WO2017222083A1 - Novel compounds - Google Patents

Novel compounds Download PDF

Info

Publication number
WO2017222083A1
WO2017222083A1 PCT/JP2017/024276 JP2017024276W WO2017222083A1 WO 2017222083 A1 WO2017222083 A1 WO 2017222083A1 JP 2017024276 W JP2017024276 W JP 2017024276W WO 2017222083 A1 WO2017222083 A1 WO 2017222083A1
Authority
WO
WIPO (PCT)
Prior art keywords
tetrahydro
benzoxazepin
amino
mmol
propan
Prior art date
Application number
PCT/JP2017/024276
Other languages
French (fr)
Inventor
Stephen Mack
Martin Teall
Alka PARMAR
Original Assignee
Takeda Pharmaceutical Company Limited
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 Takeda Pharmaceutical Company Limited filed Critical Takeda Pharmaceutical Company Limited
Publication of WO2017222083A1 publication Critical patent/WO2017222083A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D267/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D267/02Seven-membered rings
    • C07D267/08Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D267/12Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D267/14Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/34Tobacco-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D267/00Heterocyclic compounds containing rings of more than six members having one nitrogen atom and one oxygen atom as the only ring hetero atoms
    • C07D267/02Seven-membered rings
    • C07D267/08Seven-membered rings having the hetero atoms in positions 1 and 4
    • C07D267/12Seven-membered rings having the hetero atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • 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
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • the present invention relates to benzoxazepine derivatives, processes for their preparation, pharmaceutical compositions containing them and their use in therapy, particularly for use in treating disorders associated with changes in one or both of the glutamatergic and GAB Aergic signalling pathways regulated in full or in part by metabotropic glutamate receptor 7 (mGluR7).
  • mGluR7 metabotropic glutamate receptor 7
  • L-Glutamate is the major neurotransmitter in the mammalian central nervous system and activates both ionotropic and metabotropic glutamate receptors. L-Glutamate plays a central role in numerous physiological functions such as learning and memory (1), sensory perception, development of synaptic plasticity, motor control, respiration and regulation of cardiovascular function. Thus an imbalance in glutamatergic neurotransmission often underlies many neuropathological conditions.
  • the metabotropic glutamate receptors are a family of G protein-coupled receptors that have been divided into three groups on the basis of sequence homology, putative signal transduction mechanisms and pharmacologic properties.
  • Group I includes mGluRl and mGluR5 and these receptors have been shown to activate phospholipase C.
  • Group II includes mGluR2 and mGluR3 whilst
  • Group III includes mGluR4, mGluR6, mGluR7 and mGluR8.
  • Group II and III receptors are linked to the inhibition of the cyclic AMP cascade but differ in their agonist selectivities.
  • mGluR7 is an inhibitory GPCR expressed pre-synaptically at the synaptic cleft on
  • mGluR7 modulators would be expected to be useful in treating a wide variety of neurological and psychiatric disorders such as Parkinson's disease (2, 3); dementia associated with Parkinson's disease (3, 4); Alzheimer's disease (5); Huntington's Chorea (6); amyotrophic lateral sclerosis and multiple sclerosis; bipolar disorder (6, 7); psychiatric diseases such as schizophrenia, post-traumatic stress disorder, anxiety disorders and depression (1,4, 6, 8-11); and addiction. They may also be useful in treating age-related hearing loss/tinnitus (12). There is a need for treatment of the above conditions and others described herein with compounds that are mGluR7 modulators.
  • the present invention provides modulators of mGluR7.
  • X 1 is absent or is -CH2- or -CH(CH3)-;
  • X 2 is -CH2-, -CHF-, -CF2- or -CH2CH2-, provided that when X 1 is -CH 2 - or -CH(CH3)-, then X 2 is -CH2- only;
  • R 1 , R 2 , R 3 and R 4 each independently represent hydrogen, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; either R 5 represents hydrogen, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy and R 6 represents hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkylsulphonyl or -NR 14 R 15 , or when X 1 is absent, R 5 and R 6 together form a linking group, -[CH2]m- 5 in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or
  • N(COCH3) heteroatom moieties provided that if the linking group contains two heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another; n is 0, 1 or 2;
  • each R independently represents halogen, cyano, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; each of R 8 , R 9 , R 10 , R 1 1 , R 12 and R 13 independently represents hydrogen, hydroxyl, cyano, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; or,
  • R 14 and R 15 each independently represent hydrogen or a C1-C6 alkyl, C3-C6 cycloalkylmethyl, C1-C6 alkylcarbonyl, C3-C6 cycloalkylcarbonyl or C1-C6
  • alkoxycarbonyl group wherein each of the preceding groups is optionally substituted by at least one halogen atom, or
  • R 14 and R 15 together with the nitrogen atom to which they are attached form a saturated 4- to 6-membered heterocyclic ring optionally comprising a further ring heteroatom selected from nitrogen and oxygen, the heterocyclic ring being unsubstituted or substituted by at least one substituent selected from halogen, oxo and C1-C3 alkyl.
  • an "alkyl" substituent group or an “alkyl” moiety in a substituent group may be linear or branched.
  • C1-C6 alkyl groups/moieties include methyl, ethyl, propyl, 2-methyl-l- propyl, 2-methyl-2-propyl, 2-methyl-l -butyl, 3 -methyl- 1 -butyl, 2-methy 1-3 -butyl, 2,2- dimethyl-1 -propyl, 2 ⁇ methyl-pentyl, 3 -methyl- 1-pentyl, 4-methyl-l-pentyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-l -butyl, 3,3-dimethyl-l- butyl, 2-ethyl-l -butyl, n-butyl, tert-butyl, n-pentyl, and n-hexyl.
  • a “cycloalkyl” substituent group or a “cycloalkyl” moiety in a substituent group refers to a saturated hydrocarbyl ring containing, for example, from 3 to 8 carbon atoms, examples of which include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • oxo refers to an oxygen atom doubly bonded to the carbon atom to which it is attached to form the carbonyl of a ketone or aldehyde.
  • halogen includes fluorine, chlorine, bromine and iodine.
  • R 14 and R 15 form a saturated 4- to 6-membered heterocyclic ring which is substituted, it should be understood that the substituent(s) may be attached to any suitable ring atom.
  • X 1 is absent or is -C3 ⁇ 4- or -CH(CH3)- and X 2 is -CH 2 -, -CHF-, -CF2- or -CH2CH2-,
  • X when X is -CH2- or -CH(CH3)-, then X is -CH2- only. Consequently, when X 1 is absent, X 2 is -CH2-, -CHF-, -CF2- or -CH2CH2-, and when X 1 is -C3 ⁇ 4- or -CH(CH3)-, then X 2 is -CH2-;
  • X 1 is absent.
  • X is -CH2-, -CF 2 - or -CH2CH2-.
  • X is -CH2-
  • X and X have the following meanings:
  • R , R , R and R each independently represent hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy.
  • R , R , R and R each independently represent hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C4, or C1-C3, or C1-C2 alkyl (e.g. methyl), cyclopropyl or C1-C4, or C1-C3, or C1-C2 alkoxy (e.g. methoxy).
  • R 1 , R 2 , R 3 and R 4 each independently represent hydrogen, fluorine, chlorine, methyl or methoxy.
  • R 1 represents hydrogen, fluorine, chlorine, methyl or methoxy
  • R , R and R each represent hydrogen.
  • R represents hydrogen, fluorine, chlorine, methyl or methoxy and R 1 , R 2 and R 4 each represent hydrogen.
  • R 5 represents hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy and R 6 represents hydrogen, C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl, C1-C6, or C1-C4, or C1-C2 alkylsulphonyl or NR 14 R 15 .
  • halogen e.g. fluorine, chlorine or bromine
  • R 5 represents hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropyl or C1-C4, or C1-C3, or C1-C2 alkoxy.
  • halogen e.g. fluorine, chlorine or bromine
  • R 5 represents hydrogen, fluorine, chlorine, bromine, C1-C2 alkyl, cyclopropyl or C1-C2 alkoxy.
  • R represents hydrogen, fluorine, chlorine, bromine, cyclopropyl, methyl or methoxy.
  • R 1 , R 2 , R 3 , R 4 and R 5 each independently represent hydrogen or halogen.
  • R 6 represents hydrogen, C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropyl, C1-C4, or C1-C3, or C1-C2 alkylsulphonyl or NR 14 R 15 .
  • R 6 represents hydrogen, C1-C2 alkyl or NR 14 R 15 .
  • R 6 represents NR 14 R 15 .
  • R 14 and R 15 each independently represent hydrogen or a C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkylmethyl, C1-C6, or C1-C4, or C1-C2 alkylcarbonyl, C3-C6, or C4-C6, or C5-C6 cycloalkylcarbonyl or C1-C6, or C1-C4, or
  • halogen such as fluorine, chlorine or bromine
  • atom e.g. one, two, three, four or five halogen atoms
  • heterocyclic ring optionally comprising a further ring heteroatom selected from nitrogen and oxygen, the heterocyclic ring being unsubstituted or substituted by at least one substituent (e.g. one, two or three substituents independently) selected from halogen (e.g. fluorine, chlorine or bromine), oxo and C1-C3 alkyl (e.g. methyl or ethyl).
  • substituent e.g. one, two or three substituents independently
  • halogen e.g. fluorine, chlorine or bromine
  • oxo and C1-C3 alkyl e.g. methyl or ethyl
  • R 14 and R 15 each independently represent hydrogen or a C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropylmethyl, C1-C4, or C1-C3, or C1-C2 alkylcarbonyl, cyclopropylcarbonyl or C1-C4, or C1-C3, or C1-C2 alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by one to five, or one to three, halogen, particularly fluorine, atoms.
  • R and R each independently represent hydrogen or a
  • each of the preceding groups is optionally substituted by one to five, or one to three, halogen, particularly fluorine, atoms.
  • R 14 and R 15 each independently represent hydrogen or a
  • C1-C4 alkoxycarbonyl group wherein the C1-C2 alkyl and C1-C3 alkylcarbonyl groups are optionally substituted by one to three halogen, particularly fluorine, atoms.
  • R 14 and R 15 together with the nitrogen atom to which they are attached form a saturated 5-membered heterocyclic ring optionally comprising a further ring heteroatom selected from nitrogen and oxygen, the heterocyclic ring being
  • -NR 14 R 15 represents any one of the following moieties or is selected from a group containing any two or more of such moieties:
  • R 1 to R 6 have the following meanings where -
  • NR R is as defined above:
  • R 5 and R 6 together form a linking group, -[03 ⁇ 4] ⁇ , in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or N(COCH3)heteroatom moieties provided that if the linking group contains two heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another.
  • linking groups include: -CH 2 CH 2 NH-, CH 2 CH2N(COCH 3 )-, -NHCH2CH2-, -N(COCH 3 )CH 2 CH2- , -CH2NHCH2-, -CH 2 N(COCH3)CH 2 -, -OCH2CH2NH- or -NHCH 2 CH20-.
  • Each R independently represents halogen (e.g. fluorine, chlorine or bromine), cyano, C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy.
  • halogen e.g. fluorine, chlorine or bromine
  • cyano e.g. fluorine, chlorine or bromine
  • each R independently represents halogen (e.g. fluorine, chlorine or bromine), cyano, C1-C4, or Ci-C 3 , or C1-C2 alkyl (e.g. methyl), cyclopropyl or C1-C4, or Ci-C 3 , or C1-C2 alkoxy (e.g. methoxy).
  • halogen e.g. fluorine, chlorine or bromine
  • C1-C4 cyano
  • Ci-C 3 or C1-C2 alkyl (e.g. methyl)
  • C1-C2 alkyl e.g. methyl
  • cyclopropyl or C1-C4, or Ci-C 3 e.g. methoxy
  • each R independently represents fluorine, chlorine, methyl or methoxy.
  • n is 1 and R represents a halogen atom, particularly a fluorine atom.
  • n 0.
  • R , R , R , R and R each independently represent hydrogen, hydroxyl, cyano, halogen (e.g. fluorine, chlorine or bromine), C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy.
  • halogen e.g. fluorine, chlorine or bromine
  • R , R , R , R and R each independently represent hydrogen, hydroxyl, cyano, halogen (e.g. fluorine,chlorine or bromine), C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropyl or C1-C4, or C1-C3, or C1-C2 alkoxy.
  • halogen e.g. fluorine,chlorine or bromine
  • R 8 , R 9 , R 10 , R 1 1 , R 12 and R 13 each independently represent hydrogen, hydroxyl, halogen (e.g. fluorine or chlorine), C1-C2 alkyl, cyclopropyl or
  • R 8 , R 9 , R 10 , R 1 1 , R 12 and R 13 each independently represent hydrogen, hydroxyl, fluorine, methyl or methoxy.
  • R 8 to R 13 are shown in the following table:
  • R and R together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring and R 10 to
  • R 13 are as defined above.
  • R and R together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring and R 8 , R 9 ,
  • R 1 ⁇ 2 and R 1 1 3 J are as defined above.
  • R are as defined above.
  • R and R together with the carbon atoms to which they are attached form a saturated 3- to 6-membered carbocyclic ring and R 8 , R 1 1 , R 12 and R 13 are as defined above.
  • R and R together form a methylene bridge and R 9 , R 10 , R 11 and R 13 are as defined above.
  • bicyclic ring systems (AB) according to the above-mentioned second to sixth aspects of the invention include the following structures:
  • the compounds of formula (I) are those in which:
  • X 1 is absent or is -CH2- or -CH(CH3>-;
  • X 2 is -CH2-, -CF2- or -CH2CH2-;
  • R 1 , R 2 , R 3 and R 4 each independently represent hydrogen, fluorine, chlorine, methyl or methoxy;
  • R 5 represents hydrogen, fluorine, chlorine, bromine, cyclopropyl, methyl or methoxy and R 6 represents hydrogen, methyl or -NR 14 R 15 , or when X 1 is absent, R 5 and R 6 together form a linking group, -[CH2]m- 5 in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or
  • N(COCH3) heteroatom moieties provided that if the linking group contains two heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another;
  • n 0 or 1 ;
  • R represents halogen, methyl or methoxy
  • each ofR 8, R 9, R 10 , R 11 , R 12 and R 13 independently represents hydrogen, hydroxyl, fluorine, methyl or methoxy or, alternatively, either R 10 and R 11 together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring, or R and R together form a methylene bridge
  • R and R each independently represent hydrogen or a C i -C2 alkyl, cyclopropylmethyl, C1-C3 alkylcarbonyl, cyclopropylcarbonyl or C1-C4 alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by at least one halogen atom, or
  • R 1 and R 15 together with the nitrogen atom to which they are attached form a saturated 5-membered heterocyclic ring, the heterocyclic ring being unsubstituted or substituted by at least one substituent selected from halogen, oxo and C1-C3 alkyl.
  • R 3a is hydrogen or fluorine
  • p is 0 or 1 ;
  • R 7a is fluorine
  • R 10a is hydrogen or fluorine
  • R 1 la is hydrogen or fluorine
  • the compounds of formula (la) contain a single fluorine substituent. If R 7a is a fluorine substituent, it is preferably attached to the 9-position of the bicyclic ring structure AB (which is shown marked with an asterisk in formula (la) above). According to another aspect, the compounds of formula (la) contain two fluorine substituents as R l0a and R l la .
  • Examples of compounds of the invention include:
  • the present invention further provides a process for the preparation of a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined above which comprises reacting a compound of formula (II), or a salt (e.g. hydrochloride salt) thereof,
  • X 1 , X 2 , R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are as defined in formula (I) above, with a compound of formula (III), or a salt (e.g. hydrochloride salt) thereof,
  • the above process may conveniently be carried out by combining the carboxylic acid of formula (II) with the amine of formula (III) in the presence of a coupling reagent such as
  • EDC l-ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • HOAt 7-aza-l- hydroxybenzotriazole
  • HATU l-[bis(dimethylamino)memylene]-lH-l,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate
  • Boc tert-butyloxycarbonyl (a protecting group)
  • Boc fert-butyloxycarbonyl (a protecting group)
  • Step (i) is analogous to methods described in the journal article by S. Has-Becker et al., Synlett 2001, No. 9, 1395-1398.
  • TEA denotes trimethylamine
  • MeCN denotes acetonitrile.
  • Step (ii) is an oxidation reaction which may conveniently be carried out using 3- chlorobenzenecarboperoxoic acid (mCPBA) in an organic solvent such as dichloromethane at a temperature in the range of from room temperature to 60°C.
  • mCPBA 3- chlorobenzenecarboperoxoic acid
  • Step (iii) is a hydrolysis reaction which may conveniently be carried out in the presence of a base such as lithium hydroxide in a polar solvent, e.g. a mixture of water and
  • tetrahydrofuran at a pressure of 8 bar (800 kPa) and at a temperature in the range of from room temperature to 60°C.
  • a compound of formula (II) in which R 1 to R 5 are each hydrogen, X 1 is absent, X is -CH2- and R is methyl sulphonyl may be prepared as shown in reaction scheme 3 except that step (i) is replaced as follows (see methods described in US
  • NaSMe sodium methanethiolate
  • a compound of formula (I) may be converted into another compound
  • a compound of formula (I) in which R represents a halogen atom may be converted to a corresponding compound of formula (I) in which R represents a C1-C6 alkyl or C3-C6 cycloalkyl group by reacting the former with a boronic acid derivative such as R 7a -B(OH)2, R 7a -B(pinacole ester) or R 7a -BF3 ⁇ K + where R 7a represents the replacement R bonded to the boron atom via a carbon-boron bond, in the presence of a base such as potassium carbonate, caesium carbonate or potassium phosphate, and a metal catalyst such as Pd(0), typically where the metal catalyst is in the form of a transition metal complex such as tetrakis(triphenylphosphine) palladium or di-tert- butyl [dichloro( ⁇ di-tert-buty 1 [4-(dimethylamino)pheny
  • a transition metal complex
  • reaction mixture is typically heated, e.g. to around 60-120°C under conventional heating or microwave irradiation.
  • a compound of formula (I) in which R 10 represents a hydroxyl group may be converted to a corresponding compound of formula (I) in which R 10 represents a cyano group according to methods described in US 2002/0188124 in the name of Fukami et al. (Banyu Pharmaceutical Co. Ltd.), for example,
  • the compounds of formula (I) above may be converted to a pharmaceutically acceptable salt thereof, preferably an acid addition salt such as a formate, hemi-formate,
  • hydrochloride hydrobromide
  • benzenesulphonate besylate
  • saccharin e.g.
  • the compounds of formula (I) are in the form of a hydrochloride salt.
  • compounds of formula (I) may bear one or more radiolabels.
  • radiolabels may be introduced by using radiolabel-containing reagents in the synthesis of the compounds of formula (I), or may be introduced by coupling the
  • any atom specified herein may also be an isotope of said atom.
  • the term "hydrogen” encompasses ! H, 2 H and 3 H.
  • carbon atoms are to be understood to include 12 C, 13 C and 14 C
  • nitrogen atoms are to be understood to include 14 N and 15 N
  • oxygen atoms are to be understood to include 16 0, 17 0 and 18 0.
  • compounds of formula (I) may be isotopically labelled.
  • an “isotopically labelled" compound is one in which the abundance of a particular nuclide at a particular atomic position within the molecule is increased above the level at which it occurs in nature.
  • Compounds of formula (I) and their salts may be in the form of hydrates or solvates which form an aspect of the present invention.
  • Such solvates may be formed with common organic solvents, including but not limited to, alcoholic solvents e.g. methanol, ethanol or isopropanol.
  • Compounds of formula (I) and their salts may be amorphous or in a polymorphic form or a mixture of any of these, each of which forms an aspect of the present invention.
  • the compounds of formula (I) and their pharmaceutically acceptable salts have activity as pharmaceuticals and may be used in treating conditions or disorders associated with changes in one or both of the glutamatergic and GABAergic signalling pathways regulated in full or in part by metabotropic glutamate receptor 7.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined for use in therapy, in particular for the treatment of conditions associated with metabotropic glutamate receptor 7.
  • the present invention also provides the use of a compound of formula (I) or a
  • the present invention still further provides a method of treating a condition associated with metabotropic glutamate receptor 7 which comprises administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I) or a
  • Prophylaxis is expected to be particularly relevant to the treatment of persons who have suffered a previous episode of, or are otherwise considered to be at increased risk of, the disorder or condition in question.
  • Persons at risk of developing a particular disorder or condition generally include those having a family history of the disorder or condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the disorder or condition or those in the prodromal phase of a disorder.
  • treat include improvement of the conditions described herein.
  • the terms “treat”, “treatment” and “treating” include all processes providing slowing, interrupting, arresting, controlling, or stopping of the state or progression of the conditions described herein, but does not necessarily indicate a total elimination of all symptoms or a cure of the condition.
  • the terms “treat”, “treatment,” and “treating” are intended to include therapeutic as well as prophylactic treatment of such conditions.
  • condition refers to any unhealthy or abnormal state.
  • conditions associated with metabotropic glutamate receptor 7 includes conditions, disorders and diseases in which the modulation of mGluR7 may provide a therapeutic benefit, examples of which include:
  • Nervous system disorders Parkinson's disease, including dementia associated with Parkinson's disease; Alzheimer's disease; Huntington's Chorea; amyotrophic lateral sclerosis; multiple sclerosis; bipolar disorder; and psychiatric disorders such as
  • schizophrenia, post-traumatic stress disorder, anxiety disorders and depression e.g. major depressive disorder
  • Hearing disorders hearing loss and/or tinnitus caused by age, noise or trauma;
  • Schizophrenia is a debilitating psychiatric disorder characterised by a combination of negative symptoms (such as social withdrawal, anhedonia, avolition and apathy) and positive symptoms (including hallucinations, delusions and paranoia) as well as marked cognitive deficits (such as impairment of executive function).
  • the executive function (EF) has been defined as "a set of abilities, which allows us to invoke voluntary control of our behavioral responses. These functions enable human beings to develop and carry out plans, make up analogies, obey social rules, solve problems, adapt to unexpected circumstances, do many tasks simultaneously, and locate episodes in time and place.
  • EF includes divided attention and sustained attention, working memory (WM), set-shifting, flexibility, planning, and the regulation of goal directed behavior and can be defined as a brain function underlying the human faculty to act or think not only in reaction to external events but also in relation with internal goals and states" (Orellana G. and Slachevsky A., 2013. Executive Functioning in Schizophrenia. Front. Psychiatry, 4, 35). Accordingly, the present invention also provides a method of treating a negative symptom, a positive symptom and/or a cognitive deficit associated with a psychiatric disorder, especially schizophrenia, which comprises administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined.
  • the daily dosage of the compound of the invention if inhaled, may be in the range from 0.05 micrograms per kilogram body weight (p.g/kg) to 100 micrograms per kilogram body weight ⁇ g/kg).
  • the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight ⁇ g/kg) to 100 milligrams per kilogram body weight (mg/kg).
  • the compounds of formula (I) and pharmaceutically acceptable salts thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the formula (I) compound/salt (active ingredient) is in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the present invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the invention still further provides a process for the preparation of a pharmaceutical composition of the invention which comprises mixing a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined with a pharmaceutically acceptable adjuvant, diluent or carrier.
  • compositions of the invention are those conventionally employed in the field of pharmaceutical formulation, and include, but are not limited to, sugars, sugar alcohols, starches, ion exchangers, alumina, aluminium stearate, lecithin, serum proteins such as human serum albumin, buffer substances such as phosphates, glycerine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes such as protamine sulphate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol
  • carboxymethylcellulose polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, rectally, nasally, buccally, vaginally or via an implanted reservoir. Oral administration is preferred.
  • the pharmaceutical compositions of the invention may contain any conventional non-toxic pharmaceutically acceptable adjuvants, diluents or carriers.
  • parenteral as used herein includes subcutaneous,
  • compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • the suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • suitable dispersing or wetting agents such as, for example, Tween 80
  • suspending agents such as, for example, Tween 80
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic
  • parenterally acceptable diluent or solvent for example, as a solution in 1,3-butanediol.
  • acceptable diluents and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long- chain alcohol diluent or dispersant.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, powders, granules, and aqueous suspensions and solutions. These dosage forms are prepared according to techniques well-known in the art of pharmaceutical formulation. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavouring and/or colouring agents may be added.
  • compositions of the invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing the active ingredient with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active ingredient.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance
  • the pharmaceutical composition will preferably comprise from 0.05 to 99 %w (per cent by weight), more preferably from 0.05 to 80 %w, still more preferably from 0.10 to 70 %w, and even more preferably from 0.10 to 50 %w, of active ingredient, all percentages by weight being based on total composition.
  • the compounds of the invention that is, compounds of formula (I) and pharmaceutically acceptable salts thereof
  • the invention therefore further relates to combination therapies wherein a compound of the invention or a pharmaceutical composition or formulation comprising a compound of the invention is administered with another therapeutic agent or agents for the treatment of one or more of the conditions previously indicated.
  • therapeutic agents may be selected from the following:
  • anti-addiction drugs including, for example, acamprosate, disulfiram, naltrexone and nalmefene for alcohol dependency, and gabapentin, modafinil, topiramate, vigabatrin and baclofen for drug, particularly cocaine, addiction;
  • antidepressants such as amitriptyline, amoxapine, bupropion, citalopram,
  • clomipramine desipramine, doxepin duloxetine, elzasonan, escitalopram, fluvoxamine, fluoxetine, gepirone, imipramine, ipsapirone, maprotiline, nortriptyline, nefazodone, paroxetine, phenelzine, protriptyline, reboxetine, robaizotan, sertraline, sibutramine, tianeptine, thionisoxetine, tranylcypromaine, trazodone, trimipramine, venlafaxine, vortioxetine and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • antipsychotics including, for example, amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, brexpiprazole, carbamazepine, cariprazine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, fluphenazine, haloperidol, iloperidone, lamotrigine, loxapine, lurasidone, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutlypiperidine, pimozide, prochlorperazine, quetiapine, risperidone, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trim
  • anxiolytics including, for example, alnespirone, azapirones, benzodiazepines, barbiturates, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof.
  • Example anxiolytics include adinazolam, alprazolam, balezepam, bentazepam, bromazepam, brotizolam, buspirone, clonazepam, clorazepate, chlordiazepoxide, cyprazepam, diazepam, diphenhydramine, estazolam, fenobam, flunitrazepam, flurazepam, fosazepam, lorazepam, lormetazepam, meprobamate, midazolam, nitrazepam, oxazepam, prazepam, prazosin, quazepam, reclazepam, tracazo
  • anticonvulsants including, for example, carbamazepine, valproate, lamotrigine, levetiracetam and gabapentin, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • Alzheimer's therapies including, for example, donepezil, galantamine, memantine, rivastigmine, tacrine, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • Parkinson's therapies including, for example, L-dopa, ropinirole, pramipexole, monoamine oxidase type B (MAO-B) inhibitors such as deprenyl, selegiline and rasagiline, catechol-O-methyl transferase (COMT) inhibitors such as entacapone or tolcapone, adenosine A-2 inhibitors, dopamine re-uptake inhibitors, NMDA antagonists, Nicotine agonists, and Dopamine agonists and inhibitors of neuronal nitric oxide synthase, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • MAO-B monoamine oxidase type B
  • COMP catechol-O-methyl transferase
  • entacapone or tolcapone adenosine A-2 inhibitors
  • dopamine re-uptake inhibitors NMDA antagonists
  • Nicotine agonists and Dopamine
  • migraine therapies including, for example, almotriptan, amantadine, botulinum toxin A, bromocriptine, butalbital, cabergoline, dichloralphenazone, dmydroergotarnine, eletriptan, frovatriptan, lisuride, naratriptan, pergolide, pramipexole, rizatriptan, ropinirole, sumatriptan, topiramate, zolmitriptan, and zomitriptan, and equivalents and
  • (ix) stroke therapies including, for example, abciximab, activase, citicoline,
  • urinary incontinence therapies including, for example, darafenacin, duloxetine, falvoxate, mirabegron, oxybutynin, propiverine, robalzotan, solifenacin, and tolterodine, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • neuropathic pain therapies including, for example, capsaicin, gabapentin, lidoderm, and pregabalin, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • nociceptive pain therapies such as celecoxib, etoricoxib, lumiracoxib, rofecoxib, valdecoxib, diclofenac, loxoprofen, naproxen, and paracetamol, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • insomnia therapies including, for example, allobarbital, alonimid, amobarbital, benzoctamine, butabarbital, capuride, chloral, cloperidone, clorethate, dexclamol, ethchlorvynol, eszopiclone, etomidate, glutethimide, halazepam, hydroxyzine, lorediplon, mecloqualone, melatonin, mephobarbital, methaqualone, midaflur, nisobamate, pentobarbital, phenobarbital, propofol, ralmeteon, roletamide, suvorexant, triclofos, secobarbital, zaleplon, and Zolpidem, zopiclone and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • mood stabilizers including, for example, carbamazepine, divalproex, gabapentin, lamotrigine, lithium, olanzapine, quetiapine, valproate, valproic acid, and verapamil, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
  • (xv) 5HT1B ligands such as, for example, compounds disclosed in WO 99/05134 and
  • alpha 7 nicotinic agonists such as, for example, compounds disclosed in
  • (xix) delta opioid agonists such as, for example, compounds disclosed in WO 97/23466 and WO 02/094794.
  • Such combination products employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active agent within approved dosage ranges.
  • Nuclear magnetic resonance (NMR) spectra were recorded at 400 MHz or 300 MHz as stated and at 300.3K, 298.2K or 293K unless otherwise stated; the chemical shifts ( ⁇ ) are reported in parts per million.
  • Spectra were recorded using a Bruker (trade mark) 400 AVANCE instrument fitted with a 5mm BBFO probe with instrument controlled by Bruker TopSpin 2.1 software, or by a Bruker 400 AVANCE-III HD instrument fitted with a 5mm BBFO smart probe or a 5mm BBFO probe with instrument controlled by Bruker TopSpin 3.2 software, or by a Bruker 400 AVANCE-III instrument fitted with a 5mm BBFO probe with instrument controlled by Bruker Topspin 3.0 software or by a Bruker 300MHz AVANCE II instrument fitted with a 5mm DUL probe with instrument controlled by Bruker TopSpin 1.3 software, or 5mm BBFO probe controlled by Bruker Topspin 3.2 software.
  • Ultra Performance Liquid Chromatography with UV (photodiode array) detection over a wide range of wavelengths, normally 220-450 nm, using a Waters (trade mark) Acquity UPLC system equipped with Acquity UPLC BEH, HSS or HSS T3 C18 columns (2.1mm id x 50mm long) operated at 50 or 60 °C.
  • Mobile phases typically consisted of acetonitrile mixed with water containing either 0.1% formic acid, 0.1% TFA or 0.025% ammonia.
  • Mass spectra were recorded with a Waters SQD single quadrupole mass spectrometer using atmospheric pressure ionisation.
  • Preparative HPLC was performed using Agilent Technologies (trade mark) 1100 Series system or a Waters autopurification LC/MS system typically using Waters 19 mm id x 250 mm long C18 columns such as XBridge (trade mark) or SunFire (trade mark) 5 ⁇ materials at room temperature.
  • Mobile phases typically consisted of acetonitrile mixed with water containing either 0.1% formic acid or 0.1% ammonia, unless otherwise stated.
  • Super Critical Fluid Chromatography (SFC) chiral separations were performed on a Waters prep30/MS system, using a flow rate of 30 mL/min, temperature of 40 °C and a pressure of 100 bar.
  • Mobile phases typically consisted of supercritical C0 2 and a polar solvent such as methanol, ethanol or isopropanol. Column type and eluent are detailed for individual examples.
  • 'Room temperature' as used in the present specification, means a temperature in the range from about 18 °C to about 25 °C.
  • DIAD Diisopropyl azodicarboxylate
  • DIBAL-H Diisobutylaluminium hydride
  • DIPEA N,N-Diisopropylethylamine
  • HATU l-[Bis(dimemylamino)memylene]-lH-l,2,3-triazolo[4,5-b]pyridinium 3- oxid hexafluorophosphate
  • T3P Propylphosphonic Anhydride
  • N-(7-fluoro-3,4-dihydro-2H-l-benzopyran-4-ylidene)hydroxylamine (7.60 g, 41.9 mmol) was dissolved in DCM (450 mL) and the solution was cooled to 0 to -5 °C.
  • DIBAL-H (1M solution in cyclohexane, 252 mL, 252 mmol) was added drop wise at 0 to -5 °C over 5 min and the reaction mixture was warmed to room temperature and stirred for 2h.
  • the reaction mixture was cooled again to 0 °C, and sodium fluoride (50.40 g, 1.20 mol) was added followed by the careful addition of water (19 mL).
  • the reaction mixture was stirred at 0 °C for 30 min.
  • the mixture was filtered through a pad of celite and was washed with EtOAc. The filtrates were evaporated in vacuo.
  • the crude product was purified by flash
  • Methyl iodide (2.95 mL, 47.1 mmol) was added to a suspension of (3S)-3- ⁇ [(tert- butoxy)carbonyl]amino ⁇ -3-phenylpropanoic (2.5 g, 9.42 mmol) and K2CO3 (2.60 g, 18.9 mmol) in DMF (50 mL) at room temperature. The mixture was heated at 80 °C under nitrogen for 24h. The mixture was concentrated in vacuo to remove most of the DMF and the residue was partitioned between water and EtOAc. The organic phase was dried (MgS0 4 ) and concentrated in vacuo.
  • N-(2-Hydroxyphenyl)methanesulfonamide (85.00 g, 0.46 mol) was dissolved in ethanol (275 mL). A solution of sodium ethoxide (21% solution in ethanol) (173 mL, 0.47 mol) in ethanol (275 mL) was added. More ethanol (200 mL) was added and the reaction was stirred under nitrogen for 30 min. The solvent was concentrated in vacuo, and azeotroped with THF (200 mL). The residue was dissolved in DMF (680 mL). ( ⁇ )-Epichlorohydrin (35.5 mL, 0.46 mol) was added and the mixture was heated to 90 °C for 7h and stirred at room temperature overnight.
  • 3M in diethyl ether 8.29 mL, 24.87 mmol
  • the reaction was quenched by cautious addition of saturated aqueous NH 4 C1 solution, extracted 3 times with EtOAc, dried (Na 2 S0 4 ) and concentrated in vacuo.
  • the crude product was purified by flash chromatography (0-75% EtOAc in petroleum ether on silica). The material was loaded onto a cation exchange cartridge, washing with methanol and eluting with 2M ammonia in methanol solution and concentrated in vacuo to afford the title compound.
  • Di-tert-butyl dicarbonate (4.56 mL, 19.6 mmol) was added to a solution of ethyl 3-amino- 2,2-difluoro-3-phenylpropanoate (2.25 g, 9.82 mmol) and sodium hydrogen carbonate (1.649 g, 19.6 mmol) in THF (10 mL) under nitrogen.
  • the reaction was heated to 60 °C for 18h.
  • Methyl iodide (3.20 mL, 51.4 mmol) was added to a suspension of 5-(methylsulfonyl)- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-ol (Intermediate 4 step (ii), 5 g, 20.6 mmol) and silver oxide (23.81 g, 103 mmol) in MeCN (40 mL) and DMF (15 mL) in a sealed tube. The reaction was covered in foil and stirred at room temperature overnight. The suspension was filtered through celite and the filtrate was concentrated in vacuo to afford the title compound.
  • HATU (0.120 g, 0.32 mmol) was added to a solution of (S)-3-((tert- butoxycarbonyl)(methyl)amino)-3-phenylpropanoic acid (Intermediate 3, 0.084 g, 0.30 mmol) and DEPEA (0.058 mL, 0.33 mmol) in DMF (1 mL) at room temperature. The mixture was stirred for 5 min. 5-methanesulfonyl-3-methyl-2,3,4,5-tetrahydro-l,5- benzoxazepin-3-ol (Intermediate 4, 0.054 g, 0.30 mmol) was added and the mixture was stirred until homogeneous and allowed to stand for 18h.
  • Soidum tert-butoxide (0.371 g, 3.86 mmol) and Pd 2 (dba) 3 (0.177 g, 0.193 mmol) were added.
  • the mixture was degassed and a nitrogen atmosphere introduced three times.
  • the mixture was heated at 60 °C for 24h.
  • the mixture was partitioned between water and EtOAc.
  • the organic phase was dried (MgS0 4 ) and concentrated in vacuo. The residue was purified by flash chromatography (15-30% EtOAc in petroleum ether) to afford the title compound.
  • HC1 (4 M in dioxane) (3.81 mL, 15.24 mmol) was added to a solution of tert-butyl N- [(2S)-4-(2-bromo-6-fluorophenoxy)butan-2-yl]carbamate (3.68 g, 10.16 mmol) in DCM (15 mL) at room temperature. The mixture was stirred at room temperature and allowed to stand for 18h. Further HC1 (4 M in dioxane) (8.89 mL, 35.6 mmol) was added and the mixture was diluted with MeOH (15 mL), stirred and allowed to stand for 18h. The mixture was concentrated in vacuo, adding DCM to assist removal of dioxane, to afford the title compound.
  • HATU (0.800 g, 2.100 mmol) was added to a stirred solution of (3S)-3- ⁇ [(tert- butoxy)carbonyl]amino ⁇ -3-phenylpropanoic (0.53 g, 2 mmol) and DIPEA (0.770 mL, 4.40 mmol) in DMF (8 mL) at room temperature. The mixture was stirred at room temperature for 5 minutes. (2S)-4-(2-bromo-6-fluorophenoxy)butan-2-amine hydrochloride (0.60 g, 2.000 mmol) was added and the mixture was stirred until homogeneous and allowed to stand for 2h. The mixture was partitioned between water and EtOAc. THF and water were added.
  • the aqueous phase was extracted with EtOAc.
  • the combined organic phases were dried (MgS0 4 ) and concentrated in vacuo to ca. 20 mL.
  • the resulting suspension was filtered, washed with ether and air-dried to afford the title compound.
  • HATU (798 mg, 2.100 mmol) was added to a solution of (3S)-3-(2-bromophenyl)-3- ⁇ [(tert-butoxy)carbonyl]amino ⁇ propanoic acid (0.688 g, 2.00 mmol) and DIPEA (0.384 mL, 2.200 mmol) in DMF (5 mL) at room temperature.
  • the mixture was stirred at room temperature for 5 minutes and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.298 g, 2.00 mmol) was added.
  • the mixture was stirred until homogeneous and allowed to stand for 18h.
  • the mixture was partitioned between water and EtOAc.
  • the organic phase was dried (MgS0 4 ) and concentrated in vacuo. The resulting residue was purified by flash
  • HATU (0.096 g, 0.25 mmol) was added to a solution of 3-acetamido-3-phenylpropanoic acid (0.050 g, 0.24 mmol), DIPEA (0.046 mL, 0.27 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.036 g, 0.24 mmol) in DMF (0.5 mL). The mixture was stirred and allowed to stand for 18h. The mixture was purified by reverse phase preparative HPLC to afford the title compound.
  • HATU (0.120 g, 0.315 mmol) was added to a solution of (3R)-3- ⁇ [(tert- butoxy)carbonyl]amino ⁇ -3-phenylpropanoic (0.080 g, 0.30 mmol) and DIPEA (0.058 mL, 0.33 mmol) in DMF (0.5 mL) at room temperature. The mixture was stirred and allowed to stand. After 5 min, 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) was added. The mixture was stirred at room temperature for 2h.
  • HATU (0.798 g, 2.10 mmol) was added to a solution of (3S)-3- ⁇ [(tert- butoxy)carbonyl]amino ⁇ -3-phenylpropanoic (531 mg, 2.00 mmol) and DIPEA (0.768 mL, 4.40 mmol) in DMF (5 mL). The mixture was stirred at room temperature. After 5 min, 8- fluoro-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (Intermediate 1, 0.334 g, 2.00 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 18h. The mixture was partitioned between water and EtOAc.
  • Example 3 Prepared as described for Example 7 using iodoethane (0.094 g, 0.60 mmol) and (3S)-3- amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l-one (Example 3, 0.148 g, 0.50 mmol) to afford the title compound.
  • Example 15 l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-2-(l,2,3,4- tetrahydroisoquinolm-l-yl)ethan-l-one Prepared as described for Example 2 using 2- ⁇ 2-[(tert-butoxy)carbonyl]-l,2,3,4- tetrahydroisoquinolin-l-yl ⁇ acetic acid (0.087 g, 0.30 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound.
  • Example 3 Prepared as described for Example 18 using cyclopropanecarbonyl chloride (0.023 g, 0.22 mmol) and (3 S)-3 -amino-3 -phenyl- 1 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan- 1 - one (Example 3, 0.059 g, 0.20 mmol) to afford the title compound.
  • TFAA (0.050 g, 0.24 mmol) was added to a solution of (3 S)-3-amino-3 -phenyl- 1 -(2,3 ,4,5- tetrahydro-l,5-benzoxazepin-5-yl)propan-l-one (Example 3, 0.059 g, 0.20 mmol) in MeCN (1 mL). The mixture was stirred at room temperature for 5 min. Further DIPEA (0.084 mL, 0.48 mmol) was added, followed by more TFAA (0.101 g, 0.48 mmol) and the reaction was allowed to stand for 5 min. The mixture was basified (DIP EA) and purified by reverse phase preparative HPLC to afford the title compound.
  • HATU (0.120 g, 0.32 mmol) was added to a solution of (3S)-3- ⁇ [(tert- butoxy)carbonyl]amino ⁇ -3-phenylpropanoic (0.080 g, 0.30 mmol) and DIPEA (0.058 mL, 0.33 mmol) in MeCN (1 mL) at room temperature. The mixture was stirred for 5 min. 6- fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepine (Intermediate 5, 0.050 g, 0.30 mmol) was added and the reaction was stirred under nitrogen until homogeneous and allowed to stand for 48h. The reaction was heated to 40-50 °C for lh.
  • HATU (0.399 g, 1.05 mmol) was added to a solution of 2- ⁇ 2-[(tert-butoxy)carbonyl]- l,2,3,4-tetrahydroisoquinolin-l-yl ⁇ acetic acid (0.291 g, 1.00 mmol) and DIPEA (0.192 mL, 1.10 mmol) in DMF (1 mL) at room temperature. The mixture was stirred for 5 min and 9- fluoro-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (Intermediate 2, 0.167 g, 1.00 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 72h.
  • Example 33 tert-butyl N-[2,2-difluoro-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyl] carbamate
  • HATU (0.28 g, 0.730 mmol) was added to a solution of 3-((tert-butoxycarbonyl)amino)- 2,2-difluoro-3-phenylpropanoic acid (Intermediate 6, 0.20 g, 0.66 mmol) and pyridine (0.054 mL, 0.66 mmol) in DMF (1 n L) under nitrogen. After 10 min 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.099 g, 0.66 mmol) was added. The reaction was stirred at room temperature for 24h. The crude reaction mixture was diluted with DCM (10 mL) and washed with saturated sodium bicarbonate solution. The organic phase was washed with water, dried (phase separator) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC to afford the title compound.
  • HATU (0.12 g, 0.32 mmol) was added to a solution of (3S)-3-phenylbutanoic acid (49.3 mg, 0.30 mmol) and DIPEA (0.058 mL, 0.33 mmol) in DMF (1 mL) at room temperature. The mixture was stirred for 5 minutes. 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 mg, 0.30 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 2h. The reaction mixture was purified by reverse phase preparative HPLC to afford the title compound.
  • Example 37 l-(3-hydroxy-3-methyl-2,3 ? 4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan-l-one
  • Oxalyl chloride (0.180 mL, 2.05 mmol) was added to a solution of 3-(2- methylphenyl)propanoic acid (0.224 g, 1.37 mmol) in DCM (2 mL). A drop of DMF was added to the reaction. The mixture was stirred at room temperature and allowed to stand. After ca. 1.5 h the mixture was concentrated in vacuo whilst maintaining the temperature below 30 °C, adding DCM to assist removal of oxalyl chloride. The residue was dissolved in DCM ⁇ ca.
  • Example 39 3-(2-methylphenyl)-l-(2,3 ) 4,5-tetrahydro-l,5-beiizoxazepin-5-yl)propan- 1-one
  • Example 42 4-amino-4-phenyl-l-(2 ⁇ ,4,5-tetrahydro-l,5-beiizoxazepin-5-yl)butan-l- one Prepared as described for Example 2 using 4- ⁇ [(te t-butoxy)carbonyl]amino ⁇ -4- phenylbutanoic acid (0.084 g, 0.30 mmol) and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound.
  • Oxalyl chloride (0.180 mL, 2.05 mmol) was added to a solution of 3-(2- methylphenyl)propanoic acid (0.224 g, 1.37 mmol) in DCM (2 mL). A drop of DMF was added to accelerate the reaction. The mixture was stirred at room temperature and allowed to stand. After ca. 1.5h the mixture was concentrated in vacuo while maintaining the temperature below 30 °C.
  • Example 45 l-(2,3,4,5-ieirahydro-l,5-bcnzoxazepin-5-yl)-2-( 1,2,3,4- tetrahydroisoquinolin-4-yl)ethan-l-one
  • HATU 0.523 g, 1.376 mmol
  • (3S)-3-(2-chlorophenyl)-3- ⁇ [(fert-butoxy)carbonyl] amino ⁇ propanoic acid (0.375 g, 1.251 mmol)
  • 9-fluoro-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine 0.209 g, 1.251 mmol
  • DIPEA 0.240 mL, 1.376 mmol
  • the crude product was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica). The residue was taken up in dioxane (1 mL) under nitrogen gas and HCl (4M in dioxane) (0.656 mL, 2.62 mmol) was added. The reaction was stirred at 80 °C for lh. The reaction was concentrated in vacuo and dried under vacuum at 80 °C for 30 min to afford the title compound.
  • HATU 0.99 g, 1.05 mmol
  • 3S 3- ⁇ [(tert- butoxy)carbonyl]amino ⁇ -3-phenylpropanoic
  • DIPEA 0.192 mL, 1.10 mmol
  • DMF 3 mL
  • 7- fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepine 0.167 g, 1.00 mmol
  • Acetyl chloride (0.026 g, 0.330 mmol) was added to a solution of l-(2,3,4,5-tetrahydro- l,5-benzoxazepin-5-yl)-2-(l,2,3,4-tetrahydroisoquinolin-4-yl)ethan-l-one (Example 45, 0.097 g, 0.300 mmol) and DIPEA (0.063 mL, 0.360 mmol) in DCM (1 mL) and stirred at room temperature for lh. The mixture was partitioned between water and DCM. The organic phase was concentrated in vacuo and the residue was purified by reverse phase preparative HPLC to afford the title compound.
  • Example 51 2-(2-acetyl-l,2 » 4-tetrahydroisoquinolin-l-yl)-l-(2,3 5 4,5-tetrahydro-l,5- benzoxazepin-5-y l)ethan-l -one
  • Acetyl chloride (0.007 g, 0.092 mmol) was added to a solution of l-(2,3,4,5-tetrahydro- l,5-benzoxazepin-5-yl)-2-(l,2,3,4-tetrahydroisoqumolin-l-yl)ethan-l-one (Example 15, 0.027 g, 0.084 mmol) and DIPEA (0.018 mL, 0.100 mmol) in MeCN (1 mL). The mixture was stirred at room temperature for 30 min. The reaction mixture was purified by reverse phase preparative HPLC to afford the title compound.
  • Example 35 was purified by chiral SFC (AD Daicel CHIRALPAK, 20% EtOH and 0.2% DEA) to afford the title compounds.
  • cyclopropylboronic acid (0.039 g, 0.450 mmol), tripotassium phosphate (0.191 g, 0.900 mmol) and tetrakis(triphenylphosphine)palladium(0) (0.010 g, 0.009 mmol) was dissolved in toluene (5 mL). Water (0.25 mL) was added and mixture was heated at 120 °C for 20 minutes in the microwave. The mixture was partitioned between water and EtOAc. The organic phase was dried (MgS0 4 ) and concentrated in vacuo. The resulting residue was purified by flash chromatography (20-50% EtOAc in petroleum ether on silica).
  • test compounds were assessed in a CRE- directed luciferase reporter gene assay, using a stable CHO cell line expressing the CRE- luc reporter and human mGluR7 genes. In this cell line, production of cAMP stimulated the transcription of the luciferase gene and luciferase activity was then measured in a luminescent enzyme assay (Steady Glo assay; Promega E2550). Activation of mGluR7 decreased the forskolin stimulated luminescence signal.
  • Forskolin (Sigma F3917) was then added to the wells (5 ⁇ of 2.5 ⁇ ) and the plate was incubated for five hours (37 °C). During this incubation, the Steady Glo Substrate reagent was warmed to 37 °C. Aliquots (11ml; stored at -20 °C) of this reagent were prepared by dissolving the contents of 1 vial of lyophilised substrate in 100 ml Steady-Glo buffer. A 25 ⁇ addition of the substrate was made to all wells and the plate was incubated for thirty minutes at RT, on a plate shaker (300 rpm; in the dark). Luminescence was then measured using the En Vision Multilabel Reader (Perkin Elmer).
  • Luminescence values were normalised to 'maximum' (forskolin alone) and 'minimum' (forskolin in the presence of tool mGluR7 agonist) controls.
  • EC 50 values were derived from this data using non-linear regression and a four parameter curve fit. The EC 5 o values for the compounds of the Examples are shown in Table 1.
  • Metabotropic glutamate 7 receptor subtype modulates motor symptoms in rodent models of Parkinson's disease. J Pharmacol Exp Ther., 332 (3), 1064-71.

Abstract

The present invention provides compounds of formula (I) and pharmaceutically acceptable salts thereof, wherein n, X1, X2, R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12and R13 are as defined in the specification, processes for their preparation, pharmaceutical compositions containing them and their use in therapy.

Description

DESCRIPTION
Title of Invention: NOVEL COMPOUNDS
The present invention relates to benzoxazepine derivatives, processes for their preparation, pharmaceutical compositions containing them and their use in therapy, particularly for use in treating disorders associated with changes in one or both of the glutamatergic and GAB Aergic signalling pathways regulated in full or in part by metabotropic glutamate receptor 7 (mGluR7).
L-Glutamate is the major neurotransmitter in the mammalian central nervous system and activates both ionotropic and metabotropic glutamate receptors. L-Glutamate plays a central role in numerous physiological functions such as learning and memory (1), sensory perception, development of synaptic plasticity, motor control, respiration and regulation of cardiovascular function. Thus an imbalance in glutamatergic neurotransmission often underlies many neuropathological conditions.
The metabotropic glutamate receptors are a family of G protein-coupled receptors that have been divided into three groups on the basis of sequence homology, putative signal transduction mechanisms and pharmacologic properties. Group I includes mGluRl and mGluR5 and these receptors have been shown to activate phospholipase C. Group II includes mGluR2 and mGluR3 whilst Group III includes mGluR4, mGluR6, mGluR7 and mGluR8. Group II and III receptors are linked to the inhibition of the cyclic AMP cascade but differ in their agonist selectivities. mGluR7 is an inhibitory GPCR expressed pre-synaptically at the synaptic cleft on
GABAergic and glutamatergic neurons. Depending on the location it can inhibit or disinhibit synaptic activity and can therefore be seen as a modulator of neuronal function. Therefore, mGluR7 modulators would be expected to be useful in treating a wide variety of neurological and psychiatric disorders such as Parkinson's disease (2, 3); dementia associated with Parkinson's disease (3, 4); Alzheimer's disease (5); Huntington's Chorea (6); amyotrophic lateral sclerosis and multiple sclerosis; bipolar disorder (6, 7); psychiatric diseases such as schizophrenia, post-traumatic stress disorder, anxiety disorders and depression (1,4, 6, 8-11); and addiction. They may also be useful in treating age-related hearing loss/tinnitus (12). There is a need for treatment of the above conditions and others described herein with compounds that are mGluR7 modulators. The present invention provides modulators of mGluR7.
In accordance with the present invention, there is provided a compound of formula (I) or a pharmaceutically acceptable salt thereof
Figure imgf000003_0001
wherein
X1 is absent or is -CH2- or -CH(CH3)-;
X2 is -CH2-, -CHF-, -CF2- or -CH2CH2-, provided that when X1 is -CH2- or -CH(CH3)-, then X2 is -CH2- only;
R1, R2, R3 and R4 each independently represent hydrogen, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; either R5 represents hydrogen, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy and R6 represents hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkylsulphonyl or -NR14R15, or when X1 is absent, R5 and R6 together form a linking group, -[CH2]m-5 in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or
N(COCH3) heteroatom moieties provided that if the linking group contains two heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another; n is 0, 1 or 2;
n
each R independently represents halogen, cyano, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; each of R 8 , R 9 , R 10 , R 1 1 , R 12 and R 13 independently represents hydrogen, hydroxyl, cyano, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; or,
alternatively,
(i) R and R together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring; or
(ii) R10 and R11 together with the carbon atom to which they are attached form a saturated
3- to 6-membered carbocyclic ring; or
(iii) R 12 and R 13 together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring; or
(iv) R9 and R10 together with the carbon atoms to which they are attached form a saturated
3- to 6-membered carbocyclic ring; or
(v) R 8 and R 12 together form a methylene bridge,
provided that no more than one carbocyclic ring or methylene bridge according to (i) to (v) above can be present in ring A; and
either R14 and R15 each independently represent hydrogen or a C1-C6 alkyl, C3-C6 cycloalkylmethyl, C1-C6 alkylcarbonyl, C3-C6 cycloalkylcarbonyl or C1-C6
alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by at least one halogen atom, or
R14 and R15 together with the nitrogen atom to which they are attached form a saturated 4- to 6-membered heterocyclic ring optionally comprising a further ring heteroatom selected from nitrogen and oxygen, the heterocyclic ring being unsubstituted or substituted by at least one substituent selected from halogen, oxo and C1-C3 alkyl. In the context of the present specification, unless otherwise stated, an "alkyl" substituent group or an "alkyl" moiety in a substituent group (such as an alkoxy group) may be linear or branched.
Examples of C1-C6 alkyl groups/moieties include methyl, ethyl, propyl, 2-methyl-l- propyl, 2-methyl-2-propyl, 2-methyl-l -butyl, 3 -methyl- 1 -butyl, 2-methy 1-3 -butyl, 2,2- dimethyl-1 -propyl, 2~methyl-pentyl, 3 -methyl- 1-pentyl, 4-methyl-l-pentyl, 2-methyl-2- pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-l -butyl, 3,3-dimethyl-l- butyl, 2-ethyl-l -butyl, n-butyl, tert-butyl, n-pentyl, and n-hexyl.
A "cycloalkyl" substituent group or a "cycloalkyl" moiety in a substituent group refers to a saturated hydrocarbyl ring containing, for example, from 3 to 8 carbon atoms, examples of which include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term "oxo" refers to an oxygen atom doubly bonded to the carbon atom to which it is attached to form the carbonyl of a ketone or aldehyde.
The term "halogen" includes fluorine, chlorine, bromine and iodine.
When R14 and R15 form a saturated 4- to 6-membered heterocyclic ring which is substituted, it should be understood that the substituent(s) may be attached to any suitable ring atom.
For the purposes of the present invention, where a combination of moieties is referred to as one group, for example, alkylsulphonyl or alkoxycarbonyl, the last mentioned moiety contains the atom by which the group is attached to the rest of the molecule.
When any chemical group or moiety in formula (I) is described as substituted, it will be appreciated that the number and nature of substituents will be selected so as to avoid sterically undesirable combinations. Further, it will be appreciated that the invention does not encompass any unstable ring or other structures (e.g. >NCH2N<, >NCH20- or aminal groupings of the type
>C(NRaRb)(NRcRd)) or any 0-0 or S-S bonds.
X1 is absent or is -C¾- or -CH(CH3)- and X2 is -CH2-, -CHF-, -CF2- or -CH2CH2-,
1 2
provided that when X is -CH2- or -CH(CH3)-, then X is -CH2- only. Consequently, when X1 is absent, X2 is -CH2-, -CHF-, -CF2- or -CH2CH2-, and when X1 is -C¾- or -CH(CH3)-, then X2 is -CH2-;
In one embodiment, X1 is absent.
2
In another embodiment, X is -CH2-, -CF2- or -CH2CH2-.
In yet another embodiment, X is -CH2-
In a still further embodiment, X and X have the following meanings:
Figure imgf000006_0001
R , R , R and R each independently represent hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy. In one embodiment, R , R , R and R each independently represent hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C4, or C1-C3, or C1-C2 alkyl (e.g. methyl), cyclopropyl or C1-C4, or C1-C3, or C1-C2 alkoxy (e.g. methoxy).
In another embodiment, R1, R2, R3 and R4 each independently represent hydrogen, fluorine, chlorine, methyl or methoxy.
In a further embodiment, R1 represents hydrogen, fluorine, chlorine, methyl or methoxy
2 3 4
and R , R and R each represent hydrogen.
In a still further embodiment, R represents hydrogen, fluorine, chlorine, methyl or methoxy and R1, R2 and R4 each represent hydrogen.
According to one aspect of the invention, R5 represents hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy and R6 represents hydrogen, C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl, C1-C6, or C1-C4, or C1-C2 alkylsulphonyl or NR14R15.
In one embodiment, R5 represents hydrogen, halogen (e.g. fluorine, chlorine or bromine), C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropyl or C1-C4, or C1-C3, or C1-C2 alkoxy.
In another embodiment, R5 represents hydrogen, fluorine, chlorine, bromine, C1-C2 alkyl, cyclopropyl or C1-C2 alkoxy. In still another embodiment, R represents hydrogen, fluorine, chlorine, bromine, cyclopropyl, methyl or methoxy.
In yet another embodiment, R1, R2, R3, R4 and R5 each independently represent hydrogen or halogen.
In one embodiment, R6 represents hydrogen, C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropyl, C1-C4, or C1-C3, or C1-C2 alkylsulphonyl or NR14R15.
In another embodiment, R6 represents hydrogen, C1-C2 alkyl or NR14R15.
In a further embodiment, R6 represents NR14R15.
R14 and R15 each independently represent hydrogen or a C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkylmethyl, C1-C6, or C1-C4, or C1-C2 alkylcarbonyl, C3-C6, or C4-C6, or C5-C6 cycloalkylcarbonyl or C1-C6, or C1-C4, or
C1-C2 alkoxycarbonyl group, wherein each of the preceding groups is optionally
substituted by at least one halogen (such as fluorine, chlorine or bromine) atom (e.g. one, two, three, four or five halogen atoms), or
R14 and R15 together with the nitrogen atom to which they are attached form a saturated 4-,
5- or 6-membered heterocyclic ring optionally comprising a further ring heteroatom selected from nitrogen and oxygen, the heterocyclic ring being unsubstituted or substituted by at least one substituent (e.g. one, two or three substituents independently) selected from halogen (e.g. fluorine, chlorine or bromine), oxo and C1-C3 alkyl (e.g. methyl or ethyl).
In one embodiment, R14 and R15 each independently represent hydrogen or a C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropylmethyl, C1-C4, or C1-C3, or C1-C2 alkylcarbonyl, cyclopropylcarbonyl or C1-C4, or C1-C3, or C1-C2 alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by one to five, or one to three, halogen, particularly fluorine, atoms.
In another embodiment, R and R each independently represent hydrogen or a
C1-C2 alkyl, cyclopropylmethyl, C1-C3 alkylcarbonyl, cyclopropylcarbonyl or Ci-
C4 alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by one to five, or one to three, halogen, particularly fluorine, atoms.
In yet another embodiment, R14 and R15 each independently represent hydrogen or a
C1-C2 alkyl, cyclopropylmethyl, C1-C3 alkylcarbonyl, cyclopropylcarbonyl or
C1-C4 alkoxycarbonyl group, wherein the C1-C2 alkyl and C1-C3 alkylcarbonyl groups are optionally substituted by one to three halogen, particularly fluorine, atoms.
In still another embodiment, R14 and R15 together with the nitrogen atom to which they are attached form a saturated 5-membered heterocyclic ring optionally comprising a further ring heteroatom selected from nitrogen and oxygen, the heterocyclic ring being
unsubstituted or substituted by at least one substituent (e.g. one, two or three substituents independently) selected from fluorine, chlorine, oxo, methyl and ethyl.
In a particular embodiment of the invention, -NR14R15 represents any one of the following moieties or is selected from a group containing any two or more of such moieties:
(i) -NH2
(ii) -NHCH3
(iii) -N(CH3)2
(iv) -NHC2H5
(v) -NHCH2CHF2
(vi) -NH(cyclopropylmethyl)
(vii) -NHC(0)CH3 (viii) -NHC(0)CF3
(ix) -NHC(0)C2H5
(x) -NHC(0)CH(CH3)2
(xi) -NHC(0)cyclopropyl
(xii) -NHC(OXtert-butoxy)
(xiii) pyrrolidin-2-one.
In a particular embodiment of the invention, R1 to R6 have the following meanings where -
NR R is as defined above:
R1 R2 R3 R4 R5 R6
H H H H H -NR14R15
H H F H H -NR14R15
H H H H F -NR14R15
F H H H H -NR14R15
CH3 H H H H -NR14R15
H H H H CH3 -NR14R15
-OCH3 H H H H -NR14R15
H H H H -OCH3 -NR14R15
CI H H H H -NR14R15
H H H H CI -NR14R15
H H H H Br -NR14R15
H H H H cyclopropyl -NR14R15
H H H H H CH3
H H H H H H
CH3 H H H H H
H H H H CH3 H According to a second aspect of the invention, when X1 is absent, R5 and R6 together form a linking group, -[0¾]ΠΓ, in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or N(COCH3)heteroatom moieties provided that if the linking group contains two heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another. Examples of such linking groups include: -CH2CH2NH-, CH2CH2N(COCH3)-, -NHCH2CH2-, -N(COCH3)CH2CH2- , -CH2NHCH2-, -CH2N(COCH3)CH2-, -OCH2CH2NH- or -NHCH2CH20-. η
Each R independently represents halogen (e.g. fluorine, chlorine or bromine), cyano, C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy.
In an embodiment of the invention, each R independently represents halogen (e.g. fluorine, chlorine or bromine), cyano, C1-C4, or Ci-C3, or C1-C2 alkyl (e.g. methyl), cyclopropyl or C1-C4, or Ci-C3, or C1-C2 alkoxy (e.g. methoxy).
In another embodiment, each R independently represents fluorine, chlorine, methyl or methoxy.
In yet another embodiment, n is 1 and R represents a halogen atom, particularly a fluorine atom.
In still another embodiment, n is 0.
According to an aspect of the present invention, R , R , R , R , R and R each independently represent hydrogen, hydroxyl, cyano, halogen (e.g. fluorine, chlorine or bromine), C1-C6, or C1-C4, or C1-C2 alkyl, C3-C6, or C4-C6, or C5-C6 cycloalkyl or C1-C6, or C1-C4, or C1-C2 alkoxy.
In one embodiment, R , R , R , R , R and R each independently represent hydrogen, hydroxyl, cyano, halogen (e.g. fluorine,chlorine or bromine), C1-C4, or C1-C3, or C1-C2 alkyl, cyclopropyl or C1-C4, or C1-C3, or C1-C2 alkoxy.
In another embodiment, R 8 , R 9 , R 10 , R 1 1 , R 12 and R 13 each independently represent hydrogen, hydroxyl, halogen (e.g. fluorine or chlorine), C1-C2 alkyl, cyclopropyl or
C1-C2 alkoxy.
In a further embodiment, R 8 , R 9 , R 10 , R 1 1 , R 12 and R 13 each independently represent hydrogen, hydroxyl, fluorine, methyl or methoxy.
Particular values of R 8 to R 13 are shown in the following table:
R10
R8 R9 R11 R12 R13
H H H H H H
H H CH3 OH H H
H H OH CH3 H H
H H H OCH3 H H
H H OCH3 H H H
H H F F H H
H H H F H H
H H F H H H
CH3 H H H H H According to a second aspect of the invention, R and R together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring and R10 to
R 13 are as defined above.
According to a third aspect of the invention, R and R together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring and R 8 , R 9 ,
R 1^2 and R 113J are as defined above.
According to a fourth aspect of the invention, R and R together with the carbon atom
o to which they are attached form a saturated 3- to 6-membered carbocyclic ring and R to
R are as defined above.
According to a fifth aspect of the invention, R and R together with the carbon atoms to which they are attached form a saturated 3- to 6-membered carbocyclic ring and R 8 , R 1 1 , R 12 and R 13 are as defined above.
According to a sixth aspect of the invention, R and R together form a methylene bridge and R9, R10, R11 and R13 are as defined above.
Examples of bicyclic ring systems (AB) according to the above-mentioned second to sixth aspects of the invention include the following structures:
Figure imgf000014_0001
In a preferred embodiment of the invention, the compounds of formula (I) are those in which:
X1 is absent or is -CH2- or -CH(CH3>-; X2 is -CH2-, -CF2- or -CH2CH2-;
R1, R2, R3 and R4 each independently represent hydrogen, fluorine, chlorine, methyl or methoxy;
either R5 represents hydrogen, fluorine, chlorine, bromine, cyclopropyl, methyl or methoxy and R6 represents hydrogen, methyl or -NR14R15, or when X1 is absent, R5 and R6 together form a linking group, -[CH2]m-5 in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or
N(COCH3) heteroatom moieties provided that if the linking group contains two heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another;
n is 0 or 1 ;
η
R represents halogen, methyl or methoxy; each ofR 8, R 9, R 10 , R 11 , R 12 and R 13 independently represents hydrogen, hydroxyl, fluorine, methyl or methoxy or, alternatively, either R10 and R11 together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring, or R and R together form a methylene bridge; and either R and R each independently represent hydrogen or a C i -C2 alkyl, cyclopropylmethyl, C1-C3 alkylcarbonyl, cyclopropylcarbonyl or C1-C4 alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by at least one halogen atom, or
R1 and R15 together with the nitrogen atom to which they are attached form a saturated 5-membered heterocyclic ring, the heterocyclic ring being unsubstituted or substituted by at least one substituent selected from halogen, oxo and C1-C3 alkyl.
In another preferred embodiment, there is provided a subset of compounds of formula (I) as represented by formula (la)
Figure imgf000015_0001
in which
R 3a is hydrogen or fluorine;
p is 0 or 1 ;
R7a is fluorine;
R10a is hydrogen or fluorine; and R1 la is hydrogen or fluorine.
According to one aspect, the compounds of formula (la) contain a single fluorine substituent. If R7a is a fluorine substituent, it is preferably attached to the 9-position of the bicyclic ring structure AB (which is shown marked with an asterisk in formula (la) above). According to another aspect, the compounds of formula (la) contain two fluorine substituents as Rl0a and Rl la.
Examples of compounds of the invention include:
N-[3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propyl]acetamide;
(3R)-3-amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l-one;
(3 S)-3-amino-3-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan-l -one;
(3 S)-3-amino- 1 -(8-fluoro-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3S)-3-amino-3-(4-fluorophenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-amino-3-(2-fluorophenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-(methylamino)-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-(dimethylamino)-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan- 1 -one;
(3 S)-3-amino- 1 -(9-fluoro-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3 -amino-3 -(2-methylphenyl)- 1 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one;
(3 S)-3 -amino-3 -(2-methoxyphenyl)- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-amino-3-(2-chlorophenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3 S)-3 -(ethylamino)-3 -phenyl- 1 -(2,3 ,4,5-tetrahydro- 1 ,5 -benzoxazepin-5 -yl)propan-
1-one;
(3S)-3-[(cyclopropylmethyl)amino]-3-phenyl-l-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propan- 1 -one;
l-(2,3,4,5-tetrahycko-l,5-benzoxazepm-5-yl)-2-(l,2,3,4-tetrahydroisoquinolin-l- yl)ethan-l-one; (3 S)- 1 -(3 -hydroxy-3 -methyl-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 - (methylamino)-3-phenylpropan- 1 -one;
N-[( 1 S)-3 -oxo- 1 -phenyl-3 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propyl]propanamide;
N-[(l S)-3-oxo-l -phenyl-3-(2,3,4,5-tetrahydro-l ,5-benzoxazepin-5- yl)propyl]cyclopropanecarboxamide;
2-methyl-N-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]propanamide;
2,2,2-trifluoro-N-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahya^o-l,5-benzoxazepin-5- yl)propyl]acetamide;
N-[(lS)-l-(2-methylphenyl)-3-oxo-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]acetamide;
N- [( 1 S)- 1 -(2-chlorophenyl)-3-oxo-3 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propyl]acetamide;
l-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]pyrrolidin-2-one;
(3 S)-3-amino- 1 -(6-fluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3S)-3-amino-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(4- fluorophenyl)propan- 1 -one;
l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-2-(l,2,3,4- tetrahydroisoquinolin- 1 -yl)ethan- 1 -one;
(3S)-3-amino-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- fluorophenyl)propan- 1 -one;
(3S)-3-[(2,2-difluoroethyl)arnino]-3-phenyl-l-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5 -yl)propan- 1 -one;
(3S)-3-amino-l-(9-fluoro-2,3,4,5-te1xahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan- 1 -one;
(3 S)- 1 -(9-fluoro-2,3,4,5-tetrahydro- 1 ,5-berizoxazepin-5-yl)-3-(methylarriino)-3- phenylpropan- 1 -one;
tert-butyl N-[2,2-difluoro-3-oxo- 1 -phenyl-3-(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin- 5 -yl)propy 1] carbamate ; 3 -amino-2,2-difluoro-3 -phenyl- 1 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-amino-l-(3-methoxy-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)butan- 1 -one;
l-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan- 1 -one;
4-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)pentan- 1 -one;
3- (2-methylphenyl)- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan- 1 -one; (3R)-3-amino-4-phenyl-l -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)butan- 1 -one; (3 S)-3-amino-4-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)butan- 1 -one;
4- amino-4-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)butan- 1 -one;
l-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan- 1 -one;
1- (2,3,4,5-te1xahydro-l,5-benzoxazepin-5-yl)-2-(l,2,3,4-tetTahydroisoquinolin-4- yl)ethan-l-one;
(3S)-3-amino-3-(2-chlorophenyl)-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin- 5-yl)propan- 1 -one;
(3 S)-3 -amino- 1 -(3 ,3 -difluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3 -amino- 1 -(7-fluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 - phenylpropan- 1 -one;
2- (2-acetyl- 1 ,2,3 ,4-tetrahydroisoquinolin-4-yl)- 1 -(2,3 ,4,5-tetrahydro- 1,5- benzoxazepin-5-yl)ethan- 1 -one;
(3S)-3-amino-l-(3-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
2-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-l-yl)-l-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)ethan- 1 -one;
(3 S)-amino- 1 - { 3 ,4-dihydro-2,4-methanobenzo [b] [ 1 ,4] oxazepin-5 -yl } -3 - phenylpropan- 1 -one
(3S)-3-amino-l-[(4S)-9-fluoro-4-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl] -3 -phenylpropan- 1 -one; (3S)-3-amino-l-(9-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3-amino- 1 -(9-chloro-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3-amino- 1 -[(4R)-9-fluoro-4-methyl-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl] -3 -phenylpropan- 1 -one;
(3 S)-3-amino- 1 - {4,5-dihydro-2H-spiro[ 1 ,5-benzoxazepine-3 , 1 '-cyclopropane] -5- yl } -3 -phenylpropan- 1 -one;
(3 S)-3-amino- 1 -(9-methoxy-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3S)-3-aniino-3-(2-bromophenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-amino-3-(2-cyclopropylphenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one; and enantiomers, diastereoisomers and mixtures thereof; and
pharmaceutically acceptable salts of any of the foregoing.
It should be noted that each of the chemical compounds listed above represents a particular and independent aspect of the invention.
The present invention further provides a process for the preparation of a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined above which comprises reacting a compound of formula (II), or a salt (e.g. hydrochloride salt) thereof,
Figure imgf000019_0001
in which X1, X2, R1, R2, R3, R4, R5 and R6 are as defined in formula (I) above, with a compound of formula (III), or a salt (e.g. hydrochloride salt) thereof,
Figure imgf000020_0001
in which n, R7, R8, R9, R10, R1 \ R12 and R13 are as defined in formula (I) above; and optionally thereafter carrying out one or more of the following procedures:
• converting a compound of formula (I) into another compound of formula (I)
• removing any protecting groups
• forming a pharmaceutically acceptable salt.
The above process may conveniently be carried out by combining the carboxylic acid of formula (II) with the amine of formula (III) in the presence of a coupling reagent such as
(1) EDC (l-ethyl-3-(3-dimethylaminopropyl)carbodiimide) and HOAt (7-aza-l- hydroxybenzotriazole) with triethylamine in dichloromethane at room temperature to 60°C, or
(2) HATU (l-[bis(dimethylamino)memylene]-lH-l,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate) with triethylamine in dichloromethane at room temperature to 60°C; or
(3) HATU with N,N-diisopropylethylamine in dimethylformamide at room temperature to 60°C; or
(4) Propylphosphonic anhydride with triethylamine in dichloromethane at room
temperature to 60°C;
followed, if necessary, to remove any protecting groups (e.g. a tert-butyloxycarbonyl group ifR6 is a -NR14R15 group such as -NH2), by reaction with hydrochloric acid in an organic solvent, e.g. a polar solvent such as 1,4-dioxane, at a temperature in the range from, for example, room temperature to 100°C. Compounds of formulae (II) and (III) are known compounds or may be prepared according to processes known in the art. For example, compounds of formula (II) in which R1 to R5 are each hydrogen, X is absent, X is -CHF- and R is -NH2 may be prepared as shown in the following reaction scheme 1 :
Scheme 1
Figure imgf000021_0001
in which the abbreviations used have the following meanings:
Boc : tert-butyloxycarbonyl (a protecting group)
LDA : lithium diisopropylamide
THF : tetrahydrofuran
NFSI : N-Fluorobenzenesulfonimide
Compounds of formula (II) in which one of R1 to R5 is a C3-C6 cycloalkyl, e.g. cyclopropyl, group and the remainder are hydrogen, X 1 is absent, X 2 i ·s -CH2- and R I is -NH2 may be prepared as shown in the following reaction scheme 2:
Scheme 2
Figure imgf000022_0001
Figure imgf000022_0002
in which the abbreviations used have the following meanings:
Boc : fert-butyloxycarbonyl (a protecting group)
MeOH : methanol Compounds of formula (II) in which R 1 to R5 are each hydrogen, X 1 is absent, X2 is -CH2- and R6 is C1-C6 alkylsulphonyl (e.g. ethyl sulphonyl) may be prepared as in the following reaction scheme 3:
Scheme 3
Figure imgf000022_0003
TEA, MeCN, 80°C
step (iii)
Figure imgf000022_0004
Step (i) is analogous to methods described in the journal article by S. Has-Becker et al., Synlett 2001, No. 9, 1395-1398. TEA denotes trimethylamine and MeCN denotes acetonitrile. Step (ii) is an oxidation reaction which may conveniently be carried out using 3- chlorobenzenecarboperoxoic acid (mCPBA) in an organic solvent such as dichloromethane at a temperature in the range of from room temperature to 60°C.
Step (iii) is a hydrolysis reaction which may conveniently be carried out in the presence of a base such as lithium hydroxide in a polar solvent, e.g. a mixture of water and
tetrahydrofuran, at a pressure of 8 bar (800 kPa) and at a temperature in the range of from room temperature to 60°C.
As an alternative, a compound of formula (II) in which R1 to R5 are each hydrogen, X1 is absent, X is -CH2- and R is methyl sulphonyl may be prepared as shown in reaction scheme 3 except that step (i) is replaced as follows (see methods described in US
2010/0048939 in the name of Dow Agrosciences LLC):
Esterification
Figure imgf000023_0001
in which the abbreviations used have the following meanings:
MeOH: methanol
AcOH: acetic acid (ethanoic acid)
NaSMe: sodium methanethiolate
MeCN: acetonitrile
In one embodiment, a compound of formula (I) may be converted into another compound
η
of formula (I). For example, a compound of formula (I) in which R represents a halogen atom may be converted to a corresponding compound of formula (I) in which R represents a C1-C6 alkyl or C3-C6 cycloalkyl group by reacting the former with a boronic acid derivative such as R7a-B(OH)2, R7a-B(pinacole ester) or R7a-BF3~K+ where R7a represents the replacement R bonded to the boron atom via a carbon-boron bond, in the presence of a base such as potassium carbonate, caesium carbonate or potassium phosphate, and a metal catalyst such as Pd(0), typically where the metal catalyst is in the form of a transition metal complex such as tetrakis(triphenylphosphine) palladium or di-tert- butyl [dichloro( { di-tert-buty 1 [4-(dimethylamino)pheny 1] -phosphaniumyl } )palladio] [4- (dimethylamino)phenyl] phosphanium. A solvent such as a dioxane/water mixture may be used and the reaction mixture is typically heated, e.g. to around 100-160°C under conventional heating or microwave irradiation. η
Further, a compound of formula (I) in which R represents a halogen atom may be converted to a corresponding compound of formula (I) in which R represents a Ci-
C6 alkoxy group by reacting the former with the desired alcohol in the presence of a base such as sodium hydride and a solvent such as anhydrous tetrahydrofuran. The reaction mixture is typically heated, e.g. to around 60-120°C under conventional heating or microwave irradiation.
Still further, a compound of formula (I) in which R10 represents a hydroxyl group may be converted to a corresponding compound of formula (I) in which R10 represents a cyano group according to methods described in US 2002/0188124 in the name of Fukami et al. (Banyu Pharmaceutical Co. Ltd.), for example,
Figure imgf000024_0001
Figure imgf000024_0002
in which the abbreviations used have the following meanings and the deprotection reaction is carried out according to procedures known in the art, for example, as described in Scheme 1 above:
TEA: trimethylamine
MsCl: methanesulphonyl chloride
DMF: dimethylformamide
It will be appreciated by those skilled in the art that in the processes of the present invention certain functional groups such as phenol, hydroxyl or amino groups in the reagents may need to be protected by protecting groups. Thus, the preparation of the compounds of formula (I) may involve, at an appropriate stage, the introduction and/or removal of one or more protecting groups.
The protection and deprotection of functional groups is described, for example, in
'Protective Groups in Organic Chemistry', edited by J.W.F. McOmie, Plenum Press (1973); 'Greene's Protective Groups in Organic Synthesis', 4th edition, T.W. Greene and P.G.M. Wuts, Wiley-Interscience (2007); and 'Protecting Groups', 3rd edition, P. J. ocienski, Thieme (2005).
The compounds of formula (I) above may be converted to a pharmaceutically acceptable salt thereof, preferably an acid addition salt such as a formate, hemi-formate,
hydrochloride, hydrobromide, benzenesulphonate (besylate), saccharin (e.g.
monosaccharin), trifluoroacetate, sulphate, nitrate, phosphate, acetate, fumarate, maleate, tartrate, lactate, citrate, pyruvate, succinate, valerate, propanoate, butanoate, malonate, oxalate, l-hydroxy-2-napthoate (xinafoate), methanesulphonate or -toluenesulphonate salt. In one embodiment of the invention, the compounds of formula (I) are in the form of a hydrochloride salt.
In one aspect of the invention, compounds of formula (I) may bear one or more radiolabels. Such radiolabels may be introduced by using radiolabel-containing reagents in the synthesis of the compounds of formula (I), or may be introduced by coupling the
compounds of formula (I) to chelating moieties capable of binding to a radioactive metal atom. Such radiolabeled versions of the compounds may be used, for example, in diagnostic imaging studies. Unless stated otherwise, any atom specified herein may also be an isotope of said atom. For example, the term "hydrogen" encompasses !H, 2H and 3H. Similarly carbon atoms are to be understood to include 12C, 13C and 14C, nitrogen atoms are to be understood to include 14N and 15N, and oxygen atoms are to be understood to include 160, 170 and 180.
In a further aspect of the invention, compounds of formula (I) may be isotopically labelled. As used herein, an "isotopically labelled" compound is one in which the abundance of a particular nuclide at a particular atomic position within the molecule is increased above the level at which it occurs in nature.
Compounds of formula (I) and their salts may be in the form of hydrates or solvates which form an aspect of the present invention. Such solvates may be formed with common organic solvents, including but not limited to, alcoholic solvents e.g. methanol, ethanol or isopropanol.
Where compounds of formula (I) are capable of existing in stereoisomeric forms, it will be understood that the invention encompasses the use of all geometric and optical isomers (including atropisomers) of the compounds of formula (I) and mixtures thereof including racemates. The use of tautomers and mixtures thereof also forms an aspect of the present invention. Enantiomerically pure forms are particularly desired. "Enantiomerically pure" denotes the presence of at least 75%w, in particular at least 90%w and, more particularly, at least 95%w of one of the two possible enantiomers of a compound.
Compounds of formula (I) and their salts may be amorphous or in a polymorphic form or a mixture of any of these, each of which forms an aspect of the present invention.
The compounds of formula (I) and their pharmaceutically acceptable salts have activity as pharmaceuticals and may be used in treating conditions or disorders associated with changes in one or both of the glutamatergic and GABAergic signalling pathways regulated in full or in part by metabotropic glutamate receptor 7. Thus, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined for use in therapy, in particular for the treatment of conditions associated with metabotropic glutamate receptor 7.
The present invention also provides the use of a compound of formula (I) or a
pharmaceutically acceptable salt thereof as hereinbefore defined for the preparation of a medicament for the treatment of conditions associated with metabotropic glutamate receptor 7.
The present invention still further provides a method of treating a condition associated with metabotropic glutamate receptor 7 which comprises administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I) or a
pharmaceutically acceptable salt thereof as hereinbefore defined.
In the context of the present specification, the term "therapy" also includes "prophylaxis" unless there are specific indications to the contrary. The terms "therapeutic" and
"therapeutically" should be construed accordingly.
Prophylaxis is expected to be particularly relevant to the treatment of persons who have suffered a previous episode of, or are otherwise considered to be at increased risk of, the disorder or condition in question. Persons at risk of developing a particular disorder or condition generally include those having a family history of the disorder or condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the disorder or condition or those in the prodromal phase of a disorder.
The terms "treat", "treatment" and "treating" include improvement of the conditions described herein. The terms "treat", "treatment" and "treating" include all processes providing slowing, interrupting, arresting, controlling, or stopping of the state or progression of the conditions described herein, but does not necessarily indicate a total elimination of all symptoms or a cure of the condition. The terms "treat", "treatment," and "treating" are intended to include therapeutic as well as prophylactic treatment of such conditions. As used herein the terms "condition," "disorder," and "disease" relate to any unhealthy or abnormal state. The term "conditions associated with metabotropic glutamate receptor 7" includes conditions, disorders and diseases in which the modulation of mGluR7 may provide a therapeutic benefit, examples of which include:
(1) Nervous system disorders: Parkinson's disease, including dementia associated with Parkinson's disease; Alzheimer's disease; Huntington's Chorea; amyotrophic lateral sclerosis; multiple sclerosis; bipolar disorder; and psychiatric disorders such as
schizophrenia, post-traumatic stress disorder, anxiety disorders and depression (e.g. major depressive disorder);
(2) Addiction disorders: alcohol, drug or nicotine addiction;
(3) Hearing disorders: hearing loss and/or tinnitus caused by age, noise or trauma; and
(4) Others: idiopathic autism; severe neonatal encephalopathy; autism spectrum disorder (ASD); X-linked intellectual disability (also known as X-linked mental
retardation); epilepsy; cerebral ischemias; eye disorders; and pain (e.g. inflammatory pain or neuropathic pain).
Schizophrenia is a debilitating psychiatric disorder characterised by a combination of negative symptoms (such as social withdrawal, anhedonia, avolition and apathy) and positive symptoms (including hallucinations, delusions and paranoia) as well as marked cognitive deficits (such as impairment of executive function). The executive function (EF) has been defined as "a set of abilities, which allows us to invoke voluntary control of our behavioral responses. These functions enable human beings to develop and carry out plans, make up analogies, obey social rules, solve problems, adapt to unexpected circumstances, do many tasks simultaneously, and locate episodes in time and place. EF includes divided attention and sustained attention, working memory (WM), set-shifting, flexibility, planning, and the regulation of goal directed behavior and can be defined as a brain function underlying the human faculty to act or think not only in reaction to external events but also in relation with internal goals and states" (Orellana G. and Slachevsky A., 2013. Executive Functioning in Schizophrenia. Front. Psychiatry, 4, 35). Accordingly, the present invention also provides a method of treating a negative symptom, a positive symptom and/or a cognitive deficit associated with a psychiatric disorder, especially schizophrenia, which comprises administering to a patient in need thereof a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined.
For the above-mentioned therapeutic uses the dosage administered will, of course, vary with the compound employed, the mode of administration, the treatment desired and the disorder indicated. For example, the daily dosage of the compound of the invention, if inhaled, may be in the range from 0.05 micrograms per kilogram body weight (p.g/kg) to 100 micrograms per kilogram body weight ^g/kg). Alternatively, if the compound is administered orally, then the daily dosage of the compound of the invention may be in the range from 0.01 micrograms per kilogram body weight ^g/kg) to 100 milligrams per kilogram body weight (mg/kg).
The compounds of formula (I) and pharmaceutically acceptable salts thereof may be used on their own but will generally be administered in the form of a pharmaceutical composition in which the formula (I) compound/salt (active ingredient) is in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
Therefore the present invention further provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined, in association with a pharmaceutically acceptable adjuvant, diluent or carrier.
The invention still further provides a process for the preparation of a pharmaceutical composition of the invention which comprises mixing a compound of formula (I) or a pharmaceutically acceptable salt thereof as hereinbefore defined with a pharmaceutically acceptable adjuvant, diluent or carrier.
Conventional procedures for the selection and preparation of suitable pharmaceutical formulations are described in, for example, "Pharmaceutics - The Science of Dosage Form Design", M. E. Aulton, Churchill Livingstone, 1988. Pharmaceutically acceptable adjuvants, diluents or carriers that may be used in the pharmaceutical compositions of the invention are those conventionally employed in the field of pharmaceutical formulation, and include, but are not limited to, sugars, sugar alcohols, starches, ion exchangers, alumina, aluminium stearate, lecithin, serum proteins such as human serum albumin, buffer substances such as phosphates, glycerine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes such as protamine sulphate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium
carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.
The pharmaceutical compositions of the present invention may be administered orally, parenterally, by inhalation spray, rectally, nasally, buccally, vaginally or via an implanted reservoir. Oral administration is preferred. The pharmaceutical compositions of the invention may contain any conventional non-toxic pharmaceutically acceptable adjuvants, diluents or carriers. The term parenteral as used herein includes subcutaneous,
intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
The pharmaceutical compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. The suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic
parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable diluents and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long- chain alcohol diluent or dispersant.
The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, powders, granules, and aqueous suspensions and solutions. These dosage forms are prepared according to techniques well-known in the art of pharmaceutical formulation. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavouring and/or colouring agents may be added.
The pharmaceutical compositions of the invention may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing the active ingredient with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active ingredient. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
The pharmaceutical compositions of this invention may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance
bioavailability, fluorocarbons, and/or other solubilising or dispersing agents known in the art.
Depending on the mode of administration, the pharmaceutical composition will preferably comprise from 0.05 to 99 %w (per cent by weight), more preferably from 0.05 to 80 %w, still more preferably from 0.10 to 70 %w, and even more preferably from 0.10 to 50 %w, of active ingredient, all percentages by weight being based on total composition. The compounds of the invention (that is, compounds of formula (I) and pharmaceutically acceptable salts thereof) may also be administered in conjunction with other compounds used for the treatment of the above conditions.
The invention therefore further relates to combination therapies wherein a compound of the invention or a pharmaceutical composition or formulation comprising a compound of the invention is administered with another therapeutic agent or agents for the treatment of one or more of the conditions previously indicated. Such therapeutic agents may be selected from the following:
(i) anti-addiction drugs including, for example, acamprosate, disulfiram, naltrexone and nalmefene for alcohol dependency, and gabapentin, modafinil, topiramate, vigabatrin and baclofen for drug, particularly cocaine, addiction;
(ii) antidepressants such as amitriptyline, amoxapine, bupropion, citalopram,
clomipramine, desipramine, doxepin duloxetine, elzasonan, escitalopram, fluvoxamine, fluoxetine, gepirone, imipramine, ipsapirone, maprotiline, nortriptyline, nefazodone, paroxetine, phenelzine, protriptyline, reboxetine, robaizotan, sertraline, sibutramine, tianeptine, thionisoxetine, tranylcypromaine, trazodone, trimipramine, venlafaxine, vortioxetine and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(iii) antipsychotics including, for example, amisulpride, aripiprazole, asenapine, benzisoxidil, bifeprunox, brexpiprazole, carbamazepine, cariprazine, clozapine, chlorpromazine, debenzapine, divalproex, duloxetine, eszopiclone, fluphenazine, haloperidol, iloperidone, lamotrigine, loxapine, lurasidone, mesoridazine, olanzapine, paliperidone, perlapine, perphenazine, phenothiazine, phenylbutlypiperidine, pimozide, prochlorperazine, quetiapine, risperidone, sertindole, sulpiride, suproclone, suriclone, thioridazine, trifluoperazine, trimetozine, valproate, valproic acid, zopiclone, zotepine, zicronapine, ziprasidone, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(iv) anxiolytics including, for example, alnespirone, azapirones, benzodiazepines, barbiturates, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof. Example anxiolytics include adinazolam, alprazolam, balezepam, bentazepam, bromazepam, brotizolam, buspirone, clonazepam, clorazepate, chlordiazepoxide, cyprazepam, diazepam, diphenhydramine, estazolam, fenobam, flunitrazepam, flurazepam, fosazepam, lorazepam, lormetazepam, meprobamate, midazolam, nitrazepam, oxazepam, prazepam, prazosin, quazepam, reclazepam, tracazolate, trepipam, temazepam, triazolam, uldazepam, and zolazepam; and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(v) anticonvulsants including, for example, carbamazepine, valproate, lamotrigine, levetiracetam and gabapentin, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(vi) Alzheimer's therapies including, for example, donepezil, galantamine, memantine, rivastigmine, tacrine, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(vii) Parkinson's therapies including, for example, L-dopa, ropinirole, pramipexole, monoamine oxidase type B (MAO-B) inhibitors such as deprenyl, selegiline and rasagiline, catechol-O-methyl transferase (COMT) inhibitors such as entacapone or tolcapone, adenosine A-2 inhibitors, dopamine re-uptake inhibitors, NMDA antagonists, Nicotine agonists, and Dopamine agonists and inhibitors of neuronal nitric oxide synthase, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(viii) migraine therapies including, for example, almotriptan, amantadine, botulinum toxin A, bromocriptine, butalbital, cabergoline, dichloralphenazone, dmydroergotarnine, eletriptan, frovatriptan, lisuride, naratriptan, pergolide, pramipexole, rizatriptan, ropinirole, sumatriptan, topiramate, zolmitriptan, and zomitriptan, and equivalents and
pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(ix) stroke therapies including, for example, abciximab, activase, citicoline,
desmoteplase, , and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof; (x) urinary incontinence therapies including, for example, darafenacin, duloxetine, falvoxate, mirabegron, oxybutynin, propiverine, robalzotan, solifenacin, and tolterodine, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(xi) neuropathic pain therapies including, for example, capsaicin, gabapentin, lidoderm, and pregabalin, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(xii) nociceptive pain therapies such as celecoxib, etoricoxib, lumiracoxib, rofecoxib, valdecoxib, diclofenac, loxoprofen, naproxen, and paracetamol, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(xiii) insomnia therapies including, for example, allobarbital, alonimid, amobarbital, benzoctamine, butabarbital, capuride, chloral, cloperidone, clorethate, dexclamol, ethchlorvynol, eszopiclone, etomidate, glutethimide, halazepam, hydroxyzine, lorediplon, mecloqualone, melatonin, mephobarbital, methaqualone, midaflur, nisobamate, pentobarbital, phenobarbital, propofol, ralmeteon, roletamide, suvorexant, triclofos, secobarbital, zaleplon, and Zolpidem, zopiclone and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(xiv) mood stabilizers including, for example, carbamazepine, divalproex, gabapentin, lamotrigine, lithium, olanzapine, quetiapine, valproate, valproic acid, and verapamil, and equivalents and pharmaceutically active isomer(s) and/or metabolite(s) thereof;
(xv) 5HT1B ligands such as, for example, compounds disclosed in WO 99/05134 and
WO 02/08212;
(xvi) mGluR2 agonists;
(xvii) alpha 7 nicotinic agonists such as, for example, compounds disclosed in
WO 96/006098, WO 97/030998, WO 99/003859, WO 00/042044, WO 01/029034, WO 01/60821, WO 01/36417, WO 02/096912, WO 03/087102, WO 03/087103, WO 03/087104, WO 2004/016617, WO 2004/016616, and WO 2004/019947;
(xviii) chemokine receptor CCRI inhibitors; and
(xix) delta opioid agonists such as, for example, compounds disclosed in WO 97/23466 and WO 02/094794.
Such combination products employ the compounds of this invention within the dosage range described herein and the other pharmaceutically active agent within approved dosage ranges.
The present invention will now be further explained by reference to the following illustrative examples, in which the starting materials and reagents used are available from commercial suppliers or prepared via literature procedures.
Nuclear magnetic resonance (NMR) spectra were recorded at 400 MHz or 300 MHz as stated and at 300.3K, 298.2K or 293K unless otherwise stated; the chemical shifts (δ) are reported in parts per million. Spectra were recorded using a Bruker (trade mark) 400 AVANCE instrument fitted with a 5mm BBFO probe with instrument controlled by Bruker TopSpin 2.1 software, or by a Bruker 400 AVANCE-III HD instrument fitted with a 5mm BBFO smart probe or a 5mm BBFO probe with instrument controlled by Bruker TopSpin 3.2 software, or by a Bruker 400 AVANCE-III instrument fitted with a 5mm BBFO probe with instrument controlled by Bruker Topspin 3.0 software or by a Bruker 300MHz AVANCE II instrument fitted with a 5mm DUL probe with instrument controlled by Bruker TopSpin 1.3 software, or 5mm BBFO probe controlled by Bruker Topspin 3.2 software.
Purity was assessed using one or more of the following:
• Ultra Performance Liquid Chromatography (UPLC) with UV (photodiode array) detection over a wide range of wavelengths, normally 220-450 nm, using a Waters (trade mark) Acquity UPLC system equipped with Acquity UPLC BEH, HSS or HSS T3 C18 columns (2.1mm id x 50mm long) operated at 50 or 60 °C. Mobile phases typically consisted of acetonitrile mixed with water containing either 0.1% formic acid, 0.1% TFA or 0.025% ammonia. Mass spectra were recorded with a Waters SQD single quadrupole mass spectrometer using atmospheric pressure ionisation.
• UPLC with UV (photodiode array) detection over a wide range of wavelengths, normally 220 - 450 nm, using Shimadzu (trade mark) Nexera X2 UPLC controlled by Lab Solution software equipped with Acquity UPLC BEH, HSS or HSS T3 CI 8 columns (2.1mm id x 50mm long) operated at 50 °C. Mobile phases typically consisted of acetonitrile mixed with water containing either 0.1% formic acid, 0.1% TFA or 0.025% ammonia. Mass spectra were recorded with a Shimadzu single quadrupole mass spectrometer using DUIS ionisation.
Compounds were purified using normal phase chromatography on silica, using Biotage (trade mark)n KP-Sil cartridges, Interchim (trade mark) PuriFlash cartridges or Kinesis (trade mark) Telos silica cartridges, or on basic silica using Biotage KP-NH cartridges, or by reverse phase chromatographic methods using Biotage KP-C18-HS cartridges or by Biotage Isolute SCX-2 or Phenomenex (trade mark) Strata ABW catch-release cartridges, or by preparative high performance liquid chromatography (UPLC).
Preparative HPLC was performed using Agilent Technologies (trade mark) 1100 Series system or a Waters autopurification LC/MS system typically using Waters 19 mm id x 250 mm long C18 columns such as XBridge (trade mark) or SunFire (trade mark) 5 μιη materials at room temperature. Mobile phases typically consisted of acetonitrile mixed with water containing either 0.1% formic acid or 0.1% ammonia, unless otherwise stated.
Super Critical Fluid Chromatography (SFC) chiral separations were performed on a Waters prep30/MS system, using a flow rate of 30 mL/min, temperature of 40 °C and a pressure of 100 bar. Mobile phases typically consisted of supercritical C02 and a polar solvent such as methanol, ethanol or isopropanol. Column type and eluent are detailed for individual examples.
'Room temperature', as used in the present specification, means a temperature in the range from about 18 °C to about 25 °C. Abbreviations
DAST: Diethylaminosulfur trifluoride
DCM: Dichloromethane
DIAD: Diisopropyl azodicarboxylate
DIBAL-H: Diisobutylaluminium hydride
DIPEA: N,N-Diisopropylethylamine
DMF: Dimethylformamide
DMP: Dess-Martin Periodinane
EtOAc: Ethyl acetate
h: hour
HATU: l-[Bis(dimemylamino)memylene]-lH-l,2,3-triazolo[4,5-b]pyridinium 3- oxid hexafluorophosphate
HPLC: High-Performance Liquid Chromatography
MeCN: Acetonitrile
MeOH: Methanol
min: minute
Pd2(dba)3 : Tris(dibenzylideneacetone)dipalladium(0)
TEA: Triethylamine
THF: Tetrahydrofuran
TFAA: Trifluoroacetic anhydride
T3P: Propylphosphonic Anhydride
1. Intermediates
Intermediate 1: 8-fluoro-2,3>4,5-tetrahydro-l ,5-benzoxazepine
Figure imgf000037_0001
Step (i): N-(7-fluoro-3,4-dihydro-2H-l-benzopyran-4-ylidene)hydroxylamine
7-Fluorochroman-4-one (10.0 g, 60.2 mmol) and hydroxylamine hydrochloride (4.60 g, 66.2 mmol) were dissolved in anhydrous methanol (100 mL) and cooled to 0 °C. TEA (10.1 mL, 72.2 mmol) was added drop wise. The reaction mixture was warmed to room temperature and refluxed for 2h. The reaction mixture was cooled to room temperature, then cooled to 0 °C in an ice-bath before quenching it with water (200 mL). The reaction mixture was stirred at 0 °C for 30 min. The solid was filtered, washed with water and dried in a vacuum oven at 50 °C overnight to afford the title compound.
1H NMR (400 MHz, CDCb) δ ppm 2.99 (t, J= 6.27 Hz, 2 H), 4.27 (t, J= 6.13 Hz, 2 H),
6.54 - 6.71 (m, 2 H), 7.75 - 7.88 (m, 1 H), 8.14 (s, 1 H)
MS ES+: 182
Step (ii): 8-fluoro-2,3J4,5-tetrahydro-l,5-benzoxazepine
N-(7-fluoro-3,4-dihydro-2H-l-benzopyran-4-ylidene)hydroxylamine (7.60 g, 41.9 mmol) was dissolved in DCM (450 mL) and the solution was cooled to 0 to -5 °C. DIBAL-H (1M solution in cyclohexane, 252 mL, 252 mmol) was added drop wise at 0 to -5 °C over 5 min and the reaction mixture was warmed to room temperature and stirred for 2h. The reaction mixture was cooled again to 0 °C, and sodium fluoride (50.40 g, 1.20 mol) was added followed by the careful addition of water (19 mL). The reaction mixture was stirred at 0 °C for 30 min. The mixture was filtered through a pad of celite and was washed with EtOAc. The filtrates were evaporated in vacuo. The crude product was purified by flash
chromatography (10-20% EtOAc in hexanes on silica) to afford the title compound.
1H NMR (400 MHz, CDC13) δ ppm 1.90 - 2.07 (m, 2 H), 3.05 - 3.23 (m, 2 H), 3.28 - 3.82 (m, 1 H), 3.98 - 4.14 (m, 2 H), 6.53 - 6.73 (m, 3 H)
MS ES+: 168
Intermediate 2: 9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepine
Figure imgf000038_0001
Prepared as described for Intermediate 1 using 8-Fluorochroman-4-one (9.50 g, 57.2 mmol) to afford the title compound.
¾ NMR (400 MHz, CDC13) δ ppm 1.96 - 2.10 (m, 2 H), 3.19 - 3.31 (m, 2 H), 3.84 (br 1 H), 4.09 - 4.20 (m, 2 H), 6.42 - 6.52 (m, 1 H), 6.54 - 6.65 (m, 1 H), 6.70 - 6.81 (m, 1 MS ES+: 168 Intermediate 3: (3S)-3-{[(teri-butoxy)carbonyl](methyl)amino}-3-phenyIpropanoic acid
Figure imgf000039_0001
Step (iii)
Figure imgf000039_0002
Step (i): methyl (3S)-3-{[(i'-?ri-butoxy)carbonyl]amino}-3-phenylpropanoate
Methyl iodide (2.95 mL, 47.1 mmol) was added to a suspension of (3S)-3-{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (2.5 g, 9.42 mmol) and K2CO3 (2.60 g, 18.9 mmol) in DMF (50 mL) at room temperature. The mixture was heated at 80 °C under nitrogen for 24h. The mixture was concentrated in vacuo to remove most of the DMF and the residue was partitioned between water and EtOAc. The organic phase was dried (MgS04) and concentrated in vacuo. The residue was purified by flash chromatography (20-30% [1 :1 EtOAcrTHF] in petroleum ether on silica) to afford the title compound. 1H NMR (400 MHz, OMSO-d6) δ ppm 1.36 (s, 9 H), 2.60 - 2.83 (m, 2 H), 3.56 (s, 3 H), 4.81 - 5.06 (m, 1 H), 7.16 - 7.26 (m, 1 H), 7.28 - 7.36 (m, 4 H), 7.42 - 7.53 (m, 1 H)
Step (ii): methyl (3S)-3-{[(teri-butoxy)carbonyl](methyl)amino}-3-phenylpropanoate
Sodium hydride (60% dispersion in mineral oil, 0.134 g, 3.35 mmol) was added to a solution of methyl (3S)-3-{[(tert-butoxy)carbonyl]amino}-3-phenylpropanoate (0.850 g, 3.04 mmol) in DMF (15 mL) and stirred for 30 min. The reaction was cooled to 0 °C and methyl iodide (0.228 mL, 3.65 mmol) was added and the reaction was stirred at room temperature for 2h. The reaction mixture was partitioned between EtOAc and water. The aqueous was further extracted with EtOAc and the combined organics then washed with brine. The organic phase was passed through a phase separator and concentrated in vacuo. The crude product was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica) to afford the title compound. lU NMR (400 MHz, DMSO-ifc) δ ppm 1.37 - 1.45 (m, 9 H), 2.55 (s, 3 H), 2.87 - 3.17 (m, 2 H), 3.60 (s, 3 H), 5.44 - 5.72 (m, 1 H), 7.26 - 7.33 (m, 3 H), 7.33 - 7.41 (m, 2 H)
Step (Hi): (3S)-3-{[(teri'-butoxy)carbonyl](methyI)amino}-3-phenylpropanoic acid
LiOH (453 mg, 18.9 mmol) was added to a stirred solution of methyl (3S)-3-{[(terr- butoxy)carbonyl](methyl)amino}-3-phenylpropanoate (0.555 g, 1.89 mmol) in dioxane (5 mL) and water (5 mL) and the mixture was stirred for 3h at room temperature. The reaction mixture was acidified with 2M HC1 to pH~5 and extracted with EtOAc. The organic phase was dried (MgS04) and concentrated in vacuo to afford the title compound. *H NMR (400 MHz, DMSO-ifc) δ ppm 1.32 - 1.48 (m, 9 H), 2.57 (s, 3 H), 2.74 - 2.89 (m, 1 H), 2.93 - 3.07 (m, 1 H), 5.43 - 5.68 (m, 1 H), 7.25 - 7.32 (m, 3 H), 7.33 - 7.41 (m, 2 H), 12.22 - 12.41 (m, 1 H)
Intermediate 4: 5-methanesulfonyl-3-methyl-2,354,5-tetrahydro-l,5-benzoxazepin-3-ol
Figure imgf000040_0001
Step (i): N-(2-hydroxyphenyl)methanesulfonamide
A solution of methanesulphonyl chloride (14.3 mL, 183 mmol) in DCM (20 mL) was added drop wise over ca. 5 min to a solution of 2-aminophenol (20 g, 183 mmol) and pyridine (44.5 mL, 550 mmol) in DCM (100 mL). The mixture was stirred at 0 °C for 5 min and allowed to warm to room temperature for lh. The mixture was concentrated in vacuo. The residue was partitioned between water and EtOAc, HC1 (2M) was added until the aqueous pH ~ 1. The organic phase was separated, washed with water and brine, dried (MgS04) and concentrated in vacuo. The residue was triturated with ether, filtered and dried to afford the title compound.
1H NMR (300 MHz, DMSO-c¾) 5 ppm 2.94 (s, 3 H), 6.73 - 6.82 (m, 1 H), 6.85 - 6.91 (m, 1 H), 6.99 - 7.09 (m, 1 H), 7.15 - 7.22 (m, 1 H), 8.66 (br. s, 1 H), 9.74 (br. s, 1 H)
Step (ii): 5-(MethyIsulfonyl)-2,3,4,5-tetrahydrobenzo[b] [l,4]oxazepin-3-ol
N-(2-Hydroxyphenyl)methanesulfonamide (85.00 g, 0.46 mol) was dissolved in ethanol (275 mL). A solution of sodium ethoxide (21% solution in ethanol) (173 mL, 0.47 mol) in ethanol (275 mL) was added. More ethanol (200 mL) was added and the reaction was stirred under nitrogen for 30 min. The solvent was concentrated in vacuo, and azeotroped with THF (200 mL). The residue was dissolved in DMF (680 mL). (±)-Epichlorohydrin (35.5 mL, 0.46 mol) was added and the mixture was heated to 90 °C for 7h and stirred at room temperature overnight. Half the amount of DMF was concentrated in vacuo and the remaining solution was partitioned between EtOAc and water. The organic layer was separated and the aqueous layer was further extracted with EtOAc. The combined organics were washed with brine and water. The organic layer was dried (Na2S04), filtered through a pad of celite and the filtrate was concentrated in vacuo. The resulting residue was triturated in ethanol (170 mL) for 15 min. The solid was collected upon filtration, washed with cold ethanol and dried in the vacuum oven at 40 °C to afford the title compound. 1H NMR (400 MHz, DMSO-tfc) δ ppm 3.11 (s, 3 H), 3.43 - 3.57 (m, 1 H), 3.57 - 3.87 (m, 2 H), 3.93 - 4.10 (m, 2 H), 5.45 - 5.53 (m, 1 H), 7.08 - 7.16 (m, 2 H), 7.24 - 7.34 (m, 1 H), 7.36 - 7.43 (m, 1 H)
MS ES+: 244
Step (iii): 5-methanesulfonyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-3-one
DMP (17.4 g, 41.0 mmol) was added to a suspension of 5-(methylsulfonyl)-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-ol (5 g, 20.6 mmol) in DCM (200 mL) under nitrogen. The reaction was stirred at room temperature for 18h. The reaction was quenched with saturated aqueous sodium thiosulfate, with vigorous stirring for 15 min. The phases were separated and the organic was washed 3 times with saturated aqueous sodium bicarbonate then brine, dried (phase separator) and concentrated in vacuo to afford the title product ¾ NMR (400 MHz, DMSO-i¾) δ ppm 3.39 (s, 3 H), 4.52 (s, 2 H), 4.71 (s, 2 H), 7.07 - 7.15 (m,'2 H), 7.24 - 7.34 (m, 1 H), 7.41 - 7.51 (m, 1 H) MS ES+: 242
Step (iv): 5-methanesulfonyl-3-methyl-2,3j4,5-tetrahydro-l,5-benzoxazepin-3- olMethylmagnesium bromide (3M in diethyl ether) (8.29 mL, 24.87 mmol) was added to suspension of 5-methanesulfonyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-3-one (2 g, 8.29 mmol) in THF (40 mL) at 0 °C under nitrogen. The reaction was stirred at 0 °C for 3h, then allowed to warm to room temperature and stirred for 18h. The reaction was quenched by cautious addition of saturated aqueous NH4C1 solution, extracted 3 times with EtOAc, dried (Na2S04) and concentrated in vacuo. The crude product was purified by flash chromatography (0-75% EtOAc in petroleum ether on silica). The material was loaded onto a cation exchange cartridge, washing with methanol and eluting with 2M ammonia in methanol solution and concentrated in vacuo to afford the title compound.
MS ES+: 258
Intermediate 5: 6-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepine
Figure imgf000042_0001
Prepared as described for Intermediate 1 using 5-Fluorochroman-4-one (2.00 g, 12.0 mmol) to afford the title compound.
lH NMR (400 MHz, CDC ) δ ppm 1.96 - 2.11 (m, 2 H), 3.21 - 3.36 (m, 2 H), 4.02 - 4.20 (m, 3 H), 6.57 - 6.80 (m, 3 H)
Intermediate 6: 3-{[(/ert-butoxy)carbonyl]amino}-2,2-difluoro-3-phenylpropanoic acid
Figure imgf000043_0001
Step (iii)
Figure imgf000043_0002
Step (vi)
Figure imgf000043_0003
Step (i): ethyl 2,2-difluoro-3-hydroxy-3-phenyIpropanoate
Zinc (8.87 g, 136 mmol) was added to a refluxing solution of THF (250 mL) under nitrogen. Ethyl 2-bromo-2,2-difluoroacetate (18.00 mL, 136 mmol) and benzaldehyde (11.49 mL, 113 mmol) were added sequentially. The reaction was heated to reflux for 15 min then cooled to room temperature. The reaction was poured into a mixture of EtOAc, aqueous sodium hydrogen sulfate and aqueous sodium bicarbonate. The mixture was stirred for 15 min. The layers were separated and the aqueous was extracted with more EtOAc. The organics were combined, dried (MgS04) and concentrated in vacuo. The crude product was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica) to afford the title compound.
1H NMR (400 MHz, CDC13) δ ppm 1.31 (t, J = 7.15 Hz, 3 H), 2.65 - 2.73 (m, 1 H), 4.33 (q, J= 7.15 Hz, 2 H), 5.20 (s, 1 H), 7.37 - 7.52 (m, 5 H)
Step (ii): ethyl 2,2-difluoro-3-(methanesulfonyloxy)-3-phenylpropanoate
A solution of methanesulfonyl chloride (4.86 mL, 62.3 mmol) was added drop wise to a solution of ethyl 2,2-difluoro-3-hydroxy-3-phenylpropanoate (12.0 g, 48.0 mmol) and TEA (8.69 mL, 62.3 mmol) in DCM (200 mL) under nitrogen. The reaction was stirred at room temperature for 2 hours. The mixture was washed with 10% HC1, water, sodium bicarbonate and brine, dried (MgS04) and concentrated in vacuo to afford the title compound.
¾ NMR (400 MHz, CDC13) δ ppm 1.33 (t, J= 7.20 Hz, 3 H), 2.91 (s, 3 H), 4.35 (q, J= 7.15 Hz, 2 H), 5.92 - 6.03 (m, 1 H), 7.44 - 7.58 (m, 5 H)
Step (iii): ethyl 3-azido-2,2-difluoro-3-phenylpropanoate
Sodium azide (5.53 g, 85 mmol) was added to a solution of ethyl 2,2-difluoro-3- (methylsulfonyloxy)-3-phenylpropanoate (13.8 g, 42.5 mmol) and tetrabutylammonium bromide (0.137 g, 0.43 mmol) in DMF (130 mL) under nitrogen. The reaction was heated to 65 °C for 48h. The reaction was cooled to room temperature. Water was added to quench the reaction. The mixture was diluted with EtOAc, washed three times with water, dried (MgS04) and concentrated in vacuo. The crude product was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica). The crude product was further purified by flash chromatography (0-50% EtOAc in petroleum ether on silica) to afford the title compound.
¾ NMR (400 MHz, CDC13) δ ppm 1.31 (t, J= 7.15 Hz, 3 H), 4.33 (q, J= 7.15 Hz, 2 H), 5.01 - 5.16 (m, 1 H), 7.38 - 7.49 (m, 5 H)
Step (iv): ethyl 3-amino-2,2-difluoro-3-phenyIpropanoate
10% Palladium on carbon (50% water) (1.06 g, 0.99 mmol) was added to a solution of ethyl 3-azido-2,2-difluoro-3-phenylpropanoate (2.53 g, 9.91 mmol) in EtOAc (45 mL) and the reaction was stirred under an atmosphere of hydrogen gas. The reaction was stirred at room temperature for 18h. The reaction was filtered and the filtrate concentrated in vacuo. The residue was dissolved in EtOAc (45 mL) and 10% palladium on carbon (50% water) (1.06 g, 0.99 mmol) was added. The reaction was placed under vacuum then an atmosphere of hydrogen and stirred vigorously for 18h. The reaction was filtered through celite and washed with EtOAc and methanol. The filtrate was concentrated in vacuo to afford the title compound.
1H NMR (400 MHz, CDCI3) δ ppm 1.25 (t, J- 7.15 Hz, 3 H), 1.81 (br. s., 2 H), 4.26 (q, J = 7.15 Hz, 2 H), 4.42 - 4.57 (m, 1 H), 7.32 - 7.43 (m, 5 H)
MS ES+: 230
Step (v): ethyl 3-{[(tert-butoxy)carbonyl]amino}-2,2-difluoro-3-pheiiylpropanoate Di-tert-butyl dicarbonate (4.56 mL, 19.6 mmol) was added to a solution of ethyl 3-amino- 2,2-difluoro-3-phenylpropanoate (2.25 g, 9.82 mmol) and sodium hydrogen carbonate (1.649 g, 19.6 mmol) in THF (10 mL) under nitrogen. The reaction was heated to 60 °C for 18h. Sodium hydrogen carbonate (0.5g) was added, followed by di-tert-butyl dicarbonate (1.35g) and the reaction was stirred at 60 °C for 30 min. The reaction was cooled to room temperature. The mixture was diluted with EtOAc, washed with water, 10% HCl, saturated sodium bicarbonate solution, and brine, dried (MgS04) and concentrated in vacuo. The crude product was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica) to afford the title compound.
'H NMR (300 MHz, DMSO-</6) δ ppm 1.21 (t, J= 7.12 Hz, 3 H), 1.38 (s, 9 H), 4.14 - 4.36 (m, 2 H), 5.17 - 5.41 (m, 1 H), 7.28 - 7.55 (m, 5 H), 8.16 - 8.30 (m, 1 H)
Step (vi): 3-{[( eri'-butoxy)carbonyl]amino}-2,2-difluoro-3-phenyIpropanoic acid
LiOH (0.480 g, 20.0 mmol) was added to a suspension of ethyl 3-{[(tert- butoxy)carbonyl]amino}-2,2-difluoro-3-phenylpropanoate (2.2 g, 6.68 mmol) in EtOH (10 mL) and water (5 mL). The reaction was stirred at room temperature for 30 min. The reaction was concentrated in vacuo and acidified with 1M HCl to pHl. The aqueous was extracted twice with EtOAc. The organics were combined, dried (MgS0 ) and
concentrated in vacuo to afford the title compound.
1H NMR (400 MHz, DMSO-^) δ ppm 1.38 (s, 9 H), 5.13 - 5.31 (m, 1 H), 7.27 - 7.50 (m, 5 H), 8.01 - 8.16 (m, 1 H)
MS ES": 300
Intermediate -methoxy-2,3j4,5-tetrahydro-l,5-beiizoxazepine
Figure imgf000045_0001
Step (i): 3-methoxy-5-(methylsulfonyl)-2,3,4,5-tetrahydrobenzo[b] [l,4]oxazepine
Methyl iodide (3.20 mL, 51.4 mmol) was added to a suspension of 5-(methylsulfonyl)- 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-ol (Intermediate 4 step (ii), 5 g, 20.6 mmol) and silver oxide (23.81 g, 103 mmol) in MeCN (40 mL) and DMF (15 mL) in a sealed tube. The reaction was covered in foil and stirred at room temperature overnight. The suspension was filtered through celite and the filtrate was concentrated in vacuo to afford the title compound.
MS ES+: 258
Step (ii): 3-methoxy-2,3,4,5-tetrahydro-l,5-benzoxazepine
Bis(2-methoxyethoxy)aluminum(III) sodium hydride (60% in toluene, 1.848 mL, 5.83 mmol) was added to a solution of 3-methoxy-5-(methylsulfonyl)-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.3 g, 1.17 mmol) in toluene (3 mL) at room
temperature and stirred for 18h. The reaction was quenched with sodium carbonate. The crude mixture was dissolved in methanol and loaded onto an SCX cartridge. The cartridge was washed with methanol and DCM, then eluted with 4M ammonia in methanol. The ammonia in methanol fractions were combined and concentrated in vacuo to afford the title compound.
Ή NMR (400 MHz, DMSO-cfc) δ ppm 3.13 - 3.23 (m, 1 H), 3.33 (s, 3 H), 3.35 - 3.44 (m, 1 H), 3.57 - 3.67 (m, 1 H), 4.02 - 4.11 (m, 1 H), 4.16 - 4.23 (m, 1 H), 5.36 - 5.43 (m, 1 H), 6.51 - 6.58 (m, 1 H), 6.64 - 6.76 (m, 3 H)
Intermediates 8 and 9: tert-butyl N-[(lS)-3-(3-hydroxy-3-methyl-2,3»4,5-tetrahydro- 1 ,5-benzoxazep -5-y l)-3-oxo-l -pheny lpropy 1] - -methy lcarbamate
Figure imgf000046_0001
HATU (0.120 g, 0.32 mmol) was added to a solution of (S)-3-((tert- butoxycarbonyl)(methyl)amino)-3-phenylpropanoic acid (Intermediate 3, 0.084 g, 0.30 mmol) and DEPEA (0.058 mL, 0.33 mmol) in DMF (1 mL) at room temperature. The mixture was stirred for 5 min. 5-methanesulfonyl-3-methyl-2,3,4,5-tetrahydro-l,5- benzoxazepin-3-ol (Intermediate 4, 0.054 g, 0.30 mmol) was added and the mixture was stirred until homogeneous and allowed to stand for 18h. The mixture was partitioned between water and EtOAc. The organic phase was concentrated in vacuo. The residue was purified by flash chromatography (20-50% EtOAc in petroleum ether on silica) to afford the two title compounds. Intermediate 8: tert-butyl N-[(lS)-3-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)-3-oxo-l-phenyIpropyI]-N-methylcarbamate
Fastest running stereoisomer
MS ES+: 441
Retention time: 1.52 min
Intermediate 9: tert-butyl N-[(lS)-3-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yI)-3-oxo-l-phenylpropyI]-N-methylcarbamate
Slowest running stereoisomer
MS ES+: 441
Retention time: 1.53 min
Intermediate 10: (S)-i'eri-butyl (3-oxo-3-(3-oxo-3,4-dihydrobenzo[b] [l,4]oxazepin- 5(2H -yl)-l-phenylpropyI)carbamate (560 mg, 1.364 mmol)
Figure imgf000047_0001
Step (i): 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-ol
Bis(2-methoxyethoxy)aluminum(III) sodium hydride (1.954 mL, 6.17 mmol) was added to a suspension of 5-(memylsulfonyl)-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-ol
(Intermediate 4 step (ii), 0.5 g, 2.055 mmol) in toluene (10 mL) under nitrogen. The reaction was heated to 85 °C for 24h. The reaction was cooled to room temperature and quenched with sodium carbonate solution. The toluene was removed in vacuo and the mixture was partitioned between EtOAc and sodium carbonate solution. The phases were separated and the aqueous was extracted three times with EtOAc. The combined organics were dried (MgS04) and concentrated in vacuo to afford the title compound.
MS ES+: 166 1H NMR (300 MHz, DMSO-ifc) δ ppm 2.95 - 3.08 (m, 1 H), 3.24 - 3.30 (m, 1 H), 3.74 - 3.94 (m, 2 H), 4.12 - 4.23 (m, 1 H), 4.91 - 5.01 (m, 1 H), 5.33 (br. s., 1 H), 6.51 - 6.61 (m, 1 H), 6.64 - 6.79 (m, 3 H)
Step (ii): tert-butyl ((lS)-3-(3-hydroxy-3,4-dihydrobenzo[b][l,4]oxazepin-5(2H)-yl)-3- oxo-l-phenylpropyl)carbamate
2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-ol (1.557 g, 9.42 mmol) was added to a solution of (3S)-3-{[(tert-butoxy)carbonyl]amino}-3-phenylpropanoic (2.5 g, 9.42 mmol) and HATU (3.94 g, 10.37 mmol) and pyridine (0.838 mL, 10.37 mmol) in DMF (10 mL) under nitrogen. The reaction was stirred at room temperature for 24h. HATU (3.94 g, 10.37 mmol) was added and the reaction was stirred at room temperature for 72h. The mixture was diluted with EtOAc, washed with saturated NaHCCb, water, and brine, dried (MgS04) and concentrated in vacuo. The crude product was purified by flash chromatography (0- 100% EtOAc in petroleum ether on silica) to afford the title compound.
MS ES+: 413
Intermediate 11; ier/-butyl N-[(lS)-3-oxo-3-(3-oxo-2,3,4,5-tetrahydro-l,5- benzoxazepi -5-yl)-l-phenylpropyl]carbamate
Figure imgf000048_0001
DMP (1.16 g, 2.74 mmol) was added to a solution of tert-butyl ((lS)-3-(3-hydroxy-3,4- dihydrobenzo [b] [ 1 ,4] oxazepin-5 (2H)-yl)-3 -oxo- 1 -pheny lpropyl)carbamate (Intermediate 10, 0.565 g, 1.370 mmol) in DCM (5 mL) under nitrogen. The reaction was stirred at room temperature for 18h. The reaction was diluted with DCM and Na2S2C<3 was added. The suspension was stirred at room temperature for 15 min. The aqueous phase was extracted with DCM, followed by EtOAc. The organics were combined, dried (MgS04) and concentrated in vacuo to afford the title compound.
MS ES": 409
Intermediate 12: 7-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepine
Figure imgf000049_0001
Prepared as described for Intermediate 1 using 6-fluoro-3,4-dihydro-2H-l-benzopyran-4- one (lg, 5.99 mmol), to afford the title compound.
1H NMR (400 MHz, CDCb) δ ppm 1.94 - 2.20 (m, 2 H), 3.20 - 3.24 (m, 2H), 3.65 - 3.78 (br. s, 1 H), 4.01 - 4.06 (m, 2 H), 6.38 - 6.45 (m, 2 H), 6.83 - 6.87 (m, 1 H)
MS ES+: 168
Intermediates 13 and 14: fer/-butyl N-[(lS)-3-(3-fluoro-2,3.4,5-tetrahydro-l,5- benzoxazepin-5-yl)-3-oxo-l-phenylpropyI] carbamate
Figure imgf000049_0002
DAST (0.801 mL, 6.07 mmol) was added to a solution of tert-butyl ((lS)-3-(3-hydroxy- 3 ,4-dihydrobenzo [b] [ 1 ,4]oxazepin-5 (2H)-yl)-3 -oxo- 1 -phenylpropyl)carbamate
(Intermediate 10, 0.695 g, 1.213 mmol) in DCM (5 mL) under nitrogen at 0 °C. The reaction was stirred at room temperature for 18h. The reaction was cooled to 0 °C and DAST (0.801 mL, 6.07 mmol) was added. The reaction was left to stir at room temperature for 2h. The reaction was quenched with water and diluted with DCM. The organic phase was collected, washed with water, dried (MgS04) and concentrated in vacuo. The crude product was purified by flash chromatography (0-100% EtOAc in petroleum ether on silica). The resulting residue was purified by chiral SFC (ChiralPak Lux-C4, 20% IP A) to afford the two title compounds.
Intermediate 13: tert-b tyl N-[(lS)-3-(3-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)-3-oxo-l-pheny!propyl]carbamate
Fastest running isomer
MS ES+: 415
Retention time = 1.49 min Intermediate 14: teri-butyl N-[(lS)-3-(3-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)-3-oxo-l-phenylpropyl]carbamate
Slowest running isomer
MS ES+: 415
Retention time = 1.47 min -tetrahydro-2,4-methanobenzo[b] [l,4]oxazepine
Figure imgf000050_0001
Step (i): fer/-butyl N-[3-(2-bromophenoxy)cyclobutyl]carbamate
DIAD (1.090 mL, 5.61 mmol) was added to a solution of tert-butyl N-(3- hydroxycyclobutyl)carbamate (1 g, 5.34 mmol), 2-bromophenol (0.924 g, 5.34 mmol) and triphenylphosphine (1.401 g, 5.34 mmol) in THF (15 mL) at 0°C. The mixture was stirred, allowed to warm to RT and then to stand for 2h. The mixture was partitioned between DCM and NaOH (1 M). The aqueous phase was extracted with DCM. The combined organic phases were dried and concentrated in vacuo. The residue was purified by flash chromatography (20-30% EtOAc in petroleum ether on silica)to afford the title compound. 1H NMR (400 MHz, DMSO-ifc) δ ppm 1.30 - 1.45 (m, 9 H), 1.94 - 2.08 (m, 2 H), 2.74 - 2.85 (m, 2 H), 3.65 - 3.78 (m, 1 H), 4.37 - 4.50 (m, 1 H), 6.82 - 6.92 (m, 1 H), 6.93 - 6.99 (m, 1 H), 7.20 - 7.27 (m, 1 H). 7.28 - 7.35 Cm. 1 H). 7.53 - 7.62 (m. 1 H)
MS ES+:
Step (ii): tert-butyl 3,4-dihydro-2,4-methanobenzo[b][l,4]oxazepine-5(2H)- carboxylate
A solution/suspension of tert-butyl (3-(2-bromophenoxy)cyclobutyl)carbamate (0.660 g, 1.929 mmol) and bis(3,5-bis(trifluoromethyl)phenyl)(2',4',6'-triisopropyl-3,6-dimethoxy- [l,r-biphenyl]-2-yl)phosphine (0.310 g, 0.386 mmol) in toluene (15 mL) was degassed and a nitrogen atmosphere introduced three times. Soidum tert-butoxide (0.371 g, 3.86 mmol) and Pd2(dba)3 (0.177 g, 0.193 mmol) were added. The mixture was degassed and a nitrogen atmosphere introduced three times. The mixture was heated at 60 °C for 24h. The mixture was partitioned between water and EtOAc. The organic phase was dried (MgS04) and concentrated in vacuo. The residue was purified by flash chromatography (15-30% EtOAc in petroleum ether) to afford the title compound.
1H NMR (400 MHz, DMSO-cfc) δ ppm 1.41 (s, 9 H), 1.72 - 1.82 (m, 2 H), 2.59 - 2.70 (m, 2 H), 4.59 - 4.67 (m, 1 H), 4.82 - 4.89 (m, 1 H), 6.95 - 7.03 (m, 2 H), 7.08 - 7.16 (m, 1 H), 7.29 - 7.36 (m, 1 H)
MS ES+: 206 (M-lBu)
Step (iii): 2,3,4,5-tetrahydro-2,4-methanobenzo[b][l,4]oxazepine
HC1 (4 M in dioxane) (0.892 mL, 3.57 mmol) was added to a solution of tert-butyl 3,4- dihydro-2,4-methanobenzo[b][l,4]oxazepine-5(2H)-carboxylate (0.233 g, 0.892 mmol) in DCM (3 mL). The mixture was stirred and allowed to stand for 2.5h The mixture was partitioned between DCM and NaOH (1 M). The aqueous phase was extracted with DCM. The combined organic phases were dried (MgS04) and concentrated in vacuo to afford the title compound.
MS ES+: 162
Intermediate 16: tert-butyl N-[(lS)-2-{[(2S)-4-(2-bromo-6-fluorophenoxy)butan-2- yl] carbamoyl}-l-phenylethyI] carbamate
Figure imgf000051_0001
Step (i): feri-butyl N-[(2S)-4-hydroxybutan-2-yl]carbamate
Di-tert-butyl dicarbonate (2.60 mL, 11.22 mmol) was added to a solution of (S)-3- aminobutan-l-ol (1.00 g, 11.2 mmol) in MeOH (10 mL) at room temperature. The mixture was stirred at room temperature and allowed to stand. The mixture was partitioned between water and DCM. The aqueous phase was extracted with DCM. The combined organic phases were dried (MgS04) and concentrated in vacuo. The residue was dissolved in DCM (10 mL), dried (MgS04) and concentrated in vacuo to afford the title compound.
!H NMR (400 MHz, CD3CN) 5 ppm 1.11 (d, J= 6.69 Hz, 3 H), 1.41 (s, 9 H), 1.44 - 1.51 (m, 1 H), 1.55 - 1.69 (m, 1 H), 2.85 - 3.05 (m, 1 H), 3.43 - 3.58 (m, 2 H), 3.61 - 3.80 (m, 1 H), 5.13 - 5.34 (m, 1 H)
Step (ii): tert-butyl N-[(2S)-4-(2-bromo-6-fluorophenoxy)butan-2-yl]carbamate
DIAD (2.29 mL, 11.76 mmol) was added to a solution of (S)-tert-butyl (4-hydroxybutan-2- yl)carbamate (2.12 g, 11.2 mmol), 2-bromo-6-fluorophenol (2.14 g, 11.20 mmol) and triphenylphosphine (2.94 g, 11.20 mmol) in THF (20 mL) at room temperature. The mixture was stirred at room temperature and allowed to stand for 18h. The mixture was partitioned between DCM and NaOH (1M). The aqueous phase was extracted with DCM. The combined organic phases were dried (MgS04) and concentrated in vacuo. The residue was purified by flash chromatography (20% EtOAc in petroleum ether) to afford the title compound.
1H NMR (400 MHz, DMSO-ck) δ ppm 1.08 (d, J= 6.60 Hz, 3 H), 1.37 (s, 9 H), 1.74 - 1.89 (m, 2 H), 3.64 - 3.82 (m, 1 H), 3.95 - 4.13 (m, 2 H), 6.69 - 6.81 (m, 1 H), 7.03 - 7.15 (m, 1 H), 7.26 - 7.37 (m, 1 H), 7.42 - 7.49 (m, 1 H)
MS ES+: 306 (M-tBu)
Step (iii): (2S)-4-(2-bromo-6-fluorophenoxy)butan-2-amine hydrochloride
HC1 (4 M in dioxane) (3.81 mL, 15.24 mmol) was added to a solution of tert-butyl N- [(2S)-4-(2-bromo-6-fluorophenoxy)butan-2-yl]carbamate (3.68 g, 10.16 mmol) in DCM (15 mL) at room temperature. The mixture was stirred at room temperature and allowed to stand for 18h. Further HC1 (4 M in dioxane) (8.89 mL, 35.6 mmol) was added and the mixture was diluted with MeOH (15 mL), stirred and allowed to stand for 18h. The mixture was concentrated in vacuo, adding DCM to assist removal of dioxane, to afford the title compound.
1H NMR (400 MHz, OMSO-de) 6 ppm 1.29 (d, J= 6.60 Hz, 3 H), 1.77 - 1.96 (m, 1 H), 2.09 - 2.25 (m, 1 H), 3.28 - 3.55 (m, 2 H), 4.06 - 4.24 (m, 2 H), 7.05 - 7.17 (m, 1 H), 7.31 - 7.41 (m, 1 H), 7.44 - 7.53 (m, 1 H), 8.09 (br. s, 3 H)
MS ES+: 262 Step (iv): ter/-butyl N-[(lS)-2-{[(2S)-4-(2-bromo-6-fluorophenoxy)butan-2- y 1] carbamoyl}- 1 -pheny lethyl] carbamate
HATU (0.800 g, 2.100 mmol) was added to a stirred solution of (3S)-3-{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (0.53 g, 2 mmol) and DIPEA (0.770 mL, 4.40 mmol) in DMF (8 mL) at room temperature. The mixture was stirred at room temperature for 5 minutes. (2S)-4-(2-bromo-6-fluorophenoxy)butan-2-amine hydrochloride (0.60 g, 2.000 mmol) was added and the mixture was stirred until homogeneous and allowed to stand for 2h. The mixture was partitioned between water and EtOAc. THF and water were added. The aqueous phase was extracted with EtOAc. The combined organic phases were dried (MgS04) and concentrated in vacuo to ca. 20 mL. The resulting suspension was filtered, washed with ether and air-dried to afford the title compound.
Ή NMR (400 MHz, DMSO-i e) δ ppm 1.03 (d, J= 6.60 Hz, 3 H), 1.34 (s, 9 H), 1.67 - 1.82 (m, 2 H), 2.39 - 2.47 (m, 2 H), 3.65 - 4.02 (m, 4 H), 4.82 - 4.94 (m, 1 H), 7.03 - 7.13 (m, 2 H), 7.17 - 7.28 (m, 4 H), 7.29 - 7.42 (m, 2 H), 7.43 - 7.51 (m, 1 H), 7.61 - 7.71 (m, 1 H) MS ES+: 509
Intermediate 17: 9-methyl-2 54,5-tetrahydro-l,5-benzoxazepine
Figure imgf000053_0001
Prepared as described for Intermediate 1 using 8-methyl-3,4-dihydro-2H-l-benzopyran-4- one (lg, 6.13 mmol), to afford the title compound.
1H NMR (400 MHz, CDC13) δ ppm 1.95 - 2.08 (m, 2 H), 2.24 (s, 3 H), 3.17 - 3.27 (m, 2 H), 3.70 (br. s, 1 H), 4.04 - 4.14 (m, 2 H), 6.57 - 6.62 (m, 1 H), 6.67 - 6.73 (m, 1 H), 6.74 - 6.82 (m, 1 H)
MS ES+: 164
Intermediate 18: 9-chloro-2,3,4,5-tetrahydro-l,5-benzoxazepine
Figure imgf000053_0002
Prepared as described for Intermediate 1 using 8-chloro-3,4-dihydro-2H-l-benzopyran-4- one (lg, 5.45 mmol), to afford the title compound.
1H NMPv (400 MHz, CDC13) δ ppm 1.97 - 2.11 (m, 2 H), 3.22 - 3.35 (m, 2 H), 3.79 (br. s., 1 H), 4.14 - 4.23 (m, 2 H), 6.55 - 6.64 (m, 1 H), 6.72 - 6.82 (m, 1 H), 6.83 - 6.91 (m, 1 H) MS ES+: 184
Intermediate 19: tert-butyl N-[(lS)-2-{[(2R)-4-(2-bromo-6-fluorophenoxy)butan-2- yl] carbamoyl}-l-phenylethyl] carbamate
Figure imgf000054_0001
Prepared as described for Intermediate 15 using (R)-3-aminobutan-l-ol (1 g, 11.22 mmol) and 2-bromo-6-fluorophenol (2.12g, 11.1 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-ik) δ ppm 0.90 - 1.00 (m, 3 H), 1.23 - 1.40 (m, 9 H), 1.67 -
1.89 (m, 2 H), 2.39 - 2.47 (m, 2 H), 3.86 - 4.06 (m, 3 H), 4.81 - 4.96 (m, 1 H), 7.04 - 7.19
(m, 2 H), 7.21 - 7.42 (m, 6 H), 7.42 - 7.49 (m, 1 H), 7.61 - 7.71 (m, 1 H)
MS ES+: 509
Intermediate 20: 4,5-dihydro-2H-spiro[l,5-benzoxazepine-3,l'-cycIopropane]
Figure imgf000054_0002
A solution of tert-butyl N-{[l-(hydroxymethyl)cyclopropyl]methyl}carbamate (1.00 g, 4.97 mmol), 2-bromophenol (0.537 mL, 4.97 mmol) and triphenylphosphine (1.30 g, 4.97 mmol) in THF (30 mL) was treated drop wise with DIAD (1.02 mL, 4.97 mmol). The reaction was stirred at room temperature for 18h under nitrogen. The reaction was partitioned between EtOAc and water and the organic phase was collected, dried (MgS04) and concentrated in vacuo. The crude material was purified by flash chromatography (0- 50% EtOAc in petroleum ether on silica) to afford the title compound.
Ή NMR (400 MHz, DMSO-Jd) δ ppm 0.47 - 0.60 (m, 4 H), 1.36 (s, 9 H), 3.09 - 3.17 (m, 2 H), 3.90 (s, 2 H), 6.81 - 6.93 (m, 2 H), 7.00 - 7.08 (m, 1 H), 7.27 - 7.38 (m, 1 H), 7.52 - 5 7.61 (m, 1 H)
MS Ε8+: 318 (Μ-Τ ι)
Step (ii) tert-butyl 4,5-dihydro-2H-spiro[l,5-benzoxazepine-3,l'-cyclopropane]-5- carboxylate
l o A suspension of tert-butyl N-( { 1 - [(2-bromophenoxy)methyl] cyclopropyl } methyl)- carbamate (0.30 g, 0.84 mmol), sodium tert-butoxide (0.162 g, 1.684 mmol), bis(3,5- , bis(trifluoromethyl)phenyl)(2',4',6,-triisopropyl-3 ,6-dimethoxy- [1,1 '-biphenyl] -2- yl)phosphine and Pd2(dba)3 (0.077 g, 0.084 mmol) in toluene (10 mL) was degassed and refilled with nitrogen gas three times. The reaction was heated to 80°C for 2.5h. The is reaction was cooled to room temperature and diluted with EtOAc, washed with water, followed by brine, dried (MgS04) and concentrated in vacuo to afford the title compound. MS ES+: 176 (M-Boc)
Step (iii) 4,5-dihydro-2H-spiro[l,5-benzoxazepine-3,l'-cyclopropane]
0 HC1 (4M in dioxane, 1.68 mL, 6.71 mmol) was added to a solution of tert-butyl 4,5- dihydro-2H-spiro[l,5-benzoxazepine-3,r-cyclopropane]-5-carboxylate (0.40 g, 1.119 mmol) in dioxane (5 mL) under nitrogen. The reaction was stirred at room temperature for 18h. HC1 (4M in dioxane, 1.68 ml, 6.71 mmol) was added and the reaction was heated to 80 °C for 2h. The reaction was concentrated in vacuo, dissolved in EtOAc, washed with5 bicarbonate solution, followed by water and brine, dried (MgS04) and concentrated in vacuo. The crude product was purified by flash chromatography (0-100% EtOAc in petroleum ether on silica) to afford the title compound.
1H NMR (400 MHz, DMSO- d) δ ppm 0.52 (s, 4 H), 2.84 - 2.95 (m, 2 H), 3.74 (s, 2 H), 5.34 (br. s, 1 H), 6.57 - 6.68 (m, 1 H), 6.74 - 6.84 (m, 3 H)
0 MS ES+: 176
Intermediate 21: 9-methoxy-2.3.4,5-tetrahvdro-l,5-benzoxazepine
Figure imgf000056_0001
Prepared as described for Intermediate 1 using 8-methoxy-3,4-dihydro-2H-l-benzopyran- 4-one (lg, 5.61 mmol), to afford the title compound.
1H NMR (400 MHz, CDC13) δ ppm 1.97 - 2.07 (m, 2 H), 3.21 - 3.30 (m, 2 H), 3.65 - 3.81 (m, 1 H), 3.84 (s, 3 H), 4.11 - 4.22 (m, 2 H), 6.32 - 6.39 (m, 1 H), 6.42 - 6.48 (m, 1 H), 6.76 - 6.83 (m, 1 H)
MS ES+: 180
Intermediate 22: tert-butyl N-[(lS)-l-(2-bromophenyl)-3-oxo-3-(2,3>4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propyI] carbamate
Figure imgf000056_0002
HATU (798 mg, 2.100 mmol) was added to a solution of (3S)-3-(2-bromophenyl)-3- {[(tert-butoxy)carbonyl]amino}propanoic acid (0.688 g, 2.00 mmol) and DIPEA (0.384 mL, 2.200 mmol) in DMF (5 mL) at room temperature. The mixture was stirred at room temperature for 5 minutes and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.298 g, 2.00 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 18h. The mixture was partitioned between water and EtOAc. The organic phase was dried (MgS04) and concentrated in vacuo. The resulting residue was purified by flash
chromatography (20-50% EtOAc in petroleum ether on Silica) to afford the title compound.
¾ NMR (400 MHz, DMSO-efc) δ ppm 0.93 - 1.51 (m, 9 H), 1.67 - 1.98 (m, 2 H), 2.02 - 2.15 (m, 1 H), 2.31 - 2.45 (m, 1 H), 2.54 - 2.85 (m, 2 H), 3.60 - 3.81 (m, 1 H), 4.25 - 4.35 (m, 1 H), 4.53 - 4.67 (m, 1 H), 5.13 - 5.36 (m, 1 H), 6.98 - 7.40 (m, 7 H), 7.46 - 7.56 (m, 1 H)
MS ES+: 475
2. Examples Example 1: N-[3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- y I)propy I] acetamide
Figure imgf000057_0001
HATU (0.096 g, 0.25 mmol) was added to a solution of 3-acetamido-3-phenylpropanoic acid (0.050 g, 0.24 mmol), DIPEA (0.046 mL, 0.27 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.036 g, 0.24 mmol) in DMF (0.5 mL). The mixture was stirred and allowed to stand for 18h. The mixture was purified by reverse phase preparative HPLC to afford the title compound.
Ή NMR (400 MHz, DMSO-efe) 5 ppm 1.65 - 1.92 (m, 4 H), 2.60 - 2.84 (m, 3 H), 3.52 - 3.78 (m, 2 H), 3.93 - 4.42 (m, 1 H), 4.48 - 4.64 (m, 1 H), 5.15 - 5.28 (m, 1 H), 6.92 - 7.40 (m, 9 H), 8.11 - 8.25 (m, 1 H)
MS ES+: 339
Example 2: (3R)-3-amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one
Figure imgf000057_0002
HATU (0.120 g, 0.315 mmol) was added to a solution of (3R)-3-{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (0.080 g, 0.30 mmol) and DIPEA (0.058 mL, 0.33 mmol) in DMF (0.5 mL) at room temperature. The mixture was stirred and allowed to stand. After 5 min, 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) was added. The mixture was stirred at room temperature for 2h. HC1 (4M in dioxane) (0.750 mL, 3.00 mmol) was added and the reaction was stirred at room temperature for lh. The mixture was stirred at 60 °C for lh. The mixture was basified with NaOH (2M) and extracted with DCM. The organic phase was concentrated in vacuo and the residue was purified by reverse phase preparative HPLC to afford the title compound.
1H NMR (400 MHz, DMSO-ifc) δ ppm 1.68 - 2.23 (m, 5 H), 2.64 - 2.83 (m, 1 H), 3.54 - 3.73 (m, 1 H), 4.05 - 4.69 (m, 4 H), 6.94 - 7.48 (m, 9 H)
MS ES+: 297 Example 3: (3S)-3-ammo-3-phenyl-l-(2 >4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one
Figure imgf000058_0001
Prepared as described for Example 2 using (3S)-3-{[(iert-butoxy)carbonyl]amino}-3- phenylpropanoic (0.080 g, 0.30 mmol) and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound.
!H NMR (400 MHz, DMSO-A) δ ppm 1.68 - 2.25 (m, 6 H), 2.69 - 2.83 (m, 1 H), 3.57 - 3.75 (m, 1 H), 4.07 - 4.19 (m, 1 H), 4.24 - 4.45 (m, 1 H), 4.54 - 4.67 (m, 1 H), 6.85 - 7.53 (m, 9 H)
MS ES+: 297
Example 4: (3S)-3-amino-l-(8-fluoro-2,3»4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride
Figure imgf000058_0002
HATU (0.798 g, 2.10 mmol) was added to a solution of (3S)-3-{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (531 mg, 2.00 mmol) and DIPEA (0.768 mL, 4.40 mmol) in DMF (5 mL). The mixture was stirred at room temperature. After 5 min, 8- fluoro-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (Intermediate 1, 0.334 g, 2.00 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 18h. The mixture was partitioned between water and EtOAc. The organic phase was washed with water, dried (MgS04) and concentrated in vacuo. The residue was purified by flash chromatography (20-50% EtOAc in petroleum ether on silica) to afford tert-butyl N-[(1S)- 3-(8-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-oxo-l-phenylpropyl]carbamate. HCI (4M in dioxane) (3.38 mL, 13.51 mmol) was added to a solution of teri-butyl N-[(1S)- 3-(8-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-oxo-l-phenylpropyl]carbamate (0.560 g, 1.35 mmol) in DCM (5 mL). The reaction was stirred at room temperature for 3.5h. The mixture was concentrated in vacuo and the residue was triturated with ether, filtered and dried. The solid was partitioned between NaOH (1M) and DCM. The aqueous phase was extracted with DCM. The combined organic phases were dried (MgS04) and concentrated in vacuo. The residue was dissolved in minimal ether and HCI (4M in dioxane, ca. 2 eq) was added. The resulting crystals were filtered, washing with ether and drying under vacuum to afford the title compound.
1H NMR (400 MHz, DMSO-<&) δ ppm 1.59 - 1.85 (m, 2 H), 1.99 - 2.18 (m, 1 H), 2.70 - 2.83 (m, 1 H), 2.89 - 3.14 (m, 1 H), 3.60 - 3.83 (m, 1 H), 3.89 - 4.20 (m, 1 H), 4.31 - 4.65 (m, 2 H), 6.83 - 7.11 (m, 2 H), 7.17 - 7.65 (m, 6 H), 8.19 - 8.51 (m, 3 H)
MS ES+: 315
Example 5: (3S)-3-amino-3-(4-fluorophenyl)-l-(2,3»4,5-tetrahydro-l,5-benzoxazepin- 5-yl)propan-l-one hydrochloride
Figure imgf000059_0001
Prepared as described for Example 4 using (3S)-3-(4-fluorophenyi)-3-{[(tert- butoxy)carbonyl] amino} propanoic acid (0.567 g, 2.00 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.298 g, 2.00 mmol) to afford the title compound. 1H NMR (400 MHz, DMSO-<fc) δ ppm 1.62 - 1.79 (m, 1 H), 1.93 - 2.13 (m, 1 H), 2.65 - 2.83 (m, 1 H), 2.93 - 3.13 (m, 1 H), 3.58 - 3.75 (m, 1 H), 3.94 - 4.14 (m, 1 H), 4.31 - 4.75 (m, 3 H), 7.00 - 7.70 (m, 8 H), 8.24 - 8.50 (m, 3 H)
MS ES+: 315
Example 6: (3S)-3-amino-3-(2-fluorophenyl)-l-(2 ,4,5-tetrahydro-l,5-beiizoxazepin- 5-yl)propan-l-one
Figure imgf000059_0002
Prepared as described for Example 2 using (3S)-3-(2-fluorophenyl)-3-{[(tert- butoxy)carbonyl]amino} propanoic acid (0.567 g, 2.00 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.298 g, 2.00 mmol) to afford the title compound. [H NMR (400 MHz, DMSO-i 6) δ ppm 1.64 - 1.91 (m, 2 H), 1.97 - 2.27 (m, 3 H), 2.65 - 2.86 (m, 1 H), 3.58 - 3.78 (m, 1 H), 4.00 - 4.27 (m, 1 H), 4.31 - 4.70 (m, 3 H), 6.93 - 7.46 (m, 8 H)
MS ES+: 315
Example 7: (3S)-3-(methylamino)-3-phenyl-l -(2,3.4,5-tetrahydro-l ,5-benzoxazepin-5- yl)propan-l-one
Figure imgf000060_0001
A solution of methyl iodide (0.023 mL, 0.36 mmol) in DMF (0.5 mL) was added to a suspension of (3S)-3-amino-3-phenyl-l-(2,3,4,5-tetrahydro-l ,5-benzoxazepin-5-yl)propan- 1-one (Example 3, 0.089 g, 0.30 mmol) and Cs2C03 (0.108 g, 0.33 mmol) in DMF (1 mL). The mixture was stirred at room temperature for 3h. The mixture was diluted with 2M NaOH and partitioned between water and EtOAc. The organic phase was concentrated in vacuo and the residue was purified by reverse phase preparative HPLC to afford the title compound.
Ή NMR (400 MHz, DMSO- ¼) δ ppm 1.93 - 2.08 (m, 4 H), 2.12 - 2.30 (m, 2 H), 2.32 - 2.47 (m, 1 H), 2.58 - 2.82 (m, 1 H), 3.53 - 4.08 (m, 3 H), 4.30 - 4.69 (m, 2 H), 6.88 - 7.43 (m, 9 H)
MS ES+: 311
Example 8: (3S)-3-(dimethylamino)-3-phenyl-l-(2 ,4,5-tetrahydro-l,5-beiizoxazepin- 5-yl)propan-l-one
Figure imgf000060_0002
A solution of methyl iodide (0.023 mL, 0.36 mmol) in DMF (0.5 mL) was added to a suspension of (3S)-3-amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan- 1-one (Example 3, 0.089 g, 0.30 mmol) and Cs2C03 (0.108 g, 0.33 mmol) in DMF (1 mL). The mixture was stirred at room temperature for 3h. The mixture was diluted with 2M NaOH and partitioned between water and EtOAc. The organic phase was concentrated and the residue was purified by reverse phase preparative HPLC to afford the title compound. 1H NMR (400 MHz, CDCb) δ ppm 1.72 - 1.86 (m, 1 H), 2.04 (s, 3 H), 2.14 (s, 3 H), 2.61 - 2.90 (m, 3 H), 3.50 - 4.00 (m, 3 H), 4.46 - 4.86 (m, 2 H), 6.97 - 7.18 (m, 4 H), 7.20 - 7.28 (m, 4 H), 7.30 - 7.37 (m, 1 H)
MS ES+: 325
Example 9: (3S)-3-amino-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yI)-3- phenylpropan-l-one
Figure imgf000061_0001
Prepared as described for Example 2 using (3S)-3-{[(tert-butoxy)carbonyl]amino}-3- phenylpropanoic (0.265 g, 1.00 mmol) and 9-fluoro-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (Intermediate 2, 0.167 g, 1.00 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-ifc) δ ppm 1.56 - 1.88 (m, 2 H), 2.01 - 2.15 (m, 1 H), 2.71 - 2.90 (m, 1 H), 2.93 - 3.18 (m, 1 H), 3.60 - 3.86 (m, 1 H), 4.05 - 4.17 (m, 1 H), 4.39 - 4.64 (m, 2 H), 6.93 - 7.50 (m, 8 H), 8.37 (br. s., 3 H)
MS ES+: 315
Example 10: (3S)-3-amino-3-(2-methylphenyl)-l-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propan-l-on
Figure imgf000061_0002
Prepared as described for Example 2 using (3S)-3-(2-methylphenyl)-3-{[(tert- butoxy)carbonyl]amino}propanoic acid (0.140 g, 0.50 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.075 g, 0.50 mmol) to afford the title compound. !H NMR (400 MHz, DMSO-ifc) δ ppm 1.63 - 1.94 (m, 4 H), 2.31 - 2.47 (m, 1 H), 2.66 - 2.82 (m, 1 H), 3.52 - 3.78 (m, 2 H), 4.17 - 4.43 (m, 2 H), 4.47 - 4.71 (m, 2 H), 6.93 - 7.22 (m, 7 H), 7.23 - 7.39 (m, 3 H) MS ES+: 311
Example 11: (3S)-3-amino-3-(2-methoxyphenyl)-l-(2,3?4,5-tetrahydro-l,5- benzoxazepin-5-y l)propan-l -on
Figure imgf000062_0001
Prepared as described for Example 2 using (3S)-3-(2-methoxyphenyl)-3-{[(teri- butoxy)carbonyl] amino} propanoic acid (0.148 g, 0.50 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.075 g, 0.50 mmol) to afford the title compound. 1H NMR (400 MHz, DMSO-ik) δ ppm 1.66 - 1.83 (m, 1 H), 1.98 - 2.10 (m, 1 H), 2.22 - 2.42 (m, 1 H), 2.65 - 2.85 (m, 1 H), 3.58 - 3.73 (m, 4 H), 4.19 - 4.39 (m, 2 H), 4.45 - 4.70 (m, 2 H), 6.77 - 7.35 (m, 10 H)
MS ES+: 327
Example 12: (3S)-3-amino-3-(2-chlorophenyl)-l-(2 54,5-tetrahydro-l,5- benzoxazepin-5-yl)propan-l-on
Figure imgf000062_0002
Prepared as described for Example 2 using (3S)-3-(2-chlorophenyl)-3-{[(tert- butoxy)carbonyl] amino} propanoic acid (0.150 g, 0.50 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.075 g, 0.50 mmol) to afford the title compound. 1H NMR (400 MHz, DMSO-<¼) δ ppm 1.71 - 1.83 (m, 1 H), 2.20 - 2.45 (m, 2 H), 2.67 - 2.83 (m, 1 H), 3.62 - 3.76 (m, 1 H), 4.27 - 4.42 (m, 1 H), 4.44 - 4.55 (m, 1 H), 4.56 - 4.73 (m, 2 H), 6.97 - 7.43 (m, 9 H), 7.43 - 7.55 (m, 1 H)
MS ES+: 331
Example 13: (3S)-3-(ethylamino)-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one
Figure imgf000063_0001
Prepared as described for Example 7 using iodoethane (0.094 g, 0.60 mmol) and (3S)-3- amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l-one (Example 3, 0.148 g, 0.50 mmol) to afford the title compound.
!H NMR (400 MHz, DMSO-</< δ ppm 0.81 - 0.98 (m, 3 H), 1.66 - 1.81 (m, 1 H), 1.88 - 2.45 (m, 5 H), 2.62 - 2.83 (m, 1 H), 3.51 - 3.73 (m, 1 H), 3.78 - 4.09 (m, 2 H), 4.32 - 4.68 (m, 2 H), 6.86 - 7.49 (m, 9 H)
MS ES+: 325
Example 14: (3S)-3-[(cyclopropylmethyl)amino]-3-phenyl-l-(2,3,4,5-tetrahyd:
benzoxazepin-5-yl)propan-l-on
Figure imgf000063_0002
Prepared as described for Example 7 using (bromomethyl)cyclopropane (0.081 g, 0.60 mmol) and (3 S)-3-amino-3-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan- 1 - one (Example 3, 0.148 g, 0.50 mmol) to afford the title compound.
¾ NMR (400 MHz, DMSO-<&) δ ppm 0.01 - 0.06 (m, 2 H), 0.31 - 0.45 (m, 2 H), 0.75 - 0.90 (m, 1 H), 1.70 - 1.91 (m, 2 H), 2.00 - 2.40 (m, 4 H), 2.70 - 2.83 (m, 1 H), 3.56 - 3.90 (m, 2 H), 3.97 - 4.16 (m, 1 H), 4.37 - 4.69 (m, 2 H), 6.94 - 7.52 (m, 9 H)
MS ES+: 351
Example 15: l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-2-(l,2,3,4- tetrahydroisoquinolm-l-yl)ethan-l-one
Figure imgf000063_0003
Prepared as described for Example 2 using 2-{2-[(tert-butoxy)carbonyl]-l,2,3,4- tetrahydroisoquinolin-l-yl} acetic acid (0.087 g, 0.30 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound. 1H NMR (400 MHz, DMSO-fik) δ ppm 1.73 - 1.85 (m, 1 H), 1.97 - 2.16 (m, 1 H), 2.23 - 2.38 (m, 1 H), 2.54 - 3.06 (m, 6 H), 3.60 - 3.82 (m, 1 H), 4.03 - 4.22 (m, 1 H), 4.25 - 4.50 (m, 2 H), 4.61 - 4.78 (m, 1 H), 6.66 - 7.48 (m, 8 H)
MS ES+: 323
Example 16: (3S)-l-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- (methylamino)-3-phenylpropan- -one
Figure imgf000064_0001
HCl (4M in dioxane, 0.062 mL, 0.246 mmol) was added to a solution of tert-butyl N-[(1S)- 3-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-oxo-l-phenylpropyl]- N-methylcarbamate (Intermediate 8, 0.018g, 0.041 mmol) in DCM (3 mL). The mixture was stirred at room temperature and allowed to stand for 18h. The mixture was partitioned between DCM and NaOH (1M). The organic phase was dried (MgS04) and concentrated in vacuo. The residue was purified by reverse phase preparative HPLC to afford the title compound.
¾ NMR (400 MHz, CDCh) δ ppm 1.16 (s, 3 H), 2.20 (s, 3 H), 2.51 - 2.63 (m, 1 H), 2.68 - 2.78 (m, 1 H), 2.85 - 2.95 (m, 1 H), 3.40 - 3.58 (m, 2 H), 3.95 - 4.06 (m, 1 H), 4.88 - 4.99 (m, 1 H), 6.88 - 6.97 (m, 2 H), 7.10 - 7.16 (m, 1 H), 7.17 - 7.28 (m, 7 H), 7.30 - 7.37 (m, 1 H)
MS ES+: 341
Retention time: 1.24 min
Example 17: (3S)-l-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepiii-5-yl)-3- (methylamino)-3-phenylpropan-l-one
Figure imgf000065_0001
Prepared as described for Example 16 using tert-butyl N-[(lS)-3-(3-hydroxy-3-methyl- 2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-oxo-l-phenylpropyl]-N-methylcarbarnate (Intermediate 9, 0.029 g, 0.066 mmol) and HCl (4M in dioxane, 0.099 mL, 0.396 mmol) to afford the title compound.
1H NMR (400 MHz, CDCh) δ ppm 1.20 (s, 3 H), 2.19 (s, 3 H), 2.36 - 2.54 (m, 2 H), 2.72 -
2.84 (m, 1 H), 3.52 - 3.59 (m, 1 H), 3.95 - 4.04 (m, 1 H), 4.07 - 4.14 (m, 1 H), 4.94 - 5.05
(m, 1 H), 5.31 (s, 1 H), 6.94 - 7.02 (m, 2 H), 7.04 - 7.15 (m, 2 H), 7.17 - 7.29 (m, 5 H),
7.30 - 7.36 (m, 1 H)
MS ES+: 341
Retention time: 1.22 min
Example 18: N-[(lS)-3-oxo-l-phenyl-3-(2^,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]propanamide
Figure imgf000065_0002
Propanoyl chloride (0.020 g, 0.22 mmol) was added to a solution of (3S)-3-amino-3- phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l-one (Example 3, 0.059 g, 0.20 mmol) and DIPEA (0.038 mL, 0.22 mmol) in MeCN (1 mL). The mixture was stirred at room temperature for 30 min. The mixture was diluted with EtOH and purified by reverse phase preparative HPLC to afford the title compound.
1H NMR (400 MHz, DMSO-<&) δ ppm 0.79 - 1.05 (m, 3 H), 1.55 - 1.85 (m, 2 H), 1.85 - 2.17 (m, 3 H), 2.30 - 2.46 (m, 1 H), 2.59 - 2.84 (m, 2 H), 3.51 - 3.77 (m, 1 H), 3.92 - 4.42 (m, 1 H), 4.51 - 4.64 (m, 1 H), 5.17 - 5.37 (m, 1 H), 6.92 - 7.44 (m, 8 H), 8.05 - 8.19 (m, 1 H)
MS ES+: 353 Example 19: N-[(lS)-3-oxo-l-phenyl-3-(2 ,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]cyclopropanecarboxamide
Figure imgf000066_0001
Prepared as described for Example 18 using cyclopropanecarbonyl chloride (0.023 g, 0.22 mmol) and (3 S)-3 -amino-3 -phenyl- 1 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan- 1 - one (Example 3, 0.059 g, 0.20 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-i 6) δ ppm 0.44 - 0.79 (m, 4 H), 1.44 - 1.56 (m, 1 H), 1.61 - 2.06 (m, 2 H), 2.31 - 2.48 (m, 1 H), 2.64 - 2.82 (m, 2 H), 3.52 - 3.80 (m, 1 H), 3.97 (d, J = 11.83 Hz, 1 H), 4.55 (d, J = 13.30 Hz, 1 H), 5.22 - 5.35 (m, 1 H), 6.94 - 7.39 (m, 9 H), 8.34 - 8.53 (m, 1 H)
MS ES+: 365
Example 20: 2-methvl-N-i(lS)-3-oxo-l-phenvl-3-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyl]propanamide
Figure imgf000066_0002
Prepared as described for Example 18 using 2-methylpropanoyl chloride (0.023 g, 0.22 mmol) and (3S)-3-amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l- one (Example 3, 0.059 g, 0.20 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-i&) δ ppm 0.83 - 1.04 (m, 6 H), 1.65 - 1.83 (m, 2 H), 2.22 -
2.46 (m, 2 H), 2.62 - 2.83 (m, 2 H), 3.53 - 3.77 (m, 1 H), 3.93 - 4.41 (m, 1 H), 4.52 - 4.63
(m, 1 H), 5.16 - 5.36 (m, 1 H), 6.91 - 7.38 (m, 9 H), 7.98 - 8.17 (m, 1 H)
MS ES+: 367
Example 21: 2,2,2-trifluoro-N-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyl]acetamide
Figure imgf000067_0001
TFAA (0.050 g, 0.24 mmol) was added to a solution of (3 S)-3-amino-3 -phenyl- 1 -(2,3 ,4,5- tetrahydro-l,5-benzoxazepin-5-yl)propan-l-one (Example 3, 0.059 g, 0.20 mmol) in MeCN (1 mL). The mixture was stirred at room temperature for 5 min. Further DIPEA (0.084 mL, 0.48 mmol) was added, followed by more TFAA (0.101 g, 0.48 mmol) and the reaction was allowed to stand for 5 min. The mixture was basified (DIP EA) and purified by reverse phase preparative HPLC to afford the title compound.
¾ NMR (400 MHz, DMSO-cfe) 6 ppm 1.62 - 1.88 (m, 2 H), 2.63 - 2.96 (m, 2 H), 3.55 - 3.81 (m, 1 H), 3.94 - 4.16 (m, 1 H), 4.32 - 4.68 (m, 2 H), 5.20 - 5.46 (m, 1 H), 6.92 - 7.50 (m, 9 H), 9.68 - 9.90 (m, 1 H)
MS ES+: 393
Example 22: N-[(lS)-l-(2-methylphenyl)-3-oxo-3-(2v3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyljacetamide
Figure imgf000067_0002
Prepared as described for Example 18 using acetyl chloride (0.013 g, 0.165 mmol) and of
(3S)-3-ammo-3-(2methylphenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l- one (Example 10, 0.047 g, 0.15 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-iid) δ ppm 1.57 - 1.85 (m, 4 H), 1.88 - 2.10 (m, 1 H), 2.14 -
2.38 (m, 3 H), 2.61 - 2.82 (m, 2 H), 3.51 - 3.78 (m, 1 H), 3.93 - 4.09 (m, 1 H), 4.30 - 4.63
(m, 2 H), 5.33 - 5.53 (m, 1 H), 6.94 - 7.45 (m, 8 H), 8.00 - 8.21 (m, 1 H)
MS ES+:353
Example 23: N-[(lS)-l-(2-chlorophenyl)-3-oxo-3-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyl]acetamide
Figure imgf000068_0001
Prepared as described for Example 18 using acetyl chloride (0.013 g, 0.17 mmol) and
(3S)-3-amino-3-(2-cWorophenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l- one (Example 12, 0.050 g, 0.15 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-i¼) δ ppm 1.63 - 1.89 (m, 4 H), 2.23 - 2.41 (m, 1 H), 2.60 -
2.92 (m, 2 H), 3.49 - 3.85 (m, 2 H), 4.18 - 4.39 (m, 1 H), 4.49 - 4.68 (m, 1 H), 5.42 - 5.63
(m, 1 H), 6.97 - 7.56 (m, 8 H), 8.09 - 8.23 (m, 1 H)
MS ES+: 373
Example 24: l-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)prdpyl]pyrrolidin-2-one
Figure imgf000068_0002
4-Chlorobutanoyl chloride (0.044 g, 0.32 mmol) was added to a stirred solution of (3S)-3- amino-3 -phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan- 1 -one (Example 3, 0.089 g, 0.30 mmol) and DIPEA (0.058 mL, 0.33 mmol) in DCM (2 mL). The mixture was stirred at room temperature for 5 min. The mixture was washed with water and the aqueous phase was extracted with DCM. The combined organic phases were dried (MgS04) and concentrated in vacuo. The residue was dissolved in DMF (2 mL). NaH (60% dispersion in mineral oil, 0.013 g, 0.33 mmol) was added and the mixture was stirred under nitrogen and then allowed to stand for 30 min. Further NaH (60% dispersion in mineral oil, 0.013 g, 0.33 mmol) was added. The mixture was stirred under nitrogen and then allowed to stand for 30 min. The mixture was diluted with EtOAc and washed with water. The organic phase was concentrated in vacuo and the residue was purified by reverse phase preparative HPLC to afford the title compound.
¾ NMR (400 MHz, DMSO-ck) 6 ppm 1.17 - 1.39 (m, 1 H), 1.52 - 2.03 (m, 4 H), 2.06 - 2.27 (m, 2 H), 2.56 - 3.10 (m, 4 H), 3.52 - 3.75 (m, 1 H), 4.25 - 4.45 (m, 1 H), 4.54 - 4.74 (m, 1 H), 5.42 - 5.59 (m, 1 H), 6.81 - 7.48 (m, 9 H) MS ES+: 365
Example 25: (3S)-3-amino-l-(6-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one
Figure imgf000069_0001
HATU (0.120 g, 0.32 mmol) was added to a solution of (3S)-3-{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (0.080 g, 0.30 mmol) and DIPEA (0.058 mL, 0.33 mmol) in MeCN (1 mL) at room temperature. The mixture was stirred for 5 min. 6- fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepine (Intermediate 5, 0.050 g, 0.30 mmol) was added and the reaction was stirred under nitrogen until homogeneous and allowed to stand for 48h. The reaction was heated to 40-50 °C for lh. The mixture was partitioned between water and EtOAc. The organic phase was dried (MgS04) and concentrated in vacuo. The residue was purified by flash chromatography (20-50% EtOAc in petroleum ether on silica). The resulting residue was taken up in DCM (1 mL) and treated with HC1 (4M in dioxane) (0.375 mL, 1.50 mmol). The mixture was stirred at room temperature and allowed to stand for 2h. The mixture was concentrated in vacuo and the residue was purified by reverse phase preparative HPLC, the combined fractions were extracted with DCM and the combined organic phases were concentrated in vacuo to afford the title compound.
1H NMR (400 MHz, CDCb) δ ppm 1.07 - 1.22 (m, 1 H), 1.36 - 1.48 (m, 1 H), 1.80 - 1.90 (m, 1 H), 2.10 - 2.47 (m, 2 H), 2.49 - 2.65 (m, 1 H), 2.72 - 2.86 (m, 1 H), 3.62 - 3.81 (m, 1 H), 4.23 - 4.58 (m, 2 H), 4.81 - 4.98 (m, 1 H), 6.80 - 6.98 (m, 2 H), 7.16 - 7.28 (m, 4 H), 7.29 - 7.33 (m, 2 H)
MS ES+: 315
Example 26: (3S)-3-amino-l-(9-fluoro-2 »4,5-tetrahydro-l,5-beiizoxazepiii-5-yl)-3-(4- fluorophenyl)propan-l-one hydrochloride
Figure imgf000069_0002
A solution of (3S)-3-(4-fluorophenyl)-3-{[(tert-butoxy)carbonyl]amino}propanoic acid (0.25 g, 0.882 mmol), 9-fluoro-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (Intermediate 2, 0.148 g, 0.882 mmol) and TEA (0.246 mL, 1.77 mmol) in DCM (5 mL) was treated with T3P (50% in EtOAc, 0.780 mL, 1.32 mmol) and stirred at room temperature for 18h. More TEA (0.246 mL, 1.77 mmol) and T3P (0.780 mL, 1.32 mmol) was added and the reaction was stirred at room temperature for a further lh. The reaction was diluted with DCM, washed with water, dried (phase separator) and concentrated in vacuo. The resulting residue was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica). A solution of the resulting residue in DCM (5 mL) was treated with HC1 (4M in dioxane, 0.927 mL, 3.71 mmol) and stirred at room temperature for lh. Ether was added and the precipitate was filtered and air-dried to afford the title compound.
Ή NMR (400 MHz, DMSO-d6) δ ppm 1.55 - 1.88 (m, 2 H), 1.92 - 2.21 (m, 1 H), 2.70 - 2.86 (m, 1 H), 2.92 - 3.13 (m, 1 H), 3.61 - 3.82 (m, 1 H), 4.01 - 4.22 (m, 1 H), 4.39 - 4.67 (m, 2 H), 6.91 - 7.69 (m, 7 H), 8.28 (br. s., 3 H)
MS ES+: 333
Examples 27 and 28: l-(9-fluoro-2 »4,5-tetrahydro-l,5-benzoxazepin-5-yl)-2-(l,2,3,4- tetrahydroisoquinolin-l-yl)eth -l-one
Figure imgf000070_0001
HATU (0.399 g, 1.05 mmol) was added to a solution of 2-{2-[(tert-butoxy)carbonyl]- l,2,3,4-tetrahydroisoquinolin-l-yl}acetic acid (0.291 g, 1.00 mmol) and DIPEA (0.192 mL, 1.10 mmol) in DMF (1 mL) at room temperature. The mixture was stirred for 5 min and 9- fluoro-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (Intermediate 2, 0.167 g, 1.00 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 72h. The mixture was partitioned between water and EtOAc. The organic phase was concentrated in vacuo and the residue was purified by flash chromatography (20-50% EtOAc in petroleum ether on silica). The resulting residue was taken up in DCM (3 mL) and treated with HC1 (4M in dioxane) (0.806 mL, 3.22 mmol). The mixture was stirred at room temperature and allowed to stand for 18h. The mixture was concentrated in vacuo and the resulting residue was partitioned between DCM and NaOH (0.5M). The organic phase was dried (MgS04) and concentrated in vacuo. The residue was purified by flash chromatography (20-50% EtOAc in petroleum ether on KP-NH). The resulting residue was purified by chiral SFC (ID Daicel CHIRALPAK, 34% MeOH + 0.2% diethylamine) to afford the title compounds. Example 27: fastest running stereoisomer
1H NMR (400 MHz, CDC13) δ ppm 1.79 - 1.95 (m, 1 H), 2.00 - 2.54 (m, 3 H), 2.59 - 2.97 (m, 5 H), 2.99 - 3.20 (m, 1 H), 3.70 - 3.88 (m, 1 H), 4.40 - 4.62 (m, 2 H), 4.83 - 4.96 (m, 1 H), 6.74 - 7.21 (m, 7 H)
MS ES+: 341
Example 28: slowest running stereoisomer
1H NMR (400 MHz, DMSO-</6) δ ppm 1.66 - 1.95 (m, 3 H), 2.07 - 2.37 (m, 2 H), 2.54 - 2.76 (m, 4 H), 2.79 - 3.12 (m, 2 H), 3.53 - 3.79 (m, 2 H), 4.21 - 4.32 (m, 1 H), 4.97 - 5.12 (m, 1 H), 6.93 - 7.13 (m, 4 H), 7.15 - 7.27 (m, 3 H), 7.29 - 7.37 (m, 2 H)
MS ES+: 341
Example 29: (3S)-3-amino-l-(9-fluoro-2,3J4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- fluorophenyl)propan-l-one hydrochloride
Figure imgf000071_0001
Prepared as described for Example 26 using (3S)-3-(2-fluorophenyl)-3-{[(tert- butoxy)carbonyl] amino} propanoic acid (0.250 g, 0.88 mmol), and 9-fluoro-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (Intermediate 2, 0.148 g, 0.88 mmol) to afford the title compound.
1H NMR (300 MHz, DMSO- e) δ ppm 1.52 - 1.94 (m, 2 H), 1.96 - 2.19 (m, 1 H), 2.56 - 2.89 (m, 2 H), 2.95 - 3.24 (m, 1 H), 3.58 - 3.85 (m, 1 H), 4.35 - 4.64 (m, 1 H), 4.71 - 4.97 (m, 1 H), 6.87 - 7.70 (m, 7 H), 8.43 (br. s., 3 H)
MS ES+: 333
Example 30: (3S)-3-[(2,2-difluoroethyl)amino]-3-phenyl-l-(2,3»4,5-tetrahydro-l,5- benzoxazepin-5-yl)propan-l-one
Figure imgf000072_0001
l,l-Difluoro-2-iodoethane (0.080 mL, 0.90 mmol) was added to a solution of (3S)-3- amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-l-one (Example 3, 0.050 g, 0.15 mmol) and DIPEA (0.157 mL, 0.90 mmol) in MeCN (1 mL). The reaction was stirred in the microwave for 30 min at 160 °C. l,l-Difluoro-2-iodoethane (0.080 mL, 0.90 mmol) was added and the mixture heated in the microwave at 170 °C for 45 min. The mixture was diluted with EtOAc, washed with water, dried (MgS04) and concentrated in vacuo. The resulting residue was purified by reverse phase preparative HPLC to afford the title compound.
1H NMR (400 MHz, CDC13) δ ppm 1.69 - 1.86 (m, 1 H), 2.08 - 2.43 (m, 3 H), 2.45 - 2.91 (m, 4 H), 3.57 - 3.78 (m, 1 H), 3.96 - 4.48 (m, 2 H), 4.77 - 4.90 (m, 1 H), 5.53 - 5.95 (m, 1 H), 6.87 - 7.27 (m, 8 H), 7.28 - 7.36 (m, 1 H)
MS ES+: 361
Example 31: (3S)-3-amino-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan-l-one h drochloride
Figure imgf000072_0002
Prepared as described for Example 26 using (3S)-3-(2-methylphenyl)-3-{[(tert- butoxy)carbonyl]amino}propanoic acid (0.25 g, 0.90 mmol) and 9-fluoro-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (Intermediate 2, 0.15 g, 0.90 mmol) to afford the title compound.
¾ NMR (400 MHz, DMSO-ifc) δ ppm 1.44 - 1.79 (m, 2 H), 1.79 - 2.09 (m, 1 H), 2.22 - 2.38 (m, 3 H), 2.57 - 2.78 (m, 1 H), 2.86 - 3.10 (m, 1 H), 3.51 - 3.78 (m, 1 H), 3.90 - 4.10 (m, 1 H), 4.32 - 4.53 (m, 1 H), 4.62 - 4.87 (m, 1 H), 6.80 - 7.42 (m, 7 H), 8.20 (br. s., 3 H) MS ES+: 329 Example 32: (3S)-l-(9-fluoro-2,354,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- (methyIamino)-3-phenylpropa -l-one hydrochloride
Figure imgf000073_0001
Prepared as described for Example 4 using (3S)-3-{[(tert- butoxy)carbonyl](methyl)amino}-3-phenylpropanoic acid (Intermediate 3, 0.263 g, 0.94 mmol) and 9-fluoro-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (Intermediate 2, 0.157 g, 0.94 mmol) to afford the title compound.
lR NMR (400 MHz, DMSO-tf6) δ ppm 1.42 - 1.88 (m, 2 H), 2.25 - 2.43 (m, 3 H), 2.56 - 2.88 (m, 2 H), 3.00 - 3.28 (m, 1 H), 3.53 - 4.09 (m, 2 H), 4.36 - 4.61 (m, 2 H), 6.86 - 7.68 (m, 8 H), 8.79 - 9.25 (m, 2 H)
MS ES+: 329
Example 33: tert-butyl N-[2,2-difluoro-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyl] carbamate
Figure imgf000073_0002
HATU (0.28 g, 0.730 mmol) was added to a solution of 3-((tert-butoxycarbonyl)amino)- 2,2-difluoro-3-phenylpropanoic acid (Intermediate 6, 0.20 g, 0.66 mmol) and pyridine (0.054 mL, 0.66 mmol) in DMF (1 n L) under nitrogen. After 10 min 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.099 g, 0.66 mmol) was added. The reaction was stirred at room temperature for 24h. The crude reaction mixture was diluted with DCM (10 mL) and washed with saturated sodium bicarbonate solution. The organic phase was washed with water, dried (phase separator) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC to afford the title compound.
Ή NMR (400 MHz, DMSO-ifc) δ ppm 1.25 - 1.43 (m, 9 H), 1.62 - 1.84 (m, 1 H), 2.78 - 2.93 (m, 1 H), 3.49 - 3.73 (m, 1 H), 3.81 - 4.15 (m, 2 H), 4.36 - 4.58 (m, 1 H), 5.35 - 5.56 (m, 1 H), 5.76 (s, 1 H), 7.01 - 7.16 (m, 3 H), 7.17 - 7.47 (m, 6 H)
MS ES+: 433 Example 34: 3-amino-2,2-difluoro-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one hydrochloride
Figure imgf000074_0001
HCl (4M in dioxane, 0.31 mL, 1.249 mmol) was added to a solution of tert-butyl N-[2,2- difluoro-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propyl]carbamate (Example 33, 0.090 g, 0.21 mmol) in dioxane (0.5 mL) under nitrogen. The reaction was heated to 100 °C for 10 min. The reaction was concentrated in vacuo and dried at 80 °C under vacuum to afford the title compound.
1H NMR (400 MHz, DMSO-cfe) δ ppm 1.42 - 1.59 (m, 1 H), 1.62 - 1.84 (m, 1 H), 2.78 - 2.99 (m, 1 H), 3.52 - 3.73 (m, 1 H), 3.88 - 4.12 (m, 1 H), 4.32 - 4.56 (m, 1 H), 5.11 - 5.38 (m, 1 H), 6.77 - 7.68 (m, 9 H), 8.58 - 9.09 (m, 3 H)
MS ES+: 333
Example 35: (3S)-3-amino-l-(3-methoxy-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride
Figure imgf000074_0002
Prepared as described for Example 34 using 3-methoxy-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.20 g, 1.12 mmol) and (3S)-3 -{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (0.31 g, 1.12 mmol) to afford the title compound.
¾ NMR (300 MHz, DMSO-i&) δ ppm 2.60 - 2.86 (m, 1 H), 3.11 - 3.42 (m, 6 H), 3.59 - 3.81 (m, 1 H), 4.05 - 4.43 (m, 1 H), 4.53 - 5.09 (m, 2 H), 6.92 - 7.65 (m, 9 H), 8.29 - 8.59 (m, 3 H)
MS ES+: 327
Example 36: (3S)-3-phenyl-l-(2,3»4,5-tetrahydro-l,5-benzoxazepiii-5-yl)butaii-l-one
Figure imgf000075_0001
HATU (0.12 g, 0.32 mmol) was added to a solution of (3S)-3-phenylbutanoic acid (49.3 mg, 0.30 mmol) and DIPEA (0.058 mL, 0.33 mmol) in DMF (1 mL) at room temperature. The mixture was stirred for 5 minutes. 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 mg, 0.30 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 2h. The reaction mixture was purified by reverse phase preparative HPLC to afford the title compound.
1H NMR (400 MHz, DMSO-fife) δ ppm 0.94 - 1.32 (m, 3 H), 1.68 - 1.82 (m, 1 H), 1.93 - 2.18 (m, 1 H), 2.26 - 2.49 (m, 2 H), 2.65 - 2.85 (m, 1 H), 2.93 - 3.25 (m, 1 H), 3.54 - 3.78 (m, 1 H), 4.05 - 4.43 (m, 1 H), 4.53 - 4.68 (m, 1 H), 6.88 - 7.39 (m, 9 H)
MS ES+: 296
Example 37: l-(3-hydroxy-3-methyl-2,3?4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan-l-one
Figure imgf000075_0002
Oxalyl chloride (0.180 mL, 2.05 mmol) was added to a solution of 3-(2- methylphenyl)propanoic acid (0.224 g, 1.37 mmol) in DCM (2 mL). A drop of DMF was added to the reaction. The mixture was stirred at room temperature and allowed to stand. After ca. 1.5 h the mixture was concentrated in vacuo whilst maintaining the temperature below 30 °C, adding DCM to assist removal of oxalyl chloride. The residue was dissolved in DCM {ca. 2 mL) and added dropwise to a solution of 3-methyl-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepin-3-ol (0.350 g, 1.37 mmol) and DIPEA (0.358 mL, 2.05 mmol) in DCM (2 mL) at 0 °C. The reaction was allowed to warm to room temperature for 24h. The crude reaction mixture was diluted with DCM and washed with saturated sodium bicarbonate solution. The organic phase was collected, washed with water, dried (phase separator) and concentrated in vacuo. The crude product was purified by flash
chromatography (0-50% EtOAc in petroleum ether on silica). The crude mixture was further purified by flash chromatography (0-50% EtOAc in petroleum ether on silica) to afford the title compound.
1H NMR (300 MHz, CDC13) δ ppm 1.20 (s, 3 H), 2.15 (s, 3 H), 2.47 - 2.64 (m, 2 H), 2.77 - 3.01 (m, 3 H), 3.43 - 3.55 (m, 2 H), 3.93 - 4.08 (m, 1 H), 4.80 - 4.95 (m, 1 H), 6.97 - 7.18 (m, 7 H), 7.23 - 7.27 (m, 1 H)
MS ES+: 326
Example 38: 4-phenyl-l-(2,3,4 -tetrahydro-l,5-benzoxazepin-5-yI)pentj
Figure imgf000076_0001
Prepared as described for Example 36 using 4-phenylpentanoic acid (0.054 g, 0.30 mmol) and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.300 mmol) to afford the title compound.
Ή NMR (400 MHz, OMSO-de) δ ppm 0.97 - 1.14 (m, 3 H), 1.55 - 1.87 (m, 4 H), 1.91 - 2.08 (m, 2 H), 2.52 - 2.60 (m, 1 H), 2.63 - 2.76 (m, 1 H), 3.56 - 3.71 (m, 1 H), 4.27 - 4.42 (m, 1 H), 4.55 - 4.67 (m, 1 H), 6.94 - 7.37 (m, 9 H)
MS ES+: 310
Example 39: 3-(2-methylphenyl)-l-(2,3)4,5-tetrahydro-l,5-beiizoxazepin-5-yl)propan- 1-one
Figure imgf000076_0002
Prepared as described for Example 36 using 3-(2-methylphenyl)propanoic acid (0.050 g, 0.30 mmol) and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound.
1H NMR (400 MHz, OMSO-de) δ ppm 1.69 - 1.82 (m, 1 H), 1.90 - 2.20 (m, 5 H), 2.32 - 2.47 (m, 1 H), 2.60 - 2.91 (m, 3 H), 3.59 - 3.78 (m, 1 H), 4.28 - 4.38 (m, 1 H), 4.55 - 4.71 (m, 1 H), 6.85 - 6.99 (m, 1 H), 6.99 - 7.21 (m, 5 H), 7.22 - 7.34 (m, 2 H)
MS ES+: 296 Example 40: (3R)-3-amino-4-phenyH-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)butan-l-one
Figure imgf000077_0001
Prepared as described for Example 2 using (3R)-3-{[(tert-butoxy)carbonyl]amino}-4- phenylbutanoic acid (0.084 g, 0.30 mmol) and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound.
¾ NMR (400 MHz, DMSO-A) δ ppm 1.37 - 1.49 (m, 1 H), 1.69 - 2.08 (m, 4 H), 2.11 - 2.45 (m, 3 H), 2.67 - 2.80 (m, 1 H), 3.11 - 3.26 (m, 1 H), 3.59 - 3.76 (m, 1 H), 4.30 - 4.47 (m, 1 H), 4.57 - 4.71 (m, 1 H), 6.96 - 7.38 (m, 9 H)
MS ES+: 311
Example 41: (3S)-3-amino-4-phenyl-l-(2,3?4,5-tetrahydro-l,5-benzoxazepm-5- yl)butan-l-one
Figure imgf000077_0002
Prepared as described for Example 2 using (3S)-3-{[(/ert-butoxy)carbonyl]amino}-4- phenylbutanoic acid (0.084 g, 0.30 mmol) and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-<k) δ ppm 1.29 - 1.50 (m, 2 H), 1.70 - 2.09 (m, 3 H), 2.13 - 2.45 (m, 2 H), 2.56 - 2.82 (m, 2 H), 3.15 - 3.26 (m, 1 H), 3.60 - 3.77 (m, 1 H), 4.33 - 4.43 (m, 1 H), 4.58 - 4.69 (m, 1 H), 6.96 - 7.34 (m, 9 H)
MS ES+: 311
Example 42: 4-amino-4-phenyl-l-(2^,4,5-tetrahydro-l,5-beiizoxazepin-5-yl)butan-l- one
Figure imgf000077_0003
Prepared as described for Example 2 using 4-{[(te t-butoxy)carbonyl]amino}-4- phenylbutanoic acid (0.084 g, 0.30 mmol) and 2,3,4,5-tetrahydrobenzo[b][l,4]oxazepine (0.045 g, 0.30 mmol) to afford the title compound.
1H NMR (300 MHz, CDC13) δ ppm 1.70 - 2.11 (m, 4 H), 2.12 - 2.34 (m, 2 H), 2.75 (br. s., 1 H), 3.61 - 3.91 (m, 2 H), 4.44 (br. s., 1 H), 4.82 (br. s., 1 H), 6.96 - 7.27 (m, 9 H) MS ES+: 311
Examples 43 and 44: l-(3-hydroxy-3-methyl-2,3»4,5-tetrahydro-l,5-benzoxazepin-5- yl)-3-(2-methylphenyl)propan-l
Figure imgf000078_0001
Oxalyl chloride (0.180 mL, 2.05 mmol) was added to a solution of 3-(2- methylphenyl)propanoic acid (0.224 g, 1.37 mmol) in DCM (2 mL). A drop of DMF was added to accelerate the reaction. The mixture was stirred at room temperature and allowed to stand. After ca. 1.5h the mixture was concentrated in vacuo while maintaining the temperature below 30 °C. The residue was dissolved in DCM (2 mL) and added drop wise to a solution of 3-methyl-2,3,4,5-tetrahydrobenzo[b][l,4]oxazepin-3-ol (0.350 g, 1.37 mmol) and DIPEA (0.358 mL, 2.05 mmol) in DCM (2 mL) at 0 °C and the reaction was allowed to warm to room temperature for 24h. The crude reaction mixture was diluted with DCM, washed with saturated sodium bicarbonate solution, followed by water, dried (phase separator) and concentrated in vacuo. The crude product was purified by flash
chromatography (0-50% EtOAc in petroleum ether on silica). The crude product was further purified by flash chromatography (0-50% EtOAc in petroleum ether on silica). The resulting residue was purified by chiral SFC (AD Daicel CHIRALPAK, 18% MeOH) to afford the title compounds.
Example 43: fastest running enantiomer
1H NMR (300 MHz, DMSO-ik) δ ppm 0.93 - 1.28 (m, 3 H), 2.06 (s, 3 H), 2.16 - 2.35 (m, 1 H), 2.37 - 2.48 (m, 1 H), 2.58 - 2.95 (m, 2 H), 3.20 - 3.31 (m, 1 H), 3.77 - 4.02 (m, 1 H), 4.55 - 4.79 (m, 1 H), 4.85 - 5.35 (m, 1 H), 6.82 - 7.38 (m, 8 H)
MS ES+: 326 Example 44: slowest running enantiomer
¾ NMR (300 MHz, DMSO-i¼) δ ppm 0.98 - 1.24 (m, 3 H), 2.01 - 2.34 (m, 3 H), 2.39 2.48 (m, 1 H), 2.59 - 3.03 (m, 3 H), 3.18 - 3.31 (m, 1 H), 3.75 - 4.02 (m, 1 H), 4.47 - 4. (m, 1 H), 4.97 - 5.26 (m, 1 H), 6.83 - 7.34 (m, 8 H)
MS ES+: 326
Example 45: l-(2,3,4,5-ieirahydro-l,5-bcnzoxazepin-5-yl)-2-( 1,2,3,4- tetrahydroisoquinolin-4-yl)ethan-l-one
Figure imgf000079_0001
Prepared as described for Example 2 using 2-{2-[(ter/-butoxy)carbonyl]-l, 2,3,4- tetrahydroisoquinolin-4-yl} acetic acid (0.291 g, 1.00 mmol) and 2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.149 g, 1.00 mmol) to afford the title compound. ¾NMR (400 MHz, DMSO-<&) δ ppm 1.69 - 1.86 (m, 1 H), 1.89 - 2.15 (m, 1 H), 2.17 - 2.43 (m, 1 H), 2.62 - 2.97 (m, 2 H), 3.07 - 3.52 (m, 3 H), 3.62 - 3.79 (m, 1 H), 3.90 - 4.10 (m, 2 H), 4.27 - 4.46 (m, 1 H), 4.58 - 4.74 (m, 1 H), 6.69 - 7.65 (m, 9 H)
MS ES+: 323
Example 46: (3S)-3-amino-3-(2-chIorophenyl)-l-(9-fluoro-2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propan-l-one hydrochloride
Figure imgf000079_0002
HATU (0.523 g, 1.376 mmol) was added to a solution of (3S)-3-(2-chlorophenyl)-3- {[(fert-butoxy)carbonyl] amino} propanoic acid (0.375 g, 1.251 mmol), 9-fluoro-2,3,4,5- tetrahydrobenzo[b][l,4]oxazepine (0.209 g, 1.251 mmol) and DIPEA (0.240 mL, 1.376 mmol) in DMF (1.50 mL). The reaction was stirred at room temperature for 72h. The mixture was partitioned between DCM and saturated NaHCCb solution. The phases were separated and the aqueous extracted with DCM. The combined organics were concentrated in vacuo. The crude product was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica). The resulting residue was taken up in DCM (5 mL) under nitrogen gas and HCl (4 M in dioxane) (1.20 mL, 4.80 mmol) was added. The reaction was stirred at room temperature for 3 hours. The reaction mixture was concentrated under a stream of dry nitrogen. The material was loaded onto a SCX cartridge, washed with methanol and eluted with 2M ammonia/methanol solution then concentrated in vacuo. The residue was dissolved in DCM (5 mL) and HCl (4 M in dioxane) (0.5 ml, 2.000 mmol) was added. The mixture was stirred at room temperature for 15 min, then concentrated in vacuo and triturated with diethyl ether to afford the title compound.
¾ NMR (400 MHz, DMSO-afe) δ ppm 1.67 - 1.85 (m, 1 H), 1.94 - 2.17 (m, 1 H), 2.56 - 2.86 (m, 2 H), 3.00 - 3.20 (m, 1 H), 3.63 - 3.84 (m, 1 H), 4.18 - 4.62 (m, 2 H), 4.88 - 5.13 (m, 1 H), 6.85 - 7.71 (m, 7 H), 8.51 (br. s., 3 H)
MS ES+: 349
Example 47: (3S)-3-amino-l-(3 -difluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)- 3-phenylpropan-l-one hydrochloride
Figure imgf000080_0001
DAST (1.803 mL, 13.64 mmol) was added to a solution of rt-butyl N-[(lS)-3-oxo-3-(3- oxo-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-l-phenylpropyl]carbamate (Intermediate 11, 0.560 g, 1.364 mmol) in DCM (10 mL) under nitrogen. The reaction was stirred at room temperature for 24h. The reaction was cooled to 0 °C and quenched with water. The mixture was diluted with DCM, washed with water, dried (MgS04) and concentrated in vacuo. The crude product was purified by flash chromatography (0-50% EtOAc in petroleum ether on silica). The residue was taken up in dioxane (1 mL) under nitrogen gas and HCl (4M in dioxane) (0.656 mL, 2.62 mmol) was added. The reaction was stirred at 80 °C for lh. The reaction was concentrated in vacuo and dried under vacuum at 80 °C for 30 min to afford the title compound.
1H NMR (400 MHz, DMSO- e) δ ppm 2.52 - 2.66 (m, 1 H), 3.01 - 3.26 (m, 1 H), 3.74 - 4.29 (m, 2 H), 4.35 - 4.70 (m, 2 H), 4.82 - 5.08 (m, 1 H), 7.03 - 7.58 (m, 9 H), 8.32 (br. s.,
3 H) MS ES+: 333
Example 48: (3S)-3-amino-l-(7-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride
Figure imgf000081_0001
HATU (0.399 g, 1.05 mmol) was added to a solution of (3S)-3-{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (0.265 g, 1.00 mmol) and DIPEA (0.192 mL, 1.10 mmol) in DMF (3 mL) at room temperature. The mixture was stirred for 5 min. 7- fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepine (Intermediate 12, 0.167 g, 1.00 mmol) was added. The mixture was stirred until homogeneous and allowed to stand for 18h. The mixture was partitioned between water and EtOAc. The aqueous phase was extracted with EtOAc. The combined organic phases were washed with water, dried (MgS04) and concentrated in vacuo. The resulting residue was purified by flash chromatography (20- 50% EtOAc in petroleum ether on silica). The resulting residue was dissolved in DCM (5 mL). HCI (4M in dioxane) (1.00 mL, 4.00 mmol) was added and the mixture stirred and allowed to stand for 18h. The mixture was concentrated in vacuo to afford the title compound.
1H NMR (400 MHz, DMSO-fifc) δ ppm 1.66 - 2.39 (m, 5 H), 2.54 - 2.64 (m, 1 H), 2.66 - 2.85 (m, 1 H), 3.51 - 3.75 (m, 1 H), 3.92 - 4.44 (m, 3 H), 4.51 - 4.71 (m, 1 H), 6.93 - 7.49 (m, 8 H)
MS ES+: 315
Example 49: 2-(2-acetyl-l,2,3,4-tetrahydroisoquinolin-4-yl)-l-(2,354,5-tetrahydro-l,5- benzoxazepin-5-yl)ethan-l-one
Figure imgf000081_0002
Acetyl chloride (0.026 g, 0.330 mmol) was added to a solution of l-(2,3,4,5-tetrahydro- l,5-benzoxazepin-5-yl)-2-(l,2,3,4-tetrahydroisoquinolin-4-yl)ethan-l-one (Example 45, 0.097 g, 0.300 mmol) and DIPEA (0.063 mL, 0.360 mmol) in DCM (1 mL) and stirred at room temperature for lh. The mixture was partitioned between water and DCM. The organic phase was concentrated in vacuo and the residue was purified by reverse phase preparative HPLC to afford the title compound.
1H NMR (400 MHz, DMSO) δ ppm 1.70 - 1.86 (m, 1 H), 1.87 - 2.17 (m, 4 H), 2.29 - 2.48 (m, 1 H), 2.66 - 2.88 (m, 1 H), 2.93 - 3.24 (m, 1 H), 3.34 - 3.52 (m, 1 H), 3.56 - 3.88 (m, 2 H), 4.03 - 4.58 (m, 3 H), 4.59 - 4.80 (m, 2 H), 6.86 - 7.42 (m, 8 H)
MS ES+: 365
Example 50: (3S)-3-amino-l-(3-fluoro-2,3»4,5-tetrahydro-l,5-beuzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride
Figure imgf000082_0001
HCI (4M in dioxane) (0.145 mL, 0.579 mmol) was added to a solution of tert-butyl N- [(1 S)-3-(3-fluoro-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3-oxo- 1- phenylpropyljcarbamate (Intermediate 14, 0.040 g, 0.097 mmol) in dioxane (0.3 mL) under nitrogen. The reaction was stirred at room temperature for 18h. The reaction was concentrated in vacuo to afford the title compound upon drying at 100 °C in a vacuum oven.
¾ NMR (400 MHz, DMSO-ck) δ ppm 2.82 - 3.11 (m, 2 H), 3.63 - 3.92 (m, 2 H), 4.39 - 4.67 (m, 2 H), 4.77 - 5.13 (m, 2 H), 6.91 - 7.65 (m, 9 H), 8.14 - 8.50 (m, 3 H)
MS ES+: 315
Example 51: 2-(2-acetyl-l,2 »4-tetrahydroisoquinolin-l-yl)-l-(2,354,5-tetrahydro-l,5- benzoxazepin-5-y l)ethan-l -one
Figure imgf000083_0001
Acetyl chloride (0.007 g, 0.092 mmol) was added to a solution of l-(2,3,4,5-tetrahydro- l,5-benzoxazepin-5-yl)-2-(l,2,3,4-tetrahydroisoqumolin-l-yl)ethan-l-one (Example 15, 0.027 g, 0.084 mmol) and DIPEA (0.018 mL, 0.100 mmol) in MeCN (1 mL). The mixture was stirred at room temperature for 30 min. The reaction mixture was purified by reverse phase preparative HPLC to afford the title compound.
1H NMR (400 MHz, CDCb) δ ppm 1.70 - 1.93 (m, 1 H), 2.11 - 3.03 (m, 10 H), 3.51 - 3.80 (m, 2 H), 4.35 - 4.49 (m, 1 H), 4.80 - 4.97 (m, 1 H), 5.40 - 5.68 (m, 1 H), 6.74 - 7.28 (m, 8 H)
MS ES+: 365
Example 52: (3S)-amino-l-{3,4-dihydro-2,4-methanobenzo[b][l,4]oxazepin-5-yl}-3- phenylpropan-l-one
Figure imgf000083_0002
Prepared as described for Example 2, using (3S)-3-{[(tert-butoxy)carbonyl]amino}-3- phenylpropanoic (0.117 g, 0.44 mmol) and 2,3,4,5-tetrahydro-2,4- methanobenzo[b][l,4]oxazepine (Intermediate 15, 0.071 g, 0.440 mmol) to afford the title compound.
1H NMR (400 MHz, CDCb) δ ppm 1.36 - 1.50 (m, 2 H), 1.76 - 2.01 (m, 2 H), 2.46 - 2.64 (m, 2 H), 2.65 - 2.79 (m, 2 H), 4.35 - 4.50 (m, 1 H), 4.78 - 5.00 (m, 2 H), 6.94 - 7.26 (m, 7 H), 7.27 - 7.40 (m, 2 H)
MS ES+: 309
Examples 53 and 54: (3S)-3-amino-l-(3-methoxy-2,3>4,5-tetrahydro-l,5- benzoxazepin-5-yl)-3-phenylpropan-l-one
Figure imgf000084_0001
Example 35 was purified by chiral SFC (AD Daicel CHIRALPAK, 20% EtOH and 0.2% DEA) to afford the title compounds.
Example 53: fastest running isomer
1H NMR (400 MHz, CDC13) δ ppm 2.38 - 2.52 (m, 1 H), 2.54 - 2.76 (m, 2 H), 3.36 - 3.83 (m, 6 H), 4.02 - 4.16 (m, 1 H), 4.36 - 4.63 (m, 2 H), 4.89 - 5.09 (m, 1 H), 6.89 - 7.11 (m, 2 H), 7.12 - 7.27 (m, 5 H), 7.29 - 7.39 (m, 2 H)
MS ES+: 327
Example 54: slowest running isomer
1H NMR (400 MHz, CDC13) δ ppm 2.35 - 2.89 (m, 4 H), 3.39 - 3.84 (m, 6 H), 4.24 - 4.56 (m, 2 H), 4.94 - 5.11 (m, 1 H), 6.85 - 7.04 (m, 2 H), 7.07 - 7.26 (m, 5 H), 7.28 - 7.33 (m, 2 H)
MS ES+: 327
Example 55: (3S)-3-amino-l-[(4S)-9-fluoro-4-methyl-2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl]-3-phenylpropan-l-one
Figure imgf000084_0002
A solution/suspension of tert-butyl ((S)-3-(((S)-4-(2-bromo-6-fluorophenoxy)butan-2- yl)amino)-3-oxo-l-phenylpropyl)carbamate (Intermediate 16, 0.050 g, 0.098 mmol) and bis(3,5-bis(trifluoromemyl)phenyl)(2',4^6'-^
yl)phosphine (0.016 g, 0.020 mmol) in toluene (3 mL) was degassed and purged with nitrogen (3 times). Sodium tert-butoxide (0.019 g, 0.196 mmol) and Pd2(dba)3 (0.009 g, 9.82 μηιοΐ) were added. The mixture, now a deep crimson colour, was degassed and purged (3 times) again. The mixture was heated at 100 °C for 4h. The mixture was partitioned between water and EtOAc. The organic phase was collected, dried (MgS04) and concentrated in vacuo. The residue was purified by flash chromatography (20-50% EtOAc in petroleum ether). The resulting residue was dissolved in DCM (0.5 mL) and HC1 (4 M in dioxane) (0.1 mL, 0.40 mmol) was added. The reaction was stirred at room temperature for lh. The mixture was concentrated in vacuo and the residue was purified by reverse phase preparative HPLC to afford the title compound.
!H NMR (400 MHz, Methanol-^) δ ppm 1.03 (d, J = 6.69 Hz, 3 H), 1.52 - 1.62 (m, 2 H), 2.03 - 2.10 (m, 1 H), 2.37 - 2.44 (m, 1 H), 2.59 - 2.65 (m, 1 H), 4.00 - 4.05 (m, 3 H), 4.28 - 4.33 (m, 1 H), 6.87 - 6.93 (m, 1 H), 6.99 - 7.10 (m, 2 H), 7.12 - 7.35 (m, 6 H)
MS ES+: 329
Example 56: (3S)-3-amino-l-(9-methyl-2,354,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride
Figure imgf000085_0001
Prepared as described for Example 4 using (3S)-3-{[(teri-butoxy)carbonyl]amino}-3- phenylpropanoic (0.265 g, 1.0 mmol) and 9-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepine
(Intermediate 17, 0.163 g, 1.0 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-^) δ ppm 1.60 - 1.81 (m, 2 H), 2.13 - 2.28 (m, 3 H), 2.43 -
2.49 (m, 1 H), 2.64 - 2.80 (m, 1 H), 2.91 - 3.19 (m, 1 H), 3.46 - 3.68 (m, 1 H), 4.05 - 4.44
(m, 1 H), 4.48 - 4.67 (m, 2 H), 6.87 - 7.26 (m, 3 H), 7.29 - 7.52 (m, 5 H), 8.25 - 8.53 (m, 3
H)
MS ES+: 311
Example 57: (3S)-3-amino-l-(9-chloro-2^,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride
Figure imgf000085_0002
Prepared as described for Example 4 using (3S)-3-{[(tert-butoxy)carbonyl]amino}-3- phenylpropanoic (0.265 g, 1.0 mmol) and 9-chloro-2,3,4,5-tetrahydro-l,5-benzoxazepine (Intermediate 18, 0.184 g, 1.0 mmol) to afford the title compound. Ή NMR (400 MHz, DMSO-fik) δ ppm 1.63 - 1.83 (m, 1 H), 2.53 - 2.61 (m, 1 H), 2.70 - 3.22 (m, 2 H), 3.31 - 3.37 (m, 2 H), 3.60 - 3.80 (m, 1 H), 4.41 - 4.72 (m, 2 H), 7.04 - 7.24 (m, 2 H), 7.27 - 7.65 (m, 6 H), 8.25 - 8.69 (m, 3 H)
MS ES+: 331
Example 58: (3S)-3-amino-l-[(4R)-9-fluoro-4-methyl-2 ,4,5-tetrahydro-l,5- benzoxazepin-5-yl]-3-phenylpropan-l-one
Figure imgf000086_0001
Prepared as described for Example 55 using tert-butyl N-[(lS)-2-{[(2R)-4-(2-bromo-6- fluorophenoxy)butan-2-yl]carbamoyl}-l-phenylethyl]carbamate (Intermediate 19, 0.06 g, 0.118 mmol) to afford the title compound.
lU NMR (400 MHz, Methanol-^) δ ppm 0.94 - 1.07 (m, 3 H), 1.56 - 1.66 (m, 1 H), 2.12 - 2.21 (m, 1 H), 2.30 - 2.38 (m, 1 H), 2.56 - 2.63 (m, 1 H), 4.11 - 4.18 (m, 1 H), 4.30 - 4.44 (m, 3 H), 4.91 - 5.01 (m, 1 H), 6.52 - 6.62 (m, 1 H), 6.93 - 7.03 (m, 1 H), 7.10 - 7.45 (m, 7 H)
MS ES+: 329
Example 59: (3S)-3-amino-l-{4,5-dihydro-2H-spiro[l,5-benzoxazepine-3,l'- cyclopropane]-5-yl}-3-phenylpropan-l-one hydrochloride
Figure imgf000086_0002
Prepared as described for Example 4 using 4,5-dihydro-2H-spiro[l,5-benzoxazepine-3, - cyclopropane]] (Intermediate 20, 0.22g, 1.19 mmol) and (3S)-3-{[(tert- butoxy)carbonyl]amino}-3-phenylpropanoic (0.316 g, 1.193 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-cfe) δ ppm 0.21 - 0.50 (m, 3 H), 0.53 - 0.84 (m, 1 H), 2.51 - 2.61 (m, 1 H), 2.87 - 3.10 (m, 2 H), 3.45 - 3.68 (m, 1 H), 3.73 - 3.80 (m, 1 H), 4.04 - 4.21 (m, 1 H), 4.51 - 4.66 (m, 1 H), 6.99 - 7.21 (m, 3 H), 7.28 - 7.47 (m, 6 H), 8.19 - 8.44 (m, 3 H)
MS ES+: 323
Example 60: (3S)-3-amino-l-(9-methoxy-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride
Figure imgf000087_0001
Prepared as described for Example 4 using 9-methoxy-2,3,4,5-tetrahydro-l,5- benzoxazepine (Intermediate 21, 0.180 g, 1.00 mmol) and (3S)-3-{[(fert- butoxy)carbonyl]amino}-3-phenylpropanoic (0.265 g, 1.00 mmol) to afford the title compound.
1H NMR (400 MHz, DMSO-^) δ ppm 1.63 - 1.77 (m, 1 H), 2.09 (s, 1 H), 2.44 - 2.49 (m, 1 H), 2.65 - 2.78 (m, 1 H), 2.92 - 3.13 (m, 1 H), 3.46 - 3.63 (m, 1 H), 3.73 - 3.87 (m, 3 H), 4.10 - 4.42 (m, 1 H), 4.46 - 4.67 (m, 2 H), 6.64 - 6.78 (m, 1 H), 6.86 - 6.94 (m, 1 H), 7.00 - 7.15 (m, 2 H), 7.27 - 7.48 (m, 4 H), 8.24 - 8.57 (m, 3 H)
MS ES+: 327
Example 61: (3S)-3-amino-3-(2-bromophenyl)-l-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propan-l-on
Figure imgf000087_0002
A solution of tert-butyl N-[(lS)-l-(2-bromophenyl)-3-oxo-3-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyl]carbamate (Intermediate 22, 0.143 g, 0.10 mmol) in MeOH (3 mL) was treated with HCI (4 M in dioxane) (0.30 ml, 1.200 mmol). The mixture was stirred and allowed to stand for 4h. The mixture was concentrated in vacuo and the resulting residue was purified by reverse phase preparative HPLC to afford the title compound. lK NMR (400 MHz, DMSO-ifc) δ ppm 1.66 - 1.87 (m, 1 H), 1.88 - 2.13 (m, 3 H), 2.18 2.44 (m, 1 H), 2.52 - 2.61 (m, 1 H), 2.65 - 2.88 (m, 1 H), 3.56 - 3.80 (m, 1 H), 4.22 - 4. (m, 3 H), 6.93 - 7.81 (m, 8 H)MS ES+: 375
Example 62: (3S)-3-amino-3-(2-cyclopropylphenyl)-l-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propan-l -on
Figure imgf000088_0001
A mixture of tert-butyl N-[(lS)-l-(2-bromophenyl)-3-oxo-3-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propyl]carbamate (Intermediate 22, 0.143 g, 0.3 mmol),
cyclopropylboronic acid (0.039 g, 0.450 mmol), tripotassium phosphate (0.191 g, 0.900 mmol) and tetrakis(triphenylphosphine)palladium(0) (0.010 g, 0.009 mmol) was dissolved in toluene (5 mL). Water (0.25 mL) was added and mixture was heated at 120 °C for 20 minutes in the microwave. The mixture was partitioned between water and EtOAc. The organic phase was dried (MgS04) and concentrated in vacuo. The resulting residue was purified by flash chromatography (20-50% EtOAc in petroleum ether on silica). The residue was further purified by flash chromatography (20-30% EtOAc in petroleum ether on silica). This material was dissolved in MeOH (3 mL) and HC1 (4 M in dioxane) (0.30 mL, 1.200 mmol) was added. The mixture was stirred and allowed to stand for 4h. The mixture was concentrated in vacuo and the residue was purified reverse phase preparative HPLC to afford the title compound
1H NMR (400 MHz, CDC13) δ ppm 0.46 - 0.76 (m, 2 H), 0.79 - 1.04 (m, 2 H), 1.66 - 1.76 (m, 1 H), 1.83 (s, 3 H), 2.17 - 2.40 (m, 1 H), 2.41 - 2.71 (m, 2 H), 2.73 - 2.91 (m, 1 H), 3.62 - 3.83 (m, 1 H), 4.31 - 4.50 (m, 1 H), 4.82 - 5.22 (m, 2 H), 6.87 - 7.26 (m, 7 H), 7.30 - 7.46 (m, 1 H)
MS ES+: 337
3. Biological efficacy of compounds of the invention
mGluR? Assay
The ability of the test compounds to activate mGluR7 was determined by their ability to reduce forskolin stimulated cAMP production. Compounds were assessed in a CRE- directed luciferase reporter gene assay, using a stable CHO cell line expressing the CRE- luc reporter and human mGluR7 genes. In this cell line, production of cAMP stimulated the transcription of the luciferase gene and luciferase activity was then measured in a luminescent enzyme assay (Steady Glo assay; Promega E2550). Activation of mGluR7 decreased the forskolin stimulated luminescence signal.
The day prior to the assay, compounds were serially diluted in DMSO (lOOx final assay concentration (FAC)), in 384-well plates which were then stored in the dark at room temperature (RT) until use. Cells were seeded at 12.5 k/well in white, clear bottom 384- well plates (Corning 3707) and left for one hour at RT followed by an overnight incubation (37 °C). The following day, the DMSO compound plate was diluted 1 :20 (5x FAC) in Opti-MEM I (Life Technologies 11058021). The growth media was removed from the cell plate and replaced with 15 μΐ Opti-MEM I, followed by a 5 μΐ addition from the 5x compound plate and a fifteen minute incubation (37 °C). Forskolin (Sigma F3917) was then added to the wells (5 μΐ of 2.5 μΜ) and the plate was incubated for five hours (37 °C). During this incubation, the Steady Glo Substrate reagent was warmed to 37 °C. Aliquots (11ml; stored at -20 °C) of this reagent were prepared by dissolving the contents of 1 vial of lyophilised substrate in 100 ml Steady-Glo buffer. A 25 μΐ addition of the substrate was made to all wells and the plate was incubated for thirty minutes at RT, on a plate shaker (300 rpm; in the dark). Luminescence was then measured using the En Vision Multilabel Reader (Perkin Elmer).
Compound activity was examined using a 10-point, half log concentration-response range and each concentration was tested in duplicate wells. Luminescence values were normalised to 'maximum' (forskolin alone) and 'minimum' (forskolin in the presence of tool mGluR7 agonist) controls. EC50 values were derived from this data using non-linear regression and a four parameter curve fit. The EC5o values for the compounds of the Examples are shown in Table 1. Results
Table 1
Ex No. EC50 (nM) Ex No. EC50 (nM) Ex No. EC50 (nM)
1 95 2 800 3 68
4 280 5 146 6 63
7 54 8 200 9 167
10 21 11 400 12 13
13 57 14 1 15 211
16 1100 17 - 18 13
19 3 20 11 21 8
22 2 23 4 24 6
25 111 26 109 27 258
28 - 29 38 30 11
31 29 32 100 33 49
34 440 35 200 36 18
37 70 38 15 39 12
40 1500 41 5800 42 730
43 - 44 178 45 1050
46 18 47 12 48 280
49 500 50 84 51 540
52 400 53 70 54 41
55 1650 56 87 57 61
58 27 59 151 60 3090
61 22 62 14 References
1. O'Connor R.M., Finger B.C., Flor P.J. and Cryan J.F., 2010. Metabotropic glutamate receptor 7: at the interface of cognition and emotion. Eur J Pharmacol., 639 (1- 3), 123-31.
2. Konieczny J. and Lenda T., 2013 Contribution of the mGluR7 receptor to
antiparkinsonian-like effects in rats: a behavioral study with the selective agonist AM 082. Pharmacol Rep., 65 (5), 1194-1203.
3. Greco B., Lopez S., van der Putten H. and Flor P.J., 2010. Amalric M.
Metabotropic glutamate 7 receptor subtype modulates motor symptoms in rodent models of Parkinson's disease. J Pharmacol Exp Ther., 332 (3), 1064-71.
4. Bradley S.R., Standaert D.G., Levey A.I. and Conn P.J., 1999. Distribution of group III mGluRs in rat basal ganglia with subtype-specific antibodies. Ann N Y Acad Sci., 868, 531-4.
5. Conn PJ. and Niswender CM., 2006. mGluR7's lucky number. Proceedings of the National Academy of Sciences of the United States of America, 103 (2), 251-2.
6. Hovels0 N., Sotty F., Montezinho L., Pinheiro P., Herrik K. and Merk A.,2012. Therapeutic Potential of Metabotropic Glutamate Receptor Modulators. Curr
Neuropharmacol., 10 (1), 12-48.
7. Kandaswamy R., McQuillin A., Curtis D. and Gurling H., 2014. Allelic Association, DNA Resequencing and Copy Number Variation at the Metabotropic Glutamate Receptor GRM7 Gene Locus in Bipolar Disorder. Am J Med Genet B Neuropsychiatr Genet., 165 (4), 365-72.
8. Palucha-Poniewiera A., Szewczyk B. and Pile A, 2014.. Activation of the mTOR signaling pathway in the antidepressant-like activity of the mGlu5 antagonist MTEP and the mGlu7 agonist AMN082 in the FST in rats. Neuropharmacology, 82, 59-68.
9. Palucha A., Klak K., Branski P., van der Putten H., Flor P.J. and Pile A., 2007.
Activation of the mGlu7 receptor elicits antidepressant-like effects in mice.
Psychopharmacology (Berl)., 194 (4), 555-62.
10. Kalinichev M., Rouillier M., Girard F., Royer-Urios I., Bournique B., Finn T. et al., 2013. ADX71743, a potent and selective negative allosteric modulator of metabotropic glutamate receptor 7: in vitro and in vivo characterization. J Pharmacol Exp Ther., 344 (3), 624-36. 11. Bolonna A. A., Kerwin R.W., Munro J., Arranz M.J., Makoff A. J., 2001.
Polymorphisms in the genes for mGluR types 7 and 8: association studies with schizophrenia. Schizophr Res., 47 (1), 99-103.
12. Friedman, R. A., et al., 2009. GRM7 variants confer susceptibility to age-related hearing impairment. Hum Mol Genet 18(4), 785-796.

Claims

92
C L A I M S
1. A compound of formula (I) or a pharmaceutically acceptable salt thereof
Figure imgf000093_0001
wherein
X1 is absent or is -C¾- or -CH(CH3)-;
X2 is -CH2-, -CHF-, -CF2- or -CH2CH2-, provided that when X1 is -CH2- or -CH(CH3)-, then X2 is -CH2- only;
R1, R2, R3 and R4 each independently represent hydrogen, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; either R5 represents hydrogen, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy and R6 represents hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkylsulphonyl or -NR14R15, or when X1 is absent, R5 and R6 together form a linking group, -[CH2]m-, in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or
N(COCH3) heteroatom moieties provided that if the linking group contains two
heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another;
n is 0, 1 or 2;
n
each R independently represents halogen, cyano, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; 93 each of R8, R9, R10, R11, R12 and R13 independently represents hydrogen, hydroxyl, cyano, halogen, C1-C6 alkyl, C3-C6 cycloalkyl or C1-C6 alkoxy; or,
alternatively,
O Q
(i) R and R together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring; or
(ii) R10 and R11 together with the carbon atom to which they are attached form a saturated
3- to 6-membered carbocyclic ring; or
(iii) R 1 and R 1 together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic ring; or
(iv) R9 and R10 together with the carbon atoms to which they are attached form a saturated 3- to 6-membered carbocyclic ring; or
(v) R and R together form a methylene bridge,
provided that no more than one carbocyclic ring or methylene bridge according to (i) to (v) above can be present in ring A; and
either R14 and R15 each independently represent hydrogen or a C1-C6 alkyl, C3-C6 cycloalkylmethyl, C1-C6 alkylcarbonyl, C3-C6 cycloalkylcarbonyl or C1-C6
alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by at least one halogen atom, or
R14 and R15 together with the nitrogen atom to which they are attached form a saturated 4- to 6-membered heterocyclic ring optionally comprising a further ring heteroatom selected from nitrogen and oxygen, the heterocyclic ring being unsubstituted or substituted by at least one substituent selected from halogen, oxo and C1-C3 alkyl.
2. A compound according to claim 1, wherein X is absent.
3. A compound according to claim 1 or claim 2, wherein X is -CH2- 94
4. A compound according to any one of the preceding claims, wherein R , R , R , R and R each independently represent hydrogen or halogen.
A compound according to any one of the preceding claims, wherein R represents
NR14R15.
6. A compound according to any one of the preceding claims, wherein R and R each independently represent hydrogen or a C1-C2 alkyl, cyclopropylmethyl,
C1-C3 alkylcarbonyl, cyclopropylcarbonyl or C1-C4 alkoxycarbonyl group, wherein the
C1-C2 alkyl and C1-C3 alkylcarbonyl groups are optionally substituted by one to three halogen atoms.
7. A compound according to claim 1, wherein
X1 is absent or is -C¾- or -CH(CH3)-;
X2 is -CH2-, -CF2- or -CH2CH2-;
R1, R2, R3 and R4 each independently represent hydrogen, fluorine, chlorine, methyl or methoxy;
either R5 represents hydrogen, fluorine, chlorine, bromine, cyclopropyl, methyl or methoxy and R6 represents hydrogen, methyl or -NR1 R15, or when X1 is absent, R5 and R6 together form a linking group, -[CH2]m-, in which m is 2, 3 or 4, wherein up to two -[CH2]- moieties may be replaced by oxygen, NH or
N(COCH3) heteroatom moieties provided that if the linking group contains two heteroatom moieties then m is 3 or 4 and the heteroatom moieties are not situated adjacent one another;
n is 0 or 1 ;
n
R represents halogen, methyl or methoxy; 95 each of R , R , R , R , R and R independently represents hydrogen, hydroxyl, fluorine, methyl or methoxy or, alternatively, either R10 and R1 1 together with the carbon atom to which they are attached form a saturated 3- to 6-membered carbocyclic
o 1
ring, or R and R together form a methylene bridge; and either R14 and R15 each independently represent hydrogen or a C1-C2 alkyl, cyclopropylmethyl, C1-C3 alkylcarbonyl, cyclopropylcarbonyl or C1-C4 alkoxycarbonyl group, wherein each of the preceding groups is optionally substituted by at least one halogen atom, or
R14 and R15 together with the nitrogen atom to which they are attached form a saturated 5-membered heterocyclic ring, the heterocyclic ring being unsubstituted or substituted by at least one substituent selected from halogen, oxo and C1-C3 alkyl.
8. A compound according to claim 1, wherein
X1 is absent;
X2 is -CH2-;
R1, R2, R4 and R5 each represent hydrogen; R represents hydrogen or fluorine;
R6 represents -NR14R15;
n is 0 or 1 ;
7
R represents fluorine;
R 8 , R 9 , R 12 and R 11 each represent hydrogen;
R10 and R11 each independently represent hydrogen or fluorine; and
R14 and R15 each represent hydrogen.
9. A compound of formula (I) as defined in claim 1 selected from:
N-[3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propyl]acetamide; (3R)-3-amino-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-berizoxazepin-5-yl)propan-l-one; 96
(3 S)-3-amino-3 -phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan- 1 -one;
(3S)-3-amino-l-(8-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3S)-3-amino-3-(4-fluorophenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3 S)-3-amino-3-(2-fluorophenyl)- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-(methylamino)-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3 S)-3-(dimethylamino)-3-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-amino-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3-amino-3-(2-methylphenyl)- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-amino-3-(2-methoxyphenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3 S)-3 -amino-3 -(2-chlorophenyl)- 1 -(2,3 ,4,5 -tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one;
(3S)-3-(ethylamino)-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)propan-
1-one;
(3S)-3-[(cyclopropylmethyl)amino]-3-phenyl-l-(2,3,4,5-tetrahydro-l,5- benzoxazepin-5-yl)propan-l -one;
1 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-2-( 1 ,2,3 ,4-tetrahydroisoquinolin- 1 - yl)ethan-l-one;
(3 S)- 1 -(3 -hydroxy-3 -methyl-2,3 ,4,5-tetrahydro- 1 , 5 -benzoxazepin-5 -yl)-3 - (methylamino)-3-phenylpropan-l -one;
N-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]propanamide;
N- [( 1 S)-3 -oxo- 1 -phenyl-3 -(2,3 ,4,5-tetrahydro- 1 , 5 -benzoxazepin-5- yl)propyl]cyclopropanecarboxamide;
2-methyl-N-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]propanamide; 97
2,2,2-trifluoro-N-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]acetamide;
N-[(lS)-l-(2-methylphenyl)-3-oxo-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]acetamide;
N-[(lS)-l-(2-chlorophenyl)-3-oxo-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]acetamide;
l-[(lS)-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propyl]pyrrolidin-2-one;
(3S)-3-arrdno-l-(6-fluoro-2,3,4,5-tetrahydxo-l,5-berizoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3-amino- 1 -(9-fluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 -(4- fluorophenyl)propan- 1 -one;
l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-2-(l,2,3,4- tetrahydroisoquinolin- 1 -yl)ethan- 1 -one;
(3 S)-3 -amino- 1 -(9-fluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 -(2- fluorophenyl)propan- 1 -one;
(3 S)-3-[(2,2-difluoroethyl)amino] -3 -phenyl- 1 -(2,3 ,4,5-tetrahydro- 1,5- benzoxazepin-5-yl)propan- 1 -one;
(3S)-3-amino-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan- 1 -one;
(3S)-l-(9-fluoro-2,3,4,5-tetrahya^o-l,5-benzoxazepin-5-yl)-3-(methylamino)-3- phenylpropan- 1 -one;
tert-butyl N-[2,2-difluoro-3-oxo-l-phenyl-3-(2,3,4,5-tetrahydro-l,5-benzoxazepin- 5-yl)propyl]carbamate;
3- amino-2,2-difluoro-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3 S)-3 -amino- 1 -(3 -methoxy-2,3 ,4,5 -tetrahydro- 1 ,5 -benzoxazepin-5 -yl)-3 - phenylpropan- 1 -one;
(3S)-3-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)butan-l-one;
l-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan- 1 -one;
4- phenyl- 1 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)pentan- 1 -one;
3-(2-methylphenyl)- 1 -(2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)propan- 1 -one; 98
(3R)-3-amino-4-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)butan- 1 -one; (3S)-3-amino-4-phenyl- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)butan-l -one; 4-amino-4-phenyl-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)butan-l-one;
l-(3-hydroxy-3-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- methylphenyl)propan- 1 -one;
1- (2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-2-(l,2,3,4-tetrahydroisoquinolin-4- yl)ethan-l-one;
(3 S)-3-amino-3-(2-chlorophenyl)- 1 -(9-fluoro-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin- 5-yl)propan- 1 -one;
(3 S)-3-amino- 1 -(3 ,3 -difluoro-2,3 ,4,5-tetrahydro- 1 ,5 -benzoxazepin-5-yl)-3 - phenylpropan- 1 -one;
(3S)-3-amino-l-(7-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
2- (2-acetyl- 1 ,2,3,4-tetrahydroisoquinolin-4-yl)- 1 -(2,3 ,4,5-tetrahydro- 1,5- benzoxazepin-5-yl)ethan- 1 -one;
(3S)-3-amino-l-(3-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
2-(2-acetyl- 1 ,2,3 ,4-tetrahydroisoquinolin- 1 -yl)- 1 -(2,3 ,4,5-tetrahydro- 1,5- benzoxazepin-5-yl)ethan- 1 -one;
(3 S)-amino- 1 - { 3 ,4-dihydro-2,4-methanobenzo [b] [ 1 ,4] oxazepin-5 -yl } -3 - phenylpropan- 1 -one
(3S)-3-amino-l-[(4S)-9-fluoro-4-methyl-2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl] -3 -phenylpropan- 1 -one;
(3 S)-3 -amino- 1 -(9-methy 1-2,3 ,4,5-tetrahydro- 1 ,5 -benzoxazepin-5 -yl)-3 - phenylpropan- 1 -one;
(3S)-3-ammo-l-(9-chloro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one;
(3 S)-3-amino- 1 -[(4R)-9-fluoro-4-methyl-2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl] -3 -phenylpropan- 1 -one;
(3 S)-3-amino- 1 - {4,5-dihydro-2H-spiro[l ,5-benzoxazepine-3, 1 '-cyclopropane]-5- yl } -3 -phenylpropan- 1 -one;
(3S)-3-amino-l-(9-methoxy-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan- 1 -one; 99
(3S)-3-amino-3-(2-bromophenyl)-l-(2,3,4,5-tetrahydro-l,5-benzoxazepin-5- yl)propan-l-one;
(3 S)-3-amino-3-(2-cyclopropylphenyl)- 1 -(2,3 ,4,5 -tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one; and enantiomers, diastereoisomers and mixtures thereof; and pharmaceutically acceptable salts of any of the foregoing.
10. A compound of formula (I) as defined in claim 1 selected from:
(3 S)-3-amino-3-(4-fluorophenyl)- 1 -(2,3,4,5-tetrahydro- 1 ,5-benzoxazepin-5- yl)propan-l-one hydrochloride;
(3 S)-3 -amino- 1 -(9-fluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 - phenylpropan- 1 -one;
(3 S)-3 -amino- 1 -(6-fluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 - phenylpropan- 1 -one;
(3 S)-3 -amino- 1 -(9-fluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 -(4- fluorophenyl)propan- 1 -one hydrochloride;
(3S)-3-amino-l-(9-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3-(2- fluorophenyl)propan- 1 -one hydrochloride;
(3 S)-3 -amino- 1 -(3 ,3 -difluoro-2,3 ,4,5-tetrahydro- 1 ,5-benzoxazepin-5-yl)-3 - phenylpropan- 1 -one hydrochloride;
(3S)-3-amino-l-(3-fluoro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride;
(3S)-3-amino-l-(9-chloro-2,3,4,5-tetrahydro-l,5-benzoxazepin-5-yl)-3- phenylpropan-l-one hydrochloride; and
pharmaceutically acceptable salts of any of the foregoing.
1 1. A process for the preparation of a compound of formula (I) or a pharmaceutically acceptable salt thereof as defined in claim 1 which comprises reacting a compound of formula (II), or a salt thereof, 100
Figure imgf000101_0001
in which X1, X2, R1, R2, R3, R4, R5 and R6 are as defined in formula (I) above, with a compound of formula (III), or a salt thereof,
Figure imgf000101_0002
in which n, R7, R8, R9, R10, R11, R12 and R13 are as defined in formula (I) above; and optionally thereafter carrying out one or more of the following procedures:
• converting a compound of formula (I) into another compound of formula (I)
• removing any protecting groups
• forming a pharmaceutically acceptable salt.
12. A pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 10, in association with a pharmaceutically acceptable adjuvant, diluent or carrier, and optionally one or more other therapeutic agents.
13. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 10, for use in therapy. 101
14. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 10, for use in treating alcohol, drug or nicotine addiction.
15. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 10, for use in treating hearing loss or tinnitus.
16. A compound of formula (I) or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 10, for use in treating schizophrenia.
PCT/JP2017/024276 2016-06-23 2017-06-23 Novel compounds WO2017222083A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1610956.3A GB201610956D0 (en) 2016-06-23 2016-06-23 Novel compounds
GB1610956.3 2016-06-23

Publications (1)

Publication Number Publication Date
WO2017222083A1 true WO2017222083A1 (en) 2017-12-28

Family

ID=56891677

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2017/024276 WO2017222083A1 (en) 2016-06-23 2017-06-23 Novel compounds

Country Status (2)

Country Link
GB (1) GB201610956D0 (en)
WO (1) WO2017222083A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2182886A1 (en) * 1972-03-22 1973-12-14 Robins Co Inc A H 5-aminoalkyl-tetrahydro-1,5-benzoxazepines - as cns-depressants from tetrahydrobenzo xazepines via 5-chloroacyl-derivs with amines
EP1408042A1 (en) * 2001-06-14 2004-04-14 Banyu Pharmaceutical Co., Ltd. Novel isoxazolopyridone derivatives and use thereof
WO2007025709A2 (en) * 2005-08-31 2007-03-08 Novartis Ag Organic compounds
WO2015106025A1 (en) * 2014-01-09 2015-07-16 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Substituted benzoxazine and related compounds

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2182886A1 (en) * 1972-03-22 1973-12-14 Robins Co Inc A H 5-aminoalkyl-tetrahydro-1,5-benzoxazepines - as cns-depressants from tetrahydrobenzo xazepines via 5-chloroacyl-derivs with amines
EP1408042A1 (en) * 2001-06-14 2004-04-14 Banyu Pharmaceutical Co., Ltd. Novel isoxazolopyridone derivatives and use thereof
WO2007025709A2 (en) * 2005-08-31 2007-03-08 Novartis Ag Organic compounds
WO2015106025A1 (en) * 2014-01-09 2015-07-16 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Substituted benzoxazine and related compounds

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHRISTINE E. GEE ET AL: "Blocking Metabotropic Glutamate Receptor Subtype 7 (mGlu7) via the Venus Flytrap Domain (VFTD) Inhibits Amygdala Plasticity, Stress, and Anxiety-related Behavior", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 289, no. 16, 4 March 2014 (2014-03-04), US, pages 10975 - 10987, XP055403873, ISSN: 0021-9258, DOI: 10.1074/jbc.M113.542654 *
K NAGARAJAN ET AL: "Synthesis of 10,11-dihydrodibenz[b,f][l,4]oxazepine derivatives as potential anticonvulsant & psychotropic agents", INDIAN JOURNAL OF CHEMISTRY - SECTION B: ORGANIC AND MEDICINAL CHEMISTRY, August 1985 (1985-08-01), pages 840 - 844, XP055403980, Retrieved from the Internet <URL:http://repository.ias.ac.in/93498/1/177p.pdf> [retrieved on 20170905] *

Also Published As

Publication number Publication date
GB201610956D0 (en) 2016-08-10

Similar Documents

Publication Publication Date Title
AU2016219247B2 (en) Fused dihydro-4H-pyrazolo[5,1-C][1,4]oxazinyl compounds and analogs for treating CNS disorders
WO2016001875A1 (en) Indane and indoline derivatives and the use thereof as soluble guanylate cyclase activators
CN103124727A (en) Substituted 3-phenyl-1,2,4-oxadiazole compounds
AU2017347549A1 (en) Therapeutic compounds and methods of use thereof
US20220048884A1 (en) Nrf2 activator
US20210340107A1 (en) Nrf2 activator
WO2018141842A1 (en) 5-(7H-PYRROLO[2,3-d]PYRIMIDIN-4-YL)-5-AZASPIRO[2.5]OCTANE-8-CARBOXYLIC ACID DERIVATIVES AS NOVEL JAK KINASE INHIBITORS
AU2017210803A1 (en) Indane derivatives as mGluR7 modulators
EP3541812B1 (en) Mglur7 agonist compounds for treating mglur7- regulated diseases, disorders, or conditions
WO2017222083A1 (en) Novel compounds
WO2018047983A1 (en) Indane derivatives useful as modulators of mglur7
WO2013128421A1 (en) Spirohydantoin compounds and their use as selective androgen receptor modulators
WO2018079862A1 (en) Mglur7 modulators

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: 17745521

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: 17745521

Country of ref document: EP

Kind code of ref document: A1