WO2019142111A1 - Salt forms of an oxazine derivative bace inhibitor - Google Patents

Salt forms of an oxazine derivative bace inhibitor Download PDF

Info

Publication number
WO2019142111A1
WO2019142111A1 PCT/IB2019/050339 IB2019050339W WO2019142111A1 WO 2019142111 A1 WO2019142111 A1 WO 2019142111A1 IB 2019050339 W IB2019050339 W IB 2019050339W WO 2019142111 A1 WO2019142111 A1 WO 2019142111A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
salt form
compound according
diffraction pattern
hemihydrate
Prior art date
Application number
PCT/IB2019/050339
Other languages
French (fr)
Inventor
Zhenxing DU
Arnaud Grandeury
Siyi Jiang
Rita Ramos
Yanxiang WU
Irene XIA
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Publication of WO2019142111A1 publication Critical patent/WO2019142111A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to salt forms, specifically the fumarate, succinate and tartrate salt forms, of the BACE inhibitor N-(6-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3, 6- dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide, and the advantageous properties associated therewith.
  • AD Alzheimer’s disease
  • pharmacological therapies available are symptomatic drugs such as cholinesterase inhibitors or other drugs used to control the secondary behavioral symptoms of AD.
  • Investigational treatments targeting the AD pathogenic cascade include those intended to interfere with the production, accumulation, or toxic sequelae of amyloid-b (Ab) species (Kramp VP, Herrling P, 2011).
  • Cerebral amyloid angiopathy is a common age related cerebral small vessel disease, characterised by progressive deposition of Ab, in particular Ab40, in the wall of small to medium sized arteries, arterioles and capillaries of the cerebral cortex and overlying leptomeninges (Charidimou A et ai, 2011).
  • CAA Cerebral amyloid angiopathy
  • Mild forms of CAA often appear asymptomatic; however, CAA may also lead to severe vascular pathologies and is a risk factor for cerebral hemorrhages ranging from silent microbleeds to spontaneous intracerebral haemorrhage, a devastating form of stroke.
  • ApoE4 is a strong genetic risk factor for both AD and CAA (Shinohara M et al, 2016).
  • Human APOE is located on chromosome 19 (gene APOE, Uniprot P02649). Three major isoforms (ApoE2, -3 and -4) are known in humans. ApoE4 (with Arg at positions 112 and 158) has an allele frequency of 5-35% in humans (Verghese PB et ai, 2011) and ApoE4 homozygotes are estimated to represent about 2 to 3% of the general population (Quintino- Santos SR et ai, 2012).
  • Beta-site-APP cleaving enzyme-1 an enzyme involved in the processing of the amyloid precursor protein [APP]
  • BACE-1 an enzyme involved in the processing of the amyloid precursor protein [APP]
  • Compound 1 N-(6-((3R,6R)-5-Amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5- fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide, referred to herein as“Compound 1”, is an orally active BACE inhibitor, previously described in WO 2012/095469 A1.
  • a pharmaceutical composition comprising the fumarate, succinate and tartrate salt forms of Compound 1.
  • Compound 1 in fumarate, succinate or tartrate salt form for use in the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • a method for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy which method comprises administering to a patient in need thereof a therapeutically effective amount of Compound 1 in fumarate, succinate or tartrate salt form.
  • a sixth aspect of the invention there is provided the use of Compound 1 in fumarate, succinate or tartrate salt form for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • a seventh aspect of the invention there is provided the use of Compound 1 in fumarate, succinate or tartrate salt form, for the manufacture of a medicament for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • Figure 1 shows shows the X-ray powder diffraction pattern for crystalline free base
  • Figure 2 shows the X-ray powder diffraction pattern for micronized crystalline free base Compound 1 Form A when measured using CuK ⁇ radiation.
  • Figure 3 shows the DSC thermogram for crystalline free base Compound 1 Form A.
  • Figure 4 shows the X-ray powder diffraction pattern for crystalline Compound 1 in hemifumarate hemihydrate salt form when measured using CuK ⁇ radiation.
  • Figure 5 shows the X-ray powder diffraction pattern for micronized crystalline Compound 1 in hemifumarate hemihydrate salt form when measured using CuK ⁇ radiation.
  • Figure 6 shows the DSC thermogram for crystalline Compound 1 in hemifumarate hemihydrate salt form.
  • Figure 7 shows the X-ray powder diffraction pattern for crystalline Compound 1 in hemisuccinate hemihydrate salt form when measured using CuK ⁇ radiation.
  • Figure 8 shows the X-ray powder diffraction pattern for micronized crystalline Compound 1 in hemisuccinate hemihydrate salt form when measured using CuK ⁇ radiation.
  • Figure 9 shows the DSC thermogram for crystalline Compound 1 in hemisuccinate hemihydrate salt form.
  • Figure 10 shows the X-ray powder diffraction pattern for crystalline Compound 1 in hemi-L- tartrate hemihydrate salt form when measured using CuK ⁇ radiation.
  • Figure 11 shows the X-ray powder diffraction pattern for micronized crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form when measured using CuK ⁇ radiation.
  • Figure 12 shows the DSC thermogram for crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form.
  • Figure 13 shows the design of a two part, open-label, two-period, fixed-sequence study in healthy subjects to evaluate the PK of Compound 1 when given alone and in combination with the strong CYP3A4 inhibitor itraconazole or the strong CYP3A4 inducer rifampicin.
  • Embodiment A1 Compound 1 in fumarate, succinate or tartrate salt form.
  • Embodiment A2 Compound 1 in fumarate salt form.
  • Embodiment A3 Compound 1 according to Embodiment A2 in hemifumarate salt form.
  • Embodiment A4 Compound 1 in succinate salt form.
  • Embodiment A5 Compound 1 according to Embodiment A4 in hemisuccinate salt form.
  • Embodiment A6 Compound 1 in tartrate salt form.
  • Embodiment A7 Compound 1 according to Embodiment A6 in L-tartrate salt form.
  • Embodiment A8 Compound 1 according to Embodiment A6 in hemitartrate salt form or A7 in hemi-L-tartrate salt form.
  • Embodiment A9 Compound 1 according to any one of Embodiments A1 to A8 in hydrated salt form.
  • Embodiment A10 Compound 1 according to Embodiment A9 in hemihydrate or
  • Embodiment A11 Compound 1 according to Embodiment A10 in hemihydrate salt form.
  • Embodiment A12 Compound 1 in hemifumarate hemihydrate salt form.
  • Embodiment A13 Compound 1 in hemisuccinate hemihydrate salt form.
  • Embodiment A14 Compound 1 in hemitartrate hemihydrate salt form.
  • Embodiment A15 Compound 1 in hemi-L-tartrate hemihydrate salt form.
  • Embodiment A16 Compound 1 according to any one of Embodiments A1 to A15 in crystalline form.
  • Embodiment A17 Compound 1 according to Embodiment A16 in substantially pure form. Detailed Compound 1 fumarate salt embodiments
  • Embodiment A18 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 11.9, 13.9 and 19.9° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A19 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 23.0, 27.9 and 30.8° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A20 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 11.9, 13.9, 16.2, 17.3 and 19.9° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A21 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 11.9, 19.9, 23.0, 27.9 and 30.8° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A22 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 4.0, 11.9, 13.9, 15.6, 16.2, 17.3, 19.9, 23.0, 27.9 and 30.8° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A23 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 4 when measured using CuK ⁇ radiation.
  • Embodiment A24 Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 14.0, 16.1 and 19.0° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A25 Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 9.1 , 11.8, 14.0, 16.1 and 19.0° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A26 Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 3.9, 7.3, 9.1 , 11.8, 14.0, 16.1 , 17.1 , 19.0, 19.7 and 22.3° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A27 Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 5 when measured using CuK ⁇ radiation.
  • Embodiment A28 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an endotherm at about 239 °C when analysed by DSC at a heating rate of 10 °C per minute.
  • Embodiment A29 Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having a DSC thermogram substantially the same as that shown in Figure 6.
  • Embodiment A30 Compound 1 according to any one of Embodiments A18 to A29 wherein Compound 1 is in hemifumarate salt form.
  • Embodiment A31 Compound 1 according to any one of Embodiments A18 to A30 wherein Compound 1 is in hydrated salt form.
  • Embodiment A32 Compound 1 according to any one of Embodiments A18 to A31 wherein Compound 1 is in hemihydrate salt form.
  • Embodiment A33 Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 12.0, 14.0 and 20.1° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A34 Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 12.0, 14.0, 17.3, 17.6 and 20.1° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A35 Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 4.0, 12.0, 14.0, 15.7, 16.2, 17.3, 17.6, 20.1 , 23.3 and 31.2° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A36 Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 7 when measured using CuK ⁇ radiation.
  • Embodiment A37 Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 10.1 , 15.5 and 19.8° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A38 Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 10.1 , 14.5, 15.5, 19.8 and 23.3° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A39 Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 9.2, 10.1 , 11.9, 12.7, 14.5, 15.5, 19.8, 22.0, 23.3 and 24.5° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A40 Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 8 when measured using CuK ⁇ radiation.
  • Embodiment A41 Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an endotherm at about 217 °C when analysed by DSC at a heating rate of 10 °C per minute.
  • Embodiment A42 Compound 1 according to Embodiment A4 in crystalline succinate salt form and having a DSC thermogram substantially the same as that shown in Figure 9.
  • Embodiment A43 Compound 1 according to any one of Embodiments A33 to A42 wherein Compound 1 is in hemisuccinate salt form.
  • Embodiment A44 Compound 1 according to any one of Embodiments A33 to A43 wherein Compound 1 is in hydrated salt form.
  • Embodiment A45 Compound 1 according to any one of Embodiments A33 to A44 wherein Compound 1 is in hemihydrate salt form. Detailed Compound 1 tartrate salt embodiments
  • Embodiment A46 Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 14.0, 17.7 and 19.8° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A47 Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 12.2, 14.0, 17.7, 19.8 and 20.4° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A48 Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 12.2, 14.0, 16.0, 17.1 , 17.3, 17.7, 19.8, 20.4, 23.5 and 31.5° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A49 Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 10 when measured using CuK ⁇ radiation.
  • Embodiment A50 Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 13.0, 17.1 and 20.3° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A51 Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 7.1 , 13.0, 17.1 , 19.8 and 20.3° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A52 Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta ( ⁇ ) peak values of 7.1 , 13.0, 13.7, 13.9, 15.9, 17.1 , 19.8, 20.3, 22.4 and 24.7° when measured using CuK ⁇ radiation, wherein said values are plus or minus 0.2° 2 ⁇ .
  • Embodiment A53 Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 11 when measured using CuK ⁇ radiation.
  • Embodiment A54 Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an endotherm at about 253 °C when analysed by DSC at a heating rate of 10 °C per minute.
  • Embodiment A55 Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having a DSC thermogram substantially the same as that shown in Figure 12.
  • Embodiment A56 Compound 1 according to any one of Embodiments A46 to A55 wherein Compound 1 is in hemitartrate salt form.
  • Embodiment A57 Compound 1 according to any one of Embodiments A46 to A56 wherein Compound 1 is in hemi-L-tartrate salt form.
  • Embodiment A58 Compound 1 according to any one of Embodiments A46 to A57 wherein Compound 1 is in hydrated salt form.
  • Embodiment A59 Compound 1 according to any one of Embodiments A46 to A58 wherein Compound 1 is in hemihydrate salt form.
  • Embodiments of the Second Aspect of the Invention Embodiment B1 A pharmaceutical composition comprising Compound 1 according to any one of Embodiments A1 to A59.
  • Embodiment B2 The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 1 to 100 mg of the base of Compound 1.
  • Embodiment B3 The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 1 to 75 mg of the base of Compound 1.
  • Embodiment B4 The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 1 , 10, 15, 25, 50 or 75 mg of the base of Compound 1.
  • Embodiment B5 The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 15 mg of the base of Compound 1.
  • Embodiment B6 The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 50 mg of the base of Compound 1.
  • Embodiment B7 The pharmaceutical composition according to any one of Embodiments B1 to B6 wherein the pharmaceutical composition comprises a gelatin capsule.
  • Embodiment B8 The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises magnesium stearate, sodium stearyl fumarate, or Aerosil 200 PH.
  • Embodiment B9 The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises magnesium stearate.
  • Embodiment B10 The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises sodium stearyl fumarate.
  • Embodiment B11 The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises Aerosil 200 PH.
  • Embodiment C1 Compound 1 according to any one of Embodiments A1 to A59 for use as a medicament.
  • Embodiment C2 Compound 1 according to any one of Embodiments A1 to A59 for use in the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • Embodiment C3 Compound 1 according to any one of Embodiments A1 to A59 for use in the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • Embodiment C4 Compound 1 for the use according to Embodiment C2 or C3 in a patient carrying one or two copies of the ApoE4 allele.
  • Embodiment C5 Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of between 10 and 30 mg of the base of Compound 1 per day.
  • Embodiment C6 Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of between 30 and 100 mg of the base of Compound 1 per day.
  • Embodiment C7 Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of between 30 and 50 mg of the base of Compound 1 per day.
  • Embodiment C8 Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of 15 mg of the base of Compound 1 per day.
  • Embodiment C9 Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of 50 mg of the base of Compound 1 per day.
  • Embodiment D1 A method for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy which method comprises administering to a patient in need thereof a therapeutically effective amount of Compound 1 according to any one of
  • Embodiments A1 to A59 are identical to Embodiments A1 to A59.
  • Embodiment D2 A method for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy which method comprises administering to a patient in need thereof a therapeutically effective amount of Compound 1 according to any one of Embodiments A1 to A59.
  • Embodiment D3 The method according to Embodiment D1 or D2 in a patient carrying one or two copies of the ApoE4 allele.
  • Embodiment D4 The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of between 10 and 30 mg per day.
  • Embodiment D5 The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of between 30 and 100 mg per day.
  • Embodiment D6 The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of between 30 and 50 mg per day.
  • Embodiment D7 The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of 15 mg per day.
  • Embodiment D8 The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of 50 mg per day.
  • Embodiment E1 Use of Compound 1 according to any one of Embodiments A1 to A59 for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • Embodiment E2 The use of Compound 1 according to Embodiment E1 for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • Embodiment E3 The use of Compound 1 according to Embodiment E1 or E2 in a patient carrying one or two copies of the ApoE4 allele.
  • Embodiment E4 The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of between 10 and 30 mg of the base of Compound 1 per day.
  • Embodiment E5 The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of between 30 and 100 mg of the base of Compound 1 per day.
  • Embodiment E6 The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of between 30 and 50 mg of the base of Compound 1 per day.
  • Embodiment E7 The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of 15 mg of the base of Compound 1 per day.
  • Embodiment E8 The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of 50 mg of the base of Compound 1 per day.
  • Embodiment F1 Use of Compound 1 according to any one of Embodiments A1 to A59, for the manufacture of a medicament for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • Embodiment F2 The use of Compound 1 according to Embodiment F1 for the manufacture of a medicament for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
  • Embodiment F3 The use of Compound 1 according to Embodiment F1 or F2 for the manufacture of a medicament for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy in a patient carrying one or two copies of the ApoE4 allele.
  • Embodiment F4 The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of between 10 and 30 mg of the base of Compound 1 per day.
  • Embodiment F5 The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of between 30 and 100 mg of the base of Compound 1 per day.
  • Embodiment F6 The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of between 30 and 50 mg of the base of Compound 1 per day.
  • Embodiment F7 The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of 15 mg of the base of Compound 1 per day.
  • Embodiment F8 The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of 50 mg of the base of Compound 1 per day.
  • the terms“Compound 1”,“Cmpd 1”,“the base of Compound 1”,“the free base of Compound 1” or“the free form of Compound 1” refer to N-(6-((3R,6R)-5-amino-3,6- dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5- (trifluoromethyl)picolinamide and having the following structural formula:
  • Example 1 using an alternative chemical naming format,“Compound 1” is also referred to as 3-chloro-5-trifluoromethyl-pyridine-2-carboxylic acid [6-((3R,6R)-5-amino-3,6-dimethyl-6- trifluoromethyl-3,6-dihydro-2H-[1 ,4]oxazin-3-yl)-5-fluoro-pyridin-2-yl]-amide.
  • Compound 1 “the free form of Compound 1” and its corresponding full chemical name are used interchangeably throughout the description of the invention.
  • Compound 1 is described in WO 2012/095469 A1 , Example 34.
  • WO 2012/095469 A1 is incorporated herewith by reference in its entirety, in particular the disclosure related to the synthesis of Example 34.
  • the term“salt” includes both hydrated and anhydrous salt forms and all stoichiometric ratios of the base of Compound 1 to its salt counterion or to molecules of water if a hydrated salt form.
  • Compound 1 refers to a salt having a stoichiometric ratio of two molecules of Compound 1 to one molecule of fumarate, succinate, or tartrate counterion, respectively.
  • the terms “hemifumarate”,“hemisuccinate”, or“hemitartrate” salt of Compound 1 include hydrated salt forms, for example, hemifumarate hemihydrate, hemisuccinate hemihydrate and hemitartrate hemihydrate salt forms.
  • the term“hemihydrate” refers to a hydrated form of a Compound 1 salt having a stoichiometric ratio of two molecules of Compound 1 to one molecule of water.
  • the hemifumarate, hemisuccinate and hemitartrate salts of Compound 1 may be
  • the hemifumarate hemihydrate, hemisuccinate hemihydrate and hemitartrate hemihydrate salts of Compound 1 may be represented as shown below.
  • hemifumarate hemihydrate, hemisuccinate hemihydrate, and hemi-L-tartrate hemihydrate salts of Compound 1 are referred to herein as:
  • salts may also be referred to as: (2E)-But-2-enedioic acid— N- ⁇ 6-[(3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6- dihydro-2H-1 ,4-oxazin-3-yl]-5-fluoropyridin-2-yl ⁇ -3-chloro-5-(trifluoromethyl)pyridine-2- carboxamide— water (1/2/1); Butanedioic acid— N- ⁇ 6-[(3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-
  • the term“tartrate salt” includes salts of Compound 1 derived from all possible stereoisomers of tartaric acid, and encompasses: 2S,3S-tartaric acid (D-tartaric acid); 2R,3R-tartaric acid (L-tartaric acid); 2 S,3R- tartaric acid tartaric acid (Meso-tartaric acid); racemic mixtures of 2S,3S-tartaric acid and 2R,3R-tartaric acid; and mixtures of 2S,3S-tartaric acid, 2R,3R-tartaric acid and 2 S,3R- tartaric acid.
  • the tartrate salt of Compound 1 is 2S,3S-tartaric acid.
  • the tartrate salt of Compound 1 is 2R,3R-tartaric acid.
  • 1.4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide means having a purity greater than 90 weight %, including greater than 90, 91 , 92, 93, 94, 95, 96, 97, 98, and 99 weight %, and also including equal to about 100 weight % of N-(6-((3R,6R)-5-amino- 3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro- 5-(trifluoromethyl)picolinamide, based on the weight of the compound in its salt form.
  • the term“Aerosil 200 PH” refers to AEROSIL ® 200 Pharma grade, a high purity amorphous anhydrous colloidal silicon dioxide used as a pharmaceutical excipient.
  • the peak positions (2Q) will show some inter-apparatus variability, typically as much as 0.2°.
  • X-ray powder diffraction analysis is typically carried out at room temperature, i.e. between 20 and 30 °C.
  • relative peak intensities will show inter-apparatus variability as well as variability due to degree of crystallinity, preferred orientation, prepared sample surface, and other factors known to those skilled in the art, and should be taken as a qualitative measure only.
  • an X-ray diffraction pattern may be obtained with a measurement error that is dependent upon the measurement conditions employed.
  • intensities in an X-ray diffraction pattern may fluctuate depending upon measurement conditions employed. It should be further understood that relative intensities may also vary depending upon experimental conditions and, accordingly, the exact order of intensity should not be taken into account. Additionally, a measurement error of diffraction angle for a conventional X-ray diffraction pattern is typically about 5% or less, and such degree of measurement error should be taken into account as pertaining to the aforementioned diffraction angles.
  • the crystal form of the instant invention is not limited to the crystal form that provides an X-ray diffraction pattern completely identical to the X-ray diffraction patterns depicted in the accompanying Figures 4, 5, 7, 8, 10 and 11 disclosed herein. Any crystal forms that provide X- ray diffraction patterns substantially identical to that disclosed in the accompanying Figures 4, 5, 7, 8, 10 and 11 fall within the scope of the present invention. The ability to ascertain substantial identities of X-ray diffraction patterns is within the purview of one of ordinary skill in the art.
  • An expression referring to a crystalline form of Compound 1 having“an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure X” may be interchanged with an expression referring to a crystalline form of Compound 1 having“an X- ray powder diffraction pattern characterised by the representative X-ray powder diffraction pattern shown in Figure X”.
  • the term“about X °C”, in the context of the endotherm temperature as measured by DSC analysis, means within a range of +/- 5 °C of temperature X, more particularly within a range of +/- 2.5 °C of temperature X.
  • Alzheimer’s disease or“AD” encompasses both preclinical and clinical Alzheimer’s disease unless the context makes clear that either only preclinical Alzheimer’s disease or only clinical Alzheimer’s disease is intended.
  • treatment of Alzheimer’s disease refers to the administration of Compound 1 of the present invention to a patient in order to ameliorate at least one of the symptoms of Alzheimer’s disease.
  • the term“prevention of Alzheimer’s disease” refers to the prophylactic treatment of AD; or delaying the onset or progression of AD. For example, the onset or progression of AD is delayed for at least 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 years.
  • “prevention of Alzheimer’s disease” refers to the prophylactic treatment of preclinical AD; or delaying the onset or progression of preclinical AD.
  • the onset or progression of preclinical AD is delayed for at least 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 years.
  • “prevention of Alzheimer’s disease” refers to the prophylactic treatment of clinical AD; or delaying the onset or progression of clinical
  • the onset or progression of clinical AD is delayed for at least
  • MCI Mild Cognitive Impairment
  • EMA European Medicines Agency
  • Diagnosis of MCI due to AD requires evidence of intra-individual decline, manifested by: a) A change in cognition from previously attained levels, as noted by self- or informant report and/or the judgment of a clinician.
  • Impairment in two or more cognitive domains although an amnestic presentation is most common, the criteria allow for diagnosis based on nonamnestic presentations (e.g. impairment in executive function and visuospatial abilities).
  • Impairment in two or more cognitive domains e.g. impairment in executive function and visuospatial abilities.
  • Absence of prominent features associated with other dementing disorders e.g. impairment in executive function and visuospatial abilities.
  • preclinical Alzheimer’s disease or“preclinical AD” refers to the presence of in vivo molecular biomarkers of AD in the absence of clinical symptoms.
  • the National Institute on Aging and Alzheimer’s Association provide a scheme, shown in Table A below, which sets out the different stages of preclinical AD (Sperling et al., 2011).
  • CAA structural magnetic resonance imaging
  • b-amyloid (Ab) proteins in the walls of cortical and leptomeningeal blood vessels.
  • Ab b-amyloid
  • CAA is a common cause of vessel wall breakdown and vascular dysfunction in older adults, making it a major contributor to fatal or disabling intracerebral hemorrhages (ICH) as well as ischemic injury and dementia (Gurol ME et al., 2016).
  • ICH intracerebral hemorrhages
  • ischemic injury and dementia ischemic injury and dementia
  • CAA-Type 1 and CAA-Type 2 unless the context makes clear that only CAA-Type 1 or CAA-Type 2 is intended.
  • CAA-Type 1 refers to capillary CAA (capCAA) characterised by Ab protein depositions in the walls of cortical capillaries (Thai et al., 2002).
  • CAA-Type 2 refers to CAA characterised by Ab protein depositions in the walls of leptomeningeal and cortical vessels, with the exception of cortical capillaries (Thai et al, 2002).
  • the term“treatment of CAA” refers to the administration of Compound 1 of the present invention to a patient in order to slow or arrest the development of CAA or at least one of the clinical symptoms of CAA, for example ICH, ischemic injury, or dementia.
  • the development of CAA may be assessed by measuring the accumulation of Ab in the walls of cortical (for example occipital cortex) and leptomeningeal blood vessels using a Positron Emission Tomography (PET) tracer, for example 18 F-florbetapir (Gurol ME et ai, 2016).
  • PET Positron Emission Tomography
  • CAA cerebral microbleeds
  • CMB cerebral microbleeds
  • MRI magnetic resonance imaging
  • SWI susceptibility-weighted imaging
  • GRE MRI T2*-weighted gradient-recalled echo imaging
  • WH white matter hyperintensities
  • CAA development may be monitored by the measurement of WMH volume using MRI (Chen YW et ai, 2006). It is expected that the “treatment of CAA” will have the resultant benefit of reducing the likelihood of a cerebral ischemic event in the patient undergoing treatment for CAA. Therefore, in one embodiment of the invention, the term“treatment of CAA” is equivalent to the term“treatment of intracerebral haemorrhage”. In another embodiment of the invention, the term“treatment of CAA” is equivalent to the term“treatment of CAA and/or intracerebral haemorrhage”. In a further embodiment of the invention, the term“treatment of CAA” is equivalent to the term “treatment of CAA and intracerebral haemorrhage associated therewith”.
  • the term“prevention of CAA” refers to the prophylactic treatment of CAA; delaying the onset or progression of CAA; or delaying the onset or progression of at least one of the clinical symptoms of CAA. For example, the onset or progression of CAA is delayed for at least 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 years.
  • the term“prevention of CAA” is equivalent to the term“prevention of intracerebral haemorrhage”.
  • the term“prevention of CAA” is equivalent to the term“prevention of CAA and/or intracerebral haemorrhage”.
  • the term“prevention of CAA” is equivalent to the term “prevention of CAA and intracerebral haemorrhage associated therewith”.
  • a genetic predisposition for the development of CAA includes, but is not limited to situations where the genetic predisposition is due to: Down’s syndrome; a mutation in the gene for amyloid precursor protein or presenilin-1 ; or the presence of one or two copies of the ApoE4 allele.
  • the term“patient” refers to a human subject.
  • a“pharmaceutical composition” comprises Compound 1 of the present invention and at least one pharmaceutically acceptable carrier or excipient, in a unit dose solid form suitable for oral administration (typically a capsule, more particularly a hard gelatin capsule).
  • a pharmaceutically acceptable carrier or excipient typically a capsule, more particularly a hard gelatin capsule.
  • a therapeutically effective amount refers to an amount of Compound 1 of the present invention that will elicit inhibition of BACE-1 in a patient as evidenced by a reduction in CSF or plasma Ab 1-40 levels relative to an initial baseline value.
  • Ab 1-40 levels may be measured using standard immunoassay techniques, for example Meso Scale Discovery (MSD) 96-well MULTI-ARRAY human/rodent (4G8) Ab40 Ultrasensitive Assay (#K110FTE- 3, Meso Scale Discovery, Gaithersburg, USA).
  • Example 1 shows how Compound 1 may be prepared and crystallised.
  • Example 2 describes the XRPD analysis of Compound 1 free Form A.
  • Example 3 describes the XRPD analysis of micronized Compound 1 in free base form.
  • Example 4 describes the DSC analysis of Compound 1 free Form A.
  • Example 5 describes the preparation of Compound 1 in hemifumarate hemihydrate salt form.
  • Example 6 describes the XRPD analysis of crystalline Compound 1 in hemifumarate hemihydrate salt form.
  • Example 7 describes the XRPD analysis of micronized Compound 1 in hemifumarate hemihydrate salt form.
  • Example 8 describes the DSC analysis of Compound 1 in hemifumarate hemihydrate salt form.
  • Example 9 describes the preparation of Compound 1 in hemisuccinate hemihydrate salt form.
  • Example 10 describes the XRPD analysis of crystalline Compound 1 in hemisuccinate hemihydrate salt form.
  • Example 11 describes the XRPD analysis of micronized Compound 1 in hemisuccinate hemihydrate salt form.
  • Example 12 describes the DSC analysis of Compound 1 in hemisuccinate hemihydrate salt form.
  • Example 13 describes the preparation of Compound 1 in hemi-L-tartrate hemihydrate salt form.
  • Example 14 describes the XRPD analysis of crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form.
  • Example 15 describes the XRPD analysis of micronized Compound 1 in hemi-L-tartrate hemihydrate salt form.
  • Example 16 describes the DSC analysis of Compound 1 in hemi-L-tartrate hemihydrate salt form.
  • Example 17 describes the stoichiometric analysis of Compound 1 in fumarate, succinate and tartrate salt form.
  • Example 18 compares the milling behavior of free base Compound 1 with the hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salt forms.
  • Example 19 describes the preparation of further alternative salt forms of Compound 1.
  • Example 20 compares the bulk stability of Compound 1 in free form with alternative salt forms and in the presence of sodium stearyl fumarate.
  • Example 21 compares the bulk stability of Compound 1 in free form with the hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salt forms in the presence of various pharmaceutical excipients.
  • Example 22 describes an in human study to assess free base Compound 1 PK when given administered in combination with a strong CYP3A4 inhibitor or inducer. Detailed Examples
  • Compound 1 may also be prepared as described below.
  • HPLC-eluent A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
  • HPLC-eluent A) water + 0.05 Vol.-% TFA, B) ACN + 0.05 Vol.-% TFA
  • HPLC-eluent A) water + 0.05 Vol.-% formic acid + 3.75 mM ammonium
  • HPLC-eluent A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
  • HPLC-column type Zorbax SB-C18, 1.8 ⁇ m
  • HPLC-eluent A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
  • HPLC-eluent A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
  • N,N- dimethylacetamide (21.87 g, 250 mmol) was added quickly, the reaction temperature rose to -57 °C.
  • the reaction mixture was stirred in a dry ice bath for 15 min and then allowed to warm to -40 °C. It was poured on a mixture of 2M aq. HCI (250 ml, 500 mmol), 250 ml water and 100 ml brine. The mixture was extracted with TBME, washed with brine, dried over MgSO 4 .H 2 O, filtered and evaporated to give a yellow oil which was purified on a silica gel column by eluting with hexane/0-5% TBME to yield 58.5 g of the title compound as a yellow liquid.
  • the catalyst solution was prepared by dissolving water (54 mg, 3.00 mmol) in 100 ml dry DCM ( ⁇ 0.001 % water). This wet DCM (44 ml, 1.32 mmol water content) was added to a well stirred solution of titanium(IV) butoxide (500 mg, 1.47 mmol) in 20 ml dry DCM. The resulting clear solution was refluxed for 1 h. This solution was then cooled to rt and 2,4-di- tert-butyl-6- ⁇ [(E)-(S)-1-hydroxymethyl-2-methyl-propylimino]-methyl ⁇ -phenol [CAS 155052- SI -6] (469 mg, 1.47 mmol) was added.
  • reaction mixture was diluted with approx. 1000 ml toluene and THF was removed by evaporation at the rotavap.
  • the resulting toluene solution of crude product was pre-purified on a silca gel column by eluting with hexanes/5-17% EtOAc. Purest fractions were combined, evaporated and crystallized from TBME/hexane to yield 29.2 g of the title compound as white crystals.
  • a glass/stainless steel autoclave was purged with nitrogen, CU2O (0.464 g, 3.24 mmol), ammonia (101 ml, 25%, aq., 648 mmol, 30 equivalents) and (2R,5R)-5-(6-Bromo-3-fluoro- pyridin-2-yl)-2,5-dimethyl-2-trifluoromethyl-5,6-dihydro-2H-[1 ,4]oxazin-3-ylamine (8 g, 21.6 mmol) in ethylene glycol (130 ml) was added. The autoclave was closed and the suspension heated up to 60 °C and the solution was stirred for about 48 hours (max.
  • the reaction mixture was diluted with ethyl acetate and washed with water and brine, dried over sodium sulfate, filtered and evaporated.
  • the crude product (12 g) was chromatographed over silicagel (cyclohexane to cyclohexane: ethyl acetate 1 : 1) to yield 5.2 g of the title compound.
  • the solvent was evaporated and the residue diluted with a suitabable organic solvent, such as ethyl acetate and aq. ammonia. Ice was added and the organic phase was washed with water and brine, dried over sodium sulfate, filtered and evaporated to yield 3.78 g of the title compound.
  • a suitabable organic solvent such as ethyl acetate and aq. ammonia. Ice was added and the organic phase was washed with water and brine, dried over sodium sulfate, filtered and evaporated to yield 3.78 g of the title compound.
  • the five most characteristic peaks of crystalline Compound 1 in free Form A are at: 10.7, 14.8, 18.7, 19.5 and 21.4° 2-theta.
  • the three most characterictic peaks of crystalline Compound 1 in free Form A are at: 10.7, 14.8 and 19.5° 2-theta.
  • X-ray powder diffraction (XRPD) analysis was performed using a Bruker D8 Advance x-ray diffractometer in reflection geometry. Measurements were taken at about 30 kV and 40 mA under the following conditions:
  • Example 3 Micronization procedure and XRPD analysis for Compound 1 free Form A
  • Crystalline Compound 1 free Form A was micronized according to the following method:
  • a spiral jet-milling instrument was used with a ring of 50 mm diameter.
  • the carrier gas was nitrogen and the energy was targeted at 1800kJ/kg (cumulative parameter considering injector and grinding nozzle number and diameter, injector and grinding nozzle pressure, and feed rate according to Midoux N et al, 1999).
  • Micronized Crystalline Form A was analysed by XRPD and the ten most characteristic peaks are shown in Table 3 (see also Figure 2).
  • micronized crystalline Compound 1 in free Form A The five most characteristic peaks of micronized crystalline Compound 1 in free Form A are at: 12.1 , 15.9, 18.5, 19.4, 24.0° 2-theta.
  • X-ray powder diffraction (XRPD) analysis was performed using a Bruker D8 Advance x-ray diffractometer in reflection geometry. Measurements were taken at about 30 kV and 40 mA under the conditions shown in Table 4.
  • the X-ray diffraction pattern was recorded between 2° and 40° (2-theta) with CuK ⁇ radiation for identification of the whole pattern.
  • Crystalline Compound 1 free Form A was analysed by differential scanning calorimetry (DSC) using a Q1000 Diffraction Scanning Calorimeter from TA Instruments and found to have an endotherm at about 171 °C when analysed at a heating rate of 10 °C per minute, see Figure 3.
  • Example 6 XRPD analysis of Compound 1 hemifumarate hemihydrate salt Crystalline Compound 1 in hemifumarate hemihydrate salt form, prepared using the large scale procedure described in Example 5, was analysed by XRPD and the ten most characteristic peaks are shown in Table 5 (see also Figure 4).
  • crystalline Compound 1 in hemifumarate hemihydrate salt form may be characterised by peaks at: 11.9, 19.9, 23.0, 27.9 and 30.8° 2-theta.
  • crystalline Compound 1 in hemifumarate hemihydrate salt form may be characterised by peaks at: 23.0, 27.9 and 30.8° 2-theta.
  • X-ray powder diffraction (XRPD) analysis was performed using a Bruker D8 Advance x-ray diffractometer in reflection geometry. Measurements were taken at about 30 kV and 40 mA under the conditions shown in Table 6. Table 6
  • Example 7 XRPD analysis of micronized Compound 1 hemifumarate hemihvdrate salt form
  • Crystalline Compound 1 in hemifumarate hemihydrate salt form prepared using the large scale procedure described in Example 5, was micronized as described in Example 3, analysed by XRPD using the methodology and equipment described in Example 3, and the ten most characteristic peaks are shown in Table 7 (see also Figure 5).
  • micronized Compound 1 in hemifumarate hemihydrate salt form are at: 9.1 , 11.8, 14.0, 16.1 and 19.0° 2-theta.
  • micronized Compound 1 in f hemifumarate hemihydrate salt form are at: 14.0, 16.1 and 19.0° 2-theta.
  • Example 8 DSC analysis of Compound 1 hemifumarate hemihvdrate salt
  • Crystalline Compound 1 in hemifumarate hemihydrate salt form prepared using the large scale procedure described in Example 5, was analysed by DSC using a Discovery(R) instrument from TA Instruments and found to have an endotherm at about 239 °C when analysed at a heating rate of 10 °C per minute, see Figure 6.
  • Example 10 XRPD analysis of Compound 1 hemisuccinate hemihvdrate salt
  • the five most characteristic peaks of crystalline Compound 1 in hemisuccinate hemihydrate salt form are at: 12.0, 14.0, 17.3, 17.6 and 20.1° 2-theta.
  • the three most characterictic peaks of crystalline Compound 1 in hemisuccinate hemihydrate salt form are at: 12.0, 14.0 and 20.1° 2-theta.
  • Example 11 XRPD analysis of micronized Compound 1 hemisuccinate hemihvdrate salt Crystalline Compound 1 in hemisuccinate hemihydrate salt form, prepared using the large scale procedure described in Example 9, was micronized as described in Example 3, analysed by XRPD using the methodology and equipment described in Example 3 and the ten most characteristic peaks are shown in Table 9 (see also Figure 8).
  • micronized Compound 1 in hemisuccinate hemihydrate salt form are at: 10.1 , 14.5, 15.5, 19.8 and 23.3° 2-theta.
  • micronized Compound 1 in hemisuccinate hemihydrate salt form are at: 10.1 , 15.5 and 19.8° 2-theta.
  • Example 12 DSC analysis of Compound 1 hemisuccinate hemihvdrate salt Crystalline Compound 1 in hemisuccinate hemihydrate salt form, prepared using the large scale procedure described in Example 9, was analysed by DSC using a Discovery(R) instrument from TA instruments and found to have an endotherm at about 217 °C when analysed at a heating rate of 10 °C per minute, see Figure 9.
  • Example 13 Preparation of Compound 1 hemi-L-tartrate hemihvdrate salt Small scale preparation
  • hemihydrate salt form are at: 14.0, 17.7 and 19.8° 2-theta.
  • Example 15 XRPD analysis of micronized Compound 1 hemi-L-tartrate hemihydrate salt
  • Crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form prepared using the large scale procedure described in Example 13, was micronized as described in Example 3, analysed by XRPD using the methodology and equipment described in Example 3 and the ten most characteristic peaks are shown in Table 11 (see also Figure 11).
  • micronized Compound 1 in hemi-L-tartrate hemihydrate salt form are at: 7.1 , 13.0, 17.1 , 19.8 and 20.3° 2-theta.
  • micronized Compound 1 in hemi-L-tartrate hemihydrate salt form are at: 13.0, 17.1 and 20.3° 2-theta.
  • Example 16 DSC analysis of Compound 1 hemi-L-tartrate hemihydrate salt
  • Crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form prepared using the large scale procedure described in Example 13, was analysed by DSC using Discovery(R) instrument from TA instruments and found to have an endotherm at about 253 °C when analysed at a heating rate of 10 °C per minute, see Figure 12.
  • Example 17 Stoichiometric analysis of Compound 1 fumarate. succinate and tartrate salt forms
  • the stoichiometric composition of the Compound 1 salt forms was examined by proton NMR spectroscopy which confirmed that the stoichiometry of the ratio between Compound 1 and the salt forming agent was 2:1 for the succinate, fumarate and tartrate salts.
  • Example 18 Milling properties of Compound 1 free base form and hemifumarate hemihvdrate, hemisuccinate hemihvdrate and hemi-L-tartrate hemihvdrate salt forms
  • the behaviour of the free base and hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salt forms were evaluated during jet milling.
  • a spiral jet milling instrument was used with a ring of 50mm diameter.
  • the carrier gas was nitrogen and the energy was targeted at 1800kJ/kg (cumulative parameter considering injector and grinding nozzle number and diameter, injector and grinding nozzle pressure, and feed rate according to Midoux N et ai, 1999).
  • the percentage yields upon micronization are shown in Table 12 below.
  • Example 19 Preparation of further alternative salt forms of Compound 1 HCI: 300mg free base Compound 1 was added to 3mL MTBE and 1 eq HCI (6. ON) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • HCI, hydrate 300mg free base
  • Compound 1 was dissolved in 3mL ethanol and 1 eq HCI (6. ON) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours.
  • the solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • H 2 SO 4 300mg free base Compound 1 was dissolved in 3mL IPAc and 1 eq H2S04 (98%) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • H 3 PO 4 300mg free base Compound 1 was added to 3mL MTBE and 1 eq H3P04 (85%) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours.
  • the solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • Acetate 300mg free base Compound 1 was dissolved in 3mL ethanol and 35.3mg acetic acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • Citrate 300mg free base Compound 1 was dissolved in 3mL IPAc and 56.1 mg citric acid (0.5 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours.
  • the solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • Butyrate 300mg free base Compound 1 was dissolved in 3mL IPAc and 51.5mg butyric acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • D-glucuronate 300mg free base Compound 1 was dissolved in 3mL acetonitrile and 113.4mg D-glucuronic acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • Galactarate 300mg free base Compound 1 was dissolved in 3mL ethanol and 61.3mg galactaric acid (0.5 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours. Glutarate: 300mg free base Compound 1 was added to 3mL MTBE and 77.1 mg glutaric acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • L-aspartate 300mg free base Compound 1 was dissolved in 3mL ethanol and 77.7mg L- aspartic acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • Glycolate 300mg free base Compound 1 was added to 3mL MTBE and 44.4mg glycolic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • N-acetylglicine 300mg free base Compound 1 was added to 3mL MTBE and 68.4mg N- acetylglycine (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • Propionate 300mg free base Compound 1 was added to 3mL MTBE and 43.3mg propionic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours. D-quinate: 300mg free base Compound 1 was dissolved in 3mL IPAc and 112.2mg D-quinic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
  • Example 20 Bulk stability of Compound 1 salts in comparison to the free form after three weeks stress exposure at 80°C/75% relative humidity and in the presence of 90% sodium stearyl fumarate
  • Example 21 Bulk stability of Compound 1 hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salts in comparison to the free form after three weeks stress exposure at 80°C/75% relative humidity and in the presence of pharmaceutical excipients
  • Example 22 In human study of pharmacokinetics of free base Compound 1 when given alone and in combination with the strong CYP3A4 inhibitor itraconazole or the strong CYP3A4 inducer rifampicin
  • DDI drug-drug interaction
  • Rifampicin at a dose of 600 mg q.d., decreased mean AUC of Compound 1 5-6-fold and mean Cmax of Compound 1 2.5-fold, when given together with Compound 1 as compared to when Compound 1 was given alone (Table 16).
  • CYP3A4 is of major importance for the elimination of Compound 1 and that the effects of co-treatment with a strong CYP3A4 inhibitor or inducer need to be taken into account when administering Compound 1.
  • n* number of subjects with non-missing values.
  • n* number of subjects with non-missing values.

Abstract

The present invention relates to salt forms of an oxazine derivative BACE-1 inhibitor for use in the treatment or prevention of Alzheimer's disease and/or cerebral amyloid angiopathy and the advantageous properties associated with said salt forms.

Description

Salt Forms of an Oxazine Derivative BACE Inhibitor
Field of the Invention
The present invention relates to salt forms, specifically the fumarate, succinate and tartrate salt forms, of the BACE inhibitor N-(6-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3, 6- dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide, and the advantageous properties associated therewith.
Background of the Invention
Alzheimer’s disease (AD) is one of the most prevalent neurological disorders worldwide and the most common and debilitating age-related condition, causing progressive amnesia, dementia, and ultimately global cognitive failure and death. Currently, the only
pharmacological therapies available are symptomatic drugs such as cholinesterase inhibitors or other drugs used to control the secondary behavioral symptoms of AD. Investigational treatments targeting the AD pathogenic cascade include those intended to interfere with the production, accumulation, or toxic sequelae of amyloid-b (Ab) species (Kramp VP, Herrling P, 2011).
Cerebral amyloid angiopathy (CAA) is a common age related cerebral small vessel disease, characterised by progressive deposition of Ab, in particular Ab40, in the wall of small to medium sized arteries, arterioles and capillaries of the cerebral cortex and overlying leptomeninges (Charidimou A et ai, 2011). CAA often coexists with AD. Mild forms of CAA often appear asymptomatic; however, CAA may also lead to severe vascular pathologies and is a risk factor for cerebral hemorrhages ranging from silent microbleeds to spontaneous intracerebral haemorrhage, a devastating form of stroke.
ApoE4 is a strong genetic risk factor for both AD and CAA (Shinohara M et al, 2016).
Human APOE is located on chromosome 19 (gene APOE, Uniprot P02649). Three major isoforms (ApoE2, -3 and -4) are known in humans. ApoE4 (with Arg at positions 112 and 158) has an allele frequency of 5-35% in humans (Verghese PB et ai, 2011) and ApoE4 homozygotes are estimated to represent about 2 to 3% of the general population (Quintino- Santos SR et ai, 2012).
Strategies that target decreasing Ab by: (1) enhancing the amyloid clearance with an active or passive immunotherapy against Ab; or (2) decreasing production through inhibition of Beta-site-APP cleaving enzyme-1 (BACE-1 , an enzyme involved in the processing of the amyloid precursor protein [APP]), are of potential therapeutic value in the treatment or prevention of AD and CAA, particularly in patients carrying one or two copies of the ApoE4 allele.
N-(6-((3R,6R)-5-Amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5- fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide, referred to herein as“Compound 1”, is an orally active BACE inhibitor, previously described in WO 2012/095469 A1.
During formulation of Compound 1 into a pharmaceutical composition, drug substance is micronized in order to increase its surface area and thereby improve its dissolution rate and bioavailability. However, micronization of Compound 1 is extremely challenging at relevant operational conditions due to poor flow and the tendency of the drug substance to adhere to the mill. These characteristics make the manufacture of a pharmaceutical composition comprising Compound 1 problematic. It is therefore an objective of the present invention to provide a solution to the problems observed when milling Compound 1.
Summary of the Invention
It has surprisingly been found that the poor milling properties of Compound 1 are overcome by use of its fumarate, succinate and tartrate salt forms.
In a first aspect of the invention, there is therefore provided the fumarate, succinate and tartrate salt forms of Compound 1.
It has further been found that the fumarate, succinate and tartrate salt forms of Compound 1 are more stable than the free form, and other tested salt forms, when in physical mixture with pharmaceutical excipients.
In a second aspect of the invention, there is therefore provided a pharmaceutical composition comprising the fumarate, succinate and tartrate salt forms of Compound 1.
In a third aspect of the invention, there is provided Compound 1 in fumarate, succinate or tartrate salt form for use as a medicament.
In a fourth aspect of the invention, there is provided Compound 1 in fumarate, succinate or tartrate salt form for use in the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
In a fifth aspect of the invention, there is provided a method for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy which method comprises administering to a patient in need thereof a therapeutically effective amount of Compound 1 in fumarate, succinate or tartrate salt form. In a sixth aspect of the invention, there is provided the use of Compound 1 in fumarate, succinate or tartrate salt form for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
In a seventh aspect of the invention, there is provided the use of Compound 1 in fumarate, succinate or tartrate salt form, for the manufacture of a medicament for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
Description of the Invention
List of Figures
Figure 1 shows shows the X-ray powder diffraction pattern for crystalline free base
Compound 1 Form A when measured using CuKα radiation.
Figure 2 shows the X-ray powder diffraction pattern for micronized crystalline free base Compound 1 Form A when measured using CuKα radiation.
Figure 3 shows the DSC thermogram for crystalline free base Compound 1 Form A.
Figure 4 shows the X-ray powder diffraction pattern for crystalline Compound 1 in hemifumarate hemihydrate salt form when measured using CuKα radiation.
Figure 5 shows the X-ray powder diffraction pattern for micronized crystalline Compound 1 in hemifumarate hemihydrate salt form when measured using CuKα radiation.
Figure 6 shows the DSC thermogram for crystalline Compound 1 in hemifumarate hemihydrate salt form. Figure 7 shows the X-ray powder diffraction pattern for crystalline Compound 1 in hemisuccinate hemihydrate salt form when measured using CuKα radiation.
Figure 8 shows the X-ray powder diffraction pattern for micronized crystalline Compound 1 in hemisuccinate hemihydrate salt form when measured using CuKα radiation.
Figure 9 shows the DSC thermogram for crystalline Compound 1 in hemisuccinate hemihydrate salt form.
Figure 10 shows the X-ray powder diffraction pattern for crystalline Compound 1 in hemi-L- tartrate hemihydrate salt form when measured using CuKα radiation.
Figure 11 shows the X-ray powder diffraction pattern for micronized crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form when measured using CuKα radiation. Figure 12 shows the DSC thermogram for crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form.
Figure 13 shows the design of a two part, open-label, two-period, fixed-sequence study in healthy subjects to evaluate the PK of Compound 1 when given alone and in combination with the strong CYP3A4 inhibitor itraconazole or the strong CYP3A4 inducer rifampicin.
Embodiments of the First Aspect of the Invention
Embodiment A1 : Compound 1 in fumarate, succinate or tartrate salt form.
Embodiment A2: Compound 1 in fumarate salt form.
Embodiment A3: Compound 1 according to Embodiment A2 in hemifumarate salt form. Embodiment A4: Compound 1 in succinate salt form.
Embodiment A5: Compound 1 according to Embodiment A4 in hemisuccinate salt form. Embodiment A6: Compound 1 in tartrate salt form.
Embodiment A7: Compound 1 according to Embodiment A6 in L-tartrate salt form.
Embodiment A8: Compound 1 according to Embodiment A6 in hemitartrate salt form or A7 in hemi-L-tartrate salt form.
Embodiment A9: Compound 1 according to any one of Embodiments A1 to A8 in hydrated salt form.
Embodiment A10: Compound 1 according to Embodiment A9 in hemihydrate or
monohydrate salt form.
Embodiment A11 : Compound 1 according to Embodiment A10 in hemihydrate salt form.
Embodiment A12: Compound 1 in hemifumarate hemihydrate salt form.
Embodiment A13: Compound 1 in hemisuccinate hemihydrate salt form.
Embodiment A14: Compound 1 in hemitartrate hemihydrate salt form.
Embodiment A15: Compound 1 in hemi-L-tartrate hemihydrate salt form.
Embodiment A16: Compound 1 according to any one of Embodiments A1 to A15 in crystalline form.
Embodiment A17: Compound 1 according to Embodiment A16 in substantially pure form. Detailed Compound 1 fumarate salt embodiments
Embodiment A18: Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 11.9, 13.9 and 19.9° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A19: Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 23.0, 27.9 and 30.8° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A20: Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 11.9, 13.9, 16.2, 17.3 and 19.9° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A21 : Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 11.9, 19.9, 23.0, 27.9 and 30.8° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A22: Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 4.0, 11.9, 13.9, 15.6, 16.2, 17.3, 19.9, 23.0, 27.9 and 30.8° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A23: Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 4 when measured using CuKα radiation.
Embodiment A24: Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 14.0, 16.1 and 19.0° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A25: Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 9.1 , 11.8, 14.0, 16.1 and 19.0° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ. Embodiment A26: Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 3.9, 7.3, 9.1 , 11.8, 14.0, 16.1 , 17.1 , 19.0, 19.7 and 22.3° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A27: Compound 1 according to Embodiment A2 in micronized fumarate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 5 when measured using CuKα radiation.
Embodiment A28: Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having an endotherm at about 239 °C when analysed by DSC at a heating rate of 10 °C per minute.
Embodiment A29: Compound 1 according to Embodiment A2 in crystalline fumarate salt form and having a DSC thermogram substantially the same as that shown in Figure 6.
Embodiment A30: Compound 1 according to any one of Embodiments A18 to A29 wherein Compound 1 is in hemifumarate salt form.
Embodiment A31 : Compound 1 according to any one of Embodiments A18 to A30 wherein Compound 1 is in hydrated salt form.
Embodiment A32: Compound 1 according to any one of Embodiments A18 to A31 wherein Compound 1 is in hemihydrate salt form.
Detailed Compound 1 succinate salt embodiments
Embodiment A33: Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 12.0, 14.0 and 20.1° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A34: Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 12.0, 14.0, 17.3, 17.6 and 20.1° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A35: Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 4.0, 12.0, 14.0, 15.7, 16.2, 17.3, 17.6, 20.1 , 23.3 and 31.2° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ. Embodiment A36: Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 7 when measured using CuKα radiation.
Embodiment A37: Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 10.1 , 15.5 and 19.8° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A38: Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 10.1 , 14.5, 15.5, 19.8 and 23.3° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A39: Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 9.2, 10.1 , 11.9, 12.7, 14.5, 15.5, 19.8, 22.0, 23.3 and 24.5° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A40: Compound 1 according to Embodiment A4 in micronized succinate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 8 when measured using CuKα radiation.
Embodiment A41 : Compound 1 according to Embodiment A4 in crystalline succinate salt form and having an endotherm at about 217 °C when analysed by DSC at a heating rate of 10 °C per minute.
Embodiment A42: Compound 1 according to Embodiment A4 in crystalline succinate salt form and having a DSC thermogram substantially the same as that shown in Figure 9.
Embodiment A43: Compound 1 according to any one of Embodiments A33 to A42 wherein Compound 1 is in hemisuccinate salt form.
Embodiment A44: Compound 1 according to any one of Embodiments A33 to A43 wherein Compound 1 is in hydrated salt form.
Embodiment A45: Compound 1 according to any one of Embodiments A33 to A44 wherein Compound 1 is in hemihydrate salt form. Detailed Compound 1 tartrate salt embodiments
Embodiment A46: Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 14.0, 17.7 and 19.8° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A47: Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 12.2, 14.0, 17.7, 19.8 and 20.4° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A48: Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 12.2, 14.0, 16.0, 17.1 , 17.3, 17.7, 19.8, 20.4, 23.5 and 31.5° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A49: Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 10 when measured using CuKα radiation.
Embodiment A50: Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 13.0, 17.1 and 20.3° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A51 : Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 7.1 , 13.0, 17.1 , 19.8 and 20.3° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A52: Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 7.1 , 13.0, 13.7, 13.9, 15.9, 17.1 , 19.8, 20.3, 22.4 and 24.7° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
Embodiment A53: Compound 1 according to Embodiment A6 in micronized tartrate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 11 when measured using CuKα radiation. Embodiment A54: Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having an endotherm at about 253 °C when analysed by DSC at a heating rate of 10 °C per minute.
Embodiment A55: Compound 1 according to Embodiment A6 in crystalline tartrate salt form and having a DSC thermogram substantially the same as that shown in Figure 12.
Embodiment A56: Compound 1 according to any one of Embodiments A46 to A55 wherein Compound 1 is in hemitartrate salt form.
Embodiment A57: Compound 1 according to any one of Embodiments A46 to A56 wherein Compound 1 is in hemi-L-tartrate salt form. Embodiment A58: Compound 1 according to any one of Embodiments A46 to A57 wherein Compound 1 is in hydrated salt form.
Embodiment A59: Compound 1 according to any one of Embodiments A46 to A58 wherein Compound 1 is in hemihydrate salt form.
Embodiments of the Second Aspect of the Invention Embodiment B1 : A pharmaceutical composition comprising Compound 1 according to any one of Embodiments A1 to A59.
Embodiment B2: The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 1 to 100 mg of the base of Compound 1.
Embodiment B3: The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 1 to 75 mg of the base of Compound 1.
Embodiment B4: The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 1 , 10, 15, 25, 50 or 75 mg of the base of Compound 1.
Embodiment B5: The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 15 mg of the base of Compound 1.
Embodiment B6: The pharmaceutical composition according to Embodiment B1 wherein the pharmaceutical composition comprises 50 mg of the base of Compound 1.
Embodiment B7: The pharmaceutical composition according to any one of Embodiments B1 to B6 wherein the pharmaceutical composition comprises a gelatin capsule. Embodiment B8: The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises magnesium stearate, sodium stearyl fumarate, or Aerosil 200 PH.
Embodiment B9: The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises magnesium stearate.
Embodiment B10: The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises sodium stearyl fumarate.
Embodiment B11 : The pharmaceutical composition according to any one of Embodiments B1 to B7 wherein the pharmaceutical composition comprises Aerosil 200 PH. Embodiments of the Third and Fourth Aspects of the Invention
Embodiment C1 : Compound 1 according to any one of Embodiments A1 to A59 for use as a medicament.
Embodiment C2: Compound 1 according to any one of Embodiments A1 to A59 for use in the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy. Embodiment C3: Compound 1 according to any one of Embodiments A1 to A59 for use in the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
Embodiment C4: Compound 1 for the use according to Embodiment C2 or C3 in a patient carrying one or two copies of the ApoE4 allele.
Embodiment C5: Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of between 10 and 30 mg of the base of Compound 1 per day.
Embodiment C6: Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of between 30 and 100 mg of the base of Compound 1 per day. Embodiment C7: Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of between 30 and 50 mg of the base of Compound 1 per day.
Embodiment C8: Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of 15 mg of the base of Compound 1 per day. Embodiment C9: Compound 1 for the use according to any one of Embodiments C1 to C4 wherein Compound 1 is used at a dose of 50 mg of the base of Compound 1 per day.
Embodiments of the Fifth Aspect of the Invention
Embodiment D1 : A method for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy which method comprises administering to a patient in need thereof a therapeutically effective amount of Compound 1 according to any one of
Embodiments A1 to A59.
Embodiment D2: A method for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy which method comprises administering to a patient in need thereof a therapeutically effective amount of Compound 1 according to any one of Embodiments A1 to A59.
Embodiment D3: The method according to Embodiment D1 or D2 in a patient carrying one or two copies of the ApoE4 allele.
Embodiment D4: The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of between 10 and 30 mg per day.
Embodiment D5: The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of between 30 and 100 mg per day.
Embodiment D6: The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of between 30 and 50 mg per day.
Embodiment D7: The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of 15 mg per day.
Embodiment D8: The method according to any one of Embodiments D1 to D3, wherein the base of Compound 1 is used at a dose of 50 mg per day.
Embodiments of the Sixth Aspect of the Invention
Embodiment E1 : Use of Compound 1 according to any one of Embodiments A1 to A59 for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
Embodiment E2: The use of Compound 1 according to Embodiment E1 for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
Embodiment E3: The use of Compound 1 according to Embodiment E1 or E2 in a patient carrying one or two copies of the ApoE4 allele. Embodiment E4: The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of between 10 and 30 mg of the base of Compound 1 per day.
Embodiment E5: The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of between 30 and 100 mg of the base of Compound 1 per day.
Embodiment E6: The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of between 30 and 50 mg of the base of Compound 1 per day.
Embodiment E7: The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of 15 mg of the base of Compound 1 per day.
Embodiment E8: The use of Compound 1 according to any one of Embodiments E1 to E3, wherein the drug substance is used at a dose of 50 mg of the base of Compound 1 per day.
Embodiments of the Seventh Aspect of the Invention
Embodiment F1 : Use of Compound 1 according to any one of Embodiments A1 to A59, for the manufacture of a medicament for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
Embodiment F2: The use of Compound 1 according to Embodiment F1 for the manufacture of a medicament for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy.
Embodiment F3: The use of Compound 1 according to Embodiment F1 or F2 for the manufacture of a medicament for the prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy in a patient carrying one or two copies of the ApoE4 allele.
Embodiment F4: The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of between 10 and 30 mg of the base of Compound 1 per day.
Embodiment F5: The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of between 30 and 100 mg of the base of Compound 1 per day. Embodiment F6: The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of between 30 and 50 mg of the base of Compound 1 per day. Embodiment F7: The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of 15 mg of the base of Compound 1 per day.
Embodiment F8: The use of Compound 1 according to any one of Embodiments F1 to F3, wherein the drug substance is used for the treatment or prevention of Alzheimer’s disease and/or cerebral amyloid angiopathy at a dose of 50 mg of the base of Compound 1 per day.
Definitions
As used herein, the terms“Compound 1”,“Cmpd 1”,“the base of Compound 1”,“the free base of Compound 1” or“the free form of Compound 1” refer to N-(6-((3R,6R)-5-amino-3,6- dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5- (trifluoromethyl)picolinamide and having the following structural formula:
Figure imgf000014_0001
In Example 1 , using an alternative chemical naming format,“Compound 1” is also referred to as 3-chloro-5-trifluoromethyl-pyridine-2-carboxylic acid [6-((3R,6R)-5-amino-3,6-dimethyl-6- trifluoromethyl-3,6-dihydro-2H-[1 ,4]oxazin-3-yl)-5-fluoro-pyridin-2-yl]-amide. The terms“Compound 1”,“Cmpd 1”,“the base of Compound 1”,“the free base of
Compound 1”,“the free form of Compound 1” and its corresponding full chemical name are used interchangeably throughout the description of the invention. Compound 1 is described in WO 2012/095469 A1 , Example 34. WO 2012/095469 A1 is incorporated herewith by reference in its entirety, in particular the disclosure related to the synthesis of Example 34. As used herein, the term“salt” includes both hydrated and anhydrous salt forms and all stoichiometric ratios of the base of Compound 1 to its salt counterion or to molecules of water if a hydrated salt form. As used herein, the terms“hemifumarate”,“hemisuccinate”, or“hemitartrate” salt of
Compound 1 refers to a salt having a stoichiometric ratio of two molecules of Compound 1 to one molecule of fumarate, succinate, or tartrate counterion, respectively. The terms “hemifumarate”,“hemisuccinate”, or“hemitartrate” salt of Compound 1 include hydrated salt forms, for example, hemifumarate hemihydrate, hemisuccinate hemihydrate and hemitartrate hemihydrate salt forms. The term“hemihydrate” refers to a hydrated form of a Compound 1 salt having a stoichiometric ratio of two molecules of Compound 1 to one molecule of water. The hemifumarate, hemisuccinate and hemitartrate salts of Compound 1 may be
represented as shown below. Compound 1 hemifumarate
Figure imgf000015_0001
Compound 1 hemisuccinate
Figure imgf000015_0002
Figure imgf000016_0001
Compound 1 hemi-L-tartrate
Figure imgf000016_0002
The hemifumarate hemihydrate, hemisuccinate hemihydrate and hemitartrate hemihydrate salts of Compound 1 may be represented as shown below. Compound 1 hemifumarate hemihvdrate
Figure imgf000017_0001
Compound 1 hemisuccinate hemihvdrate
Figure imgf000017_0002
Compound 1 hemi-L-tartrate hemihvdrate
Figure imgf000018_0001
The hemifumarate hemihydrate, hemisuccinate hemihydrate, and hemi-L-tartrate hemihydrate salts of Compound 1 are referred to herein as:
N-(6-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5- fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide hemifumarate hemihydrate;
N-(6-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5- fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide hemisuccinate hemihydrate; and
N-(6-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5- fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide hemi-L-tartrate hemihydrate; respectively. In accordance with IUPAC recommendations, the foregoing salts may also be referred to as: (2E)-But-2-enedioic acid— N-{6-[(3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6- dihydro-2H-1 ,4-oxazin-3-yl]-5-fluoropyridin-2-yl}-3-chloro-5-(trifluoromethyl)pyridine-2- carboxamide— water (1/2/1); Butanedioic acid— N-{6-[(3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-
1.4-oxazin-3-yl]-5-fluoropyridin-2-yl}-3-chloro-5-(trifluoromethyl)pyridine-2-carboxamide— water (1/2/1); and
(2R,3R)-2,3-Dihydroxybutanedioic acid— N-{6-[(3R,6R)-5-amino-3,6-dimethyl-6- (trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl]-5-fluoropyridin-2-yl}-3-chloro-5- (trifluoromethyl)pyridine-2-carboxamide— water (1/2/1); respectively, and the different naming formats of the Compound 1 salts may be used interchangeably.
As used herein, the term“tartrate salt” includes salts of Compound 1 derived from all possible stereoisomers of tartaric acid, and encompasses: 2S,3S-tartaric acid (D-tartaric acid); 2R,3R-tartaric acid (L-tartaric acid); 2 S,3R- tartaric acid tartaric acid (Meso-tartaric acid); racemic mixtures of 2S,3S-tartaric acid and 2R,3R-tartaric acid; and mixtures of 2S,3S-tartaric acid, 2R,3R-tartaric acid and 2 S,3R- tartaric acid. In one embodiment, the tartrate salt of Compound 1 is 2S,3S-tartaric acid. In another embodiment, the tartrate salt of Compound 1 is 2R,3R-tartaric acid.
The term“substantially pure,” when used in reference to crystalline fumarate, succinate and tartrate salt forms of N-(6-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-
1.4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide, means having a purity greater than 90 weight %, including greater than 90, 91 , 92, 93, 94, 95, 96, 97, 98, and 99 weight %, and also including equal to about 100 weight % of N-(6-((3R,6R)-5-amino- 3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro- 5-(trifluoromethyl)picolinamide, based on the weight of the compound in its salt form.
As used herein, the term“Aerosil 200 PH” refers to AEROSIL® 200 Pharma grade, a high purity amorphous anhydrous colloidal silicon dioxide used as a pharmaceutical excipient.
The term“substantially the same” with reference to X-ray diffraction patterns and peak positions means that typical peak position and intensity variability are taken into account.
For example, one skilled in the art will appreciate that the peak positions (2Q) will show some inter-apparatus variability, typically as much as 0.2°. X-ray powder diffraction analysis is typically carried out at room temperature, i.e. between 20 and 30 °C. Further, one skilled in the art will appreciate that relative peak intensities will show inter-apparatus variability as well as variability due to degree of crystallinity, preferred orientation, prepared sample surface, and other factors known to those skilled in the art, and should be taken as a qualitative measure only. One of ordinary skill in the art will also appreciate that an X-ray diffraction pattern may be obtained with a measurement error that is dependent upon the measurement conditions employed. In particular, it is generally known that intensities in an X-ray diffraction pattern may fluctuate depending upon measurement conditions employed. It should be further understood that relative intensities may also vary depending upon experimental conditions and, accordingly, the exact order of intensity should not be taken into account. Additionally, a measurement error of diffraction angle for a conventional X-ray diffraction pattern is typically about 5% or less, and such degree of measurement error should be taken into account as pertaining to the aforementioned diffraction angles.
Consequently, it is to be understood that the crystal form of the instant invention is not limited to the crystal form that provides an X-ray diffraction pattern completely identical to the X-ray diffraction patterns depicted in the accompanying Figures 4, 5, 7, 8, 10 and 11 disclosed herein. Any crystal forms that provide X- ray diffraction patterns substantially identical to that disclosed in the accompanying Figures 4, 5, 7, 8, 10 and 11 fall within the scope of the present invention. The ability to ascertain substantial identities of X-ray diffraction patterns is within the purview of one of ordinary skill in the art. An expression referring to a crystalline form of Compound 1 having“an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure X” may be interchanged with an expression referring to a crystalline form of Compound 1 having“an X- ray powder diffraction pattern characterised by the representative X-ray powder diffraction pattern shown in Figure X”.
As used herein, the term“about X °C”, in the context of the endotherm temperature as measured by DSC analysis, means within a range of +/- 5 °C of temperature X, more particularly within a range of +/- 2.5 °C of temperature X.
As used herein, the term“Alzheimer’s disease” or“AD” encompasses both preclinical and clinical Alzheimer’s disease unless the context makes clear that either only preclinical Alzheimer’s disease or only clinical Alzheimer’s disease is intended.
As used herein, the term“treatment of Alzheimer’s disease” refers to the administration of Compound 1 of the present invention to a patient in order to ameliorate at least one of the symptoms of Alzheimer’s disease.
As used herein, the term“prevention of Alzheimer’s disease” refers to the prophylactic treatment of AD; or delaying the onset or progression of AD. For example, the onset or progression of AD is delayed for at least 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 years. In one embodiment,“prevention of Alzheimer’s disease” refers to the prophylactic treatment of preclinical AD; or delaying the onset or progression of preclinical AD. In a further
embodiment, the onset or progression of preclinical AD is delayed for at least 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 years. In another embodiment,“prevention of Alzheimer’s disease” refers to the prophylactic treatment of clinical AD; or delaying the onset or progression of clinical
AD. In a further embodiment, the onset or progression of clinical AD is delayed for at least
O.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 years.
As used herein, the term“clinical Alzheimer’s disease” or“clinical AD” encompasses both Mild Cognitive Impairment (MCI) due to AD and dementia due to AD, unless the context makes clear that either only MCI due to AD or dementia due to AD is intended. The
European Medicines Agency (EMA) in its‘Draft guidelines on the clinical investigation of medicines for the treatment of AD and other dementias’ (EMA/Committee for Medicinal Products for Human Use (CHMP)/539931/2014) summarises the National Institute on Aging criteria for the diagnosis of MCI due to AD and AD dementia as set out below.
Diagnosis of MCI due to AD requires evidence of intra-individual decline, manifested by: a) A change in cognition from previously attained levels, as noted by self- or informant report and/or the judgment of a clinician.
b) Impaired cognition in at least one domain (but not necessarily episodic memory) relative to age-and education-matched normative values; impairment in more than one cognitive domain is permissible.
c) Preserved independence in functional abilities, although the criteria also accept‘mild problems’ in performing instrumental activities of daily living (IADL) even when this is only with assistance (i.e. rather than insisting on independence, the criteria allow for mild dependence due to functional loss).
d) No dementia, which nominally is a function of c (above).
e) A clinical presentation consistent with the phenotype of AD in the absence of other
potentially dementing disorders. Increased diagnostic confidence may be suggested by
1) Optimal: A positive Ab biomarker and a positive degeneration biomarker
2) Less optimal:
i. A positive Ab biomarker without a degeneration biomarker
ii. A positive degeneration biomarker without testing for Ab biomarkers
Diagnosis of AD dementia requires:
a) The presence of dementia, as determined by intra-individual decline in cognition and function.
b) Insidious onset and progressive cognitive decline.
c) Impairment in two or more cognitive domains; although an amnestic presentation is most common, the criteria allow for diagnosis based on nonamnestic presentations (e.g. impairment in executive function and visuospatial abilities). d) Absence of prominent features associated with other dementing disorders.
Increased diagnostic confidence may be suggested by the biomarker algorithm discussed in the MCI due to AD section above.
As used herein, the term“preclinical Alzheimer’s disease” or“preclinical AD” refers to the presence of in vivo molecular biomarkers of AD in the absence of clinical symptoms. The National Institute on Aging and Alzheimer’s Association provide a scheme, shown in Table A below, which sets out the different stages of preclinical AD (Sperling et al., 2011).
Table A: Preclinical AD staging categories
Figure imgf000022_0001
s MRI = structural magnetic resonance imaging As used herein, the term“Cerebral Amyloid Angiopathy” or“CAA” refers to a disease characterised by the accumulation of b-amyloid (Ab) proteins in the walls of cortical and leptomeningeal blood vessels. CAA is a common cause of vessel wall breakdown and vascular dysfunction in older adults, making it a major contributor to fatal or disabling intracerebral hemorrhages (ICH) as well as ischemic injury and dementia (Gurol ME et al., 2016). As used herein, the term“Cerebral Amyloid Angiopathy” or“CAA” encompasses both
CAA-Type 1 and CAA-Type 2 unless the context makes clear that only CAA-Type 1 or CAA- Type 2 is intended.
As used herein, the term“CAA-Type 1” refers to capillary CAA (capCAA) characterised by Ab protein depositions in the walls of cortical capillaries (Thai et al., 2002). As used herein, the term“CAA-Type 2” refers to CAA characterised by Ab protein depositions in the walls of leptomeningeal and cortical vessels, with the exception of cortical capillaries (Thai et al, 2002).
As used herein, the term“treatment of CAA” refers to the administration of Compound 1 of the present invention to a patient in order to slow or arrest the development of CAA or at least one of the clinical symptoms of CAA, for example ICH, ischemic injury, or dementia. The development of CAA may be assessed by measuring the accumulation of Ab in the walls of cortical (for example occipital cortex) and leptomeningeal blood vessels using a Positron Emission Tomography (PET) tracer, for example 18F-florbetapir (Gurol ME et ai, 2016). Alternatively, the development of CAA may be assessed by monitoring cerebral microbleeds (CMB) as a haemorrhagic marker of CAA (Greenberg SM et ai, 2014). Suitable techniques for the monitoring of CMB include, for example, magnetic resonance imaging (MRI) susceptibility-weighted imaging (SWI) and MRI T2*-weighted gradient-recalled echo imaging (GRE), and are described in Cheng AL et ai, 2013. In addition, white matter hyperintensities (WMH) occur at much greater volume in patients diagnosed with CAA than in healthy aged individuals or in patients suffering from AD or mild cognitive impairment (MCI) (Greenberg SM et ai, 2014). Therefore, CAA development may be monitored by the measurement of WMH volume using MRI (Chen YW et ai, 2006). It is expected that the “treatment of CAA” will have the resultant benefit of reducing the likelihood of a cerebral ischemic event in the patient undergoing treatment for CAA. Therefore, in one embodiment of the invention, the term“treatment of CAA” is equivalent to the term“treatment of intracerebral haemorrhage”. In another embodiment of the invention, the term“treatment of CAA” is equivalent to the term“treatment of CAA and/or intracerebral haemorrhage”. In a further embodiment of the invention, the term“treatment of CAA” is equivalent to the term “treatment of CAA and intracerebral haemorrhage associated therewith”.
As used herein, the term“prevention of CAA” refers to the prophylactic treatment of CAA; delaying the onset or progression of CAA; or delaying the onset or progression of at least one of the clinical symptoms of CAA. For example, the onset or progression of CAA is delayed for at least 0.5, 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 years. In one embodiment of the invention, the term“prevention of CAA” is equivalent to the term“prevention of intracerebral haemorrhage”. In another embodiment of the invention, the term“prevention of CAA” is equivalent to the term“prevention of CAA and/or intracerebral haemorrhage”. In a further embodiment of the invention, the term“prevention of CAA” is equivalent to the term “prevention of CAA and intracerebral haemorrhage associated therewith”.
As used herein, the term“a genetic predisposition for the development of CAA” includes, but is not limited to situations where the genetic predisposition is due to: Down’s syndrome; a mutation in the gene for amyloid precursor protein or presenilin-1 ; or the presence of one or two copies of the ApoE4 allele.
As used herein, the term“patient” refers to a human subject.
As used herein, a“pharmaceutical composition” comprises Compound 1 of the present invention and at least one pharmaceutically acceptable carrier or excipient, in a unit dose solid form suitable for oral administration (typically a capsule, more particularly a hard gelatin capsule). A list of pharmaceutically acceptable carriers and excipients can be found in Remington’s Pharmaceutical Sciences.
The term "a therapeutically effective amount" refers to an amount of Compound 1 of the present invention that will elicit inhibition of BACE-1 in a patient as evidenced by a reduction in CSF or plasma Ab 1-40 levels relative to an initial baseline value. Ab 1-40 levels may be measured using standard immunoassay techniques, for example Meso Scale Discovery (MSD) 96-well MULTI-ARRAY human/rodent (4G8) Ab40 Ultrasensitive Assay (#K110FTE- 3, Meso Scale Discovery, Gaithersburg, USA).
List of abbreviations
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Examples
The following Examples illustrate and provide comparisons for various aspects of the invention. Example summary
Example 1 shows how Compound 1 may be prepared and crystallised.
Example 2 describes the XRPD analysis of Compound 1 free Form A.
Example 3 describes the XRPD analysis of micronized Compound 1 in free base form.
Example 4 describes the DSC analysis of Compound 1 free Form A. Example 5 describes the preparation of Compound 1 in hemifumarate hemihydrate salt form.
Example 6 describes the XRPD analysis of crystalline Compound 1 in hemifumarate hemihydrate salt form.
Example 7 describes the XRPD analysis of micronized Compound 1 in hemifumarate hemihydrate salt form.
Example 8 describes the DSC analysis of Compound 1 in hemifumarate hemihydrate salt form.
Example 9 describes the preparation of Compound 1 in hemisuccinate hemihydrate salt form. Example 10 describes the XRPD analysis of crystalline Compound 1 in hemisuccinate hemihydrate salt form. Example 11 describes the XRPD analysis of micronized Compound 1 in hemisuccinate hemihydrate salt form.
Example 12 describes the DSC analysis of Compound 1 in hemisuccinate hemihydrate salt form. Example 13 describes the preparation of Compound 1 in hemi-L-tartrate hemihydrate salt form.
Example 14 describes the XRPD analysis of crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form.
Example 15 describes the XRPD analysis of micronized Compound 1 in hemi-L-tartrate hemihydrate salt form.
Example 16 describes the DSC analysis of Compound 1 in hemi-L-tartrate hemihydrate salt form.
Example 17 describes the stoichiometric analysis of Compound 1 in fumarate, succinate and tartrate salt form. Example 18 compares the milling behavior of free base Compound 1 with the hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salt forms.
Example 19 describes the preparation of further alternative salt forms of Compound 1.
Example 20 compares the bulk stability of Compound 1 in free form with alternative salt forms and in the presence of sodium stearyl fumarate. Example 21 compares the bulk stability of Compound 1 in free form with the hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salt forms in the presence of various pharmaceutical excipients.
Example 22 describes an in human study to assess free base Compound 1 PK when given administered in combination with a strong CYP3A4 inhibitor or inducer. Detailed Examples
Example 1 : Preparation of free base Compound 1
The preparation of Compound 1 is described in WO 2012/095469 A1 (Example 34).
Compound 1 may also be prepared as described below.
NMR Methodology Proton spectra are recorded on a Bruker 400 MHz ultrashield spectrometer unless otherwise noted. Chemical shifts are reported in ppm relative to methanol (d 3.31), dimethyl sulfoxide (d 2.50), or chloroform (d 7.29). A small amount of the dry sample (2-5 mg) is dissolved in an appropriate deuterated solvent (0.7 mL). The shimming is automated and the spectra obtained in accordance with procedures well known to the person of ordinary skill in the art.
General chromatography information
HPLC method H1 (RtH1):
HPLC-column dimensions 3.0 x 30 mm
HPLC-column type: Zorbax SB-C18, 1.8 μm
HPLC-eluent: A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
HPLC-gradient: 30-100 % B in 3.25 min, flow = 0.7 ml / min
LCMS method H2 (RtH2):
HPLC-column dimensions: 3.0 x 30 mm
HPLC-column type: Zorbax SB-C18, 1.8 μm
HPLC-eluent: A) water + 0.05 Vol.-% TFA, B) ACN + 0.05 Vol.-% TFA
HPLC-gradient: 10-100 % B in 3.25 min, flow = 0.7 ml / min
UPLCMS method H3 (RtH3):
HPLC-column dimensions: 2.1 x 50 mm
HPLC-column type: Acquity UPLC HSS T3, 1.8 μm
HPLC-eluent: A) water + 0.05 Vol.-% formic acid + 3.75 mM ammonium
acetate B) ACN + 0.04 Vol.-% formic acid
HPLC-gradient: 2-98 % B in 1.4 min, 98% B 0.75 min, flow = 1.2 ml / min
HPLC-column temperature: 50 °C
LCMS method H4 (RtH4):
HPLC-column dimensions: 3.0 x 30 mm
HPLC-column type: Zorbax SB-C18, 1.8 μm
HPLC-eluent: A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
HPLC-gradient: 70 - 100 % B in 3.25 min, flow = 0.7 ml / min
LCMS method H5 (RtH5):
HPLC-column dimensions: 3.0 x 30 mm
HPLC-column type: Zorbax SB-C18, 1.8 μm HPLC-eluent: A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
HPLC-gradient: 80 - 100 % B in 3.25 min, flow = 0.7 ml / min
LCMS method H6 (RtH6):
HPLC-column dimensions: 3.0 x 30 mm
HPLC-column type: Zorbax SB-C18, 1.8 μm
HPLC-eluent: A) water + 0.05 Vol.-% TFA; B) ACN + 0.05 Vol.-% TFA
HPLC-gradient: 40 - 100 % B in 3.25 min, flow = 0.7 ml / min a) 2-Bromo-5-fluoro-4-triethylsilanyl-pyridine
A solution of diisopropylamine (25.3 g, 250 mmol) in 370 ml THF was cooled with a dry-ice acetone bath at -75 °C. BuLi (100 ml, 250 mmol, 2.5 M in hexanes) was added dropwise while maintaining the temperature below -50 °C. After the temperature of the mixture had reached -75 °C again, a solution of 2-bromo-5-fluoropyridine (36.7 g, 208 mmol) in 45 ml THF was added dropwise. The mixture was stirred for 1 h at -75 °C. Triethylchlorosilane (39.2 g, 260 mmol) was added quickly. The temperature stayed below -50 °C. The cooling bath was removed and the reaction mixture was allowed to warm to -15 °C, poured onto aq. NH4CI (10%). TBME was added and the layers were separated. The organic layer was washed with brine, dried with MgSO4.H2O, filtered and evaporated to give a brown liquid which was distilled at 0.5 mm Hg to yield the title compound as a slightly yellow liquid (b.p. 105-1 11 °C). HPLC: RtH4 = 2.284 min; ESIMS: 290, 292 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCl3): 8.14 (s, 1 H), 7.40 (d, 1 H), 1.00-0.82 (m, 15H). b) 1-(6-Bromo-3-fluoro-4-triethylsilanyl-pyridin-2-yl)-ethanone
A solution of diisopropylamine (25.4 g, 250 mmol) in 500 ml THF was cooled to -75 °C. BuLi (100 ml, 250 mmol, 2.5 M in hexanes) was added dropwise while maintaining the temperature below -50 °C. After the reaction temperature had reached -75 °C again, a solution of 2-bromo-5-fluoro-4-triethylsilanyl-pyridine (56.04 g, 193 mmol) in 60 ml THF was added dropwise. The mixture was stirred in a dry ice bath for 70 minutes. N,N- dimethylacetamide (21.87 g, 250 mmol) was added quickly, the reaction temperature rose to -57 °C. The reaction mixture was stirred in a dry ice bath for 15 min and then allowed to warm to -40 °C. It was poured on a mixture of 2M aq. HCI (250 ml, 500 mmol), 250 ml water and 100 ml brine. The mixture was extracted with TBME, washed with brine, dried over MgSO4.H2O, filtered and evaporated to give a yellow oil which was purified on a silica gel column by eluting with hexane/0-5% TBME to yield 58.5 g of the title compound as a yellow liquid. TLC (Hex/TBME 99/1): Rf = 0.25; HPLC: RtH4 = 1.921 min; ESIMS: 332, 334 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCl3): 7.57 (d, 1 H), 2.68 (s, 3H), 1.00-0.84 (m, 15H). c) (S)-2-(6-Bromo-3-fluoro-4-triethylsilanyl-pyridin-2-yl)-2-trimethylsilanyloxy- propionitrile
At first, the catalyst solution was prepared by dissolving water (54 mg, 3.00 mmol) in 100 ml dry DCM (≤ 0.001 % water). This wet DCM (44 ml, 1.32 mmol water content) was added to a well stirred solution of titanium(IV) butoxide (500 mg, 1.47 mmol) in 20 ml dry DCM. The resulting clear solution was refluxed for 1 h. This solution was then cooled to rt and 2,4-di- tert-butyl-6-{[(E)-(S)-1-hydroxymethyl-2-methyl-propylimino]-methyl}-phenol [CAS 155052- SI -6] (469 mg, 1.47 mmol) was added. The resulting yellow solution was stirred at rt for 1 h. This catalyst solution (0.023 M, 46.6 ml, 1.07 mmol) was added to a solution of 1-(6-bromo- 3-fluoro-4-triethylsilanyl-pyridin-2-yl)-ethanone (35.53 g, 107 mmol) and trimethylsilyl cyanide (12.73 g, 128 mmol) in 223 ml dry DCM. The mixture was stirred for 2 days and evaporated to give 47 g of the crude title compound as an orange oil. HPLC: RtH5 = 2.773 min; ESIMS: 431 , 433 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCl3): 7.46 (d, 1 H), 2.04 (s, 3H), 1.00 (t, 9H), 1.03-0.87 (m, 15H), 0.20 (s, 9H). d) (R)-1-Amino-2-(6-bromo-3-fluoro-4-triethylsilanyl-pyridin-2-yl)-propan-2-ol hydrochloride
Borane dimethyl sulfide complex (16.55 g, 218 mmol) was added to a solution of crude (S)- 2-(6-bromo-3-fluoro-4-triethylsilanyl-pyridin-2-yl)-2-trimethylsilanyloxy-propionitrile (47 g, 109 mmol) in 470 ml THF. The mixture was refluxed for 2 h. The heating bath was removed and the reaction mixture was quenched by careful and dropwise addition of MeOH. After the evolution of gas had ceased, aq. 6M HCI (23.6 ml, 142 mmol) was added slowly. The resulting solution was evaporated and the residue was dissolved in MeOH and evaporated (twice) to yield 44.5 g of a yellow foam, pure enough for further reactions. HPLC: RtH1 =
2.617 min; ESIMS: 363, 365 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCI3): 7.93 (s, br, 3H), 7.53 (d, 1 H), 6.11 (s, br, 1 H), 3.36-3.27 (m, 1 H), 3.18-3.09 (m, 1 H), 1.53 (s, 3H), 0.99-0.81 (m, 15H). e) (R)-N-(2-(6-bromo-3-fluoro-4-(triethylsilyl)pyridin-2-yl)-2-hydroxypropyl)-4- nitrobenzenesulfonamide
To a solution of crude (R)-1-amino-2-(6-bromo-3-fluoro-4-triethylsilanyl-pyridin-2-yl)-propan- 2-ol hydrochloride (43.5 g, 109 mmol) in 335 ml THF was added a solution of NaHCO3 (21.02 g, 250 mmol) in 500 ml water. The mixture was cooled to 0-5 °C and a solution of 4- nitrobenzenesulfonyl chloride (26.5 g, 120 mmol) in 100 ml THF was added in a dropwise. The resulting emulsion was stirred overnight while allowing the temperature to reach rt. The mixture was extracted with TBME. The organic layer was dried with MgSO4.H2O, filtered and evaporated to give an orange resin which was purified on a silca gel column by eluting with Hexanes/10-20% EtOAc to yield 37.56 g of the title compound as a yellow resin. TLC (Hex/EtOAc 3/1): Rf = 0.34; HPLC: RtH4 = 1.678 min; ESIMS: 548, 550 [(M+H)+, 1 Br]; 1 H- NMR (400 MHz, DMSO-d6): 8.40 (d, 2H), 8.06 (t, 1 H), 7.97 (d, 2H), 7.45 (d, 1 H), 5.42 (s,
1 H), 3.23 (d, 2H), 1.44 (s, 3H) 0.97-0.81 (m, 15H); Chiral HPLC (Chiralpak AD-H 1213, UV 210 nm): 90% ee. f) 6-Bromo-3-fluoro-2-[(S)-2-methyl-1-(4-nitro-benzenesulfonyl)-aziridin-2-yl]-4- triethylsilanyl-pyridine
A solution of triphenylphosphine (21.55 g, 82 mmol) and (R)-N-(2-(6-bromo-3-fluoro-4- (triethylsilyl)pyridin-2-yl)-2-hydroxypropyl)-4-nitrobenzenesulfonamide (37.56 g, 69 mmol) in 510 ml THF was cooled to 4 °C. A solution of diethyl azodicarboxylate in toluene (40% by weight, 38.8 g, 89 mmol) was added in a dropwise while maintaining the temperature below 10 °C. The cooling bath was removed and the reaction mixture was stirred at rt for 1 h. The reaction mixture was diluted with approx. 1000 ml toluene and THF was removed by evaporation at the rotavap. The resulting toluene solution of crude product was pre-purified on a silca gel column by eluting with hexanes/5-17% EtOAc. Purest fractions were combined, evaporated and crystallized from TBME/hexane to yield 29.2 g of the title compound as white crystals. HPLC: RtH4 = 2.546 min; ESIMS: 530, 532 [(M+H)+, 1 Br]; 1H- NMR (400 MHz, CDCI3): 8.40 (d, 2H), 8.19 (d, 2H), 7.39 (d, 1 H), 3.14 (s, 1 H), 3.02 (s, 1 H), 2.01 (s, 3H) 1.03 - 0.83 (m, 15H); a[D] -35.7° (c = 0.97, DCM). g) 6-Bromo-3-fluoro-2-[(S)-2-methyl-1-(4-nitro-benzenesulfonyl)-aziridin-2-yl]-pyridine
Potassium fluoride (1.1 g, 18.85 mmol) was added to a solution of 6-bromo-3-fluoro-2-[(S)-2- methyl-1-(4-nitro-benzenesulfonyl)-aziridin-2-yl]-4-triethylsilanyl-pyridine (5 g, 9.43 mmol) and AcOH (1.13 g, 9.43 mmol) in 25 ml THF. DMF (35 ml) was added and the suspension was stirred for 1 h at rt. The reaction mixture was poured onto a mixture of sat. aq. NaHCC>3 and TBME. The layers were separated and washed with brine and TBME. The combined organic layers were dried over MgSO4.H2O, filtered and evaporated to give a yellow oil which was crystallized from TBME/hexane to yield 3.45 g of the title compound as white crystals. HPLC: RtH6 = 2.612 min; ESIMS: 416, 418 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCl3): 8.41 (d, 2H), 8.19 (d, 2H), 7.48 (dd, 1 H), 7.35 (t, 1 H), 3.14 (s, 1 H), 3.03 (s, 1 H), 2.04 (s, 3H); a[D] -35.7° (c = 0.89, DCM). h) (R)-2-[(R)-2-(6-Bromo-3-fluoro-pyridin-2-yl)-2-(4-nitro-benzenesulfonylamino)- propoxy]-3,3,3-trifluoro-2-methyl-propionic acid ethyl ester A solution of (R)-3,3,3-trifluoro-2-hydroxy-2-methyl-propionic acid ethyl ester (11.93 g, 64.1 mmol) in DMF (158 ml) was evacuated/flushed with nitrogen twice. A solution of KOtBu (6.21 g, 55.5 mmol) in DMF (17 ml) was added dropwise while maintaining a reaction temperature of ca 25 °C using cooling with a water bath. After 15 min solid 6-bromo-3-fluoro-2-[(S)-2- methyl-1-(4-nitro-benzenesulfonyl)-aziridin-2-yl]-pyridine (17.78 g, 42.7 mmol) was added and stirring was continued for 3 h. The reaction mixture was poured onto a mixture of 1M HCI (56 ml), brine and TBME. The layers were separated, washed with brine and TBME.
The combined organic layers were dried over MgSO4.H2O, filtered and evaporated. The crude reaction product was purified via chromatography on silica gel (hexanes/25-33% TBME) to yield 16.93 g of the title compound as a yellow resin that was contaminated with an isomeric side-product (ratio 70:30 by 1H-NMR).
HPLC: RtH6 = 2.380 min; ESIMS: 602, 604 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCl3): 8.32 (d, 2H), 8.07 (d, 2H), 7.46 - 7.41 (m, 1 H), 7.30 - 7.23 (m, 1 H), 6.92 (s, 1 H), 3.39 - 4.30 (m, 2H), 3.95 (d, 1 H), 3.84 (d, 1 H), 1.68 (s, 3H), 1.56 (s, 3H), 1.40-1.34 (m, 3H) + isomeric side- product. i) (R)-2-[(R)-2-(6-Bromo-3-fluoro-pyridin-2-yl)-2-(4-nitro-benzenesulfonylamino)- propoxy]-3,3,3-trifluoro-2-methyl-propionamide
A solution of (R)-2-[(R)-2-(6-bromo-3-fluoro-pyridin-2-yl)-2-(4-nitro-benzenesulfonylamino)- propoxy]-3,3,3-trifluoro-2-methyl-propionic acid ethyl ester (16.93 g, 28.1 mmol) in a
NH3/MeOH (7M, 482 ml) was stirred at 50 °C in a sealed vessel for 26 h. The reaction mixture was evaporated and the residue was crystallized from DCM to yield 9.11 g of the title compound as colorless crystals.
HPLC: RtH6 = 2.422 min; ESIMS: 573, 575 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCl3): 8.33 (d, 2H), 8.06 (d, 2H), 7.42 (dd, 1 H), 7.30 - 7.26 (m, 1 H), 7.17 (s, br, 1 H), 6.41 (s, 1 H), 5.57 (s, br, 1 H), 4.15 (m, 2H), 1.68 (s, 3H), 1.65 (s, 3H). j) N-[(R)-1-(6-Bromo-3-fluoro-pyridin-2-yl)-2-((R)-1-cyano-2,2,2-trifluoro-1-methyl- ethoxy)-1-methyl-ethyl]-4-nitro-benzenesulfonamide
A suspension of (R)-2-[(R)-2-(6-bromo-3-fluoro-pyridin-2-yl)-2-(4-nitro- benzenesulfonylamino)-propoxy]-3,3,3-trifluoro-2-methyl-propionamide (8.43 g, 14.70 mmol) and triethylamine (5.12 ml, 36.8 mmol) in 85 ml DCM was cooled to 0-5 °C. Trifluoroacetic anhydride (2.49 ml, 17.64 mmol) was added dropwise over 30 min. Additional triethylamine (1.54 ml, 11.07 mmol) and trifluoroacetic anhydride (0.75 ml, 5.29 mmol) were added to complete the reaction. The reaction mixture was quenched by addition of 14 ml aqueous ammonia (25%) and 14 ml water. The emulsion was stirred for 15 min, more water and DCM were added and the layers were separated. The organic layer was dried with MgSO4 H2O, filtered and evaporated. Purification by column chromatography on a silica gel (hexanes/10- 25% EtOAc) gave 8.09 g of the title compound as a yellow resin.
HPLC: RtH6 = 3.120 min; ESIMS: 555, 557 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, CDCl3): 8.35 (d, 2H), 8.11 (d, 2H), 7.50 (dd, 1 H), 7.32 (dd, 1 H), 6.78 (s, 1 H), 4.39 (d 1 H), 4.22 (d, 1 H),
1.68 (s, 6H). k) (2R, 5R)-5-(6-Bromo-3-fluoro-pyridin-2-yl)-2,5-dimethyl-2-trifluoromethyl-5, 6-dihydro- 2H-[1 ,4]oxazin-3-ylamine
A solution of N-[(R)-1-(6-bromo-3-fluoro-pyridin-2-yl)-2-((R)-1-cyano-2,2,2-trifluoro-1-methyl- ethoxy)-1-methyl-ethyl]-4-nitro-benzenesulfonamide (9.18 g, 16.53 mmol) and N- acetylcysteine (5.40 g, 33.10 mmol) in 92 ml ethanol was evacuated and flushed with nitrogen. K2CO3 (4.57 g, 33.1 mmol) was added and the mixture was stirred at 80 °C for 3 days. The reaction mixture was concentrated in vacuo to about 1/4 of the original volume and partitioned between water and TBME. The organic layer was washed with 10% aq. K2CO3 solution, dried over Na2SC>4, filtered and evaporated to give a yellow oil. Column chromatography on silica (hexanes/14-50% (EtOAc:MeOH 95:5)) gave 4.55 g of the title compound as an off-white solid.
HPLC: RtH2 = 2.741 min; ESIMS: 370, 372 [(M+H)+, 1 Br]; 1H-NMR (400 MHz, DMSO-d6): 7.71 - 7.62 (m, 2H), 5.97 (s, br, 2H), 4.02 (d 1 H), 3.70 (d, 1 H), 1.51 (s, 3H), 1.47 (s, 3H).
L) (2 R, 5R)-5-(6-Amino-3-fluoro-pyridin-2-yl)-2,5-dimethyl-2-trifluoromethyl-5, 6-dihydro- 2H-[1 ,4]oxazin-3-yl amine
A glass/stainless steel autoclave was purged with nitrogen, CU2O (0.464 g, 3.24 mmol), ammonia (101 ml, 25%, aq., 648 mmol, 30 equivalents) and (2R,5R)-5-(6-Bromo-3-fluoro- pyridin-2-yl)-2,5-dimethyl-2-trifluoromethyl-5,6-dihydro-2H-[1 ,4]oxazin-3-ylamine (8 g, 21.6 mmol) in ethylene glycol (130 ml) was added. The autoclave was closed and the suspension heated up to 60 °C and the solution was stirred for about 48 hours (max. pressure 0.7 bar, inside temperature 59-60 °C). The reaction mixture was diluted with ethyl acetate and water. The organic phase was washed with water and 4 times with 12% aq. ammonia and finally with brine, dried over sodium sulfate, filtered and evaporated. The crude product (7 g, containing some ethylen glycol, quantitative yield) was used in the next step without further purification.
HPLC: RtH3 = 0.60 min; ESIMS: 307 [(M+H)+] m) [(2 R, 5R)-5-(6-Amino-3-fluoro-pyridin-2-yl)-2,5-dimethyl-2-trifluoromethyl-5,6- dihydro-2H-[1 ,4]oxazin-3-yl]-carbamic acid tert-butyl ester A solution of (2 R, 5R)-5-(6-amino-3-fluoro-pyridin-2-yl)-2,5-dimethyl-2-trifluoromethyl-5,6- dihydro-2H-[1 ,4]oxazin-3-yl amine (6.62 g, 21.6 mmol), Boc2O (4.72 g, 21.6 mmol) and Hunig’s base (5.66 ml, 32.4 mmol) in dichloromethane (185 ml) was stirred at rt for 18 hours. The reaction mixture was washed with sat. aq. NaHCO3 and brine. The aqueous layers were back extracted with dichloromethane and the combined organic layers were dried over sodium sulfate, filtered and evaporated to give a light green solid (14 g). The crude product was chromatographed over silicagel (cyclohexane: ethyl acetate 95:5 to 60:40) to afford 7.68 g of the title compound.
TLC (cyclohexane: ethyl acetate 3: 1): Rf = 0.21 ; HPLC: RtH3= 1.14 min; ESIMS: 408
[(M+H)+]; 1H-NMR (400 MHz, CDCI3): 1 1.47 (br. s, 1 H), 7.23 (dd, J=10.42, 8.78 Hz, 1 H), 6.45 (dd, J= 8.78, 2.64 Hz, 1 H), 4.50 (br. s, 2H), 4.32 (d, J= 2.38 Hz, 1 H), 4.10 (d, J=11.80 Hz, 1 H), 1.69 (s, 3H, CH3), 1.65 (s, 3H, CH3), 1.55 (s, 9H). n) ((2 R, 5R)-5-{6-[(3-Chloro-5-trifluoromethyl-pyridine-2-carbonyl)-amino]-3-fluoro- pyridin-2-yl}-2,5-dimethyl-2-trifluoromethyl-5,6-dihydro-2H-[1,4]oxazin-3-yl)-carbamic acid tert-butyl ester
A mixture of [(2 R, 5R)-5-(6-amino-3-fluoro-pyridin-2-yl)-2,5-dimethyl-2-trifluoromethyl-5,6- dihydro-2H-[1 ,4]oxazin-3-yl]-carbamic acid tert-butyl ester (3.3 g, 8.12 mmol), 3-chloro-5- trifluoromethylpicolinic acid (2.2 g, 9.74 mmol), HOAt (1.99 g, 14.62 mmol) and EDC hydrochloride (2.33 g, 12.18 mmol) was stirred in DMF (81 ml) at rt for 48 hours. The reaction mixture was diluted with ethyl acetate and washed with water and brine, dried over sodium sulfate, filtered and evaporated. The crude product (12 g) was chromatographed over silicagel (cyclohexane to cyclohexane: ethyl acetate 1 : 1) to yield 5.2 g of the title compound.
TLC (silica, cyclohexane:ethyl acetate 3: 1): Rf=0.47; HPLC: RtH3 = 1.40 min; ESIMS: 615, 616 [(M+H)+, 1 Cl]; 1H-NMR (400 MHz, CDCI3): 11.68 (s, 1 H), 10.41 (s, 1 H), 8.81 (dd, J=1.82, 0.69 Hz, 1 H), 8.45 (dd, J=8.91 , 3.14 Hz, 1 H), 8.19 (dd, J=1.88, 0.63 Hz, 1 H), 7.59 (dd,
J= 9.79, 9.16
Hz, 1 H), 4.38 (d, J=2.13 Hz, 1 H), 4.18 (d, J=1 1.80 Hz, 1 H), 1.75 (s, 3H), 1.62 (s, 3H), 1.60 (s, 9H). o) 3-Chloro-5-trifluoromethyl-pyridine-2-carboxylic acid [6-((3R,6R)-5-amino-3,6- dimethyl-6-trifluoromethyl-3,6-dihydro-2H-[1,4]oxazin-3-yl)-5-fluoro-pyridin-2-yl]-amide
A mixture of ((2 R, 5R)-5-{6-[3-chloro-5-trifluoromethyl-pyridine-2-carbonyl)-amino]-3-fluoro- pyridin-2-yl}-2,5-dimethyl-2-trifluoromethyl-5,6-dihydro-2H-[1 ,4]oxazin-3-yl)-carbamic acid tert-butyl ester (4.99 g, 8.13 mmol) and TFA (6.26 ml, 81 mmol) in dichloromethane (81 ml) was stirred at rt for 18 hours. The solvent was evaporated and the residue diluted with a suitabable organic solvent, such as ethyl acetate and aq. ammonia. Ice was added and the organic phase was washed with water and brine, dried over sodium sulfate, filtered and evaporated to yield 3.78 g of the title compound.
HPLC: RtH3 = 0.87 min; ESIMS: 514, 516 [(M+H)+, 1CI]; 1H-NMR (400 MHz, DMSO-d6): d 11.11 (s, 1 H), 9.06 (s, 1 H), 8.69 (s, 1 H), 8.13 (dd, J = 8.8, 2.6 Hz, 1 H), 7.80 - 7.68 (m, 1 H), 5.88 (br. s, 2H), 4.12 (d, J = 11.5 Hz, 1 H), 3.72 (d, J = 11.4 Hz, 1 H), 1.51 (s, 3H), 1.49 (s, 3H). p) Crystallisation procedure for free base Compound 1 (Form A) 1 wt of Compound 1 was dissolved in 5.11 wt of IPAc at 70-80 °C. The solution was filtered
(filter <2μm) and then 1.52 wt of n-heptane added. The solution was cooled to 55 °C, and seeded with 0.5% w/w of Compound 1. The suspension was held at 55 °C for 30-60 mins and then cooled to 35 °C over 2 hours. The suspension was aged for 1 hour and then 8.2 wt of n-heptane were added over 3 hours. The suspension was aged for 1 hour and then cooled to 0-5 °C over 2 hours and aged for at least 2 hours. The suspension was filtered under vacuum, and the cake washed with 10/90 w/w isopropyl acetate/n-heptane. The cake was dried under vacuum at 40-45 °C until dry, to produce Form A.
Example 2: XRPD analysis of free base Compound 1 Form A
Crystalline free base Compound 1 Form A was analysed by XRPD and the ten most characteristic peaks are shown in Table 1 (see also Figure 1).
Table 1
Figure imgf000035_0001
The five most characteristic peaks of crystalline Compound 1 in free Form A are at: 10.7, 14.8, 18.7, 19.5 and 21.4° 2-theta. The three most characterictic peaks of crystalline Compound 1 in free Form A are at: 10.7, 14.8 and 19.5° 2-theta.
X-ray powder diffraction (XRPD) analysis was performed using a Bruker D8 Advance x-ray diffractometer in reflection geometry. Measurements were taken at about 30 kV and 40 mA under the following conditions:
Table 2
Figure imgf000036_0001
The X-ray diffraction pattern was recorded between 2° and 40° (2-theta) with CuKα radiation for identification of the whole pattern. Example 3: Micronization procedure and XRPD analysis for Compound 1 free Form A
Crystalline Compound 1 free Form A was micronized according to the following method:
A spiral jet-milling instrument was used with a ring of 50 mm diameter. The carrier gas was nitrogen and the energy was targeted at 1800kJ/kg (cumulative parameter considering injector and grinding nozzle number and diameter, injector and grinding nozzle pressure, and feed rate according to Midoux N et al, 1999).
Micronized Crystalline Form A was analysed by XRPD and the ten most characteristic peaks are shown in Table 3 (see also Figure 2).
Table 3
Figure imgf000036_0002
The five most characteristic peaks of micronized crystalline Compound 1 in free Form A are at: 12.1 , 15.9, 18.5, 19.4, 24.0° 2-theta.
The three most characterictic peaks of micronized crystalline Compound 1 in free Form A are at: 12.1 , 19.4, 24.0° 2-theta. X-ray powder diffraction (XRPD) analysis was performed using a Bruker D8 Advance x-ray diffractometer in reflection geometry. Measurements were taken at about 30 kV and 40 mA under the conditions shown in Table 4.
Table 4
Figure imgf000037_0001
The X-ray diffraction pattern was recorded between 2° and 40° (2-theta) with CuKα radiation for identification of the whole pattern.
Example 4: DSC analysis of Compound 1 free Form A
Crystalline Compound 1 free Form A was analysed by differential scanning calorimetry (DSC) using a Q1000 Diffraction Scanning Calorimeter from TA Instruments and found to have an endotherm at about 171 °C when analysed at a heating rate of 10 °C per minute, see Figure 3.
Example 5: Preparation of Compound 1 hemifumarate hemihvdrate salt
Small scale preparation
300 mg of free base Compound 1 was dissolved in 3mL acetonitrile and 33.9 mg fumaric acid (0.5 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Large scale preparation
1 wt of free base Compound 1 was dissolved in 12 vol of acetone at 30°C. 0.5 equivalent of fumaric acid was dissolved into 12 vol. of EtOH. 1 wt of the ethanol fumaric acid solution was added to the Compound 1 solution and seeded with 0.5% w/w of Compound 1 fumarate salt. The suspension was aged for 1 hour and the remaining ethanol solution added over 6 hours. The suspension was aged at 30°C until filtration. The solid was filtered and dried under full vacuum at 45°C.
Example 6: XRPD analysis of Compound 1 hemifumarate hemihydrate salt Crystalline Compound 1 in hemifumarate hemihydrate salt form, prepared using the large scale procedure described in Example 5, was analysed by XRPD and the ten most characteristic peaks are shown in Table 5 (see also Figure 4).
Table 5
Figure imgf000038_0001
The five most characteristic peaks of crystalline Compound 1 in hemifumarate hemihydrate salt form are at: 11.9, 13.9, 16.2, 17.3 and 19.9° 2-theta.
Alternatively, crystalline Compound 1 in hemifumarate hemihydrate salt form may be characterised by peaks at: 11.9, 19.9, 23.0, 27.9 and 30.8° 2-theta.
The three most characterictic peaks of crystalline Compound 1 in hemifumarate hemihydrate salt form are at: 11.9, 13.9 and 19.9° 2-theta.
Alternatively, crystalline Compound 1 in hemifumarate hemihydrate salt form may be characterised by peaks at: 23.0, 27.9 and 30.8° 2-theta.
X-ray powder diffraction (XRPD) analysis was performed using a Bruker D8 Advance x-ray diffractometer in reflection geometry. Measurements were taken at about 30 kV and 40 mA under the conditions shown in Table 6. Table 6
Figure imgf000039_0001
The X-ray diffraction pattern was recorded between 2° and 40° (2-theta) with CuKα radiation for identification of the whole pattern. Example 7: XRPD analysis of micronized Compound 1 hemifumarate hemihvdrate salt form
Crystalline Compound 1 in hemifumarate hemihydrate salt form, prepared using the large scale procedure described in Example 5, was micronized as described in Example 3, analysed by XRPD using the methodology and equipment described in Example 3, and the ten most characteristic peaks are shown in Table 7 (see also Figure 5).
Table 7
Figure imgf000039_0002
The five most characteristic peaks of micronized Compound 1 in hemifumarate hemihydrate salt form are at: 9.1 , 11.8, 14.0, 16.1 and 19.0° 2-theta.
The three most characterictic peaks of micronized Compound 1 in f hemifumarate hemihydrate salt form are at: 14.0, 16.1 and 19.0° 2-theta.
Example 8: DSC analysis of Compound 1 hemifumarate hemihvdrate salt
Crystalline Compound 1 in hemifumarate hemihydrate salt form, prepared using the large scale procedure described in Example 5, was analysed by DSC using a Discovery(R) instrument from TA Instruments and found to have an endotherm at about 239 °C when analysed at a heating rate of 10 °C per minute, see Figure 6.
Example 9: Preparation of Compound 1 hemisuccinate hemihvdrate salt
Small scale preparation
300 mg of free base Compound 1 was dissolved in 3 mL ethanol and 69 mg succinic acid (1 equivalent) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m. at 25°C and dried at 25°C for 6 hours.
Large scale preparation
1 wt of free base Compound 1 and 1 equivalent of succinic acid were dissolved in 12 vol of MeOH at 60°C. The solution was cooled to 45°C and seeded with 0.5% w/w of Compound 1 succinate salt. The suspension was aged for 1 hour and cooled to 25°C over 2 hours. 13.3 vol of water was added over 2 hours. The solid was then filtered and dried under full vacuum at 45°C.
Example 10: XRPD analysis of Compound 1 hemisuccinate hemihvdrate salt
Crystalline Compound 1 in hemisuccinate hemihydrate salt form, prepared using the large scale procedure described in Example 9, was analysed by XRPD using the methodology and equipment described in Example 6 and the ten most characteristic peaks are shown in Table 8 (see also Figure 7).
Table 8
Figure imgf000040_0001
The five most characteristic peaks of crystalline Compound 1 in hemisuccinate hemihydrate salt form are at: 12.0, 14.0, 17.3, 17.6 and 20.1° 2-theta. The three most characterictic peaks of crystalline Compound 1 in hemisuccinate hemihydrate salt form are at: 12.0, 14.0 and 20.1° 2-theta.
Example 11 : XRPD analysis of micronized Compound 1 hemisuccinate hemihvdrate salt Crystalline Compound 1 in hemisuccinate hemihydrate salt form, prepared using the large scale procedure described in Example 9, was micronized as described in Example 3, analysed by XRPD using the methodology and equipment described in Example 3 and the ten most characteristic peaks are shown in Table 9 (see also Figure 8).
Table 9
Figure imgf000041_0001
The five most characteristic peaks of micronized Compound 1 in hemisuccinate hemihydrate salt form are at: 10.1 , 14.5, 15.5, 19.8 and 23.3° 2-theta.
The three most characterictic peaks of micronized Compound 1 in hemisuccinate hemihydrate salt form are at: 10.1 , 15.5 and 19.8° 2-theta.
Example 12: DSC analysis of Compound 1 hemisuccinate hemihvdrate salt Crystalline Compound 1 in hemisuccinate hemihydrate salt form, prepared using the large scale procedure described in Example 9, was analysed by DSC using a Discovery(R) instrument from TA instruments and found to have an endotherm at about 217 °C when analysed at a heating rate of 10 °C per minute, see Figure 9.
Example 13: Preparation of Compound 1 hemi-L-tartrate hemihvdrate salt Small scale preparation
300mg free base Compound 1 was dissolved in 3 mL acetonitrile and 43.8 mg L-tartaric acid (0.5 equivalent) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Large scale preparation
1 wt of free base Compound 1 was dissolved in 12 vol of acetone at 30°C. 0.5 equivalent of L-tartaric acid was dissolved in 12 vol of water. 5 g of the L-tartaric aqueous solution was added to the Compound 1 solution, which was then seeded with 0.5% w/w of Compound 1 L-tartrate salt. The suspension was aged for 1 hour, then the remainder of the aqueous solution added over 6 hours. The suspension was aged at 30°C until filtration. The solid was dried under full vacuum at 45°C. Example 14: XRPD analysis of Compound 1 hemi-L-tartrate hemihydrate salt
Crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form, prepared using the large scale procedure described in Example 13, was analysed by XRPD using the methodology and equipment described in Example 6 and the ten most characteristic peaks are shown in Table 10 (see also Figure 10). Table 10
Figure imgf000042_0001
The five most characteristic peaks of crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form are at: 12.2, 14.0, 17.7, 19.8 and 20.4° 2-theta.
The three most characterictic peaks of crystalline Compound 1 in hemi-L-tartrate
hemihydrate salt form are at: 14.0, 17.7 and 19.8° 2-theta. Example 15: XRPD analysis of micronized Compound 1 hemi-L-tartrate hemihydrate salt
Crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form, prepared using the large scale procedure described in Example 13, was micronized as described in Example 3, analysed by XRPD using the methodology and equipment described in Example 3 and the ten most characteristic peaks are shown in Table 11 (see also Figure 11).
Table 11
Figure imgf000043_0001
The five most characteristic peaks of micronized Compound 1 in hemi-L-tartrate hemihydrate salt form are at: 7.1 , 13.0, 17.1 , 19.8 and 20.3° 2-theta.
The three most characterictic peaks of micronized Compound 1 in hemi-L-tartrate hemihydrate salt form are at: 13.0, 17.1 and 20.3° 2-theta.
Example 16: DSC analysis of Compound 1 hemi-L-tartrate hemihydrate salt
Crystalline Compound 1 in hemi-L-tartrate hemihydrate salt form, prepared using the large scale procedure described in Example 13, was analysed by DSC using Discovery(R) instrument from TA instruments and found to have an endotherm at about 253 °C when analysed at a heating rate of 10 °C per minute, see Figure 12.
Example 17: Stoichiometric analysis of Compound 1 fumarate. succinate and tartrate salt forms The stoichiometric composition of the Compound 1 salt forms was examined by proton NMR spectroscopy which confirmed that the stoichiometry of the ratio between Compound 1 and the salt forming agent was 2:1 for the succinate, fumarate and tartrate salts.
The solvated/hydrated nature of the different salts was examined experimentally by analysis of residual organic solvent, thermogravimetric analysis of loss on drying, and storage behaviour at different relative humidities in combination with dynamic vapour sorption. In addition, for the succinate and fumarate salts, the natures of the solvents/hydrates were also supported by structural investigation based on single crystal X-ray data analysis. The results of these orthogonal techniques provide evidence that the fumarate, succinate and tartrate salt forms of Compound 1 obtained as described above contain water in their crystal structures consistent with a composition ratio of 2:1 :1 (Compound 1 : salt coformer: water).
Example 18: Milling properties of Compound 1 free base form and hemifumarate hemihvdrate, hemisuccinate hemihvdrate and hemi-L-tartrate hemihvdrate salt forms The behaviour of the free base and hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salt forms were evaluated during jet milling. A spiral jet milling instrument was used with a ring of 50mm diameter. The carrier gas was nitrogen and the energy was targeted at 1800kJ/kg (cumulative parameter considering injector and grinding nozzle number and diameter, injector and grinding nozzle pressure, and feed rate according to Midoux N et ai, 1999). The percentage yields upon micronization are shown in Table 12 below.
Table 12
Figure imgf000044_0001
* Process stopped after delivery of 40g free base Compound 1 into mill due to clogging of grinding chamber. In terms of micronization capacity, the free form was not processable and blocked the mill. However, yields higher than 90% were achieved for the three salts. The yield and processability upon micronization at small scale indicates unambiguously the improved properties of the three salts compared to the free form of Compound 1.
Example 19: Preparation of further alternative salt forms of Compound 1 HCI: 300mg free base Compound 1 was added to 3mL MTBE and 1 eq HCI (6. ON) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
HCI, hydrate: 300mg free base Compound 1 was dissolved in 3mL ethanol and 1 eq HCI (6. ON) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours.
The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
H2SO4: 300mg free base Compound 1 was dissolved in 3mL IPAc and 1 eq H2S04 (98%) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
H3PO4: 300mg free base Compound 1 was added to 3mL MTBE and 1 eq H3P04 (85%) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours.
The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Acetate: 300mg free base Compound 1 was dissolved in 3mL ethanol and 35.3mg acetic acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Citrate: 300mg free base Compound 1 was dissolved in 3mL IPAc and 56.1 mg citric acid (0.5 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours.
The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Butyrate: 300mg free base Compound 1 was dissolved in 3mL IPAc and 51.5mg butyric acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
D-glucuronate: 300mg free base Compound 1 was dissolved in 3mL acetonitrile and 113.4mg D-glucuronic acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Galactarate: 300mg free base Compound 1 was dissolved in 3mL ethanol and 61.3mg galactaric acid (0.5 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours. Glutarate: 300mg free base Compound 1 was added to 3mL MTBE and 77.1 mg glutaric acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
L-aspartate: 300mg free base Compound 1 was dissolved in 3mL ethanol and 77.7mg L- aspartic acid (1 eq) was added to the solution at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Pyruvate: 300mg free base Compound 1 was added to 3mL MTBE and 51.4mg pyruvic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Maleate: 300mg free base Compound 1 was added to 3mL MTBE and 67.8mg maleic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Glycolate: 300mg free base Compound 1 was added to 3mL MTBE and 44.4mg glycolic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
N-acetylglicine: 300mg free base Compound 1 was added to 3mL MTBE and 68.4mg N- acetylglycine (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Propionate: 300mg free base Compound 1 was added to 3mL MTBE and 43.3mg propionic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours. D-quinate: 300mg free base Compound 1 was dissolved in 3mL IPAc and 112.2mg D-quinic acid (1 eq) was added to the suspension at 50°C. The mixture was stirred at 50°C for 4 hours and cooled to 25°C within a 4 hour period. The mixture was kept stirring at 25°C for 14 hours. The solids were separated by centrifuge at 4000 r.p.m.at 25°C and dried at 25°C for 6 hours.
Example 20: Bulk stability of Compound 1 salts in comparison to the free form after three weeks stress exposure at 80°C/75% relative humidity and in the presence of 90% sodium stearyl fumarate
In order to assess the potential of the salts to provide improved properties over the free form, the discrimination parameters were set as follow: bulk stability (3 weeks 80°C/75%RH open dish) and physical mixture 1 :9 (w/w) with sodium stearyl fumarate (Table 13).
Table 13
Figure imgf000047_0001
As pure bulk, some salts, namely, pyruvate, galactarate, glucoronate, glutarate N- acetylgycinate and quinate are chemically unstable when exposed for 3 weeks at
80°C/75%RH. No degradation was observed for the free form.
As physical mixture with sodium stearyl fumarate (90% w/w), the discrimination power of this stress experiment was more pronounced. Almost all salts are more sensitive than the free form. The main degradation product was characterized by LC-MS.
A similar sensitivity compared to the free form was observed for the butarate and propionate, and enhanced stability was observed for the hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salts. Example 21 : Bulk stability of Compound 1 hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salts in comparison to the free form after three weeks stress exposure at 80°C/75% relative humidity and in the presence of pharmaceutical excipients
In order to assess the potential of the salts to provide improved properties over the free form in the presence of pharmaceutical excipients, the stability of Compound 1 in hemifumarate hemihydrate, hemisuccinate hemihydrate and hemi-L-tartrate hemihydrate salt form was tested relative to the free base form in an open dish for 3 weeks 80°C/75%RH (Table 14).
Table 14
Figure imgf000049_0001
Under the test conditions, the free form is significantly more sensitive to degradation than the salts. Its chemical degradation is very significant, particularly in the presence of magnesium stearate and sodium stearyl fumarate. Such chemical sensitivity is somewhat limited in milder stress conditions (4 weeks, 50 degrees Celsius, 75% RH). Some incompatibility with the free form is also clearly identified with Aerosil 200 PH. Example 22: In human study of pharmacokinetics of free base Compound 1 when given alone and in combination with the strong CYP3A4 inhibitor itraconazole or the strong CYP3A4 inducer rifampicin
In a drug-drug interaction (DDI) study in healthy volunteers, the effect of a strong CYP3A4 inhibitor (itraconazole) and a strong CYP3A4 inducer (rifampicin) on the PK of Compound 1 was evaluated. The DDI study design is outlined in Figure 13. Itraconazole, at a dose of 200 mg q.d., increased mean AUC of Compound 1 2-3-fold and mean Cmax of Compound 1 by 25%, when given together with Compound 1 as compared to when Compound 1 was given alone (Table 15). Rifampicin, at a dose of 600 mg q.d., decreased mean AUC of Compound 1 5-6-fold and mean Cmax of Compound 1 2.5-fold, when given together with Compound 1 as compared to when Compound 1 was given alone (Table 16). In conclusion, the effect of a strong CYP3A4 inducer and a strong CYP3A4 inhibitor on Compound 1 exposure in a Phase 1 study has shown that CYP3A4 is of major importance for the elimination of Compound 1 and that the effects of co-treatment with a strong CYP3A4 inhibitor or inducer need to be taken into account when administering Compound 1.
Table 15: Pharmacokinetic results - Statistical analysis of the effect of itraconazole on the plasma PK parameters of Compound 1 : Compound 1 30 mg SD + itraconazole 200 mg QD vs Compound 1 30 mg SD
Figure imgf000050_0001
n* = number of subjects with non-missing values.
An ANOVA model with fixed effects for treatment and subject was fitted to each log- transformed PK parameter. Results were back transformed to obtain 'Adjusted geo mean', 'Geo-mean ratio' and '90% Cl'. Table 16: Pharmacokinetic results - statistical analysis of the effect of rifampicin on the plasma PK parameters of Compound 1 : Compound 1 100 mg SD + rifampicin 600 mg QD vs Compound 1 100 mg SD
Figure imgf000051_0001
n* = number of subjects with non-missing values.
An ANOVA model with fixed effects for treatment and subject was fitted to each log- transformed PK parameter. Results were back transformed to obtain 'Adjusted geo mean', 'Geo-mean ratio' and '90% Cl'.
References
Charidimou A et al. (2011) Sporadic cerebral amyloid angiopathy revisited: recent insights into pathophysiology and clinical spectrum. J. Neurol. Neurosurg. Psychiatry; 83(2): 124- 137.
Cheng AL et al. (2013) Susceptibility-Weighted Imaging is More Reliable Than T2*-Weighted Gradient-Recalled Echo MRI for Detecting Microbleeds. Stroke; 44(10):2782-2786.
Chen YW et at. (2006) Progression of white matter lesions and hemorrhages in cerebral amyloid angiopathy. Neurology; 67(1):83-87.
Greenberg SM et at. (2014) Outcome Markers for Clinical Trials in Cerebral Amyloid
Angiopathy. Lancet Neurol.; 13(4):419-428.
Gurol ME et al. (2016) Florbetapir-PET to diagnose cerebral amyloid angiopathy: A prospective study. Neurology; 87(19):2043-2049.
Kramp VP, Herrling P (2011) List of drugs in development for neurodegenerative diseases: Update June 2010. Neurodegenerative Dis.; 8:44-94.
Midoux N et at. (1999) Micronization of pharmaceutical substances in a spiral jet mill.
Powder Technol.; 104:113-120.
Quintino-Santos SR et al. (2012) Homozygosity for the APOE E4 allele is solely associated with lower cognitive performance in Brazilian community dwelling older adults - The Bambui Study. Rev. Bras. Psiquiatr.; 34(4):440-445. Shinohara M et al. (2016) Impact of sex and APOE4 on cerebral amyloid angiopathy in Alzheimer’s disease. Acta Neuropathol.; 132(2):225-234.
Sperling RA et al. (2011) Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging and the Alzheimer’s Association workgroup. Alzheimers Dement.; 7(3): 1 -13.
Thai DR et at. (2002) Two types of sporadic cerebral amyloid angiopathy. J. Neuropathol. Exp. Neurol.; 61 : 282-293.
Verghese PB et al. (2011) Roles of Apolipoprotein E in Alzheimer’s disease and Other Neurological Disorders. Lancet Neurol.; 10(3): 241-252.
All references, e.g., a scientific publication or patent application publication, cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each reference was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims.

Claims

Claims
1. The compound N-(6-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-
1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5-(trifluoromethyl)picolinamide in fumarate, succinate or tartrate salt form.
2. The compound according to Claim 1 in fumarate salt form.
3. The compound according to Claim 1 in succinate salt form.
4. The compound according to Claim 1 in tartrate salt form.
5. The compound according to any one of Claims 1 to 4 in hemi salt form.
6. The compound according to any one of Claims 1 to 5 in hydrated salt form.
7. The compound according to any one of Claims 1 to 6 in hemihydrate salt form.
8. The compound according to Claim 1 which is N-(6-((3R,6R)-5-amino-3,6-dimethyl-6- (trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5- (trifluoromethyl)picolinamide hemifumarate hemihydrate.
9. The compound according to Claim 1 which is N-(6-((3R,6R)-5-amino-3,6-dimethyl-6- (trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5- (trifluoromethyl)picolinamide hemisuccinate hemihydrate.
10. The compound according to Claim 1 which is N-(6-((3R,6R)-5-amino-3,6-dimethyl-6- (trifluoromethyl)-3,6-dihydro-2H-1 ,4-oxazin-3-yl)-5-fluoropyridin-2-yl)-3-chloro-5- (trifluoromethyl)picolinamide hemi-L-tartrate hemihydrate.
11. The compound according to any one of Claims 1 to 10 in crystalline salt form.
12. The compound according to Claim 11 in substantially pure form.
13. The compound according to Claim 1 in crystalline fumarate salt form and having an X- ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 11.9, 13.9 and 19.9° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
14. The compound according to Claim 1 in crystalline succinate salt form and having an X- ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 12.0, 14.0 and 20.1° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
15. The compound according to Claim 1 in crystalline tartrate salt form and having an X-ray powder diffraction pattern with angle of refraction 2 theta (θ) peak values of 14.0, 17.7 and 19.8° when measured using CuKα radiation, wherein said values are plus or minus 0.2° 2θ.
16. The compound according to Claim 1 in crystalline fumarate salt form and having an X- ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 4 when measured using CuKα radiation.
17. The compound according to Claim 1 in crystalline succinate salt form and having an X- ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 7 when measured using CuKα radiation.
18. The compound according to Claim 1 in crystalline tartrate salt form and having an X-ray powder diffraction pattern substantially the same as the X-ray powder diffraction pattern shown in Figure 10 when measured using CuKα radiation.
19. A pharmaceutical composition comprising a compound according to any one of Claims 1 to 18.
20. A compound according to any one of Claims 1 to 18 for use as a medicament.
21. A compound according to any one of Claims 1 to 18 for use in the treatment or
prevention of Alzheimer’s disease.
PCT/IB2019/050339 2018-01-18 2019-01-16 Salt forms of an oxazine derivative bace inhibitor WO2019142111A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CNPCT/CN2018/073248 2018-01-18
CN2018073248 2018-01-18

Publications (1)

Publication Number Publication Date
WO2019142111A1 true WO2019142111A1 (en) 2019-07-25

Family

ID=65494464

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2019/050339 WO2019142111A1 (en) 2018-01-18 2019-01-16 Salt forms of an oxazine derivative bace inhibitor

Country Status (1)

Country Link
WO (1) WO2019142111A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012095469A1 (en) 2011-01-13 2012-07-19 Novartis Ag Novel heterocyclic derivatives and their use in the treatment of neurological disorders
WO2018134761A1 (en) * 2017-01-20 2018-07-26 Novartis Ag A free base oxazine derivative in crystalline form

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012095469A1 (en) 2011-01-13 2012-07-19 Novartis Ag Novel heterocyclic derivatives and their use in the treatment of neurological disorders
WO2018134761A1 (en) * 2017-01-20 2018-07-26 Novartis Ag A free base oxazine derivative in crystalline form

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
CHARIDIMOU A ET AL.: "Sporadic cerebral amyloid angiopathy revisited: recent insights into pathophysiology and clinical spectrum", J. NEUROL. NEUROSURG. PSYCHIATRY, vol. 83, no. 2, 2011, pages 124 - 137
CHEN YW ET AL.: "Progression of white matter lesions and hemorrhages in cerebral amyloid angiopathy", NEUROLOGY, vol. 67, no. 1, 2006, pages 83 - 87
CHENG AL ET AL.: "Susceptibility-Weighted Imaging is More Reliable Than T2*-Weighted Gradient-Recalled Echo MRI for Detecting Microbleeds", STROKE, vol. 44, no. 10, 2013, pages 2782 - 2786
GREENBERG SM ET AL.: "Outcome Markers for Clinical Trials in Cerebral Amyloid Angiopathy", LANCET NEUROL., vol. 13, no. 4, 2014, pages 419 - 428
GUROL ME ET AL.: "Florbetapir-PET to diagnose cerebral amyloid angiopathy: A prospective study", NEUROLOGY, vol. 87, no. 19, 2016, pages 2043 - 2049
KRAMP VP; HERRLING P: "List of drugs in development for neurodegenerative diseases: Update", NEURODEGENERATIVE DIS., vol. 8, June 2010 (2010-06-01), pages 44 - 94
MIDOUX N ET AL.: "Micronization of pharmaceutical substances in a spiral jet mill", POWDER TECHNOL., vol. 104, 1999, pages 113 - 120
QUINTINO-SANTOS SR ET AL.: "Homozygosity for the APOE E4 allele is solely associated with lower cognitive performance in Brazilian community dwelling older adults - The Bambui Study", REV. BRAS. PSIQUIATR., vol. 34, no. 4, 2012, pages 440 - 445
RICHARD J BASTIN ET AL: "Salt Selection and Optimisation Procedures for Pharmaceutical New Chemical Entities", ORGANIC PROCESS RESEARCH AND DEVELOP, AMERICAN CHEMICAL SOCIETY, US, vol. 4, no. 5, 19 July 2000 (2000-07-19), pages 427 - 435, XP008154792, ISSN: 1083-6160, [retrieved on 20000719], DOI: 10.1021/OP000018U *
SHINOHARA M ET AL.: "Impact of sex and APOE4 on cerebral amyloid angiopathy in Alzheimer's disease", ACTA NEUROPATHOL., vol. 132, no. 2, 2016, pages 225 - 234, XP036006025, DOI: doi:10.1007/s00401-016-1580-y
SPERLING RA ET AL.: "Toward defining the preclinical stages of Alzheimer's disease: Recommendations from the National Institute on Aging and the Alzheimer's Association workgroup", ALZHEIMERS DEMENT., vol. 7, no. 3, 2011, pages 1 - 13
THAI DR ET AL.: "Two types of sporadic cerebral amyloid angiopathy", J. NEUROPATHOL. EXP. NEUROL., vol. 61, 2002, pages 282 - 293, XP055448476, DOI: doi:10.1093/jnen/61.3.282
VERGHESE PB ET AL.: "Roles of Apolipoprotein E in Alzheimer's disease and Other Neurological Disorders", LANCET NEUROL., vol. 10, no. 3, 2011, pages 241 - 252, XP055291491, DOI: doi:10.1016/S1474-4422(10)70325-2

Similar Documents

Publication Publication Date Title
EP3573983B1 (en) N-[4-fluoro-5-[[(2s,4s)-2-methyl-4-[(5-methyl-1,2,4-oxadiazol-3-yl)methoxy]-1-piperidyl]methyl]thiazol-2-yl]acetamide as oga inhibitor
CN109379891B (en) Polymorphs of a compound as kallikrein inhibitor
JP5853035B2 (en) 3,4-Dihydro-pyrrolo [1,2-a] pyrazin-1-ylamine derivatives useful as inhibitors of β-secretase (BACE)
WO2019191620A1 (en) Crystalline forms of modulators of cftr
JP5853033B2 (en) 6,7-Dihydro-pyrazolo [1,5-a] pyrazin-4-ylamine derivatives useful as inhibitors of β-secretase (BACE)
US20200317639A1 (en) Polymorphs of n-[(2,6-difluoro-3-methoxyphenyl)methyl]-3-(methoxymethyl)-1-({4-[(2-oxopyridin-1yl)methyl]phenyl}methyl)pyrazole-4-carboxamide
US10752607B2 (en) Polymorphs of N-[(6-cyano-2-fluoro)-3-methoxyphenyl)methyl]-3-(methoxymethyl)-1-({4-[(2-oxopyridin-1-yl)methyl]phenyl}methyl)pyrazole-4-carboxamide as kallikrein inhibitors
CA2799635A1 (en) 5-amino-3,6-dihydro-1h-pyrazin-2-one derivatives useful as inhibitors of beta-secretase (bace)
AU2015345257A1 (en) 2-amino-3,5-difluoro-3,6-dimethyl-6-phenyl-3,4,5,6-tetrahydropyridines as BACE1 inhibitors for treating Alzheimer&#39;s disease
CN113784954A (en) Polymorphic compounds and uses thereof
US20200048237A1 (en) A Free Base Oxazine Derivative in Crystalline Form
WO2019142111A1 (en) Salt forms of an oxazine derivative bace inhibitor
US9694004B2 (en) Crystal form
KR20220155575A (en) Salt and crystalline forms of TAAR1 agonists
US20060281690A1 (en) Crystalline (2E,4S)-4-[(N-{[(2R)-1-isopropylpiperidin-2-yI]-carbonyl}-3-methyl-L-valyl)(methyl)amino]-2,5-dimethylhex-2-enoic acid and its pharmaceutical uses
JP2023507180A (en) OGA inhibitor compounds
JP2023536911A (en) Crystal forms of O-glycoprotein-2-acetamido-2-deoxy-3-D-glucopyranosidase inhibitors
NZ754849B2 (en) N-[4-fluoro-5-[[(2s,4s)-2-methyl-4-[(5-methyl-1,2,4-oxadiazol-3-yl)methoxy]-1-piperidyl]methyl]thiazol-2-yl]acetamide as oga inhibitor

Legal Events

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

Ref document number: 19706010

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19706010

Country of ref document: EP

Kind code of ref document: A1