) '"b' :: ·. Jj" In a solution of N - ((4aS, 5S, 7aS) -7a- (2-Auorophenyl) -5- (triRuoromethyl) -4a, 5,7,7a- tetrahydro-4H-mro [3,4-d] [1,3] thiazin-2-yl) benzmide (250.2 g, 589.5 mmol) in methanol (1.25 L) was added K, CO3 (81 , 5 g, 590.0 mmol). The suspension was heated to 65 ° C for 6 h. On drying at room temperature, the solvent was evaporated in vacuo. To the resulting residue, 1.0 N of aq. (1.18 L) and THF (502 mL). The heterogeneous mixture was heated at 45 ° C for 1 h. The mixture was cooled to room temperature and EtOAc (1.38 L) was added. The aqueous layer was extracted with EtOAc (0.75 L). The organics were combined, washed with NaljCO, saturated aq. (500 niL) and saturated aq. (500 mL). The organics were concentrated in vacuo to produce the title compound as a brown oil (184.1 g, 91.6 ° /) yield taking into account residual solvents). | H NMR (500 MHz, DMSO) 6 ppm 7.49 - 7.42 (m, 1 H), 7.40 - 7.33 (m, IH), 7.26 - 7.15 (m, 2 H ), 6.26 (s, 2 H), 4.77 - 4.54 (m, 1 H), 4.40 (d, J = 8.0 HZ, IH), 3.80 (dd, j = 7.9.2.3 Hz, 1 H), 3.24 - 3.17 (m, 1 H), 3.00 (dd, J = 13.9, 3.2 Hz, 1 H), 2, 85 (dd, J = 13.9, 3.9 Hz, IH); "C NMR (125 MFIz, DMSO) 6 ppm 159.75 (d, Jc, = 245.1 Hz), 149.51, 131.31 (d, Jc, = 3.9 Hz), J30, l3 (d , Jcf = 8.8 HZ), 128.08 (d, Jc, = 10.4 Hz), 128.28 (q, Jcf = 282.1 Hz), 124.87 (d, J ,, = 3, 0 Hz), 116.80 (d, J = 23.8 Hz), 78.77, 76.80 (q, J, = 30.8 Hz), 66.31, 36.37, 23.27. HRMS Calculated by C ,, H ,, F, N, OS [M + H] "321.0685; observed 321.0677. 1- (24) Synthesis of (4aS, 5S, 7aS) -7a- (2Aoro) hydrochloride -5- - 5 nitrophenyl) -5- (triBuoromethyl) -4a, 5,7,7a-tetrahydro-4H-hro [3,4-d] [i, 3] ti & in-2-amine I ~ 1. hno, , V 'A "' y" 'h2so4 "^ t))) °: H2Hc ° A ~' f, GH '" "' µ F, t H To the cooled vessel containing (4aS, 5S, 7ag-7a- (2- nuorophenyl) -5- (trinuoromethyl) -4a, 5,7,7a-tetrai & o-4H-hro [3,4-d] [1,3] thiazin-2-amine (184.1 g, 574.8 mmol) triijuoroacetic acid (0.954 kg) 10 was added in portions while the temperature was kept below 20 ° C. The mixture was stirred at 3.5 ° C and sulhiric acid (146 niL, 2.73 mol) was added over 20 min while the temperature was kept below 5 ° C. Fumigation of nitric acid (39.8 ml, 0.948 mol) was added over 30 min, while the temperature was kept below 10 ° C. After 1.5 ha O-10 ° C, the reaction mixture was slowly quenched by transferring in an aqueous solution of NaOH (575 g, 14.4 mol) in water (4.6 L) at 5 ° C. The resulting suspension was added per 1 l The suspension was then filtered and the solid was rinsed with cold water (920 mL). The solid was dried under vacuum to constant weight and then dissolved in ethanol (1.05 L). heated to 20 35 ° C and conc. (55.6 ml, 0.690 mol) was added while maintaining the temperature below 40 ° C. The suspension was then cooled to -5 ° C, maintained for 1 br and filtered. The solid was rinsed with cold ethanol (420 ml) and dried to constant weight to obtain the title compound (185.0 g, 87.3 ° 4). 1 H NMR (500 MHZ, DMSO) t ppm 11.80 (s, 2 H), 8.45 - 8.36 (m, 1 H), 8.31 25 (dd, J = 6.6, 2.5 Hz, 1 H), 7.66 (dd, J = 1l, l, 9.3 Hz, l H), 4.96 - 4.72 (m, 1 H), 4.58 (d, j = 10, Ol1z, 1H), 4.27 (d, j = 9.9W, 1H), 3.76-3.66 (m, IH), 3.39 ( dd, J = 14.9, 3.6 Hz, 1 H), 3.24 (dd, J = 14.3, 4.6 Hz, IH); "C NMR (125 MHz, DMSO) ppm 168.34, 163.33 (d, Jc, = 257.8 Hz), 144.58, 127.61 (d, Jc, = 11.6 Hz), 125 , 84, 124.10, 119.28 (d, Jc, = 26.5 HZ), 77.38 (q, 5 Jc, = 31.5 Hz), 75.99, 65.88 (d, Jc, = 4.8 Hz), 40.36, 23.98. HRMS Calculated by C ,, H ,, F, N, O, S [M + H] "366.0536; observed 366.0523. 1- (25) Synthesis of (4U, 5S, 7 © -7a- (5-amino-2-Auorophenyl) -5- (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4H-fúro [3,4 -d] [1,3] thiazin-2-amine a j) ,, ,,, ,,, ,,,, jj'ÍH, ° JÁ> ,, OH f, C hf, G h O ethanol (0.975 L ) was added to the iron powder (62.5g, 1.12 mol) under a nitrogen atmosphere. Concentrated HCl (9.03 mL) was added at room temperature and the suspension was heated to 65 ° C for 1.5 h. The suspension was then cooled to 50 ° C and sat. aq. (299 g) were added. The temperature of the reaction mixture was allowed to reach 50 ° C and (4 ^, 5S, 7 © -7a- (2-nuoro-5-nitrofeml) -5- (tnnuoromethyl) - 4a, 5,7,7a- hydrochloride tetrahydro-4HAtro [3,4-d] [1,3] thiazin-2-amine (75.0 g, 187.0 mol) was added in portions while maintaining the temperature below 68 ° C. After 30 min, ethanol (0.45 L) was added and the reaction mixture was stirred at 20-25 ° C for L h. The suspension was added for 2 h and filtered over Celite (75 g) rinsing with ethanol (0.972 L). The solution was concentrated in vacuo to a brown solid. Water (0.9 L) was added followed by 3.0 N NaOlil (0.187 L, 560 mmol) while maintaining the temperature below 35 ° C. The resulting suspension was added for 1 h at 20-25 ° C. The suspension was filtered and the solid was rinsed with Ria water (0.38 L). The solid was dried under vacuum at 40-45 ° C for 24 h to obtain the title compound 11 (57.7 g, 95.5 ° 4). 'H NMR (500 MHz, DMSO) δ ppm 6.81 (dd, J = 12.5, 8.6 w, lm, 6.62 (dd, j = 7.0.2.9w, 1H), 6 , 50-6.42 (m, lm, 6, 16 (s, 2H), 4.96 (s, 2 H), 4.72-4.54 (m, IH), 4.35 (d, j = 7.8 Hz, IH), 3.74 (dd, j = 7.8, 2.5 Hz, 1 H), 3.18-3.08 (m, IH), 3.01 (dd, j = 13.9, 3.0 Hz, 1H), 2.84 (dd, j 5 = 13.8, 3.8 Hz, IH); "C NMR (125 MHz, DMSO) D ppm 156.20 (d , Jcf = 243.0 Hz), 148.73, 145.49, 127.86 (d, Jc, = 11.0 Hz), 116.79 (d, J ,, = 24.8 Hz), 116, 10 (d, Jc, = 3.3 Hz), 114.10 (d, Jcf = 8.0 Hz), 78.89, 76.57 (q, Jcf = 31, OHz), 66.35.36, 35,23,11. HRMS Calculated by C ,, H ,, F, N, OS [M + H] "336.0794; 10 observed 336.0789. The title compound was subjected to an Ames test (strains examined Salmonella Nphimurium TA98, TAIOO, TA1535 and TA1537 and strain examined Escherichia coli WP2 uv. Mutation Research 1975, 31, 347; Mutation Research 1976, 38, 3; Proc. Nat. Acad. Sci. USA 1976, 73, 950; 15 Proc. Nat. Acad. Sci. USA 1975, 72, 5135) in the absence and presence of the S9 rat liver. The compound was negative until the tested concentration / highest dose (5000 µg / plate). 1- (26) Synthesis of N- (3 - ((4aS, 5S, 7aS) -2-amino-5- "(trinuoromethyl) -4a, 5,7,7a-tetrahydro-4H-hro [3,4- d] [1,3] thiazin-7a-yl) -4- 20 nuorophenyl) -5-methoxypyrazine-2-carboxamide <N ~ yOMe H, c'NmoMe "'' ° jj" Nj í) r'íj "NA 'À:' "r" "'" 4'y "' CÁ s SOCl ,," osf, C h dmi F3C h A suspension of 5-methoxypyrazine-2-carboxylic acid (26.29 g, 0.17 mol) in N, W-dimethylimidazoline-2-one (160 ml) was stirred at room temperature for 15 min, then cooled at 2.2 ° C. Thionyl chloride (14.7 niL, 0.202 mol) was added while maintaining temperature 25 under 5 ° C. The resulting suspension was stirred at 0-10Â ° C for 2 h while it was already transitioned into a clear solution. In another container, (4aS, 5S, 7aS) -7a- (5-amino-2-nuorophenyl) -5- (trinuoromethyl) -4a, 5,7,7a-teKai & o-4H-hro [3,4-. d] [1,3] thiazin-2-amine (52.0 g, 0.155 mol) was dissolved in N, N'-dimethylimidazoline-2-one (160 ml). The resulting solution was added to an acyl chloride solution while maintaining the temperature below 10 ° C. The reaction mixture was added for 30 min. The water (780 mL) was charged while maintaining the temperature below 30 ° C. The resulting mixture was added for 30 min and then EtOAc (780 ml) was added. To this mixture was added, 50 ° / aq. (84.8 g) until the pH of the aqueous layer 10 reaches 11. The aqueous layer was extracted with EtOAc (260 niL). The organics were combined, washed with sat. NaCl. aq. (260 ml) and water (260 ml). The organics were filtered through a Celite pad (26 g) and rinsed with EtOAc (260 ml). The organics were concentrated in vacuo to produce a solid. To the solid was added l-propanol (728 ML) 15 and the suspension was heated to 75 ° C until a clear, phoned solution. The solution was spun at -10 ° C and maintained for 1 h. The solid was filtered, rinsed with Eio 1-propanol (104 niL) and dried under vacuum (35 ° C) to constant weight to produce the title compound (62.1 g, 84.9 ° 4). 'H NMR (500 MHz, DMSO) 4 Dppm10.56 (s, 2W, 8.88 (d, j = 1.2W,! W, 8.39 (d, j = 1.2W, 1H), 7.95 20 - 7.83 (m, 2 H), 7.18 (dd, J = i2.0, 8.8 Hz, 1 H), 6.25 (s, 2 H), 4.76 - 4.60 (m, 1H), 4, 36 (d, j = 8.1 &, 1H), 4.01 (s, 3m, 3.88 (dd, j = 7,9,2,3w,] m, 3.23 3, II (m, 2 H), 2.91 (dd, J 13.8, 3.6 Hz, IH); "C NMR (125 MHz, DMSO) at ppm 162.11, 161.93, 156.13 (d, Jc, = 242.9 Hz), 149.38, 142.01, 138.35, 135.09, 133.98, 128.53 (d, Jc, = 11.6 Hz), 126.06 (q, Jc, "282.0 HZ), 25 123.32, 121.93 (d, Jc, = 8.6 Hz), 116.76 (d, Jc, = 25.1 Hz), 78.86 (d, Jc, = 6.9 Hz), 76.94 (q, Jcf = 30.5 FIZ), 66.37, 54.75, 36.44, 23.53. HRMS Calculated by C ,, H ,, F, N, O, S [M + H] "472,1066; observed 472,1052. Specific optical rotation Md +110.5 (c 0.519, MeOH) Specific optical rotation parameters: Equipment: Polarimeter: Perkin Elmer, model 341 or equivalent. Cell: Microglass cell, 100 mm path / ength, 5 1.0 mL capacity, Perkin-Elmer Cat. # BOO1-7047. Scale: Calibrated analytical weighing pan ± 0.1 mg Water bath: NESLAB RTE 1121 Chiller or OJ equivalent. Volumetric glassware: Class A. Standard Quartz Number JD 098799, or equivalent. Polarimeter: Perkin Elmer, model 341 or equivalent. Reagents: Methanol: Grade HPLC, Baker (catalog No. 9093-03) or equivalent Instrument parameters: Lamp: Na / Hal, Perkin-Elmer Cat. # BOO0-8754. Cell: Microcell (100mm), Perkin-Elmer Cat. # BO04-1693. Cell path: 100 mm (1 decimeter) Mode: OROT Wavelength: 589 nm Cell temperature: 20 ° C Integration time: 2 seconds Opening: MICRO Water bath temperature: 20 ± rc Alternative preparation of N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (trif] uoromethyl) -4a, 5,7,7a-tetrai & o-4H-nlro [3,4-d] [1,3] thiazin-7a-yl) -4-fluorophenyl) -5- (nuoromethi)) pyrazine-2-carboxamide (Example 8) f fjh, '°]' "'ÁJJ ° j — Á' ,, O ,,,,,, "o'jjj yf, C hf,!" i "s 5- (Fluoromethyl) pyrazine-2-carboxylic acid (32.6 g, 1.05 5 equiv) e (4aS, 5S, 7aS) -7a- (5-amino-2-Ruorophenyl) -5- (tnnuoromethyl) -4a, 5,7,7a- tetrahydro-4H-hro [3,4-d] [1,3] Thiazin-2-amine (70.0 g, 1.0 equiv) 'was loaded into a reactor and ethyl acetate (EtOAc, 630 nL) was added to the mixture to give a suspension. A solution of n-propane phosphonic acid anhydride (T3P, 146 g, 1.10 equiv, 50 ° /) by weight in EtOAc) was added at room temperature while controlling the temperature below 30 ° C. The reaction mixture was stirred at 40-45 ° C> 3 hours and monitored by HPLC. The reaction mixture was run at 15-20 ° C and water (140 niL) was charged. After 10-15 minutes charged 28 ° /, of hydroxide. ammonium (175 mL) while controlling the temperature below 30 ° C. EtOAc 15 (245 niL was added and the reaction mixture was added for 30 minutes at room temperature. The aqueous phase was separated and extracted again with EtOAc (490 ml). The organic phases were combined and washed with 15% aqueous NaCl (140 ml) and water (140 ml). The organic layer was filtered through Celite (1.0 Wt) and rinsed with EtOAc (140 niL). The solution was concentrated in vacuo to obtain a beige solid (quantitative crude yield) which was recrystallized from l-propanol to produce N- (3- ((4aS, 5S, 7aS) -2-mino-5- (tnnuoromethyl ) -4a, 5,7,7a-teÜaidro-4H-mro [3,4- d] [1,3] thiazin-7a-yl) -4-nuoropheni]) - 5- (Huoromethyl) phazine-2-cmboxmide as a white solid (70.0 g). ,,, 'H NMR (500 MHz, DMSO) (j 10.89 (s, 1 H), 9.30 (s, 1 H), 8.89 (s, 1 H), 7.95 (dd, J = 7.3.2.7 Hz, IH), 7.94-7.89 (m, IH), 7.21 (dd, jj = 12.0, 8.8 Hz, IH), 6.22 (S, 2 H), 5.71 (d, J = 46.3 Hz, 2 H), 4.77-4.61 (m, 1Hj), 437 (d, j = 8, lHL1H), 3, 87 (dd, j = 8,0,2,7Hz, lH), 3,20 (dt, j = 5 7,0,3,5 Hz, 1H), 3,15 (dd, j = 13,9, 3.1 Hz, 1H), 2.91 (àd, j = l3.8,3.8Hjz, 1H), "C NMR (126 MHZ, DMSO) δ 161.32 (S), 155.82 (d, J = 243.4 Hz), 153.71 (d, J = 18.7 Hz), 148.77 (S), 144.71 (d, J = 1.9 Hz), 143.30 (S), 141.01 (d, J = 5.6 Hz), 134.36 (d, J = 2.0 Hz), 128.20 (d, J = 12.1 Hz), 125.57 (q, J = 283.0 Hz), 123.12 (d, J = 3.6 Hz), 121.64 (d, J = 8.6 Hz), 116.35 (d, J = 25.2 Hz), 82, 55 (d, J = 165.8 HZ), 78.37 (S), 76.44 (q, J = 30.6 Hz), 65.89 (d, J = 5.3 Hz), 35.89 (S), 23.01 (S). HRMS Calculated by C ,, H ,, F, N, O, S [M + H] "474.1023; observed 474.1032. Specific optical rotation" [a] D20 - +1 02.4 'The preparation of (4aS5S, 7aS) -7a {5-amino-2-fluorophenyl) -5- (trinuoromethyl!) - 4a, 5,7,7a-tetrahydro-4H-hro [3, 4-d] [1, 3] thiuin-2-amine is des described above in step 1- (25) in the alternative preparation of N- (3- ((4aS, 5S, 7aS) -2-amino-5- (trinuoromethyl) -4aÁ7,7a-tetrai & o-4H-nlro [3, 4- d] [1,3] thiazin-7a-yl) -4-fluorophenyl) -5-methoxypyrazine-2-carboxamide (Example 1). In vitro cell assay: Quantification of the A3 peptide in the culture of the neurons of the rat fetus brain (1) Primary neuronal culture of the rat Primary neuronal cultures were prepared from the cerebral cortex of embryonic 18-day Wistar rats (Charles River, UK ). Specifically, the embryos were aseptically removed from the pregnant rats under ether anesthesia. The brain was isolated from the embryo and immersed in HBSS (Sigma Aldrich # H9269) containing 10 niM of HEPES (Gibco # 15630-056). The cerebral cortex was collected from the isolated brain under a stereoscopic microscope. The collected Eagles from the cerebral cortex were enzymatically treated in an enzyme solution containing 0.05 ° /} trypsin-EDTA solution (GIBCO, # 25300) at 5 37 ° C for 20 minutes to disperse the cells. The cells were then washed twice and then gently resuspended in Neurobasal medium (Gibco # 21103) supplemented with 2 ° /) supplement B27 (GIBCO # 17504-044), 0.5 mM L-glutainin (GIBCO # 25030 ),) x N2 (GTBCO # 17502-048), 100 µg / ml Pen / Strep (GIBCO l5140-i22) and 5 °) of 10 heat-inactivated FCS (PAA # A15-70l). The cell dispersion was filtered through a 40-µm nylon web (BD Falcon # 352340) to remove the remaining cell mass and thus a neuronal cell suspension was obtained. The neuronal cell suspension was diluted with the above medium and then placed in a volume of 100 µL / reservoir in a cell density 15 initial of 3.25 x 105 cells / ml in the 96 well culture plate coated with po11-D-lysine (Greiner # 655940). Placed cells were cultured on the culture plate at 37 ° C in 5 ° /) CO2-95 ° /) air for 24 hrs. The total amount of the medium was replaced with 'Neurobasal test medium' (as above excluding heat-inactivated FCS) and then the cells were I, 20 grown for an additional five days. (2) Compound addition The drug was added to the culture plate on day 6 of the culture as follows. 8 points of serial dilutions of the compound were generated in DMSO at a concentration of x 100 of the concentration of the final assay (FAC). Compound solutions were then prepared by adding 999 µl of the 'assay Neurobasal medium' (as described in the section above) to 1 µl of the DMSO compound stock. The total amount of the medium was "removed from each of the cell plate reservoirs and 140µL / reservoir of 'assay Neurobasal medium' was added followed ! 25 by 60 ul of the compound solution. The final DMSO concentration was 0.1 ° /. (3) Sampling The cells were cultured for 1 or 3 days after adding the compound by tests ABx-40 and ABx-42 respectively. ! 50 µ1 of sample medium 5 was collected and used as the ELISA sample. (4) Assessment of cell survival Cell survival was assessed using an AJamar assay according to the following procedure. After sampling of the sample to be used in the ELISA assay, 50 µ1 of 20 ° /) of the Alamar blue solution (Invitrogen 10 # DAL11OO) in the Neurobasai test medium, was added to 50 µ1 of the remaining sample inside each reservoir. The cells were then incubated at 37 ° C in 5 ° 4) of CO, -95 ° 4) for 1 hr. The measurement of fluorescence intensity for each reservoir was performed at 540/590 nm using a Pherastar plate reader 15 plus (BMG labtech). In the measurement, the reservoirs without the cells in place and containing only the Alamar medium and solution were presented as a background (bkg). (5) Af3 ELJSA Human amyloid / Rat B (42) ELISA Kit Wako (# 290- . 20 62601) and Human amyloid / Rat B (40) ELISA Kit Wako (# 294-62501) from Wako Pure Chemical Industries, Ltd. were used by Aj3 ELISA. Aj3 ELISA was performed according to the protocols recommended by the manufacturers, described in the documents accompanying the kits. The results were shown as a percentage of the control groups and the IC50 values 25 for each compound were determined using four logistic parameter adjustment model using the XLFIT5 software package (JDBS). The compounds of the present invention have an effect on reducing Afj42 production. The compound of the general formula (I) or the pharmaceutically acceptable salt thereof according to the present invention has a production reduction effect A | 342. Thus, the present invention can panicularly provide a prophylactic or therapeutic agent for a "neurodegenerative disease caused by Aj3 such as Alzheimer's-type dementia or Down's syndrome. As measured by the in vitro assay above, the compounds of examples 1 to 18 showed IC 50 values of less than 0.1 µM as shown in table 5: Table 5: Example IC, q (uM) Example IC, q (ulld) l! 0.008 II) 0.010 2! 0.004 I 12 I 0.006 3! 0.004 I 13! O, G44 4 i 0,008 I 14: 0,002 5 | 0.012 I 15! 0.007 6! 0.006 I 16 I 0.009 7! 0.008 I 17 I 0.051 8 l 0.006! 18 l 0.015 9 l 0.007 i I 0.010
权利要求:
Claims (14) [1] CLAIMS l. Compound, characterized by the fact that it is of the formula (I): YÂ) / O) "N-J XNH IJ) '"' F, Ê 'or a pharmaceutically acceptable salt thereof, where X is hydrogen or fluorine; AéCHouN; Y is methyl, ethyl, monofluoromethyl, difluoromethyl, trifluoromethyl, difluoroethyl, methoxy, ethoxy, methoxymethyl or -C" N. [2] A compound according to claim 1, characterized by the fact that X is hydrogen, or a pharmaceutically acceptable salt thereof. [3] Compound according to claim 1 or 2, characterized in that A is N or a pharmaceutically acceptable salt thereof. [4] Compound according to any one of claims 1 to 3, characterized in that Y is methyl, monofluoromethyl, difluoromethyl, trifluoromethyl or methoxy or a pharmaceutically acceptable salt! this. [5] 5. Compound according to any of the claims of! to 4, characterized by the fact that the compound is selected from: N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a- teUhydro-4H-Wro [3,4-d1 [I, 3] thiuin-7a-yl) -4-nuorophenyl) -5-metonpyrazine-2-carboxamide; N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (triQuoromethyl) -4a, 5,7,7a tetrai & o-4H-Nro [3,4-d] [1,3] ti = in-7a-yl) -4-nuorophenyl) -5-cimopicolinamide; N- (3 - ((4aS, 5 S, 7aS) -2-aInino-5- (triyl fluoromethyl) -4a, 5,7,7a teUai & o-4H-mro [3,4-d] [1,3] ti ^ -7a-yl) -4-nuorofeml) -5- (difluoromethyl) pyrazine-2-carboxamide; N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (tnfluoromethyl) -4a, 5,7,7a tetrahydro-4H-Nro [3,4-d] [1,3] ti = in-7a-yl) -4-nuorophenij) -5- (triRuoromethyl) picolinamide; N- (3 - ((4aS, 5S, 7aS) -2- = ino-5- (trinuoromethyl) -4a, 5,7,7a- 10 tetrahydro-4H-fiiro [3,4-d] [1,3] tiwin-7a-yl) -4-nuorofeml) -5-methyl] pyrazine-2-carboxamide; N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (tnnuoromethyl) -4a, 5,7,7a tetrahydro-4H-hro [3,4-d] [1,3] thiazine -7a-yl) -4-nuorophenyl) -5-methylpicolinamide; N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a- tetrahydro-4H-mro [3,4-d] [1,3, thiazin-7a-yl) -4-AuorofeMl) -5-ethylpicolin = ida; N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (trinuoromethyl) -4a, 5,7,7a tetrahydro-4H-fur [3,4-d] [1,3] thiazine -7a-il) -4muoropheni]) - 5- (nuoromethyl) pyrazine-2-carboxylic acid; N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (triAuoromethyl) -4a, 5,7,7a- tetrahydro-4HAi] ro [3,4-d] [L3] thiazin-7a-yl) -4-nuorophenyl) -5-methoxypicolinamide; N- (3 - ((4aS, 5 S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tetrahydro-4FLfúro [3,4-djj 1, 3jtiuin-7a-yl) -4-nuoropheni |) -5-ethoxypyrazine-2- carboxamide; N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a- tetrahydro-4HAüro [3,4-d] [1,3] ti = in- 7a-yl) -4-nuorophenyl) -5- (1,1-difluoroethyl) pyrazine-2-carboxamide; N {3 {(4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4H ^ 1 [3,4-d] [1,3] thiazin-7a -il) -4-f1uorophenyl) -5- (trinuoromethyl) pyrazine-2-carboxamide; N- (3 - ((4aS, 5S, 7aS) -2-amino4- (triMorometij) -4a, 5,7,7a- tehydro-4H-Hro [3,4-d] [1,3] ti = in -7a-yl) -4-nuorofeM!) - 5- (methoxymethyl) pyrazine-2-carboxamide; N- {3 - [(4aS, 5S, 7aS) -2-amino-5- (tnnuoromethyl) -4a, 5-di-i & o- 4H-fur [3.4 d] [1,3,] thiazin-7a ( 7H) yl] -4-nuorophenyl} -5 - [('H,) methyl] pyruino-2-carboxamide; N {3 {(4aS, 5S, 7aS) -2-amino-5 {tMuoromethyl) -4a, 5,7,7a- tetrahydro-4HAlro [3,4-d] [L3] tiazin-7a-yl) -4 , 5-dinuorophenyl) -5- (difluoromethyl) pyrazine-2-carboxamide; N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a tetrai & o-4H-hro [3,4-d] [1,3] tiwin- 7a-yl) -4,5-dinuorophenyl) -5-metonpyrazine-2-carboxamide; N {3 {(4aS, 5S, 7aS) -2-amino-5- (tMuorometij) -4a, 5,7,7a- tetrai & o-4H-mro [3,4-d] [1,3] tiwin-7a -yl) -4,5-dinuorophenyl) -5-methylpyrazine-2-carboxamide; N- (3 - ((4aS, 5S, 7aS) -2-amino-5- (trinuoromethi]) - 4a, 5,7,7a tehydro-4H-hro [3,4-d] [1,3] ti & in-7a-yl) -4,5-dinuorophenyl) -5- (Auoromethyl) -pyrazine-2-carboxamide; or a pharmaceutically acceptable salt thereof. [6] 6. Compound according to claim 1, characterized by the fact that it is N- (3 - ((4aS, 5S, 7aS) -2- = ino-5- (ünuoromethyl) -4a, 5,7,7a-tetrahydro 4H-fiiro [3,4-d] [1,3] thiazin-7ayl) 4-fluorophenyl) -5 methoxypyrazine-2-carboxamide, or a pharmaceutically acceptable salt thereof. [7] A compound according to claim 1, characterized by the fact that it is N {3 - ((4aS, 5S, 7aS) -2-amino-5- (trifluoromethyl) -4a, 5,7,7a-tetrahydro-4H -Wro [3,4-d] [1,3] thiain-7a-yl) -4-nuorophenyl) -5- (fluoromethyl) pyrazine-2-carboxamide or a pharmaceutically acceptable salt thereof. · ', And 4 [8] A compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, characterized in that it is for use in therapy. [9] Compound according to any one of claims 5 to 1, or a pharmaceutically acceptable salt thereof, characterized in that it is to inhibit Jocal beta 1 (BACEI) amyloid precursor protein cleavage enzyme. [10] A compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, characterized by the fact that it is for the treatment of a neurodegenerative disease such as dementia of the Alzheimer type (AD), Down syndrome, cerebrovascular amyloid angiopathy (CAA), mild cognitive impairment (MCI), memory loss, pre-senile dementia, senile dementia, hereditary cerebral hemorrhage with amyloidosis and other degenerative dementias such as dementias of vascular origin of mixed degeneratives, such as dementias of vascular origin of mixed degeneratives, dementia associated with basal cortical degeneration, dementia associated with Parkinson's disease (PD) and dementia associated with diffuse Lewy body type AD. [11] 11. Use of a compound as defined in any of the 20 claims 1 to 7, or a pharmaceutically acceptable salt thereof, characterized by the fact that it is for the manufacture of a medicament for the treatment or prevention of a neurodegenerative disease such as dementia Alzheimer's type (AD), Down syndrome, cerebrovascular amyloid angiopathy (CAA), mild cognitive impairment (MCI), memory loss, pre-senile dementia, senile dementia, hereditary cerebral hemorrhage with amyloidosis and other degenerative dementias such as dementias of mixed degenerative vascular origin, such as dementia of origin, mixed degenerative vascular, dementia associated with basal cortical degeneration, dementia associated with Parkinson's disease (PD) and dementia '· T 5 associated with diffuse Lewy type AD type. [12] A compound according to any one of the claims: from 1 to 7, or a pharmaceutically acceptable salt thereof, characterized by the fact that it is for the treatment of type 2 diabetes. [13] 13. Use of a compound as defined in any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, characterized by the fact that it is for the manufacture of a medicament for the treatment or prevention of type 2 diabetes. [14] 14. Pharmaceutical composition, characterized in that 10 comprises the compound as defined in any one of claims 1 to 7, or a pharmaceutically acceptable salt thereof, as an active ingredient in association with a pharmaceutically acceptable carrier. i5. Pharmaceutical product, characterized in that it comprises, in combination, a first active ingredient that is compound 15 as defined in any one of claims 1 to 7 or a pharmaceutically acceptable salt thereof and at least one additional active ingredient useful in the treatment of neurodegenerative disease. ~ r P 1/1 "", Figure 1: A 1- (20) of the Compound chromatogram typical of a Chiral HPLC isolation u # = = o6 ~ S c: cS ~ C> => · Kr Ç'KI = C> Ri = = =: ~ COZ "6P q = = Q ¢ S = = J u5" 8 - = = ¥ = = "m" - = = c · j - e = i - @ cD <> <> <C - <> = to 2 cN to G> Z5 <b, b = ^ <> & ct> =. ,, L.CJ to S <5 "G = <> Z" D Ó = D =: q = Í q mv
类似技术:
公开号 | 公开日 | 专利标题 BR112013018602A2|2020-09-01|compound, use of a compound, pharmaceutical composition, and pharmaceutical product US9175013B2|2015-11-03|Fused aminodihydrothiazine derivatives IL228239A|2016-06-30|3,4-dihydro--pyrrolo[1,2-a]pyrazin-1-ylamine derivatives useful as inhibitors of beta-secretase | JP6491679B2|2019-03-27|Imidazopyridazine derivatives as modulators of GABA A receptor activity MX2007011110A|2008-02-21|Crystal forms of an imidazole derivative. US10442818B2|2019-10-15|Chromone oxime derivative and its use as allosteric modulator of metabotropic glutamate receptors AU2009330131A1|2011-08-11|Compounds and methods for the treatment of pain and other diseases JP2021525272A|2021-09-24|Tetrahydrobenzoflo [2,3-C] pyridine and beta-carboline compounds for the treatment, alleviation, or prevention of disorders associated with tau aggregates WO2014015125A1|2014-01-23|Fused aminodihydrothiazine derivative salts and uses thereof MXPA05001245A|2005-09-12|Novel derivatives of cycloalcanodiones, method for the production thereof and their pharmacological applications. GB2504615A|2014-02-05|Fused aminodihydrothiazine derivatives NZ611365B2|2015-07-28|Fused aminodihydrothiazine derivatives useful as bace inhibitors EP3621620A1|2020-03-18|Substituted heterocyclic compounds as allosteric modulators of group ii metabotropic glutamate receptors
同族专利:
公开号 | 公开日 JP2014502987A|2014-02-06| UA109927C2|2015-10-26| TW201309710A|2013-03-01| NZ611365A|2015-04-24| EP2665732B1|2016-03-30| AR084903A1|2013-07-10| CL2013002083A1|2013-12-06| CA2828738A1|2012-07-26| RU2013138717A|2015-02-27| ES2575914T3|2016-07-04| WO2012098213A1|2012-07-26| US20120190672A1|2012-07-26| US8940734B2|2015-01-27| US8592408B2|2013-11-26| CN103328486A|2013-09-25| ZA201303801B|2014-07-30| US8338407B2|2012-12-25| PE20141182A1|2014-09-14| MX2013007917A|2013-08-29| JP5947815B2|2016-07-06| IL226596A|2016-04-21| KR101906466B1|2018-10-10| US20140142091A1|2014-05-22| AU2012208526A1|2013-05-02| EP2665732A1|2013-11-27| RU2593756C2|2016-08-10| KR20140002724A|2014-01-08| US20130203740A1|2013-08-08| SG190908A1|2013-07-31| CN103328486B|2016-08-17| AU2012208526B2|2015-09-24| GB201101140D0|2011-03-09|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题 US3227713A|1966-01-04|Azine derivatives | US3235551A|1966-02-15|Novel derivatives of | RU2128175C1|1994-08-09|1999-03-27|Эйсай Ко., Лтд.|Condensed pyridazine or its pharmaceutically acceptable salt, an agent showing inhibitory activity with respect to cyclic gmp-phosphodiesterase| JPH0967355A|1995-08-31|1997-03-11|Tokyo Tanabe Co Ltd|Thiazine derivative, thiazole derivative and their production| ES2295068T3|1999-11-24|2008-04-16|MERCK & CO., INC.|GAMMA-HIDROXI-2- -1-PIPERAZINPENTANAMIDS AS INHIBITORS OF HIV PROTEASE.| EP1283039A4|2000-05-19|2007-05-30|Takeda Pharmaceutical|-secretase inhibitors| US6562783B2|2001-05-30|2003-05-13|Neurologic, Inc.|Phosphinylmethyl and phosphorylmethyl succinic and glutauric acid analogs as β-secretase inhibitors| CA2484599A1|2002-05-03|2003-11-13|Israel Institute For Biological Research|Methods and compositions for treatment of central and peripheral nervous system disorders and novel compounds useful therefor| AU2003254844A1|2002-08-09|2004-02-25|Takeda Chemical Industries, Ltd.|Substituted amino compounds and use thereof| WO2004037212A2|2002-10-24|2004-05-06|Sepracor, Inc.|Compositions comprising zopiclone derivatives and methods of making and using the same| JP2004149429A|2002-10-29|2004-05-27|Takeda Chem Ind Ltd|Indole compound and application thereof| JP4494212B2|2002-11-12|2010-06-30|メルク・シャープ・エンド・ドーム・コーポレイション|Phenylcarboxamide beta-secretase inhibitor for the treatment of Alzheimer's disease| RU2405774C9|2003-12-15|2011-03-20|Шеринг Корпорейшн|Heterocyclic aspartyl protease inhibitors| US7700603B2|2003-12-15|2010-04-20|Schering Corporation|Heterocyclic aspartyl protease inhibitors| US7763609B2|2003-12-15|2010-07-27|Schering Corporation|Heterocyclic aspartyl protease inhibitors| JP2007530696A|2004-03-30|2007-11-01|メルク エンド カムパニー インコーポレーテッド|2-Aminothiazole compounds useful as aspartic protease inhibitors| PE20060664A1|2004-09-15|2006-08-04|Novartis Ag|BICYCLE AMIDAS AS KINASE INHIBITORS| US20090221579A1|2004-10-15|2009-09-03|Jeffrey Scott Albert|Substituted Amino-Compounds and Uses Thereof| US20090062282A1|2004-10-15|2009-03-05|Astrazeneca Ab|Substituted Amino-Pyrimidones and Uses Thereof| CA2594946A1|2005-01-19|2006-07-27|Merck & Co., Inc|Tertiary carbinamines having substituted heterocycles, which are active as inhibitors of beta-secretase, for the treatment of alzheimer's disease| US7868000B2|2005-06-14|2011-01-11|Schering Corporation|Aspartyl protease inhibitors| WO2007011810A1|2005-07-18|2007-01-25|Merck & Co., Inc.|Spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease| KR101052122B1|2005-10-25|2011-07-26|시오노기세야쿠 가부시키가이샤|Aminodihydrothiazine derivatives| US20090099217A1|2006-04-05|2009-04-16|Astex Therapeutics Ltd.|2-Aminopyrimidin-4-Ones And Their Use For Treating Or Preventing Alpha Beta-Related Pathologies| EP2025672B1|2006-05-31|2013-09-18|Daiichi Sankyo Company, Limited|Process for producing 1-carbamoyl-3,7-dioxo-1,4-diazepane compounds| JP2009539983A|2006-06-12|2009-11-19|シェーリングコーポレイション|Heterocyclic aspartyl protease inhibitors| AU2007332754A1|2006-12-12|2008-06-19|Schering Corporation|Aspartyl protease inhibitors| MX2009011498A|2007-04-24|2009-11-10|Shionogi & Co|Aminodihydrothiazine derivatives substituted with cyclic groups.| WO2008133273A1|2007-04-24|2008-11-06|Shionogi & Co., Ltd.|Pharmaceutical composition for treatment of alzheimer's disease| PE20091236A1|2007-11-22|2009-09-16|Astrazeneca Ab|PYRIMIDINE DERIVATIVES AS IMMUNOMODULATORS OF TLR7| PL2233474T3|2008-01-18|2015-12-31|Eisai R&D Man Co Ltd|Condensed aminodihydrothiazine derivative| WO2009131975A1|2008-04-22|2009-10-29|Schering Corporation|Phenyl-substituted 2-imino-3-methyl pyrrolo pyrimidinone compounds as bace-1 inhibitors, compositions, and their use| TWI431004B|2008-05-02|2014-03-21|Lilly Co Eli|Bace inhibitors| US8637504B2|2008-06-13|2014-01-28|Shionogi & Co., Ltd.|Sulfur-containing heterocyclic derivative having beta secretase inhibitory activity| CN102105475B|2008-07-28|2014-04-09|卫材R&D管理有限公司|Spiroaminodihydrothiazine derivatives| WO2010013302A1|2008-07-28|2010-02-04|エーザイ・アール・アンド・ディー・マネジメント株式会社|Spiroaminodihydrothiazine derivative| EP2324032B1|2008-08-19|2014-10-01|Vitae Pharmaceuticals, Inc.|Inhibitors of beta-secretase| AR073406A1|2008-09-30|2010-11-03|Eisai R&D Man Co Ltd|AMINODIHYDROTIAZINES FUSED WITH TETRAHYDROPIRANS, INHIBITORS OF BACE1 AND THE PRODUCTION OF ABETA AMILOID, PHARMACEUTICAL COMPOSITIONS CONTAINING THEMSELVES AND USES OF THE SAME FOR THE TREATMENT OF NEURODEGENERATIVE DISEASES, TALES.| EP2801570A1|2009-03-13|2014-11-12|Vitae Pharmaceuticals, Inc.|Inhibitors of beta-secretase| AR077277A1|2009-07-09|2011-08-17|Lilly Co Eli|BICYCLE COMPOUNDS TIAZIN-2-AMINA PHARMACEUTICAL FORMULATION THAT INCLUDES IT AND ITS USE FOR THE MANUFACTURE OF A USEFUL MEDICINAL PRODUCT FOR THE TREATMENT OF ALZHEIMER'S DISEASE| GB0912778D0|2009-07-22|2009-08-26|Eisai London Res Lab Ltd|Fused aminodihydro-oxazine derivatives| GB0912777D0|2009-07-22|2009-08-26|Eisai London Res Lab Ltd|Fused aminodihydropyrimidone derivatives| JP2013530238A|2010-07-01|2013-07-25|アムジエン・インコーポレーテツド|Heterocyclic compounds and their use as inhibitors of PI3K activity| GB201100181D0|2011-01-06|2011-02-23|Eisai Ltd|Fused aminodihydrothiazine derivatives| WO2012100179A1|2011-01-21|2012-07-26|Eisai R&D Management Co., Ltd.|Methods and compounds useful in the synthesis of fused aminodihydrothiazine derivatives| GB201101140D0|2011-01-21|2011-03-09|Eisai Ltd|Fused aminodihydrothiazine derivatives| GB201101139D0|2011-01-21|2011-03-09|Eisai Ltd|Fused aminodihydrothiazine derivatives|KR101052122B1|2005-10-25|2011-07-26|시오노기세야쿠 가부시키가이샤|Aminodihydrothiazine derivatives| MX2009011498A|2007-04-24|2009-11-10|Shionogi & Co|Aminodihydrothiazine derivatives substituted with cyclic groups.| WO2008133273A1|2007-04-24|2008-11-06|Shionogi & Co., Ltd.|Pharmaceutical composition for treatment of alzheimer's disease| PL2233474T3|2008-01-18|2015-12-31|Eisai R&D Man Co Ltd|Condensed aminodihydrothiazine derivative| US8637504B2|2008-06-13|2014-01-28|Shionogi & Co., Ltd.|Sulfur-containing heterocyclic derivative having beta secretase inhibitory activity| EP2360155A4|2008-10-22|2012-06-20|Shionogi & Co|2-aminopyridin-4-one and 2-aminopyridine derivative both having bace1-inhibiting activity| GB0912777D0|2009-07-22|2009-08-26|Eisai London Res Lab Ltd|Fused aminodihydropyrimidone derivatives| GB0912778D0|2009-07-22|2009-08-26|Eisai London Res Lab Ltd|Fused aminodihydro-oxazine derivatives| WO2011044187A1|2009-10-08|2011-04-14|Schering Corporation|Iminothiadiazine dioxide compounds as bace inhibitors, compositions, and their use| WO2011044185A2|2009-10-08|2011-04-14|Schering Corporation|Pentafluorosulfur imino heterocyclic compounds as bace-1 inhibitors, compositions, and their use| US8999980B2|2009-12-11|2015-04-07|Shionogi & Co., Ltd.|Oxazine derivatives| EP2634186A4|2010-10-29|2014-03-26|Shionogi & Co|Naphthyridine derivative| JP5766198B2|2010-10-29|2015-08-19|塩野義製薬株式会社|Condensed aminodihydropyrimidine derivatives| GB201100181D0|2011-01-06|2011-02-23|Eisai Ltd|Fused aminodihydrothiazine derivatives| GB201101139D0|2011-01-21|2011-03-09|Eisai Ltd|Fused aminodihydrothiazine derivatives| GB201101140D0|2011-01-21|2011-03-09|Eisai Ltd|Fused aminodihydrothiazine derivatives| WO2012100179A1|2011-01-21|2012-07-26|Eisai R&D Management Co., Ltd.|Methods and compounds useful in the synthesis of fused aminodihydrothiazine derivatives| CN103608345A|2011-04-26|2014-02-26|盐野义制药株式会社|Oxazine derivative and BACE 1 inhibitor containing same| EP2763676B1|2011-10-03|2019-12-25|The University of Utah Research Foundation|Application of 5-ht6 receptor antagonists for the alleviation of cognitive deficits of down syndrome| WO2014015125A1|2012-07-19|2014-01-23|Eisai R&D Management Co., Ltd.|Fused aminodihydrothiazine derivative salts and uses thereof| GB201212816D0|2012-07-19|2012-09-05|Eisai Ltd|Novel compounds| EP2893042B1|2012-09-05|2018-02-14|Emory University|Diagnostic testing in dementia and methods related thereto| WO2014065434A1|2012-10-24|2014-05-01|Shionogi & Co., Ltd.|Dihydrooxazine or oxazepine derivatives having bace1 inhibitory activity| TWI593692B|2013-03-12|2017-08-01|美國禮來大藥廠|Tetrahydropyrrolothiazine compounds| TWI639607B|2013-06-18|2018-11-01|美國禮來大藥廠|Bace inhibitors| TWI599358B|2014-09-16|2017-09-21|美國禮來大藥廠|Combination therapy| AR103680A1|2015-02-23|2017-05-24|Lilly Co Eli|BACE1 SELECTIVE INHIBITORS| CN107257795A|2015-03-19|2017-10-17|伊莱利利公司|Selective BACE1 inhibitor| AR104241A1|2015-04-29|2017-07-05|Lilly Co Eli|THIAZINE TETRAHYDROFIDE DERIVATIVES AS SELECTIVE BACE1 INHIBITORS| KR20170108203A|2016-03-16|2017-09-27|주식회사 피플바이오|Method for Detecting Aggregate Form of Aggregate-Forming Polypeptides| TW201740954A|2016-03-16|2017-12-01|美國禮來大藥廠|Combination therapy| TWI675034B|2016-05-20|2019-10-21|美商美國禮來大藥廠|Tetrahydrofurooxazine compounds and their use as selective bace1 inhibitors| WO2018034977A1|2016-08-18|2018-02-22|Eli Lilly And Company|Combination therapy of bace-1 inhibitor and anti-n3pglu abeta antibody| AR110470A1|2016-10-21|2019-04-03|Lilly Co Eli|COMBINATION THERAPY TO TREAT ALZHEIMER'S DISEASE| US10889579B2|2016-12-15|2021-01-12|Amgen Inc.|Thiazine derivatives as β-secretase inhibitors and methods of use| WO2018112084A1|2016-12-15|2018-06-21|Amgen Inc.|Bicyclic thiazine and oxazine derivatives as beta-secretase inhibitors and methods of use| US11021493B2|2016-12-15|2021-06-01|Amgen Inc.|1,4-thiazine dioxide and 1,2,4-thiadiazine dioxide derivatives as beta-secretase inhibitors and methods of use| WO2018112086A1|2016-12-15|2018-06-21|Amgen Inc.|Cyclopropyl fused thiazine derivatives as beta-secretase inhibitors and methods of use| EP3555080A1|2016-12-15|2019-10-23|Amgen Inc.|Oxazine derivatives as beta-secretase inhibitors and methods of use|
法律状态:
2020-09-15| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]| 2020-09-15| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]|Free format text: DE ACORDO COM O ARTIGO 229-C DA LEI NO 10196/2001, QUE MODIFICOU A LEI NO 9279/96, A CONCESSAO DA PATENTE ESTA CONDICIONADA A ANUENCIA PREVIA DA ANVISA. CONSIDERANDO A APROVACAO DOS TERMOS DO PARECER NO 337/PGF/EA/2010, BEM COMO A PORTARIA INTERMINISTERIAL NO 1065 DE 24/05/2012, ENCAMINHA-SE O PRESENTE PEDIDO PARA AS PROVIDENCIAS CABIVEIS. | 2020-10-20| B08F| Application dismissed because of non-payment of annual fees [chapter 8.6 patent gazette]|Free format text: REFERENTE A 8A ANUIDADE. | 2021-02-09| B08K| Patent lapsed as no evidence of payment of the annual fee has been furnished to inpi [chapter 8.11 patent gazette]|Free format text: EM VIRTUDE DO ARQUIVAMENTO PUBLICADO NA RPI 2598 DE 20-10-2020 E CONSIDERANDO AUSENCIA DE MANIFESTACAO DENTRO DOS PRAZOS LEGAIS, INFORMO QUE CABE SER MANTIDO O ARQUIVAMENTO DO PEDIDO DE PATENTE, CONFORME O DISPOSTO NO ARTIGO 12, DA RESOLUCAO 113/2013. | 2021-11-23| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 GB1101140.0|2011-01-21| GBGB1101140.0A|GB201101140D0|2011-01-21|2011-01-21|Fused aminodihydrothiazine derivatives| PCT/EP2012/050833|WO2012098213A1|2011-01-21|2012-01-20|Fused aminodihydrothiazine derivatives useful as bace inhibitors| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|