Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
OREXIN 1 RECEPTOR ANTAGONISTS
Document Type and Number:
WIPO Patent Application WO/2020/120994
Kind Code:
A1
Abstract:
The disclosures herein relate to novel compounds of formula (I): and salts thereof, wherein W; X; Y; Z; R1; R2; R3 and R4 are defined herein, and their use in treating, preventing, ameliorating, controlling or reducing the risk of neurological or psychiatric disorders associated with orexin receptors.

Inventors:
BROWN GILES ALBERT (GB)
CONGREVE MILES STUART (GB)
CHRISTOPHER JOHN ANDREW (GB)
SWAIN NIGEL ALAN (GB)
BUCKNELL SARAH JOANNE (GB)
STEPHENSON ANNE MARY (GB)
TEHAN BENJAMIN GERALD (GB)
MILLS MARK (GB)
PATEL ANIL (GB)
Application Number:
PCT/GB2019/053557
Publication Date:
June 18, 2020
Filing Date:
December 16, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HEPTARES THERAPEUTICS LTD (GB)
International Classes:
C07D239/26; A61K31/4439; A61K31/497; C07D241/12; C07D241/16; C07D249/06; C07D401/04
Domestic Patent References:
WO2017039331A12017-03-09
Other References:
YU YOSHIDA ET AL: "Discovery of (1 R ,2 S )-2-{[(2,4-Dimethylpyrimidin-5-yl)oxy]methyl}-2-(3-fluorophenyl)- N -(5-fluoropyridin-2-yl)cyclopropanecarboxamide (E2006): A Potent and Efficacious Oral Orexin Receptor Antagonist", JOURNAL OF MEDICINAL CHEMISTRY, vol. 58, no. 11, 11 June 2015 (2015-06-11), US, pages 4648 - 4664, XP055389586, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.5b00217
AINOLEENA TURKU ET AL: "Pharmacophore Model To Discover OX 1 and OX 2 Orexin Receptor Ligands", JOURNAL OF MEDICINAL CHEMISTRY, vol. 59, no. 18, 8 September 2016 (2016-09-08), US, pages 8263 - 8275, XP055670296, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.6b00333
PEYRON ET AL., J. NEUROSCI., vol. 18, 1998, pages 9996 - 10015
ARRIGONI, E ET AL., NEUROPHARMACOLOGY, 2018, Retrieved from the Internet
SAKURAI ET AL., CELL, vol. 92, 1998, pages 573 - 585
TSUJINOSAKURAI, PHARMACOL. REV., vol. 61, 2009, pages 162 - 176
JOHREN ET AL., ENDOCRINOLOGY, vol. 142, 2001, pages 3324 - 3331
THANNICKAL ET AL., SCI. TRANSL. MED., vol. 10, 2018, pages eaao4953
JAMES MH ET AL., BIOLOGICAL PSYCHIATRY, Retrieved from the Internet
BOUTREL ET AL., FRONT. BEHAV. NEUROSCI., vol. 7, 2013, pages 1 - 10
HOLLANDER ET AL., FRONT. BEHAV. NEUROSCI., vol. 6, 2012, pages 1 - 9
MUSCHAMP ET AL., PROC. NATL. ACAD. SCI. U.S.A., 2014, pages e1648 - e1655
BOUTREL ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 102, 2005, pages 19168 - 19173
LAWRENCE ET AL., BR. J. PHARMACOL., vol. 148, 2006, pages 752 - 759
HOLLANDER ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 105, 2008, pages 19480 - 19485
LAORDEN ET AL., PLOS ONE, vol. 7, 2012, pages e36871
AZIZI H ET AL., NEUROSCIENCE LETTERS, vol. 482, 2010, pages 255 - 259
HEYDEND ET AL., PHYSIOL. BEHAV., vol. 130, 2013, pages 182 - 190
BONAVENTURE ET AL., FRONT. PHARMACOL., vol. 8, 2017, pages 1 - 13
JOHNSON ET AL., NAT. MED., vol. 16, 2010, pages 111 - 118
FLORES, TRENDS NEUROSCI., vol. 38, 2015, pages 550 - 559
VALIANTE ET AL., BIOCHEM. BIOPHYS. RES COMM., vol. 464, 2015, pages 1290 - 1296
BRYN ET AL.: "Solid-State Chemistry of Drugs", 1999, SSCI, INC OF WEST LAFAYETTE
M. B. SMITH: "Remington's Pharmaceutical Sciences", 1994, MACK PUBLISHING COMPANY
J. MARCH: "Advanced Organic Chemistry", 1992, JOHN WILEY & SONS
HAVEAUX ET AL., ORG. SYNTH., vol. 59, 1980, pages 26
CORDOVILLA ET AL., ACS CATAL., vol. 5, 2015, pages 3040 - 3053
SWAMY ET AL., CHEM. REV., vol. 109, 2009, pages 2551 - 2561
MIYAURA ET AL., CHEM. REV., vol. 95, 1995, pages 2457 - 2483
CHRISTOPHER ET AL., MEDCHEMCOMM, vol. 6, 2015, pages 947 - 955
WERTZ ET AL., ANGEW. CHEM. INT. ED., vol. 50, 2011, pages 11511 - 11515
Attorney, Agent or Firm:
STRATAGEM INTELLECTUAL PROPERTY MANAGEMENT LIMITED (GB)
Download PDF:
Claims:
Claims

1. A compound of formula (1 )

or a salt thereof, wherein

W is N or CR5;

X is N or CR6;

Y is CH2;

Z is CR7 or N;

Ri is H or a halogen;

R2 is a 5 or 6.membered heteroaryl ring containing two or three nitrogen atoms which is optionally substituted with one or more fluorine atoms;

R3 is H or a halogen;

R4 is H or a halogen;

R5 is selected from H; halogen; C1 -C3 alkyl optionally substituted with OMe or one or more halogen groups; and C1 -C3 alkoxy optionally substituted with one or more halogen groups;

R6 is selected from H; halogen; C1 -C3 alkyl optionally substituted with OMe or one or more halogen groups; and C1 -C3 alkoxy, optionally substituted with one or more halogen groups; and

Ry is H or a halogen.

2. The compound according to claim 1 according to the formula (2)

3. The compound according to claim 1 or claim 2 wherein X is CR6.

4. The compound according to claim 3 wherein W is N.

5. The compound according to claim 3 or claim 4 wherein R6 is selected from H, F, OMe, OCHF2, OCD3 or CH2OMe.

6. The compound according to claim 1 or claim 2 wherein W is CR5.

7. The compound according to claim 6 wherein X is N.

8. The compound according to claim 6 or claim 7 wherein R5 is H, Me, CH2OMe or OMe.

9. The compound according to any one of claims 1-8 wherein Z is CH.

10. The compound according to any one of claims 1-9 wherein Ri is H.

1 1. The compound according to any one of claims 1-10 wherein R2 is a 1 ,2,3- triazole ring optionally substituted with one or more fluorine atoms; a pyrimidine ring optionally substituted with one or more fluorine atoms; or a pyrazine ring optionally substituted with one or more fluorine atoms.

12. The compound according to claim 1 1 wherein R2 is selected from the group consisting of:

13. The compound according to claim 12 wherein R2 is

14. The compound according to any one of claims 1-13 wherein R3 is H.

15. The compound according to any one of claims 1 -14 wherein R4 is F.

16. The compound according to claim 1 or claim 2 which is selected from the group consisting of:

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(triazol-2-yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-5-methyl-2-pyrimidin-2-yl- benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-5-methyl-2-(triazol-2- yl)benzamide;

2-fluoro-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-(triazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-(triazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-2-methoxy-6-(triazol-2- yl)benzamide;

N-[(1 S,2S)-2-[dideuterio-(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-

(triazol-2-yl)benzamide;

2-(difluoromethoxy)-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-

(triazol-2-yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-pyrimidin-2- yl-benzamide;

2-fluoro-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-pyrimidin-2-yl- benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-pyrazin-2-yl- benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-pyrimidin-4- yl-benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(5-fluoropyrimidin-2-yl)-

6-methoxy-benzamide; N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(triazol-2-yl)-6-

(trideuteriomethoxy)benzamide;

2-(difluoromethoxy)-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6- pyrimidin-2-yl-benzamide;

3-fluoro-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methoxy-2- (triazol-2-yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(methoxymethyl)-6-

(triazol-2-yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-(methoxymethyl)-3-

(triazol-2-yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-6-(methoxymethyl)-3-

(triazol-2-yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methoxy-3-(triazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-6-methoxy-3-(triazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methyl-3-(triazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-6-methyl-3-(triazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methyl-3-pyrimidin-2-yl- pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-3-pyrimidin-2-yl-pyridine-

2-carboxamide;

N-[(1 S,2S)-2-[(2,4-difluorophenoxy)methyl]cyclopentyl]-3-pyrimidin-2-yl- pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-3-pyrimidin-2-yl-pyridine-

2-carboxamide;

N-[(1 S,2S)-2-[(3,4-difluorophenoxy)methyl]cyclopentyl]-3-pyrimidin-2-yl- pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-4-(triazol-2- yl)pyridine-3-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-4-pyrimidin-2- yl-pyridine-3-carboxamide; N-[(1 S,2S)-2-[(5-fluoro-2-pyridyl)oxymethyl]cyclopentyl]-2-methoxy-6-(triazol-

2-yl)benzamide;

N-[(1 S,2S)-2-[(5-fluoro-2-pyridyl)oxymethyl]cyclopentyl]-2-methoxy-6- pyrimidin-2-yl-benzamide;

and

N-[(1 S,2S)-2-[(5-fluoro-2-pyridyl)oxymethyl]cyclopentyl]-2-(triazol-2-yl)-6-

(trideuteriomethoxy)benzamide.

17. A pharmaceutically acceptable salt of a compound according to any one of claims 1-16.

18. A pharmaceutical composition comprising the compound according to any one of claims 1-17 and a pharmaceutically acceptable excipient.

19. The compound according to any one of claims 1-17 for use as an antagonist of the orexin OXi receptor.

20. The compound according to any one of claims 1-17 for use in treating, preventing, ameliorating, controlling or reducing the risk of neurological or psychiatric disorders.

21. The compound according to any one of claims 1-17 for use in the treatment or prevention of substance related and addictive disorders (including opioid use disorder, opioid intoxication, opioid withdrawal, other opioid induced disorder, unspecified opioid related disorder, stimulant use disorder, stimulant intoxication, stimulant withdrawal, other stimulant induced disorders, unspecified stimulant related disorder, caffeine related disorders, caffeine intoxication, caffeine withdrawal, unspecified caffeine related disorders, tobacco related disorders, tobacco use disorder, tobacco withdrawal, other tobacco induced disorders, unspecified tobacco related disorder, alcohol use disorder, alcohol intoxication, alcohol withdrawal, unspecified alcohol related disorder, cannabis related disorders, cannabis use disorder, cannabis intoxication, cannabis withdrawal, unspecified cannabis related disorders, hallucinogen related disorders, phencyclidine use disorder, phencyclidine intoxication, other hallucinogen use disorder, hallucinogen persisting perception disorder, unspecified hallucinogen related disorder, inhalant related disorders, inhalant use disorder, inhalant intoxication, other inhalant induced disorders, unspecified inhalant related disorder, sedative, hypnotic or anxiolytic related disorders (including use disorder, intoxication and withdrawal), gambling disorder, internet gaming disorder, addiction to sex or internet use), anxiety disorders (including separation anxiety disorder, specific phobia, social anxiety disorder (social phobia), panic disorder, agoraphobia, generalized anxiety disorder, substance/medication induced anxiety disorder, anxiety disorder due to another medical condition), disruptive mood dysregulation disorder, major depressive disorder (including when specified with anxious distress, mixed features, atypical features, peripartum onset or seasonal pattern), persistent depressive disorder (dysthymia) (including when specified with anxious distress, mixed features, atypical features, peripartum onset or seasonal pattern), premenstrual dysphoric disorder, substance/medication-induced depressive disorder, other specified depressive disorder, unspecified depressive disorder bipolar and related disorders (including bipolar I disorder and bipolar II disorder, particularly, but not exclusively, when these are specified with anxious distress, cyclothymic disorder, substance/medication-induced bipolar and related disorder or bipolar and related disorder due to another medical condition), schizophrenia spectrum and other disorders (including schizotypal personality, delusional disorder, schizophreniform disorder, schizophrenia, schizoaffective disorder, and substance /medication-induced psychotic disorder), conditions associated with trauma and stress (including post traumatic stress disorder, acute stress disorder, adjustment disorders (including when specified with anxiety or with mixed anxiety and depressed mood), obsessive compulsive and related disorders (including obsessive compulsive disorder, body dysmorphia, trichlotillomania, excoriation and obsessive-compulsive and related disorders due to another medical condition), feeding and eating disorders (including binge eating disorder, anorexia nervosa, bulimia nervosa, cachexia, obesity, Prader Willi syndrome)), sleep-wake disorders, neurodegenerative disorders (including dementia), behavioural symptoms of neurodegenerative and other disorders, movement disorders, diabetes, impaired glucose tolerance, cardiovascular disease, diseases related to modulation of sympathetic outflow including hypertension, hypothalamic/pituitary disorders, neuropathic pain, restless leg syndrome, migraine, cluster headache, tension-type headache, trigeminal autonomic cephalalgias, hemicrania continua, trigeminal neuralgia, other headache disorders, hyperalgesia, pain, hyperalgesia, causalgia, and allodynia, acute pain, burn pain, atypical facial pain, back pain, complex regional pain syndrome I and II, arthritic pain, sports injury pain, pain related to infection, irritable bowel syndrome, angina pain, inflammatory disorders, renal/urinary disorders, respiratory disorders, cancer (including prostate cancer, liver cancer, colon cancer, endometrial cancer, pancreatic cancer and cancers associated with other organs of the body including the central nervous system and peripheral nervous system).

22. The compound according to any one of claims 1-17 for use in the treatment or prevention of substance related and addictive disorders, post traumatic stress disorder, panic disorder, major depressive disorder with anxious distress, diseases related to modulation of sympathetic outflow including hypertension, pain, headache, cancer.

Description:
OREXIN 1 RECEPTOR ANTAGONISTS

This application relates to novel compounds and their use as orexin receptor antagonists. Compounds described herein may be useful in the treatment or prevention of diseases in which orexin receptors are involved. The application is also directed to pharmaceutical compositions comprising these compounds and the manufacture and use of these compounds and compositions in the prevention or treatment of such diseases in which orexin receptors are involved.

BACKGROUND OF THE INVENTION

Neuroanatomical, immunohistochemical and pharmacological evidence suggests that the orexins play important roles in the regulation of a wide variety of physiological functions and complex behaviours. Orexinergic ( aka hypocretinergic) neurones are located in the lateral hypothalamus and perifornical area of the mammalian mid-brain and from here they project widely throughout the central nervous system and spinal cord via ascending and descending pathways (Peyron et a\, J. Neurosci., 1998, 18, 9996-10015). Orexinergic neurons project to forebrain structures involved in cognition and emotion, such as the cortex, amygdala, hippocampus and septum. In addition, they strongly target ascending neuronal systems involved in the control of arousal, movement, sleep wake cycles and reward processing, notably the noradrenergic locus coeruleus, serotonergic Raphe nuclei, dopaminergic ventral tegmental area and Substantia nigra, cholinergic

Pedunculopontine nuclei and histaminergic Tuberomammillary bodies. Similarly, orexinergic neurons project to brain areas involved in the control of feeding in the hypothalamus such as MCH, NPY and leptin positive neurons in the arcuate nucleus (Arrigoni, E et al 2018, Neuropharmacology doi: https://doi.Org/10.1016/j.neuropharm.2018.1 1.017). Orexinergic innervation of the dorsal horn of the spinal cord has been reported and evidence for the presence of orexin in peripheral tissues including plasma, sympathetic ganglia, myenteric plexus, endocrine cells of the gastrointestinal tract, adrenal gland, pancreas, placenta, testes, pituitary, kidney, adrenal gland, thyroid and lung amongst others suggest important roles in the regulation of a wide variety of physiological functions. The orexin peptides orexin A and orexin B are produced in the hypothalamus and exert their effect through activation of the orexin receptors: orexin 1 (OXiR) and orexin 2 (OX2R). OX1 R and OX2R share 64 % amino acid identity. Orexin A and B have equal affinity for OX2R receptors whilst the affinity of orexin B is around one order of magnitude lower at the OX1R receptor (Sakurai et al, Cell , 1998, 92, 573- 585). The orexin receptors are predominately linked to G q/ n G proteins with activation of the receptors leading to activation of the phospholipase C pathway triggering influx of extracellular sodium and calcium and depolarisation of neurones.

Whilst OX1 R and OX 2 R show some overlapping distribution, they largely show distinct (but complementary) patterns of distribution within the central nervous system which suggests that each receptor subtype plays different roles in controlling physiological function. Outside of the hypothalamus in the rat brain the highest levels of OX1 R are found in the locus coreleus, tenia tecta, hippocampal formation, dorsal raphe nucleus - areas which have been linked to anxiety, arousal, stress and panic disorders and reward-seeking behaviour. OX2R is found in the cerebral cortex, nucleus accumbens, subthalamic nucleus, paraventricular thalamic nuclei, anterior pretectal nucleus raphe nuclei and tuberomammillary bodies (Tsujino and Sakurai, Pharmacol. Rev., 2009, 61, 162-176). Orexin receptors are also expressed in peripheral tissues, for example OX1 R can be found in the kidney, testis, thyroid, ovaries, jejunum and adrenal glands (Johren et al, Endocrinology, 2001 , 142, 3324- 3331 ).

Post mortem studies of human brains and the brains of rats trained to self-administer cocaine show functional upregulation of the orexin system. In human post mortem brain 54% more immunohistochemically detected neurons producing orexin were found in the brains of heroin addicts compared to brains from neurologically normal subjects. Similar increases in orexin producing cells could be induced in wild-type mice by administration of morphine (Thannickal et al., 2018 Sci. Transl. Med. 10, eaao4953 2018). Furthermore, it has been shown that self-administation of cocaine by rats was accompanied by an increase in number and activity of orexin-expressing neurons within the lateral hypothalamus, an increase which persisted during protracted withdrawal from cocaine. (James MH, et al, Biological Psychiatry, https://doi.Org/10.1016/i.biopsych. 2018.07.022). Taken together these data suggest that an upregulation of orexinergic neuronal activity is associated with drug abuse

There is substantial preclinical evidence supporting the hypothesis that OXi R receptor antagonists are efficacious in preclinical models of drug abuse (Boutrel et al, Front. Behav. Neurosci., 2013, 7, 1-10). Intravenous self-administration of cocaine is attenuated in OXiR knock out mice (Hollander et at, Front. Behav. Neurosci., 2012, 6, 1-9; Muschamp et al, Proc. Natl. Acad. Sci. U.S.A., 2014, e1648- e1655). Intracerebroventricular administration of orexin A leads to a dose-related reinstatement of cocaine seeking and dramatically elevates intracranial self stimulation thresholds in rodent self-administration studies (Boutrel et al, Proc. Natl. Acad. Sci. U.S.A., 2005, 102, 19168-19173). Administration of a partially selective OXiR antagonist SB-334867 decreases alcohol (Lawrence et al, Br. J. Pharmacol., 2006, 148, 752-759) and nicotine (Hollander et al, Proc. Natl. Acad. Sci. U.S.A., 2008, 105, 19480-19485) self-administration in rats. In rats intraperitoneal administration of SB-334867 has also been demonstrated to significantly reduce opiate withdrawal symptoms (Laorden et al, PLoS One, 2012, 7, e36871 ; Azizi H, et al. Neuroscience Letters, 2010, 482, 255-259).

Selective OXi receptor antagonists have potential utility therefore to treat a number of Substance Related and Addictive Disorders such as Opioid Use Disorder (including but not limited to Opioid Use Disorder, Opioid Intoxication, Opioid withdrawal, Other Opioid Induced Disorder and Unspecified Opioid Related Disorder): Stimulant Related disorders (including but not limited to Stimulant Use Disorder, Stimulant Intoxication, Stimulant Withdrawal, Other Stimulant Induced Disorders and Unspecified Stimulant Related Disorder where such disorders are associated with the abuse of stimulant drugs exemplified but not limited to cocaine or related structures and, amphetamine-like substances): Caffeine Related Disorders (including but not limited to Caffeine intoxication, Caffeine withdrawal, and Unspecified Caffeine Related disorders): Tobacco Related Disorders (including but not limited to Tobacco Use Disorder, Tobacco Withdrawal, Other Tobacco Induced Disorders and Unspecified Tobacco Related Disorder where such disorders are associated with the use of Tobacco, Tobacco products or the inhalation of nicotine and related compounds): Alcohol Use Disorder, Alcohol Intoxication, Alcohol Withdrawal and Unspecified Alcohol Related Disorder: Cannabis Related Disorders (including but not limited to Cannabis Use Disorder, Cannabis Intoxication, Cannabis Withdrawal, and Unspecified Cannabis Related Disorders whether associated with the use of cannabis, cannabis extracts or synthetic cannabinoids): Hallucinogen Related Disorders (including but not limited to Phencyclidine Use Disorder, Phencyclidine intoxication, Other Hallucinogen Use Disorder, Hallucinogen Persisting Perception Disorder and Unspecified Hallucinogen Related Disorder): Inhalant Related Disorders (including but not limited to Inhalant Use Disorder, Inhalant Intoxication, Other Inhalant Induced Disorders and Unspecified Inhalant Related Disorder where such disorders are associated with the use of compounds such as volatile hydrocarbons, nicotine or nicotine derivatives): Sedative, Hypnotic or Anxiolytic Related Disorders (including but not limited to Use Disorder, Intoxication and Withdrawal). Selective OXi receptor antagonists also have potential utility to treat a number of Non-Substance Related Disorders such as Gambling Disorder, Internet Gaming Disorder or addiction to sex or internet use.

There is emerging evidence of the orexin systems involvement in anxiety, panic and fear associated learning. Orexin neurones in perifornical and lateral hypothalamic areas are highly reactive to anxiogenic stimuli and optogenetic stimulation of orexin neurons in rodents increases anxiety-like states (Heydend et al, Physiol. Behav., 2013, 130, 182-190). Using two models of panic (sodium lactate infusion and C0 2 challenge models) blockade of OXi R with the highly selective antagonist JNJ- 54717793 attenuated panic-like behaviours and cardiovascular responses in rats without altering baseline locomotor or autonomic activity and without sedation (Bonaventure et al, Front. Pharmacol., 2017, 8, 1-13). In a study of 53 medication- free patients with suicidal behaviour increased orexin levels were observed in the cerebral spinal fluid versus control (Johnson et al, Nat. Med., 2010, 16, 1 11-1 18) suggesting a link with orexin levels and anxiety.

Selective OXi receptor antagonists therefore have potential utility to treat Anxiety Disorders (including but not limited to Separation Anxiety Disorder, Specific Phobia, Social Anxiety Disorder (Social Phobia), Panic Disorder, Agoraphobia, Generalized Anxiety Disorder, Substance/Medication Induced Anxiety Disorder and Anxiety Disorder due to Another Medical Condition): Disruptive Mood Dysregulation Disorder, Major Depressive Disorder particularly, but not exclusively, when specified with Anxious Distress, mixed features, atypical features, peripartum onset or seasonal pattern: Persistent Depressive Disorder (Dysthymia) particularly, but not exclusively, when specified with Anxious Distress, mixed features, atypical features, peripartum onset or seasonal pattern: Premenstrual Dysphoric Disorder, Substance/Medication-Induced Depressive Disorder, Other Specified Depressive Disorder or Unspecified Depressive Disorder. Similarly, OXi receptor antagonists have potential utility to treat the symptoms of Bipolar and Related Disorders (including but not restricted to Bipolar I Disorder and Bipolar II Disorder, particularly, but not exclusively, when these are specified with Anxious Distress, Cyclothymic Disorder, Substance/Medication-induced Bipolar and Related Disorder or Bipolar and Related Disorder due to Another Medical Condition): Schizophrenia Spectrum and other disorders (including but not limited to Schizotypal Personality, Delusional Disorder, Schizophreniform Disorder, Schizophrenia, Schizoaffective Disorder, and Substance /Medication-Induced Psychotic Disorder).

It has been shown that orexin plays a role in mediating the extinction of fear in animal models of stress and trauma (Flores et at, Trends Neurosci., 2015, 38, 550- 559). OXi receptor antagonists therefore have potential utility to treat conditions associated with trauma and stress including but not limited to Post Traumatic Stress Disorder, Acute Stress Disorder, Adjustment Disorders particularly, but not exclusively, when specified with Anxiety or with Mixed Anxiety and Depressed Mood: Obsessive Compulsive and Related Disorders including but not limited to Obsessive Compulsive Disorder, Body Dysmorphia, Trichlotillomania, Excoriation and Obsessive Compulsive and Related Disorders due to another medical condition: Feeding and Eating Disorders including but not limited to Binge Eating Disorder, Anorexia Nervosa, Bulimia Nervosa, Cachexia, Obesity and Prader Willi syndrome.

Some Sleep-Wake Disorders may also be treated with selective OXi receptor antagonists and these disorders include but are not limited to Insomnia Disorder, Rapid Eye Movement Disorder, Sleep disturbances associated with diseases, Sleep Apnoea, Narcolepsy and Circadian Rhythm Sleep-Wake Disorders. Similarly, Neurodegenerative disorders (including but not limited to Parkinson’s Disease, Alzheimer’s Disease, Dementia, Lewy-body Dementia, Frontotemporal Dementia, Multiple System Atrophy, Perry Syndrome, Klein-Levin Syndrome, Amyotrophic Lateral Sclerosis, Niemann-Pick disease and Multiple Sclerosis): Behavioural Symptoms of neurodegenerative and other disorders (such as Positive Symptoms, Psychosis, Agitation, Anhedonia, Apathy): Movement disorders (such as Akinesias, Dyskinesia, Drug-induced Parkinsonism, Dystonia): other CNS disorders (such as Affective Neurosis, Delirium, Sexual Dysfunction, Psychosexual Dysfunction, Rett Syndrome, Attention-Deficit Disorder, Attention-Deficit Hyperactivity Disorder, Autism (including but not limited to autism spectrum disorder, autism spectrum condition, atypical autism, classic autism, Kanner autism, pervasive developmental disorder, high-functioning autism and Asperger syndrome), Fragile X syndrome, Disruptive behaviour disorder, Severe mental retardation, Vomiting.

Pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass may be amenable to treatment with selective OXi receptor antagonists, such conditions include, but are not limited to Diabetes, impaired glucose tolerance; Cardiovascular Disease including but not limited to Acute and Congestive Heart Failure, Hypotension, Hypertension, Angina Pectoris, Myocardial infarction.

Hypothalamic/pituitary disorders may be amenable to treatment with selective OXi receptor antagonists, including but not limited to Cushing's Syndrome/disease, Basophile adenoma, Prolactinoma, hyperprolactinemia, Hypophysis Tumour/adenoma, Hypothalamic diseases, Froehlich's syndrome, Adrenohypophysis disease, Hypophysis disease, Adrenohypophysis hypofunction, Adrenohypophysis hyperfunction, Hypothalamic hypogonadism, Kallman's syndrome (anosmia, hyposmia), functional or psychogenic Amenorrhea, Hypopituitarism, Hypothalamic Hypothyroidism, Hypothalamic-adrenal dysfunction, Idiopathic Hyperprolactinemia, Hypothalamic Disorders of growth hormone deficiency, Idiopathic growth deficiency, Dwarfism, Gigantism, Acromegaly.

Pain disorders may be amenable to treatment with selective OXi receptor antagonists, including but not limited to Neuropathic pain, Restless Leg Syndrome, Migraine, Cluster headache, Tension-type headache, Trigeminal autonomic Cephalalgias, Hemicrania Continua, Trigeminal neuralgia, other headache disorders, pain, enhanced or exaggerated sensitivity to pain such as Hyperalgesia, Causalgia, and Allodynia, Acute pain, Burn pain, Atypical facial pain, Back pain, Complex regional pain syndrome I and II, Arthritic pain, Sports injury pain, pain related to infection e.g. HIV, Post-chemotherapy pain, Post-stroke pain, Post-operative pain, neuralgia, conditions associated with Visceral pain such as Irritable bowel syndrome, and Angina pain; Inflammatory disorders (including Inflammatory bowel disease), Renal/urinary disorders (including urinary retention, benign prostatic hypertrophy, chronic renal failure, renal disease); Respiratory disorders (such as Chronic obstructive pulmonary disease, Asthma).

Orexin-A/OXi receptor interactions have been shown to modulate activity of the androgen receptor (Valiante et al, Biochem. Biophys. Res Comm., 2015, 464, 1290- 1296) which regulates the onset and progression of prostate cancer, suggesting that selective OXi receptor antagonists have potential therapeutic benefit in treating some forms of cancer including, but not limited to, Prostate cancer, Liver Cancer, Colon cancer, Endometrial cancer, Pancreatic cancer and cancers associated with other organs of the body including the central nervous system and peripheral nervous system. SUMMARY OF THE INVENTION

The invention relates to novel compounds. The invention also relates to the use of novel compounds as antagonists of orexin receptor OXi. The invention further relates to the use of novel compounds in the manufacture of medicaments for use as orexin receptor OXi antagonists or for the treatment of orexin system dysfunction. The invention further relates to compounds, compositions and medicaments for the treatment of neurological or psychiatric disorders. Embodiments of the invention may be compounds of the formula (1 ): or a salt thereof, wherein

W is N or CR 5 ;

X is N or CR 6 ;

Y is CH 2 ;

Z is CR7 or N;

Ri is H or a halogen;

R 2 is a 5 or 6.membered heteroaryl ring containing two or three nitrogen atoms which is optionally substituted with one or more fluorine atoms;

R 3 is H or a halogen;

R 4 is H or a halogen;

R 5 is selected from H; halogen; C1 -C3 alkyl optionally substituted with OMe or one or more halogen groups; and C1 -C3 alkoxy optionally substituted with one or more halogen groups;

R 6 is selected from H; halogen; C1-C3 alkyl optionally substituted with OMe or one or more halogen groups; and C1 -C3 alkoxy, optionally substituted with one or more halogen groups; and

Ry is H or a halogen.

The compounds may be for use as antagonists of orexin receptor OX1. The compounds may be used in the manufacture of medicaments. The compounds or medicaments may be for use in treating, preventing, ameliorating, controlling or reducing the risk of neurological or psychiatric disorders.

The compounds or medicaments may be for use in the treatment or prevention of substance related and addictive disorders (including opioid use disorder, opioid intoxication, opioid withdrawal, other opioid induced disorder, unspecified opioid related disorder, stimulant use disorder, stimulant intoxication, stimulant withdrawal, other stimulant induced disorders, unspecified stimulant related disorder, caffeine related disorders, caffeine intoxication, caffeine withdrawal, unspecified caffeine related disorders, tobacco related disorders, tobacco use disorder, tobacco withdrawal, other tobacco induced disorders, unspecified tobacco related disorder, alcohol use disorder, alcohol intoxication, alcohol withdrawal, unspecified alcohol related disorder, cannabis related disorders, cannabis use disorder, cannabis intoxication, cannabis withdrawal, unspecified cannabis related disorders, hallucinogen related disorders, phencyclidine use disorder, phencyclidine intoxication, other hallucinogen use disorder, hallucinogen persisting perception disorder, unspecified hallucinogen related disorder, inhalant related disorders, inhalant use disorder, inhalant intoxication, other inhalant induced disorders, unspecified inhalant related disorder, sedative, hypnotic or anxiolytic related disorders (including use disorder, intoxication and withdrawal), gambling disorder, internet gaming disorder, addiction to sex or internet use), anxiety disorders (including separation anxiety disorder, specific phobia, social anxiety disorder (social phobia), panic disorder, agoraphobia, generalized anxiety disorder, substance/medication induced anxiety disorder, anxiety disorder due to another medical condition), disruptive mood dysregulation disorder, major depressive disorder (including when specified with anxious distress, mixed features, atypical features, peripartum onset or seasonal pattern), persistent depressive disorder (dysthymia) (including when specified with anxious distress, mixed features, atypical features, peripartum onset or seasonal pattern), premenstrual dysphoric disorder, substance/medication-induced depressive disorder, other specified depressive disorder, unspecified depressive disorder bipolar and related disorders (including bipolar I disorder and bipolar II disorder, particularly, but not exclusively, when these are specified with anxious distress, cyclothymic disorder, substance/medication- induced bipolar and related disorder or bipolar and related disorder due to another medical condition), schizophrenia spectrum and other disorders (including schizotypal personality, delusional disorder, schizophreniform disorder, schizophrenia, schizoaffective disorder, and substance /medication-induced psychotic disorder), conditions associated with trauma and stress (including post traumatic stress disorder, acute stress disorder, adjustment disorders (including when specified with anxiety or with mixed anxiety and depressed mood), obsessive compulsive and related disorders (including obsessive compulsive disorder, body dysmorphia, trichlotillomania, excoriation and obsessive-compulsive and related disorders due to another medical condition), feeding and eating disorders (including binge eating disorder, anorexia nervosa, bulimia nervosa, cachexia, obesity, Prader Willi syndrome)), sleep-wake disorders, neurodegenerative disorders (including dementia), behavioural symptoms of neurodegenerative and other disorders, movement disorders, diabetes, impaired glucose tolerance, cardiovascular disease, diseases related to modulation of sympathetic outflow including hypertension, hypothalamic/pituitary disorders, neuropathic pain, restless leg syndrome, migraine, cluster headache, tension-type headache, trigeminal autonomic cephalalgias, hemicrania continua, trigeminal neuralgia, other headache disorders, hyperalgesia, pain, hyperalgesia, causalgia, and allodynia, acute pain, burn pain, atypical facial pain, back pain, complex regional pain syndrome I and II, arthritic pain, sports injury pain, pain related to infection, irritable bowel syndrome, angina pain, inflammatory disorders, renal/urinary disorders, respiratory disorders, cancer (including prostate cancer, liver cancer, colon cancer, endometrial cancer, pancreatic cancer and cancers associated with other organs of the body including the central nervous system and peripheral nervous system).

The compounds or medicaments may be for use in the treatment or prevention of substance related and addictive disorders, post traumatic stress disorder, panic disorder, major depressive disorder with anxious distress, diseases related to modulation of sympathetic outflow including hypertension, pain, headache, cancer.

The compounds may be formulated as a pharmaceutical composition comprising the compounds and a pharmaceutically acceptable excipient.

The compounds may be produced using any chemical synthesis method.

DETAILED DESCRIPTION OF THE INVENTION

The invention relates to novel compounds. The invention also relates to the use of novel compounds as antagonists of orexin receptor OXi. The invention further relates to the use of novel compounds in the manufacture of medicaments for use as orexin receptor OXi antagonists or for the treatment of orexin system dysfunction. The invention further relates to compounds, compositions and medicaments which are selective OXi receptor antagonists.

The invention further relates to compounds, compositions and medicaments for the treatment of Substance Related and Addictive Disorders such as Opioid Use Disorder (including but not limited to Opioid Use Disorder, Opioid Intoxication, Opioid withdrawal, Other Opioid Induced Disorder and Unspecified Opioid Related Disorder): Stimulant Related disorders (including but not limited to Stimulant Use Disorder, Stimulant Intoxication, Stimulant Withdrawal, Other Stimulant Induced Disorders and Unspecified Stimulant Related Disorder where such disorders are associated with the abuse of stimulant drugs exemplified but not limited to cocaine or related structures and, amphetamine-like substances): Caffeine Related Disorders (including but not limited to Caffeine intoxication, Caffeine withdrawal, and Unspecified Caffeine Related disorders): Tobacco Related Disorders (including but not limited to Tobacco Use Disorder, Tobacco Withdrawal, Other Tobacco Induced Disorders and Unspecified Tobacco Related Disorder where such disorders are associated with the use of Tobacco, Tobacco products or the inhalation of nicotine and related compounds): Alcohol Use Disorder, Alcohol Intoxication, Alcohol Withdrawal and Unspecified Alcohol Related Disorder: Cannabis Related Disorders (including but not limited to Cannabis Use Disorder, Cannabis Intoxication, Cannabis Withdrawal, and Unspecified Cannabis Related Disorders whether associated with the use of cannabis, cannabis extracts or synthetic cannabinoids): Hallucinogen Related Disorders (including but not limited to Phencyclidine Use Disorder, Phencyclidine intoxication, Other Hallucinogen Use Disorder, Hallucinogen Persisting Perception Disorder and Unspecified Hallucinogen Related Disorder): Inhalant Related Disorders (including but not limited to Inhalant Use Disorder, Inhalant Intoxication, Other Inhalant Induced Disorders and Unspecified Inhalant Related Disorder where such disorders are associated with the use of compounds such as volatile hydrocarbons, nicotine or nicotine derivatives): Sedative, Hypnotic or Anxiolytic Related Disorders (including but not limited to Use Disorder, Intoxication and Withdrawal). Selective OXi receptor antagonists also have potential utility to treat a number of Non-Substance Related Disorders such as Gambling Disorder, Internet Gaming Disorder or addiction to sex or internet use. The invention further relates to compounds, compositions and medicaments for the treatment of Anxiety Disorders (including but not limited to Separation Anxiety Disorder, Specific Phobia, Social Anxiety Disorder (Social Phobia), Panic Disorder, Agoraphobia, Generalized Anxiety Disorder, Substance/Medication Induced Anxiety Disorder and Anxiety Disorder due to Another Medical Condition): Disruptive Mood Dysregulation Disorder, Major Depressive Disorder particularly, but not exclusively, when specified with Anxious Distress, mixed features, atypical features, peripartum onset or seasonal pattern: Persistent Depressive Disorder (Dysthymia) particularly, but not exclusively, when specified with Anxious Distress, mixed features, atypical features, peripartum onset or seasonal pattern: Premenstrual Dysphoric Disorder, Substance/Medication-Induced Depressive Disorder, Other Specified Depressive Disorder or Unspecified Depressive Disorder. Similarly, OXi receptor antagonists have potential utility to treat the symptoms of Bipolar and Related Disorders (including but not restricted to Bipolar I Disorder and Bipolar II Disorder, particularly, but not exclusively, when these are specified with Anxious Distress, Cyclothymic Disorder, Substance/Medication-induced Bipolar and Related Disorder or Bipolar and Related Disorder due to Another Medical Condition): Schizophrenia Spectrum and other disorders (including but not limited to Schizotypal Personality, Delusional Disorder, Schizophreniform Disorder, Schizophrenia, Schizoaffective Disorder, and Substance /Medication-Induced Psychotic Disorder).

The invention further relates to compounds, compositions and medicaments for the treatment of conditions associated with trauma and stress including but not limited to Post Traumatic Stress Disorder, Acute Stress Disorder, Adjustment Disorders particularly, but not exclusively, when specified with Anxiety or with Mixed Anxiety and Depressed Mood: Obsessive Compulsive and Related Disorders including but not limited to Obsessive Compulsive Disorder, Body Dysmorphia, Trichlotillomania, Excoriation and Obsessive Compulsive and Related Disorders due to another medical condition: Feeding and Eating Disorders including but not limited to Binge Eating Disorder, Anorexia Nervosa, Bulimia Nervosa, Cachexia, Obesity and Prader Willi syndrome.

The invention further relates to compounds, compositions and medicaments for the treatment of Sleep-Wake Disorders. These disorders include but are not limited to Insomnia Disorder, Rapid Eye Movement Disorder, Sleep disturbances associated with diseases, Sleep Apnoea, Narcolepsy and Circadian Rhythm Sleep-Wake Disorders. Similarly, Neurodegenerative disorders (including but not limited to Parkinson’s Disease, Alzheimer’s Disease, Dementia, Lewy-body Dementia, Frontotemporal Dementia, Multiple System Atrophy, Perry Syndrome, Klein-Levin Syndrome, Amyotrophic Lateral Sclerosis, Niemann-Pick disease and Multiple Sclerosis): Behavioural Symptoms of neurodegenerative and other disorders (such as Positive Symptoms, Psychosis, Agitation, Anhedonia, Apathy): Movement disorders (such as Akinesias, Dyskinesia, Drug-induced Parkinsonism, Dystonia): other CNS disorders (such as Affective Neurosis, Delirium, Sexual Dysfunction, Psychosexual Dysfunction, Rett Syndrome, Attention-Deficit Disorder, Attention- Deficit Hyperactivity Disorder, Autism (including but not limited to autism spectrum disorder, autism spectrum condition, atypical autism, classic autism, Kanner autism, pervasive developmental disorder, high-functioning autism and Asperger syndrome), Fragile X syndrome, Disruptive behaviour disorder, Severe mental retardation, Vomiting.

The invention further relates to compounds, compositions and medicaments for the treatment of Pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat-free mass. Such conditions include, but are not limited to Diabetes, impaired glucose tolerance; Cardiovascular Disease including but not limited to Acute and Congestive Heart Failure, Hypotension, Hypertension, Angina Pectoris, Myocardial infarction.

The invention further relates to compounds, compositions and medicaments for the treatment of Hypothalamic/pituitary disorders, including but not limited to Cushing's Syndrome/disease, Basophile adenoma, Prolactinoma, hyperprolactinemia, Hypophysis Tumour/adenoma, Hypothalamic diseases, Froehlich's syndrome, Adrenohypophysis disease, Hypophysis disease, Adrenohypophysis hypofunction, Adrenohypophysis hyperfunction, Hypothalamic hypogonadism, Kallman's syndrome (anosmia, hyposmia), functional or psychogenic Amenorrhea, Hypopituitarism, Hypothalamic Hypothyroidism, Hypothalamic-adrenal dysfunction, Idiopathic Hyperprolactinemia, Hypothalamic Disorders of growth hormone deficiency, Idiopathic growth deficiency, Dwarfism, Gigantism, Acromegaly. The invention further relates to compounds, compositions and medicaments for the treatment of Pain disorders, including but not limited to Neuropathic pain, Restless Leg Syndrome, Migraine, Cluster headache, Tension-type headache, Trigeminal autonomic Cephalalgias, Hemicrania Continua, Trigeminal neuralgia, other headache disorders, pain, enhanced or exaggerated sensitivity to pain such as Hyperalgesia, Causalgia, and Allodynia, Acute pain, Burn pain, Atypical facial pain, Back pain, Complex regional pain syndrome I and II, Arthritic pain, Sports injury pain, pain related to infection e.g. HIV, Post-chemotherapy pain, Post-stroke pain, Post- operative pain, neuralgia, conditions associated with Visceral pain such as Irritable bowel syndrome, and Angina pain; Inflammatory disorders (including Inflammatory bowel disease), Renal/urinary disorders (including urinary retention, benign prostatic hypertrophy, chronic renal failure, renal disease); Respiratory disorders (such as Chronic obstructive pulmonary disease, Asthma).

The invention further relates to compounds, compositions and medicaments for the treatment of androgen receptor mediated cancers, including, but not limited to, Prostate cancer, Liver Cancer, Colon cancer, Endometrial cancer, Pancreatic cancer and cancers associated with other organs of the body including the central nervous system and peripheral nervous system.

Compounds of the invention include compounds according to the formula (1 )

or a salt thereof, wherein

W is N or CR 5 ;

X is N or CR 6 ;

Y is CH 2 ; Z is CR7 or N;

Ri is H or a halogen;

R 2 is a 5 or 6.membered heteroaryl ring containing two or three nitrogen atoms which is optionally substituted with one or more fluorine atoms;

R 3 is H or a halogen;

R 4 is H or a halogen;

R 5 is selected from H; halogen; C1-C3 alkyl optionally substituted with OMe or one or more halogen groups; and C1-C3 alkoxy optionally substituted with one or more halogen groups;

R 6 is selected from H; halogen; C C 3 alkyl optionally substituted with OMe or one or more halogen groups; and C1-C3 alkoxy, optionally substituted with one or more halogen groups; and

Ry is H or a halogen.

Compounds of the invention include compounds according to the formula (2)

or a salt thereof, wherein

W is N or CR 5 ;

X is N or CR 6 ;

Y is CH 2 ;

Z is CR7 or N;

Ri is H or a halogen;

R 2 is a 5 or 6.membered heteroaryl ring containing two or three nitrogen atoms which is optionally substituted with one or more fluorine atoms;

R 3 is H or a halogen; R 4 is H or a halogen;

R 5 is selected from H; halogen; C1-C3 alkyl optionally substituted with OMe or one or more halogen groups; and C1-C3 alkoxy optionally substituted with one or more halogen groups;

R 6 is selected from H; halogen; C1-C3 alkyl optionally substituted with OMe or one or more halogen groups; and C1-C3 alkoxy, optionally substituted with one or more halogen groups; and

Ry is H or a halogen.

Specific embodiments include the following substituents, which can be combined in any particular combination without limitation:

W can be N. W can be CR 5 . Where W is N, X is CR 6 rather than N, thereby X and W are not both N.

X can be N. X can be CR 6 . Where X is N, W is CR5 rather than N, thereby X and W are not both N.

Y can be CH 2 . Y can be CD 2 .

Z can be CR7. Z can be CH. Z can be CF. Z can be N.

Ri can be H. Ri can be a halogen. Ri can be F.

R 2 can be a 5 or 6_membered heteroaryl ring containing two or three nitrogen atoms which is substituted with one or more fluorine atoms.

R 2 can be a 5-membered heteroaryl ring containing three nitrogen atoms.

R 2 can be a 6-membered heteroaryl ring containing two nitrogen atoms.

R 2 can be a triazole ring. R 2 can be a 1 ,2,3-triazole ring. R 2 can be a pyrimidine ring. R 2 can be a pyrazine ring.

R 2 can be selected from the group consisting of:

R 2 can be: R 2 can be: R 2 can be:

R 2 can be: can be:

R 3 can be H. R 3 can be a halogen. R 3 can be F.

R 4 can be H. R 4 can be a halogen. R 4 can be F. R 4 can be Cl.

R 5 can be H. R 5 can be halogen. R 5 can be F. R 5 can be Ci-C 3 alkyl or Ci-C 3 alkoxy optionally substituted with one or more halogen groups. R 5 can be Ci-C 3 alkyl optionally substituted with OMe. R 5 can be Ci-C 3 alkyl or Ci-C 3 alkoxy. R 5 can be Me. R 5 can be OMe. R 5 can be CH 2 OMe.

R 6 can be H. R 6 can be C C 3 alkyl or C C 3 alkoxy optionally substituted with one or more halogen groups. R 6 can be Ci-C 3 alkyl optionally substituted with OMe. R 6 can be Ci-C 3 alkyl or Ci-C 3 alkoxy. R 6 can be a halogen. R 6 can be F. R 6 can be Me. R 6 can be OCHF 2 . R 6 can be OMe. R 6 can be CH 2 OMe. R 6 can be OCD 3 .

Ry can be H. R 7 can be a halogen. R 7 can be F.

The compound can be selected from the group consisting of:

The compound can be selected from the group consisting of:

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(triazol-2-y l)benzamide; N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-5-methyl-2-pyr imidin-2-yl- benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-5-methyl-2-(tr iazol-2- yl)benzamide;

2-fluoro-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-(triazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-(t riazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-2-methoxy-6-(t riazol-2- yl)benzamide; N-[(1 S,2S)-2-[dideuterio-(4-fluorophenoxy)methyl]cyclopentyl]-2-m ethoxy-6-(triazol-

2-yl)benzamide;

2-(difluoromethoxy)-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-(triazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-py rimidin-2-yl- benzamide;

2-fluoro-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-pyrimidin-2- yl- benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-py razin-2-yl- benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6-py rimidin-4-yl- benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(5-fluoropyr imidin-2-yl)-6- methoxy-benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(triazol-2-y l)-6-

(trideuteriomethoxy)benzamide;

2-(difluoromethoxy)-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-pyrimidin-

2-yl-benzamide;

3-fluoro-N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methoxy-2-(t riazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(methoxymeth yl)-6-(triazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-(methoxymeth yl)-3-(triazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-6-(methoxymeth yl)-3-(triazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methoxy-3-(t riazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-6-methoxy-3-(t riazol-2- yl)pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methyl-3-(tr iazol-2-yl)pyridine-

2-carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-6-methyl-3-(tr iazol-2-yl)pyridine-

2-carboxamide; N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6-methyl-3-pyr imidin-2-yl- pyridine-2-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-3-pyrimidin-2- yl-pyridine-2- carboxamide;

N-[(1 S,2S)-2-[(2,4-difluorophenoxy)methyl]cyclopentyl]-3-pyrimidi n-2-yl-pyridine-2- carboxamide;

N-[(1 S,2S)-2-[(4-chlorophenoxy)methyl]cyclopentyl]-3-pyrimidin-2- yl-pyridine-2- carboxamide;

N-[(1 S,2S)-2-[(3,4-difluorophenoxy)methyl]cyclopentyl]-3-pyrimidi n-2-yl-pyridine-2- carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-4-(t riazol-2- yl)pyridine-3-carboxamide;

N-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-4-py rimidin-2-yl- pyridine-3-carboxamide;

N-[(1 S,2S)-2-[(5-fluoro-2-pyridyl)oxymethyl]cyclopentyl]-2-methox y-6-(triazol-2- yl)benzamide;

N-[(1 S,2S)-2-[(5-fluoro-2-pyridyl)oxymethyl]cyclopentyl]-2-methox y-6-pyrimidin-2-yl- benzamide;

N-[(1 S,2S)-2-[(5-fluoro-2-pyridyl)oxymethyl]cyclopentyl]-2-(triaz ol-2-yl)-6-

(trideuteriomethoxy)benzamide.

Definitions

In this application, the following definitions apply, unless indicated otherwise.

The term “treatment”, in relation to the uses of any of the compounds described herein, including those of the formula (1 ) or formula (2), is used to describe any form of intervention where a compound is administered to a subject suffering from, or at risk of suffering from, or potentially at risk of suffering from the disease or disorder in question. Thus, the term “treatment” covers both preventative (prophylactic) treatment and treatment where measurable or detectable symptoms of the disease or disorder are being displayed.

The term“effective therapeutic amount” as used herein (for example in relation to methods of treatment of a disease or condition) refers to an amount of the compound which is effective to produce a desired therapeutic effect. For example, if the condition is pain, then the effective therapeutic amount is an amount sufficient to provide a desired level of pain relief. The desired level of pain relief may be, for example, complete removal of the pain or a reduction in the severity of the pain.

To the extent that any of the compounds described have chiral centres, the present invention extends to all optical isomers of such compounds, whether in the form of racemates or resolved enantiomers. The invention described herein relates to all crystal forms, solvates and hydrates of any of the disclosed compounds however so prepared. To the extent that any of the compounds disclosed herein have acid or basic centres such as carboxylates or amino groups, then all salt forms of said compounds are included herein. In the case of pharmaceutical uses, the salt should be seen as being a pharmaceutically acceptable salt.

Salts or pharmaceutically acceptable salts that may be mentioned include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.

Examples of pharmaceutically acceptable salts include acid addition salts derived from mineral acids and organic acids, and salts derived from metals such as sodium, magnesium, or preferably, potassium and calcium.

Examples of acid addition salts include acid addition salts formed with acetic, 2,2- dichloroacetic, adipic, alginic, aryl sulfonic acids (e.g. benzenesulfonic, naphthalene- 2-sulfonic, naphthalene-1 ,5-disulfonic and p-toluenesulfonic), ascorbic (e.g. L- ascorbic), L-aspartic, benzoic, 4-acetamidobenzoic, butanoic, (+) camphoric, camphor-sulfonic, (+)-(1S)-camphor-10-sulfonic, capric, caproic, caprylic, cinnamic, citric, cyclamic, dodecylsulfuric, ethane-1 , 2-disulfonic, ethanesulfonic, 2- hydroxyethanesulfonic, formic, fumaric, galactaric, gentisic, glucoheptonic, gluconic (e.g. D-gluconic), glucuronic (e.g. D-glucuronic), glutamic (e.g. L-glutamic), a- oxoglutaric, glycolic, hippuric, hydrobromic, hydrochloric, hydriodic, isethionic, lactic (e.g. (+)-L-lactic and (±)-DL-lactic), lactobionic, maleic, malic (e.g. (-)-L-malic), malonic, (±)-DL-mandelic, metaphosphoric, methanesulfonic, 1-hydroxy-2-naphthoic, nicotinic, nitric, oleic, orotic, oxalic, palmitic, pamoic, phosphoric, propionic, L- pyroglutamic, salicylic, 4-amino-salicylic, sebacic, stearic, succinic, sulfuric, tannic, tartaric (e.g.(+)-L-tartaric), thiocyanic, undecylenic and valeric acids.

Particular examples of salts are salts derived from mineral acids such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric and sulfuric acids; from organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, arylsulfonic acids; and from metals such as sodium, magnesium, or preferably, potassium and calcium.

Also encompassed are any solvates of the compounds and their salts. Preferred solvates are solvates formed by the incorporation into the solid state structure (e.g. crystal structure) of the compounds of the invention of molecules of a non-toxic pharmaceutically acceptable solvent (referred to below as the solvating solvent). Examples of such solvents include water, alcohols (such as ethanol, isopropanol and butanol) and dimethylsulfoxide. Solvates can be prepared by recrystallising the compounds of the invention with a solvent or mixture of solvents containing the solvating solvent. Whether or not a solvate has been formed in any given instance can be determined by subjecting crystals of the compound to analysis using well known and standard techniques such as thermogravimetric analysis (TGE), differential scanning calorimetry (DSC) and X-ray crystallography.

The solvates can be stoichiometric or non-stoichiometric solvates. Particular solvates may be hydrates, and examples of hydrates include hemihydrates, monohydrates and dihydrates. For a more detailed discussion of solvates and the methods used to make and characterise them, see Bryn et at, Solid-State Chemistry of Drugs, Second Edition, published by SSCI, Inc of West Lafayette, IN, USA, 1999, ISBN 0-967- 06710-3. The term “pharmaceutical composition” in the context of this invention means a composition comprising an active agent and comprising additionally one or more pharmaceutically acceptable carriers. The composition may further contain ingredients selected from, for example, diluents, adjuvants, excipients, vehicles, preserving agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavouring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispersing agents, depending on the nature of the mode of administration and dosage forms. The compositions may take the form, for example, of tablets, dragees, powders, elixirs, syrups, liquid preparations including suspensions, sprays, inhalants, tablets, lozenges, emulsions, solutions, cachets, granules, capsules and suppositories, as well as liquid preparations for injections, including liposome preparations.

The compounds of the invention may contain one or more isotopic substitutions, and a reference to a particular element includes within its scope all isotopes of the element. For example, a reference to hydrogen includes within its scope 1 H, 2 H (D), and 3 H (T). Similarly, references to carbon and oxygen include within their scope respectively 12 C, 13 C and 14 C and 16 0 and 18 0. In an analogous manner, a reference to a particular functional group also includes within its scope isotopic variations, unless the context indicates otherwise. For example, a reference to an alkyl group such as an ethyl group or an alkoxy group such as a methoxy group also covers variations in which one or more of the hydrogen atoms in the group is in the form of a deuterium or tritium isotope, e.g. as in an ethyl group in which all five hydrogen atoms are in the deuterium isotopic form (a perdeuteroethyl group) or a methoxy group in which all three hydrogen atoms are in the deuterium isotopic form (a trideuteromethoxy group). A group shown for example as CH 2 may be CD 2 or CHD. The isotopes may be radioactive or non-radioactive.

Therapeutic dosages may be varied depending upon the requirements of the patient, the severity of the condition being treated, and the compound being employed. Determination of the proper dosage for a particular situation is within the skill of the art. Generally, treatment is initiated with the smaller dosages which are less than the optimum dose of the compound. Thereafter the dosage is increased by small increments until the optimum effect under the circumstances is reached. For convenience, the total daily dosage may be divided and administered in portions during the day if desired.

The magnitude of an effective dose of a compound will, of course, vary with the nature of the severity of the condition to be treated and with the particular compound and its route of administration. The selection of appropriate dosages is within the ability of one of ordinary skill in this art, without undue burden. In general, the daily dose range may be from about 10 pg to about 30 mg per kg body weight of a human and non-human animal, preferably from about 50 pg to about 30 mg per kg of body weight of a human and non-human animal, for example from about 50 pg to about 10 mg per kg of body weight of a human and non-human animal, for example from about 100 pg to about 30 mg per kg of body weight of a human and non-human animal, for example from about 100 pg to about 10 mg per kg of body weight of a human and non-human animal and most preferably from about 100 pg to about 1 mg per kg of body weight of a human and non-human animal.

Pharmaceutical Formulations

While it is possible for the active compound to be administered alone, it is preferable to present it as a pharmaceutical composition (e.g. formulation).

Accordingly, in another embodiment of the invention, there is provided a pharmaceutical composition comprising at least one compound of the formula (1 ) as defined above together with at least one pharmaceutically acceptable excipient.

The composition may be a tablet composition.

The composition may be a capsule composition.

The pharmaceutically acceptable excipient(s) can be selected from, for example, carriers (e.g. a solid, liquid or semi-solid carrier), adjuvants, diluents (e.g. solid diluents such as fillers or bulking agents; and liquid diluents such as solvents and co solvents), granulating agents, binders, flow aids, coating agents, release-controlling agents (e.g. release retarding or delaying polymers or waxes), binding agents, disintegrants, buffering agents, lubricants, preservatives, anti-fungal and antibacterial agents, antioxidants, buffering agents, tonicity-adjusting agents, thickening agents, flavouring agents, sweeteners, pigments, plasticizers, taste masking agents, stabilisers or any other excipients conventionally used in pharmaceutical compositions.

The term “pharmaceutically acceptable” as used herein means compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject (e.g. a human subject) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. Each excipient must also be“acceptable” in the sense of being compatible with the other ingredients of the formulation.

Pharmaceutical compositions containing compounds of the formula (1 ) can be formulated in accordance with known techniques, see for example, Remington’s Pharmaceutical Sciences, Mack Publishing Company, Easton, PA, USA.

The pharmaceutical compositions can be in any form suitable for oral, parenteral, topical, intranasal, intrabronchial, sublingual, ophthalmic, otic, rectal, intra-vaginal, or transdermal administration.

Pharmaceutical dosage forms suitable for oral administration include tablets (coated or uncoated), capsules (hard or soft shell), caplets, pills, lozenges, syrups, solutions, powders, granules, elixirs and suspensions, sublingual tablets, wafers or patches such as buccal patches.

Tablet compositions can contain a unit dosage of active compound together with an inert diluent or carrier such as a sugar or sugar alcohol, eg; lactose, sucrose, sorbitol or mannitol; and/or a non-sugar derived diluent such as sodium carbonate, calcium phosphate, calcium carbonate, or a cellulose or derivative thereof such as microcrystalline cellulose (MCC), methyl cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, and starches such as corn starch. Tablets may also contain such standard ingredients as binding and granulating agents such as polyvinylpyrrolidone, disintegrants (e.g. swellable crosslinked polymers such as crosslinked carboxymethylcellulose), lubricating agents (e.g. stearates), preservatives (e.g. parabens), antioxidants (e.g. BHT), buffering agents (for example phosphate or citrate buffers), and effervescent agents such as citrate/bicarbonate mixtures. Such excipients are well known and do not need to be discussed in detail here.

Tablets may be designed to release the drug either upon contact with stomach fluids (immediate release tablets) or to release in a controlled manner (controlled release tablets) over a prolonged period of time or with a specific region of the Gl tract.

The pharmaceutical compositions typically comprise from approximately 1 % (w/w) to approximately 95%, preferably% (w/w) active ingredient and from 99% (w/w) to 5% (w/w) of a pharmaceutically acceptable excipient (for example as defined above) or combination of such excipients. Preferably, the compositions comprise from approximately 20% (w/w) to approximately 90% (w/w) active ingredient and from 80% (w/w) to 10% of a pharmaceutically excipient or combination of excipients. The pharmaceutical compositions comprise from approximately 1 % to approximately 95%, preferably from approximately 20% to approximately 90%, active ingredient. Pharmaceutical compositions according to the invention may be, for example, in unit dose form, such as in the form of ampoules, vials, suppositories, pre-filled syringes, dragees, powders, tablets or capsules.

Tablets and capsules may contain, for example, 0-20% disintegrants, 0-5% lubricants, 0-5% flow aids and/or 0-99% (w/w) fillers/ or bulking agents (depending on drug dose). They may also contain 0-10% (w/w) polymer binders, 0-5% (w/w) antioxidants, 0-5% (w/w) pigments. Slow release tablets would in addition typically contain 0-99% (w/w) release-controlling (e.g. delaying) polymers (depending on dose). The film coats of the tablet or capsule typically contain 0-10% (w/w) polymers, 0-3% (w/w) pigments, and/or 0-2% (w/w) plasticizers.

Parenteral formulations typically contain 0-20% (w/w) buffers, 0-50% (w/w) cosolvents, and/or 0-99% (w/w) Water for Injection (WFI) (depending on dose and if freeze dried). Formulations for intramuscular depots may also contain 0-99% (w/w) oils. The pharmaceutical formulations may be presented to a patient in“patient packs” containing an entire course of treatment in a single package, usually a blister pack.

The compounds of the formula (1 ) will generally be presented in unit dosage form and, as such, will typically contain sufficient compound to provide a desired level of biological activity. For example, a formulation may contain from 1 nanogram to 2 grams of active ingredient, e.g. from 1 nanogram to 2 milligrams of active ingredient. Within these ranges, particular sub-ranges of compound are 0.1 milligrams to 2 grams of active ingredient (more usually from 10 milligrams to 1 gram, e.g. 50 milligrams to 500 milligrams), or 1 microgram to 20 milligrams (for example 1 microgram to 10 milligrams, e.g. 0.1 milligrams to 2 milligrams of active ingredient).

For oral compositions, a unit dosage form may contain from 1 milligram to 2 grams, more typically 10 milligrams to 1 gram, for example 50 milligrams to 1 gram, e.g. 100 milligrams to 1 gram, of active compound.

The active compound will be administered to a patient in need thereof (for example a human or animal patient) in an amount sufficient to achieve the desired therapeutic effect (effective amount). The precise amounts of compound administered may be determined by a supervising physician in accordance with standard procedures.

EXAMPLES

The invention will now be illustrated, but not limited, by reference to the specific embodiments described in the following examples.

EXAMPLES 1 TO 33

The compounds of Examples 1 to 33 shown in Table 1 below have been prepared. Their NMR and LCMS properties and the methods used to prepare them are set out in Table 3. Starting materials and intermediates used in the synthesis of each of the Examples are listed in Table 2.

Preparation of the compounds of the invention

Compounds of the invention may be prepared by routes including those in Figure 1. Details of many of the standard transformations such as those in the routes below and others which could be used to perform the same transformations can be found in standard reference textbooks such as Organic Synthesis”, M. B. Smith, McGraw-Hill (1994) or“Advanced Organic Chemistry”, 4 th edition, J. March, John Wiley & Sons (1992).

Compounds of the invention may be prepared by an amide coupling reaction between a substituted 2-(phenoxymethyl)cyclopentan-1 -amine or 2-((pyridin-2- yloxy)methyl)cyclopentan-1 -amine and a substituted benzoic, nicotinic or picolinic acid. The reaction conditions will typically use a coupling agent or agents, for example EDCI and HOBt, or HATU, with a suitable base such as DIPEA, in a solvent such as DCM, typically at room temperature. Alternatively, amide couplings may be performed by reacting the amine with an acid chloride in the presence of a base, for example Et 3 N or DIPEA, in a suitable solvent such as DCM, typically at room temperature. Acid chlorides can be prepared from the corresponding acid via a number of methods, for example reaction with oxalyl chloride in a solvent system such as DCM and DMF, typically at room temperature, or reaction with 1-chloro- /V,/V,2-trimethyl-1-propenylamine, commonly known as Ghosez’ reagent (Haveaux et at, Org. Synth., 1980, 59, 26) in a suitable solvent such as DCM, at a suitable temperature, for example room temperature (rt).

Certain compounds of the invention may be prepared by first reacting a suitable amine with an acid to form the corresponding amide, then further reacting the amide product. An example of a subsequent reaction is a palladium-catalysed carbon- carbon coupling reaction between an aryl halide, for example an aryl bromide, and a trialkylstannane, commonly known as a Stille coupling (Cordovilla et at, ACS Catai, 2015, 5, 3040-3053). The trialkylstannane may be for example a tributyl- or trimethylstannane, and the palladium catalyst may be for example Pd(PPh3) 4 or Pd(P f Bu 3 ) 2 , and the reaction may or may not be carried out in the presence of an additive such as CsF, Cul or CuCI. Typically the Stille coupling reaction will be performed in a solvent such as DMF or 1 ,4-dioxane, at elevated temperature, for example 85°C, 100°C or 120°C. A further example of subsequent reaction is the reduction of an ester, for example a methyl ester, to the corresponding primary alcohol, which is then itself further reacted to form an ether. The reduction may be performed using a reducing agent such as LiBH 4 in a suitable solvent, for example THF, at a suitable temperature range, for example 0°C to rt. The ether formation may be performed under nucleophilic aromatic substitution (SnAr) conditions, wherein the alcohol is deprotonated by a suitable strong base such as NaH, then reacted with a 2-halopyridine, for example a 2-fluoropyridine, in a solvent such as DMF, at a suitable temperature range, for example rt to 50°C.

Substituted 2-(phenoxymethyl)cyclopentan-1 -amines or 2-((pyridin-2- yloxy)methyl)cyclopentan-1 -amines may be prepared by a number of methods, using transformations that will be familiar to those with skill in the art of organic synthesis. One method of preparation of amines that are subsequently used for preparation of compounds of the invention is reduction of a carboxylic acid group present on the cyclopentane ring to the corresponding primary alcohol, followed by conversion to an ether using triphenylphosphine, an aryl alcohol, and an azodi carboxyl ate such as DIAD in a solvent such as THF at a suitable temperature, for example rt. Such a reaction is commonly known as a Mitsonobu coupling (Swamy et at, Chem. Rev., 2009, 109, 2551-2561 ). The reduction of a carboxylic acid may be performed using borane-THF complex, or the deutero borane-THF version, in a solvent such as THF at a suitable temperature range, for example 0°C to rt. Alternatively the primary alcohol may be converted to a methane sulfonate ester using methane sulfonyl chloride and a base such as Et 3 N, in a suitable solvent, for example DCM, at room temperature. The methane sulfonate ester can be substituted with the anion of an aryl alcohol formed by deprotonation with a suitable base, for example cesium carbonate, typically in a solvent such as DMF at an elevated temperature, for example 80°C. As a further alternative the alcohol may be deprotonated, for example for KO f Bu, and reacted with a 2-halopyridine under SnAr conditions as detailed above. Typically the amine functionality is protected during these transformations, for example as the terf-butylcarbamate (commonly known as Boc protection) which can be deprotected under acidic conditions, for example HCI in 1 ,4-dioxane at rt. Alternatively the amine may be protected as the bis-para-methoxybenzyl ether by reaction of the amine with para-methoxybenzyl chloride in the presence of an additive such as tefra-butylammonium iodide, with a base such as DIPEA in a solvent such as MeCN at rt. Deprotection of this protecting group can be achieved by treatment under reductive conditions, for example using a palladium on charcoal catalyst in the presence of ammonium formate in a solvent such as EtOH, at an elevated temperature, such as the reflux temperature of EtOH.

Substituted benzoic, nicotinic or picolinic acids can be prepared in a number of different ways using standard transformations which will be known to those skilled in the art. For example, the acid functionality can be prepared by hydrolysis of the corresponding ester or nitrile group under acidic or basic conditions, and in some instances hydrolysis may occur concomitantly with another functional group transformation under the reaction conditions. A 2/-/-1 ,2,3-triazol-2-yl group may be introduced by reaction of an aryl halide or heteroaryl halide functionality, for example an iodide or bromide, with 2/-/-1 , 2, 3-triazole, in the presence or absence of a copper reagent such as Cul, Cu 2 0 or CuCI, and the presence or absence of an amine such as a dimethylcyclohexane-1 , 2-diamine, in the presence of a base such as cesium carbonate or potassium carbonate in a suitable solvent such as DMF, H 2 0, DMSO or 1 ,4-dioxane, or a mixture of solvents. Typically, such a reaction is performed at an elevated temperature, for example in the range 80°C to 120°C. An aryl or heteroaryl group, for example pyrimidin-2-yl, may be introduced using Stille coupling conditions as detailed above, or introduced using an aryl or heteroaryl halide, for example 2- bromopyrimidine, an aryl or heteroaryl boronic acid or ester, a suitable palladium catalyst such as PdCI 2 (dtbpf), a base such as Na 2 CC>3, in a suitable solvent system such as aqueous THF, at an elevated temperature, for example 80°C (commonly known as a Suzuki-Miyaura coupling, Miyaura et at, Chem. Rev., 1995, 95, 2457- 2483). Where intermediates are commercially available they are identified by their chemical abstracts service (CAS) reference number in Table 2, where not commercially available the synthesis of the intermediates using standard transformations is detailed herein. Commercial reagents were utilized without further purification.

General procedures

Room temperature (rt) refers to approximately 20-27°C. 1 H NMR spectra were typically recorded at 300, 400 or 500 MHz at ambient temperature unless otherwise specified. Chemical shift values are expressed in parts per million (ppm), i.e. (d)- values. Standard abbreviations, or their combinations, are used for the multiplicity of the NMR signals, for example: s=singlet, br=broad, d=doublet, t=triplet, q=quartet, quin=quintet or p=pentet, h=heptet, dd=doublet of doublets, dt=doublet of triplets, m=multiplet. Coupling constants are listed as J values, measured in Hz. NMR and mass spectroscopy results were corrected to account for background peaks. Chromatography refers to column chromatography performed using silica or C18 silica and executed under positive pressure (flash chromatography) conditions.

LCMS methods

LCMS experiments were carried out using electrospray conditions under the conditions below (Solvents: A1 = 0.1 % HCO2H in H 2 0; A2 = 0.1 % HCO2H and 10 mM ammonium bicarbonate in H 2 0; A3 = 2.5 L H 2 0 + 2.5 mL 28% ammonia in H 2 0 solution; A4 = 0.1 % HC0 2 H in H 2 0:MeCN (95:5); B1 = 0.1 % HC0 2 H in H 2 0:MeCN (1 :9); B2 = MeCN; B3 = 2.5 L MeCN + 135 mL H 2 0 + 2.5 mL 28% ammonia in H 2 0 solution; B4 = 0.1 % HC0 2 H in MeCN; B5 = 2.5 L MeCN + 125 mL H 2 0 + 2.5 mL HCO2H). LCMS data are given in the format: Mass ion, electrospray mode (positive or negative), retention time (experimental text and Table 1 ); Mass ion, electrospray mode (positive or negative), retention time, approximate purity (Table 2).

Method 1. Instruments: Hewlett Packard 1 100 with G1315A DAD, Micromass ZQ; Column: Waters X-Bridge C18, 2.5 micron, 2.1 x 20 mm or Phenomenex Gemini-NX C18, 3 micron, 2.0 x 30 mm; Gradient [time (min)/solvent B3 in A3 (%)]: 0.00/2, 0.10/2, 8.40/95, 10.00/95; Injection volume 1 pL; Instruments: Hewlett Packard 1 100 with G1315A DAD, 45°C; Flow rate 1.5 mL/min.

Method 2. Instruments: Waters Acquity UPLC with photodiode array detector and QDa mass detector; Column: C18, 1.6 micron, 2.1 x 50 mm; Gradient [time (min)/solvent B1 in A1 (%)]:0.00/10, 0.75/10, 2.80/90, 4.50/100, 4.60/100, 4.70/10, 6.00/10; column temperature 35°C; Flow rate 0.8 mL/min. Method 3. Instruments: Waters 2690 HPLC with 996 Photodiode Array Detector and Acquity QDA mass detector; Column: Waters X-Bridge C18, 5 micron, 4.6 x 100 mm; Gradient [time (min)/solvent B2 in A2 (%)]: 0.00/10, 1.00/10, 5.00/100, 7.00/100, 7.50/10, 8.00/10; Flow rate 1.2 mL/min.

Method 4. Instruments: Waters Acquity UPLC with PDA detector and QDA; Column: C18, 1.6 micron, 2.1 x 50 mm; Gradient [time (min)/solvent B1 in A1 (%)]: 0.00/3, 0.20/3, 2.70/98, 3.00/100, 3.50/100, 3.51/3, 4.00/3; Column temperature 35°C; Flow rate 0.8-1.0 mL/min.

Method 5. Instruments: Agilent Technologies 1260 LC with Chemstation software, Diode Array Detector, Agilent 6120 Quadrupole MS with APCI and ES Source; Column: Phenomenex Gemini-NX C18, 3 micron, 2 x 30 mm; Gradient [time (min)/solvent B3 in A3 (%)]:0.00/5, 2.00/95, 2.50/95, 2.60/5, 3.00/5; Injection volume 0.5 pL; UV detection 190-400 nm; column temperature 40°C; Flow rate 1.5 mL/min. Method 6. Instruments: Waters Acquity UPLC; Column: Waters X-Select C18, 2.5 micron, 4.6 x 30 mm or 1.7 micron, 2.1 x 30 mm; Gradient [time (min)/solvent B4 in A1 (%)]: 0.00/5, 0.1 1/5, 2.15/95, 2.56/95, 2.83/5; Injection volume 3 pL; UV detection 210-400 nm; column temperature 40°C; Flow rate 0.77 mL/min.

Method 7. Instruments: Agilent Technologies 1290 Infinity II Series LC with 6125 Quadrupole MSD; Column: Zorbax XDB C18, 5 micron; Gradient [time (min)/solvent B2 in A4 (%)]:0.00/5, 2.50/95, 4.00/95, 4.50/5, 6.00/5; Injection volume 1 pL; UV detection 210-400 nm; column temperature 25°C; Flow rate 1.5 mL/min.

Method 8. Instruments: Agilent 1260 Infinity, PE Sciex API2000 LC/MS/MS; Column: Agilent Poroshell 120 C18, 2.7 micron, 3 x 50; Gradient [time (min)/solvent B4 in A1 (%)]:0.00/5, 2.00/5, 3.20/95, 3.25/5, 3.50/5; Injection volume 0.5 pL; UV detection 210 to 315 nM; column temperature 40°C; Flow rate 0.8 mL/min.

Method 9. Instruments: Agilent Technologies 1260 LC with Chemstation software, Diode Array Detector, Agilent 6120 Quadrupole MS with APCI and ES Source; Column: Phenomenex Kinetex C18, 5 micron, 2.1 x 50 mm; Gradient [time (min)/solvent B5 in A1 (%)]:0.00/5, 2.00/95, 2.50/95, 2.60/5, 3.00/5; Injection volume 0.5 pL; UV detection 190-400 nm; column temperature 40°C; Flow rate 1.5 mL/min.

Abbreviations

DBU 1 ,8-Diazabicyclo(5.4.0)undec-7-ene

DCM dichloromethane

DIAD di/sopropyl azodicarboxylate

DIPEA A/./V-diisopropylethylamine

DMF A/./V-dimethylformamide

DMSO dimethylsulfoxide

EDCI A/-(3-dimethylaminopropyl)-/\/'-ethylcarbodiimide hydrochloride

ES electrospray

EtOAc ethyl acetate

EtOH ethanol

h hour(s)

HOBt 1-hydroxybenzotriazole hydrate

L litre

LC liquid chromatography

LCMS liquid chromatography mass spectrometry

MeCN acetonitrile

MeOH methanol

min = minute(s) MS mass spectrometry

NMR nuclear magnetic resonance

ref relative centrifugal force

rpm revolutions per minute

rt room temperature

s second(s)

THF tetrahydrofuran

Prefixes n-, s-, /-, t- and tert- have their usual meanings: normal, secondary, iso, and tertiary.

SYNTHESIS OF INTERMEDIATES

Preparation of substituted (1 S.2S)-2-(phenoxymethyl)cvclopentan-1-amine and

(1 S.2S)-2-((pyridin-2-yloxy)methvHcvclopentan-1 -amine intermediates

Typical procedure 1, exemplified by the preparation of Intermediate 2, (1 S,2S)- 2-((4-fluorophenoxy)methyl)cyclopentan-1 -amine hydrochloride

Step 1. A solutio entane carboxylic acid (Intermediate 1 , 50 g, 0.218 mol) in dry THF (500 ml_) was stirred under N 2 and cooled to -15 to -20°C. All solids dissolved leaving a colourless solution, to which borane-THF complex (1 M, 545 ml_, 0.545 mol) was added dropwise over 1.5 h at such a rate as to maintain the temperature of the now milky solution at <0°C. Once addition was complete the reaction mixture was allowed to stir at 0°C for 2 h; the cooling bath was removed and reaction mixture allowed to warm to rt overnight. The reaction mixture was then cooled to -15 to -20°C and cautiously quenched with MeOH (0.9 L) over 30 min maintaining the vessel temperature at <0°C. The reaction mixture was then warmed to rt over 2 h and concentrated in vacuo. MeOH (400 mL) was added and the mixture concentrated in vacuo. Purification by gradient flash chromatography eluting with 20-25% EtOAc in hexane yielded tert-butyl ((1 S,2S)-2- (hydroxymethyl)cyclopentyl)carbamate (23.0 g, 106.8 mmol) as a colourless, brittle solid.

LCMS (Method 2): m/z 160.1 (ES+, M-55), at 1.88 min.

1 H NMR: (400 MHz, DMSO-de) d 6.79 (d, J = 7.8 Hz, 1 H), 4.40 (t, J = 5.2 Hz, 1 H), 3.51-3.35 (m, 2H), 3.21 (ddd, J = 10.5, 6.8, 5.4 Hz, 1 H), 1.84-1.64 (m, 4H), 1.61- 1.41 (m, 2H), 1.37 (s, 9H), 1.33-1.20 (m, 1 H).

A further batch of the above material (303.0 g, 1.41 mol) was prepared from (1 S,2S)- 2-(tert-butoxycarbonylamino)cyclopentane carboxylic acid (Intermediate 1 , 400 g, 1.74 mol) and borane-THF complex (1 M, 4.34 L, 4.34 mol) in THF (4.0 L) using the methods above. Purification by gradient flash chromatography eluting with 10-30% EtOAc in hexane yielded material with LCMS purity of approximately 100% that was used directly in the next step. Step 2. Tert- butyl ((1 S,2S)-2-(hydroxymethyl)cyclopentyl)carbamate (214 g, 994 mmol), triphenyl phosphine (261 g, 994 mmol) and 4-fluorophenol (1 12 g, 994 mmol) in dry THF (4 L) was stirred at rt under N 2 for 10 min until all solids had dissolved. The solution was then cooled to -10 to 0°C and DIAD (196 mL, 994 mmol) was added dropwise over 30 min at such a rate as to maintain the vessel temperature between -5 to 0°C. Once addition was complete the reaction mixture was allowed to stir at 0-5°C for 1 h before warming to rt and stirring for 4 h. Solvent was removed in vacuo and the resulting pale yellow crude slurry was stored in the fridge overnight. The slurry was combined with material from an analogous reaction performed on 100 g scale of tert-butyl ((1 S,2S)-2-(hydroxymethyl)cyclopentyl)carbamate and purification by gradient flash chromatography eluting with 5-10% EtOAc in hexane yielded tert-butyl ((1 S,2S)-2-((4-fluorophenoxy)methyl)cyclopentyl)carbamate (228 g, 737 mmol).

LCMS (Method 2): m/z 210.2 (ES+, M-99), at 2.77 min.

1 H NMR: (400 MHz, DMSO-de) d 7.18-7.04 (m, 2H), 6.98-6.83 (m, 3H), 3.93 (dd, J = 9.4, 4.8 Hz, 1 H), 3.80 (dd, J = 9.4, 7.5 Hz, 1 H), 3.60 (p, J = 7.6 Hz, 1 H), 2.16-2.02 (m, 1 H), 1.93-1.76 (m, 2H), 1.69-1.48 (m, 2H), 1.48-1.39 (m, 2H), 1.37 (s, 9H).

A second batch of material (177 g) with approximately 6% contamination with 4- fluorophenol was isolated from the gradient column chromatography and used separately to the 228 g batch in the subsequent step.

Step 3. Tert- butyl ((1 S,2S)-2-((4-fluorophenoxy)methyl)cyclopentyl)carbamate (6.0 g, 19.4 mmol) was dissolved in 1 ,4-dioxane (80 mL) and cooled to 0-5°C under N 2 . 4M HCI in 1 ,4-dioxane (60 mL, 240 mmol) was added dropwise over 15 min, stirring was continued at 0-5°C for 30 min and then at rt for 3 h. The resulting colourless slurry was concentrated in vacuo before pentane (100 mL) was added, the mixture was stirred overnight, then filtered and washed with cold pentane (10 mL). Air-drying until constant weight yielded the title compound (Batch 1 , 3.2 g, 15.3 mmol). Further batches of material were prepared using the above method; batches 2 and 3 were prepared from tert-butyl ((1 S,2S)-2-((4-fluorophenoxy)methyl)cyclopentyl)carbamate (60.0 g, 194 mmol and 168 g, 543 mmol respectively), 4M HCI in 1 ,4-dioxane (600 mL, 2.4 mol and 1.68 L, 6.7 mol respectively) in 1 ,4-dioxane (800 mL and 2L respectively); batch 4 was prepared from tert-butyl ((1 S,2S)-2-((4- fluorophenoxy)methyl)cyclopentyl)carbamate (177 g of 94% purity), 4M HCI in 1 ,4- dioxane (1.66 L, 6.64 mol) and 1 ,4-dioxane (2 L). In each case the batch 2, 3 and 4 reaction mixtures were seeded with isolated batch 1 material after 4M HCI in 1 ,4- dioxane had been added and the reaction mixtures were stirred at 0-5°C for 30-60 min. Following seeding with batch 1 material, the reaction mixtures were then stirred at rt overnight before isolation as above. 1 H NMR analysis of isolated batches confirmed that they were identical and a single final quantity of the title compound (285 g, 1.36 mol) was isolated by blending individual batches.

Data in Table 2. Intermediate 3, (1 S,2S)-2-((4-chlorophenoxy)methyl)cyclopentan-1 -amine hydrochloride Cl

Step 1. Terf-butyl ((1 S,2S)-2-(hydroxymethyl)cyclopentyl)carbamate (Intermediate 2, Step 1 ; 6.00 g, 27.9 mmol), 4-chlorophenol (3.94 g, 30.7 mmol) and triphenylphosphine (7.32 g, 27.9 mmol) were dissolved in THF (8.7 mL) at rt and sonicated for 10 min before the dropwise addition of DIAD (6.04 mL, 30.7 mmol). The reaction mixture was sonicated at rt for 15 min before concentration in vacuo. Purification by gradient flash chromatography eluting with 0-25% EtOAc in hexane yielded ferf-butyl ((1 S,2S)-2-((4-chlorophenoxy)methyl)cyclopentyl)carbamate (9.00 g) as a yellow solid which was used in the subsequent step without further purification.

LCMS (Method 3): m/z 226.2 (ES+, M-99), at 2.97 min, approximate purity 79%.

Step 2. A solution of ferf-butyl ((1 S,2S)-2-((4- chlorophenoxy)methyl)cyclopentyl)carbamate (9.00 g) in DCM (120 mL) was cooled to 0°C and 3N HCI in dioxane (3N, 50 mL) was added drop wise. The reaction mixture was stirred at rt for 18 h before concentration in vacuo. Trituration with diethyl ether yielded the title compound (5.50 g, 24.4 mmol) as an off white solid.

Data in Table 2.

Intermediate 4, (1 S,2S)-2-((4-fluorophenoxy)methyl-c/ 2 )cyclopentan-1-amine

The title compound (342 mg, 1.62 mmol) was prepared in three steps from (1 S,2S)- 2-(tert-butoxycarbonylamino)cyclopentane carboxylic acid (Intermediate 1 , 1.00 g, 4.36 mmol) and cf 3 - bo rane-THF complex (1 M, 9.60 mL, 9.60 mmol) in THF (30 mL) at 30°C for 3 h; DIAD (0.47 mL, 2.41 mmol), triphenylphosphine 632 mg, 2.41 mmol) and 4-fluorophenol (225 mg, 2.01 mmol) in THF (10 mL) at rt for 4 h; and HCI in 1 ,4- dioxane (4M, 4.65 mL, 18.6 mmol) in MeOH (10 mL) at rt overnight, using the methods of Intermediate 2, steps 1 to 3. After completion of step 3, the title compound was isolated as a white solid by concentration in vacuo, azeotroping with toluene, and purification by loading onto an SCX cartridge, which was washed with MeOH and eluted with NH 3 (approximately 1 M in MeOH).

Data in Table 2.

Intermediate 5, (1 S,2S)-2-((3,4-difluorophenoxy)methyl)cyclopentan-1-amine The title compound (376 mg, 1.65 mmol) was prepared in two steps from tert- butyl ((1 S,2S)-2-(hydroxymethyl)cyclopentyl)carbamate (Intermediate 2, Step 1 ; 495 mg, 2.30 mmol), DIAD (0.49 mL, 2.51 mmol), triphenylphosphine (658 mg, 2.51 mmol) and 3,4-difluorophenol (272 mg, 2.09 mmol) in THF (10 mL) at rt for 3 h; and HCI in 1 ,4-dioxane (4M, 4.93 mL, 19.7 mmol) in MeOH (6 mL) at rt overnight, using the methods of Intermediate 2, steps 2 and 3. After completion of step 2, the title compound was isolated as an off-white solid by concentration in vacuo, azeotroping with toluene, and purification by loading onto an SCX cartridge, which was washed with MeOH and eluted with NH 3 (approximately 1 M in MeOH).

Data in Table 2.

Typical procedure 2, exemplified by the preparation of Intermediate 6, (1 S,2S)- 2-((2,4-difluorophenoxy)methyl)cyclopentan-1 -amine hydrochloride

Step 1. Mesyl chloride (4.54 mL, 39.6 mmol) was added dropwise to a solution of tert- butyl ((1 S,2S)-2-(hydroxymethyl)cyclopentyl)carbamate (Intermediate 2, Step 1 ; 7.1 1 g, 33.0 mmol) and triethylamine (9.21 mL, 66.1 mmol) in DCM (80 mL) at 0°C. The solution was stirred at rt for 3 h before partitioning between H 2 0 (300 mL) and DCM (200 mL). The aqueous phase was extracted with DCM (2 x 150 mL) and the combined organic phases were dried (Na 2 S04) and concentrated in vacuo. Purification by gradient flash chromatography eluting with 0-10% EtOAc in hexane yielded ((1 S,2S)-2-((fe/ -butoxycarbonyl)amino)cyclopentyl)methyl methanesulfonate (9.00 g, 30.7 mmol) as an off white solid.

LCMS (Method 4): m/z 194.1 (ES+, M-99), at 1.74 min.

1 H NMR: (400 MHz, DMSO- cfe) d 6.95 (d, J = 8.1 Hz, 1 H), 4.19 (dd, J = 9.7, 5.1 Hz, 1 H), 4.07 (dd, J = 9.7, 7.3 Hz, 1 H), 3.53 (p, J = 7.9 Hz, 1 H), 3.15 (s, 3H), 2.02 (dt, J = 15.2, 7.8 Hz, 1 H), 1.91-1.72 (m, 2H), 1.68-1.47 (m, 2H), 1.46-1.27 (m, 2H), 1.37 (s, 9H).

Step 2. A mixture of ((1 S,2S)-2-((fe/ -butoxycarbonyl)amino)cyclopentyl)methyl methanesulfonate (7.00 g, 23.8 mmol), 2,4-difluorophenol (6.20 g, 47.7 mmol) and Cs 2 C0 3 (15.5 g, 47.7 mmol) in DMF (100 mL) was stirred at 80°C for 6 h. After partitioning between water (1 L) and EtOAc (200 mL) the aqueous layer was extracted with EtOAc (2 x 150 mL) and the combined organic phases were dried (Na 2 S04) and concentrated in vacuo. Purification by gradient flash chromatography eluting with 0-8% EtOAc in hexane yielded tert- butyl ((1 S,2S)-2-((2,4- difluorophenoxy)methyl)cyclopentyl)carbamate (6.00 g, 18.3 mmol) as a light yellow solid.

LCMS (Method 4): m/z 228.2 (ES+, M-99), at 2.37 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d 7.26 (ddd, J = 11.7, 8.8, 3.1 Hz, 1 H), 7.16 (td, J = 9.4, 5.5 Hz, 1 H), 7.05-6.88 (m, 2H), 4.02 (dd, J = 9.5, 4.7 Hz, 1 H), 3.88 (dd, J = 9.5, 7.4 Hz, 1 H), 3.60 (p, J = 7.7 Hz, 1 H), 2.1 1 (q, J = 7.1 Hz, 1 H), 1.96-1.79 (m, 2H), 1.70-1.48 (m, 2H), 1.48-1.38 (m, 2H), 1.36 (s, 9H).

Step 3. The title compound (4.00 g, 15.2 mmol) was prepared as an off-white solid from tert- butyl ((1 S,2S)-2-((2,4-difluorophenoxy)methyl)cyclopentyl)carbamate (6.00 g, 18.3 mmol), 4N HCI in dioxane (70 mL) and 1 ,4-dioxane (80 mL) at rt for 3 h, using the methods of Intermediate 3, step 3.

Data in Table 2.

Typical procedure 3 for the preparation of substituted (1 S,2S)-2-(((pyridin-2- yl)oxy)methyl)cyclopentan-1 -amines, as exemplified by the preparation of Intermediate 8, (1 S,2S)-2-(((5-fluoropyridin-2-yl)oxy)methyl)cyclopentan-1 - amine

Step 1. 4-Methoxybenzyl chloride (2.1 mL, 15.4 mmol) was added to a solution of methyl (1 S,2S)-2-aminocyclopentanecarboxylate hydrochloride (Intermediate 7, 1.00 g, 5.57 mmol), tetrabutylammonium iodide (5.16 g, 14.0 mmol) and DIPEA (7.26 mL, 41.9 mmol) in MeCN (30 mL) and the mixture was stirred at rt for 24 h. Saturated aqueous NaHCC>3 solution (20 mL) was added, resulting in the formation of a white precipitate which was removed by filtration. The filtrate was partially concentrated in vacuo to remove MeCN and then extracted with EtOAc (3 x 50 mL). The combined organic phases were washed with aqueous NaHCC>3 solution and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-20% EtOAc in iso- hexane yielded methyl (1 S,2S)-2-(bis(4- methoxybenzyl)amino)cyclopentane-1-carboxylate (1.75 g, 4.56 mmol) as a clear gum.

LCMS (Method 5): m/z 384.4 (ES+), at 1.90 min.

1 H NMR: (400 MHz, DMSO-cfe) d 7.23-7.17 (m, 4H), 6.88-6.83 (m, 4H), 3.72 (s, 6H), 3.60 (s, 3H), 3.51 (d, J = 13.8 Hz, 2H), 3.38 (d, J = 13.7 Hz, 2H), 3.34-3.28 (m, 1 H), 2.84 (q, J = 8.4 Hz, 1 H), 1.82 (dt, J = 8.5, 6.1 Hz, 1 H), 1.76-1.44 (m, 4H), 1.35-1.20 (m, 1 H).

Step 2. Lithium borohydride (2M in THF, 4.56 mL, 9.13 mmol) was added to a solution of methyl (1 S,2S)-2-(bis(4-methoxybenzyl)amino)cyclopentane-1- carboxylate (1.75 g, 4.56 mmol) in THF (25 mL) at 0°C. The mixture was allowed to warm to rt and stirred for 12 h before the addition of further lithium borohydride (2M in THF, 4.56 mL, 9.13 mmol) and stirring continued for 72 h at rt, then 40°C for 24 h. Further lithium borohydride (2M in THF, 4.56 mL, 9.13 mmol) and stirring continued at 40°C for 16 h before cooling to 0°C and addition of water (40 mL). The mixture was extracted with EtOAc (3 x 40 mL) and the combined organic phases were washed with brine and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-40% EtOAc in iso- hexane yielded ((1 S,2S)-2-(bis(4- methoxybenzyl)amino)cyclopentyl)methanol (880 mg, 2.48 mmol) as a clear gum. LCMS (Method 5): m/z 356.4 (ES+), at 1.79 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d: 7.29-7.16 (m, 4H), 6.93-6.77 (m, 4H), 4.39 (dd, J = 5.7, 4.4 Hz, 1 H), 3.70 (s, 6H), 3.57 (d, J = 13.6 Hz, 2H), 3.52-3.42 (m, 1 H), 3.36- 3.23 (m, 2H), 3.06 (ddd, J = 10.1 , 8.0, 5.6 Hz, 1 H), 2.70 (q, J = 7.8 Hz, 1 H), 2.01 (dt, J = 7.7, 3.9 Hz, 1 H), 1.75-1.46 (m, 4H), 1.46-1.29 (m, 1 H), 1.23 (dq, J = 13.2, 7.1 Hz, 1 H). Step 3. Potassium terf-butoxide (379 mg, 3.38 mmol) was added to a solution of ((1 S,2S)-2-(bis(4-methoxybenzyl)amino)cyclopentyl)methanol (800 mg, 2.25 mmol) in THF (15 ml_) and the reaction stirred for 10 min under N 2 at room temperature. 2,5-Difluoropyridine (0.22 ml_, 2.48 mmol) was added and the reaction stirred for 4 h at rt before the addition of water (40 ml_). After extraction with EtOAc (3 x 70 ml_) the combined organic phases were washed with brine and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-15% EtOAc in iso- hexane yielded (1 S,2S)-2-(((5-fluoropyridin-2-yl)oxy)methyl)-/\/,/\/-bis(4- methoxybenzyl)cyclopentan-1-amine (830 mg, 1.84 mmol) as a clear gum.

LCMS (Method 5): m/z 451.4 (ES+), at 2.23 min.

1 H NMR: (400 MHz, DMSO-de) d: 8.09 (dt, J = 3.2, 0.6 Hz, 1 H), 7.69-7.52 (m, 1 H), 7.26-7.15 (m, 4H), 6.83-6.74 (m, 4H), 6.69 (ddd, J = 9.1 , 3.7, 0.6 Hz, 1 H), 4.24 (dd, J = 10.4, 4.4 Hz, 1 H), 3.99-3.95 (m, 1 H), 3.67 (d, J = 0.6 Hz, 6H), 3.59 (d, J = 13.6 Hz, 2H), 3.35-3.24 (m, 2H), 2.91 (q, J = 7.9 Hz, 1 H), 2.35-2.22 (m, 1 H), 1.72 (dq, J = 14.7, 8.0, 7.5 Hz, 1 H), 1.67-1.52 (m, 3H), 1 .52-1.40 (m, 1 H), 1.38-1.26 (m, 1 H).

Step 4. (1 S,2S)-2-(((5-Fluoropyridin-2-yl)oxy)methyl)-/\/,/\/-bis(4- methoxybenzyl)cyclopentan-1 -amine (354 mg, 0.33 mmol) was added to a mixture of 10% Palladium on carbon (55% w/w water, 354 mg, 0.33 mmol) and ammonium formate (2.1 g, 33.3 mmol) in EtOH (15 ml_) under N 2 and the mixture was heated at reflux for 6 h. After cooling to rt the mixture was filtered through celite and concentrated in vacuo. DCM (20 mL) was added and the mixture was extracted with 1 M aqueous HCI (2 x 30 mL). The combined aqueous phases were basified with 10M aqueous NaOH solution and then extracted with EtOAc (3 x 70 mL). The combined organic phases were dried (MgS0 4 ) and concentrated in vacuo. The isolated crude title compound (750 mg) was used without further purification in the synthesis of Examples 32 and 33.

Data in Table 2.

Preparation of substituted benzoic, nicotinic or picolinic acid intermediates

Intermediate 15, 2-(difluoromethoxy)-6-(2H-1,2,3-triazol-2-yl)benzoic acid

Step 1. KOH (7.43 g, 132 mmol) was added to a solution of methyl 2-bromo-6- hydroxybenzoate (Intermediate 14, 1.80 g, 7.79 mmol) in MeCN/H 2 0 (1 :1 , 25 mL) and the mixture cooled to -78°C. Diethyl (bromodifluoromethyl)phosphonate (2.77 mL, 15.6 mmol) was added in one portion and the reaction mixture was warmed to rt and stirred under N 2 for 6 h. H 2 0 (50 mL) and DCM (100 mL) were added and the phases were separated before the organic phase was concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-50% EtOAc in iso- hexane yielded methyl 2-bromo-6-(difluoromethoxy)benzoate (1.82 g, 6.15 mmol) as a colourless oil.

LCMS (Method 6): m/z not observed in ES+ or ES- mode, at 1.40 min.

1 H NMR: (500 MHz, DMSO-de) d 7.62 (dd, J = 8.1 , 0.9 Hz, 1 H), 7.52 (t, J = 8.3 Hz, 1 H), 7.38 (dq, J = 8.4, 0.9 Hz, 1 H), 7.31 (t, J = 72.9 Hz, 1 H), 3.89 (s, 3H). Step 2. A mixture of (1 R,2R)-/V 1 ,/V 2 -dimethylcyclohexane-1 ,2-diamine (9.54 pl_, 0.06 mmol), methyl 2-bromo-6-(difluoromethoxy)benzoate (85 mg, 0.30 mmol), copper(l) iodide (2.88 mg, 0.02 mmol), 2/-/-1 , 2, 3-triazole (36.1 mI_, 0.61 mmol) and cesium carbonate (197 mg, 0.61 mmol) in 1 ,4-dioxane (1 ml_) and water (40 mI_) was heated at 100°C for 16 h. The reaction mixture was diluted with DMF (1 mL) and heated at 120°C for 4 h before cooling to rt and partitioning between EtOAc (10 mL) and H 2 0 (10 mL). The phases were separated, the aqueous phase was washed with EtOAc (10 mL), acidified, and extracted with EtOAc (2 x 10 mL). The combined organic phases were dried (MgS0 4 ) and concentrated in vacuo to yield the crude title compound (35.0 mg, 0.12 mmol) as an orange oil. 1 H NMR analysis confirmed that ester hydrolysis had occurred concomitantly with triazole substitution, and the title compound (approximately 90% purity by 1 H NMR) was used without further purification in the synthesis of Example 8.

Data in Table 2.

Intermediate 17, 2-methoxy-6-(pyrimidin-2-yl)benzoic acid

Step 1. 2-Bromopyrimidine (4.97 g, 31.3 mmol) and a solution of Na 2 CC>3 (5.40 g, 50.9 mmol) in water (50 mL) were added to a stirred solution of methyl 2-methoxy-6- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)benzoate (Intermediate 16, 6.00 g, 18.9 mmol) in THF (120 mL). After degassing with N 2 for 10 min [1 ,1 '-bis(di-fe/ - butylphosphino)ferrocene]dichloropalladium(ll) (288 mg, 0.44 mmol) was added and the reaction mixture was heated at 80°C for 2.5 h. After cooling to rt H 2 0 (250 mL) and DCM (200 mL) were added and the phases were separated. The aqueous phase was extracted with DCM (4 x 200 mL) and the combined organic phases were dried (Na 2 S0 4 ) and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 20-70% EtOAc in heptane yielded methyl 2-methoxy-6- (pyrimidin-2-yl)benzoate (2.67 g) which contained approximately 25% w/w pinacol. LCMS (Method 8): m/z 244.9 (ES+), at 2.14 min.

Step 2. Lithium hydroxide monohydrate (0.96 g, 23.0 mmol) and H 2 0 (40 mL) were added to a stirred solution of methyl 2-methoxy-6-(pyrimidin-2-yl)benzoate (2.67 g, approximately 75% purity) in THF (90 mL). The resulting mixture was heated at 60°C for 5 d before cooling to rt and being acidified with 1 N aqueous HCI. After concentration in vacuo the resulting crude paste was freeze dried before H 2 0 (150 mL) and diethyl ether (100 mL) were added and the phases were separated. The aqueous phase was washed with diethyl ether (2 x 100 mL), concentrated in vacuo and freeze dried to yield the title compound (2.17 g) as an off-white solid. 1 H NMR analysis indicated that the title compound was approximately 53% pure (with approximately 3% pinacol and the remainder LiCI), and this was used without further purification in the synthesis of Examples 9 and 32.

Data in Table 2. Intermediate 19, 2-(methoxy-d 3 )-6-(2H-1,2,3-triazol-2-yl)benzoic acid

Step 1. 2/-/-1 ,2,3-T riazole (13.4 g, 194 mmol), CS 2 CO 3 (63.4 g, 194 mmol) and N N 2 - dimethylcyclohexane-1 ,2-diamine (13.8 g, 97.2 mmol) were added to a solution of 2- bromo-6-hydroxybenzoic acid (Intermediate 18, 21.0 g, 97.2 mmol) in DMF (250 ml_) and the mixture was degassed at rt with N 2 for 20 min. Cu 2 0 (4.17 g, 19.4 mmol) was added and the reaction mixture stirred at 80°C for 16 h before cooling to rt and filtering through celite, washing with EtOAc (1 L). The filtrate was acidified with 1 N aqueous HCI (70 ml_) to adjust to approximately pH 2 and extracted with EtOAc (3 x 500 ml_). The combined organic phases were concentrated in vacuo, purification by gradient flash column chromatography eluting with 0-3% MeOH in DCM yielded 2- hydroxy-6-(2/-/-1 ,2,3-triazol-2-yl)benzoic acid (10.4 g, 50.4 mmol) as light brown solid.

LCMS (Method 4): m/z 206.1 (ES+), at 1.02 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d 12.85 (br. s, 1 H), 10.45 (br. s, 1 H), 8.05 (s, 2H), 7.38 (t, J = 8.1 Hz, 1 H), 7.27 (d, J = 8.0 Hz, 1 H), 6.97 (dd, J = 8.2, 1.1 Hz, 1 H).

Step 2. DBU (7.67 g, 50.4 mmol) was added dropwise to a solution of 2-hydroxy-6- (2/-/-1 ,2,3-triazol-2-yl)benzoic acid (10.4 g, 50.4 mmol) in THF (1 10 ml_) at 0°C. After stirring at 0°C for 30 min methyl iodide (9.42 ml_, 151 mmol) was added at 0°C and reaction mixture stirred at rt for 16 h. Saturated aqueous NaHC0 3 solution (150 ml_) and EtOAc (70 ml_) were added, the phases were separated, and the aqueous phase was extracted with EtOAc (2 x 70 ml_). Concentration in vacuo yielded methyl 2-hydroxy-6-(2/-/-1 ,2,3-triazol-2-yl)benzoate (8.80 g, 40.2 mmol) as a brown sticky solid which was used without further purification in the next step.

LCMS (Method 4): m/z 220.1 (ES+), at 1.36 min.

1 H NMR: (400 MHz, DMSO-de) d 10.46 (s, 1 H), 8.07 (s, 2H), 7.58-7.34 (m, 2H), 6.99 (dd, J = 8.1 , 1.2 Hz, 1 H), 3.67 (s, 3H).

Step 3. A mixture of methyl 2-hydroxy-6-(2/-/-1 ,2,3-triazol-2-yl)benzoate (8.80 g, 40.2 mmol), cf 4 - MeOD (4.93 mL, 121 mmol) and Ph 3 P (15.8 g, 60.3 mmol) in THF (80 mL) was sonicated at rt for 30 min. DIAD (1 1.8 mL, 60.3 mmol) was added dropwise at 0°C and the reaction mixture then stirred at rt for 16 h. After concentration in vacuo, purification by gradient flash column chromatography eluting with 0-12% EtOAc in hexane yielded methyl 2-(methoxy-d 3 )-6-(2/-/-1 ,2,3-triazol-2-yl)benzoate (18.9 g) as a white solid which was used in the next step without further purification.

LCMS (Method 4): m/z 205.1 (ES+, M-31 ), at 1.60 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d 8.1 1 (s, 2H), 7.81-7.35 (m, 2H), 7.22 (dd, J = 8.1 , 1.2 Hz, 1 H), 3.71 (s, 3H).

Step 4. 1 N aqueous NaOH (100 mL) was added to a solution of methyl 2-(methoxy- d 3 )-6-(2/-/-1 ,2,3-triazol-2-yl)benzoate (18.9 g) in 1 ,4-dioxane (400 mL) and the reaction mixture was heated at 100°C for 16 h. After acidification with 1 N aqueous HCI (200 ml_) to approximately pH 1 , EtOAc (200ml_) was added and the phases were separated. The aqueous phase was extracted with EtOAc (2 x 200 ml_), the combined organic phases were dried (Na 2 S04) and concentrated in vacuo. Trituration with n-pentane yielded the title compound (6.46 g, 29.1 mmol) as an off white solid.

Data in Table 2.

Intermediate 21, 2-(difluoromethoxy)-6-(pyrimidin-2-yl)benzoic acid

Step 1. A suspension of 2-bromo-6-(difluoromethoxy)benzonitrile (Intermediate 20, 2.00 g, 8.06 mmol), 2-(tributylstannyl)pyrimidine (3.58 ml_, 11.3 mmol) and copper(l) chloride (0.96 g, 9.68 mmol) in 1 ,4-dioxane (3 ml_) was degassed for 20 min before the addition of tetrakis(triphenylphosphine)palladium(0) (0.47 g, 0.40 mmol) and heating at 85°C for 3 h. The reaction mixture was cooled and filtered through celite, washing with DCM. The filtrate was adsorbed onto silica and purification by gradient flash column chromatography eluting with 0-100% EtOAc in isohexane yielded 2- (difluoromethoxy)-6-(pyrimidin-2-yl)benzonitrile (1.60 g, 6.34 mmol) as a pale yellow solid.

LCMS (Method 6): m/z 248.2 (ES+), at 1.10 min.

1 H NMR: (500 MHz, DMSO-cfe) d 9.05 (d, J = 4.9 Hz, 2H), 8.15 (dd, J = 7.9, 1.1 Hz, 1 H), 7.93 (t, J = 8.2 Hz, 1 H), 7.70-7.60 (m, 2H), 7.51 (t, J = 72.5 Hz, 1 H).

Step 2. MeCN (0.03 ml_) was added to 2-(difluoromethoxy)-6-pyrimidin-2-yl- benzonitrile (100 mg, 0.40 mmol) followed by cone hydrogen chloride (0.1 ml_, 3.24 mmol) and the reaction mixture stirred at rt overnight. The reaction mixture was cooled to 0°C and 2 drops of brine were added. After sitting at 0°C for 1 h the aqueous phase was removed by pipette, and ice cold water (5 drops) added, then removed by pipette. The process was repeated with ice cold water (5 drops) then ice cold EtOAc (5 drops) and the resulting solid was dried in vacuo to yield the title compound (36 mg, 0.14 mmol) as a pale yellow solid.

Data in Table 2.

Intermediate Acid 27, 2-methoxy-4-(pyrimidin-2-yl)nicotinic acid

Step 1. A solution of methyl 2-fluoro-4-iodonicotinate (7.00 g, 24.9 mmol) in MeOH (70 ml_) was cooled to 0°C and sodium methoxide (30% in MeOH, 4.46 ml_, 24.9 mmol) was added drop wise. After stirring at rt for 16 h the reaction mixture was concentrated in vacuo and partitioned between H 2 0 (300 ml_) and EtOAc (200 ml_). The aqueous phase was extracted with EtOAc (3 x 150 ml_) and the combined organic phases were dried (Na 2 S04) and concentrated in vacuo to yield 4-iodo-2- methoxynicotinate (6.78 g, 23.1 mmol) as a light yellow liquid that was used without further purification.

LCMS (Method 4): m/z 293.9 (ES+), at 2.18 min. 1 H NMR: (400 MHz, DMSO-de) d: 7.95 (d, J = 5.4 Hz, 1 H), 7.53 (d, J = 5.4 Hz, 1 H), 3.86 (s, 3H), 3.85 (s, 3H).

Step 2. A solution of 4-iodo-2-methoxynicotinate (6.60 g, 22.5 mmol) and 2- (tributylstannyl)pyrimidine (12.5 g, 33.8 mmol) in 1 ,4-dioxane (75 ml_) at rt was degassed with N 2 for 20 min before the addition of tetrakis(triphenylphosphine)palladium(0) (2.60 g, 2.25 mmol) and Cul (0.42 g, 2.25 mmol). The reaction mixture heated at 100°C for 16 h before cooling to rt, filtering through celite and the celite was washed with EtOAc (100 ml_). The filtrate was partitioned between water (250 ml_) and EtOAc (150 ml_) and the aqueous phase was extracted with EtOAc (3 x 100 ml_). The combined organic phases were dried (Na 2 S04) and concentrated in vacuo. Purification by reverse phase gradient flash column chromatography on C18 Silica, eluting with 0-30% MeCN in H 2 0 yielded methyl 2-methoxy-4-(pyrimidin-2-yl)nicotinate (3.30 g, 13.5 mmol) as a white solid. LCMS (Method 4): m/z 246.2 (ES+), at 1.49 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d: 8.97 (d, J = 4.9 Hz, 2H), 8.42 (d, J = 5.3 Hz, 1 H), 7.82 (d, J = 5.3 Hz, 1 H), 7.59 (t, J = 4.9 Hz, 1 H), 3.94 (s, 3H), 3.75 (s, 3H).

Step 3. NaOH (0.80 g, 20.2 mmol) was added to a solution of methyl 2-methoxy-4- (pyrimidin-2-yl)nicotinate (3.30 g, 13.5 mmol) in EtOH (30 ml_) and H 2 0 (8 ml_). After stirring at 80°C for 4 h the reaction mixture was concentrated in vacuo and partitioned between water (250 ml_) and EtOAc (100 ml_). The aqueous phase was washed with EtOAc (2 x 70 ml_), then acidified with 2N aqueous HCI (50 ml_) to approximately pH 1 and extracted with EtOAc (3 x 70 ml_). The combined organic layers were dried (Na 2 S04) and concentrated in vacuo to yield the title compound (2.23 g, 9.65 mmol) as a light brown solid which was used without further purification.

Data in Table 2.

Intermediate Acid 29, 6-(methoxymethyl)-3-(2H-1,2,3-triazol-2-yl)picolinic acid

Step 1. m-CPBA (approximately 55-70%, 27.5 g, approximately 160 mmol) was added to a solution of (5-bromopyridin-2-yl)methanol (Intermediate 28, 20.0 g, 106 mmol) in DCM (200 ml_) at 0°C and stirred at rt for 2 h. The reaction mixture was concentrated in vacuo and the residue purified by gradient flash column chromatography, eluting with 0-90% EtOAc in hexane to yield 5-bromo-2- (hydroxymethyl) pyridine 1 -oxide (17.0 g, 83.8 mmol) as an off-white solid.

LCMS (Method 3): m/z 206.0 (ES+), at 1.32 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d 8.60 (d, J = 1.7 Hz, 1 H), 7.65 (dd, J = 8.5, 1.8 Hz, 1 H), 7.46 (dd, J = 8.6, 1.2 Hz, 1 H), 5.68 (t, J = 5.7 Hz, 1 H), 4.52 (d, J = 5.7 Hz, 2H).

Step 2. NaH (60% in mineral oil, 6.70 g, 168 mmol) was added to a solution of 5- bromo-2-(hydroxymethyl)pyridine 1 -oxide (17.0 g, 83.8 mmol) in THF (170 ml_) at 0°C and the reaction mixture stirred for 1 h at rt. lodomethane (7.60 ml_, 123 mmol) was added at 0°C and after stirring at 70°C for 1 h the reaction mixture was cooled to 0°C and quenched by the addition of MeOH. Solvent was removed in vacuo, purification by gradient flash column chromatography, eluting with 0-45% EtOAc in hexane, yielded 5-bromo-2-(methoxymethyl) pyridine 1 -oxide (15.0 g, 69.1 mmol) as off-white solid.

LCMS (Method 3): m/z not observed (ES+), at 1.31 min.

1 H NMR: (400 MHz, DMSO-de) d 8.64 (d, J = 1.8 Hz, 1 H), 7.63 (dd, J = 8.5, 1.8 Hz, 1 H), 7.39 (d, J = 8.5 Hz, 1 H), 4.49 (s, 2H), 3.44 (s, 3H).

Step 3. A solution of 5-bromo-2-(methoxymethyl)pyridine 1 -oxide (1.00 g, 4.60 mmol), Et 3 N (3.20 ml_, 23.0 mmol) and trimethyl silyl cyanide (2.00 ml_, 16.1 mmol) in MeCN (10 ml_) was heated at 120°C for 18 h in a sealed tube. The reaction was performed a total of 13 times on 1.00 g scale and all crude reaction mixtures combined before H 2 0 was added and the MeCN was removed in vacuo. The crude material was partitioned between H 2 0 (300 ml_) and EtOAc (100 ml_) and the aqueous phase was extracted with EtOAc (3 x 80 ml_). The combined organic phases were dried (Na 2 S0 4 ), concentrated in vacuo, and purified by gradient flash column chromatography, eluting with 0-8% EtOAc in hexane, to yield 3-bromo-6- (methoxymethyl)picolinonitrile (6.50 g, 28.8 mmol) as an off-white solid.

LCMS (Method 3): m/z 229.0 (ES+), at 2.29 min

1 H NMR: (400 MHz, DMSO-d 6 ) d 8.39 (d, J = 8.5 Hz, 1 H), 7.68 (d, J = 8.5 Hz, 1 H), 4.52 (s, 2H), 3.35 (s, 3H).

Step 4. K 2 C0 3 (5.50 g, 39.8 mmol) and 2/-/-1 , 2, 3-triazole (2.30 mL, 39.8 mmol) were added to a solution of 3-bromo-6-(methoxymethyl)picolinonitrile (6.00 g, 26.6 mmol) in DMF (60 mL) and the mixture was heated at 1 10°C for 24 h. After partitioning between cold H 2 0 (200 mL) and EtOAc (80 mL) the aqueous phase was extracted with EtOAc (2 x 50 mL). The combined organic phases were dried (Na 2 S0 4 ) and concentrated in vacuo. Purification by reverse phase gradient flash column chromatography on C18 silica, eluting with 0-35% MeCN in H 2 0 (containing 0.1 % HC0 2 H as modifier) yielded 6-(methoxymethyl)-3-(2/-/-1 ,2,3-triazol-2-yl)picolinonitrile (1.60 g, 7.44 mmol) as an orange solid.

LCMS (Method 3): m/z 216.1 (ES+), at 1.87 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d 8.59 (d, J = 8.6 Hz, 1 H), 8.37 (s, 2H), 7.94 (d, J = 8.7 Hz, 1 H), 4.63 (s, 2H), 3.42 (s, 3H).

Step 5. A solution of KOH (8.34 g, 148 mmol) in H 2 0 (15 mL) was added to a solution of 6-(methoxymethyl)-3-(2/-/-1 ,2,3-triazol-2-yl)picolinonitrile (1.60 g, 7.43 mmol) in MeOH (30 mL) and reaction mixture heated at 1 10°C for 12 h. After partitioning between dilute aqueous HCI (100 mL) and EtOAc (50 mL) the phases were separated, and the aqueous phase was extracted with EtOAc (2 x 25 mL). The combined organic phases were dried (Na 2 S0 4 ) ad concentrated in vacuo to yield the title compound (1.01 g, 4.31 mmol) as an off-white solid which was used without further purification.

Data in Table 2. Intermediate 31, 6-methoxy-3-(2H-1,2,3-triazol-2-yl)picolinic acid

2H-1 ,2,3-Triazole (450 mg, 6.49 mmol), Cs 2 C0 3 (2.82 g, 8.66 mmol) and N N 2 - dimethylcyclohexane-1 ,2-diamine (60 mg, 0.43 mmol) were added to a solution of 6- methoxy-3-bromopicolinic acid (Intermediate 30, 1.00 g, 4.44 mmol) in DMSO (10 mL). The reaction mixture was degassed with N 2 at rt for 20 min, then Cul (83 mg, 0.43 mmol) was added and the reaction mixture was heated at 120°C for 45 min in a microwave reactor. Two further reactions on the same scale were performed, the combined crude mixtures were partitioned between EtOAc (100 mL) and 1 N aqueous HCI (100 mL), and the aqueous phase was extracted with EtOAc (4 x 100 mL). The combined organic phases were dried (Na 2 SC>4), concentrated in vacuo, and purified by reverse phase gradient flash column chromatography on C18 silica, eluting with 0-50% MeCN in 0.1 N aqueous formic acid to yield the title compound (0.78 g, 3.54 mmol) as a white solid.

Data in Table 2.

Intermediate Acid 33, 2-methoxy-4-(2H-1,2,3-triazol-2-yl)nicotinic acid

A mixture of (1 R,2R)-/\/ 1 ,/\/ 2 -dimethylcyclohexane-1 , 2-diamine (0.017 mL, 0.1 1 mmol), 4-bromo-2-methoxynicotinic acid (Intermediate 32, 0.50 g, 2.16 mmol), Cu 2 0 (0.031 g, 0.22 mmol), 2/-/-1 ,2,3-triazole (0.39 mL, 6.46 mmol) and Cs 2 C0 3 (1.47 g, 4.53 mmol) in DMF (5 mL) was heated at 80°C for 3 h before concentration in vacuo to give a blue solid. 1 N aqueous HCI (9 mL) was added to take the pH to approximately 3, followed by the addition of MeCN (40 mL) and vigorous shaking. The organic phase was decanted away from an aqueous suspension and the process repeated with MeCN (2 x 20mL). The combined organic phases were dried (MgS0 4 ) and concentrated in vacuo onto silica. Purified by gradient flash column chromatography, eluting with 0-10% MeOH containing 1 % AcOH, in DCM, yielded the title compound (170 mg, 0.66 mmol) in approximately 85 % purity as a white solid, which was used without further purification.

Data in Table 2.

Intermediate 35, 3-fluoro-6-methoxy-2-(2H-1,2,3-triazol-2-yl)benzoic acid

A mixture of 2-bromo-3-fluoro-6-methoxybenzoic acid (Intermediate 34, 1.75 g, 7.02 mmol), 2/-/-1 , 2, 3-triazole (1.46 g, 21.1 mmol), Cs 2 C0 3 (4.80 g, 14.7 mmol) and dimethylcyclohexane-l , 2-diamine (0.10 g, 0.70 mmol) in DMF (20 mL) was degassed with N 2 for 20 min at rt. Cu 2 0 (0.10 g, 0.70 mmol) was added and the reaction mixture was heated at 80°C for 16 h. 1 N aqueous NaOH (30 mL) and EtOAc (100 mL) were added, the phases were separated and the aqueous layer was washed with EtOAc (2 x 75 mL). The aqueous phase was acidified with 1 N aqueous HCI (25 mL) to approximately pH 2 and extracted with EtOAc (5 x 100 mL). The combined organic phases were dried (Na 2 S0 4 ) and concentrated in vacuo. Purification by gradient reverse phase flash column chromatography on C18 silica, eluting with 0-30% MeCN in H 2 0 yielded the title compound (0.25 g, 1.05 mmol) as an off white solid.

Data in Table 2.

Intermediate 37, 2-(methoxymethyl)-6-(2H-1,2,3-triazol-2-yl)benzoic acid

Step 1. A mixture of Me 2 S0 4 (1.00 mL, 10.6 mmol) and 7-bromoisobenzofuran-1- (3/-/)-one (Intermediate 36, 0.45 g, 2.10 mmol) in 10% aqueous NaOH (16.2 mL) and H 2 0 (4.5 mL) was heated at 130°C for 16 h. H 2 0 (100 mL) was added and the mixture was acidified to approximately pH 1with 3N aqueous HCI (30 mL). The aqueous phase was extracted with EtOAc (100 mL). The organic phase was dried (Na 2 S0 4 ), concentrated in vacuo, and purified by gradient flash column chromatography, eluting with 0-15% EtOAc in hexane to yield 2-bromo-6- (methoxymethyl)benzoic acid (0.16 g, 0.66 mmol) as a white solid.

LCMS (Method 4): m/z 243.2 (ES-), at 1.07 min.

1 H NMR: (400 MHz, DMSO-cf 6 ) d 13.60 (s, 1 H), 7.60 (dd, J = 7.9, 1.1 Hz, 1 H), 7.42 (dd, J = 7.7, 1.1 Hz, 1 H), 7.34 (t, J = 7.8 Hz, 1 H), 4.42 (s, 2H), 3.25 (s, 3H).

Step 2. A mixture of 2-bromo-6-(methoxymethyl)benzoic acid (0.15 g, 0.61 mmol), 2/-/-1 , 2, 3-triazole (63 mg, 0.91 mmol), Cs 2 C0 3 (0.40 g, 1.20 mmol) and N N 2 - dimethylcyclohexane-1 ,2-diamine (86 mg, 0.60 mmol) in Dioxane (8 mL) was degassed with N 2 for 20 min at rt. Cu 2 0 (26 mg, 0.06 mmol) was added and the reaction mixture was heated at 110°C for 16 h. The mixture was acidified to approximately pH 2 with 1 N aqueous HCI (5 mL), EtOAc (100 mL) was added and the phases were separated. The aqueous phase was extracted with EtOAc (2 x 50 mL), the combined organic phases were dried (Na 2 S0 4 ) and concentrated in vacuo. Purification by reverse phase gradient flash column chromatography on C18 silica, eluting with 0-35% MeCN in H 2 0 yielded the title compound (0.10 g, 0.43 mmol) as white solid.

Data in Table 2.

Preparation of additional intermediates

Intermediate 39, 2-bromo-AM(1 S,2S)-2-((4-fluorophenoxy)methyl)cvclopentyl)-

6-methoxybenzamide

DIPEA (82 mg, 0.64 mmol) and HATU (90 mg, 0.25 mmol) were added to a stirred solution of 2-bromo-6-methoxybenzoic acid (Intermediate 38, 50.0 mg, 0.21 mmol) and (1 S,2S)-2-((4-fluorophenoxy)methyl)cyclopentan-1-amine hydrochloride (Intermediate 2, 58.0 mg, 0.23 mmol) in DMF (5 mL) at 0°C and the reaction mixture was stirred for 2 h at rt. EtOAc (20 mL) and H 2 0 (20 mL) were added and the phases were separated, washed with brine (10 mL), dried (Na 2 SC>4) and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-30% EtOAc in petroleum ether yielded the title compound as a colourless gum.

Data in Table 2.

SYNTHESIS OF EXAMPLES

Typical procedures for the preparation of examples via amide coupling, as exemplified by the preparation of the below examples in Procedures 1, 2 and 3. Procedure 1 :

Example 30, W-[( 1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-4- pyrimidin-2-yl-pyridine-3-carboxamide

To a solution of 2-methoxy-4-(pyrimidin-2-yl)nicotinic acid (Intermediate 27, 600 mg, 2.60 mmol) and (1 S,2S)-2-((4-fluorophenoxy)methyl)cyclopentan-1 -amine hydrochloride (Intermediate 2, 701 mg, 2.85 mmol) in DCM (15 mL) was added EDCI (746 mg, 3.89 mmol) and HOBt (745 mg, 3.89 mmol) followed by DIPEA (1.80 mL, 10.4 mmol) and the reaction was stirred at rt for 18 h. DCM (100 mL) was added, the phases were separated and the organic phase was washed with saturated aqueous NaHC0 3 (100 mL), dried (MgSO^, filtered and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-100% EtOAc in isohexane yielded the title compound which was dissolved in MeOH:H 2 0 (1 :1 , 10 mL) and seeded with a previous batch of material. The resulting suspension was filtered to yield the title compound (420 mg, 0.99 mmol) as a white solid. A second crop of material (100 mg, 0.24 mmol) was isolated from the mother liquor after preparative reversed phase HPLC (Phenomenex Gemini-NX 5pm C18 column, 100 x 30 mm, eluting with 40 to 70% MeCN/Solvent B over 12.5 min at 30 mL/min [where solvent B is 0.2% of (28% NH 3 /H 2 0) in H 2 0], collecting fractions by monitoring at 205 nm).

Data in Table 3.

Procedure 2:

Example 10, 2-fluoro-/V-[(1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-6- pyrimidin-2-yl-benzamide

HATU (207 mg, 0.54 mmol) was added to a solution of 2-fluoro-6-(pyrimidin-2- yl)benzoic acid (Intermediate 13, 79.0 mg, 0.36 mmol), (1 S,2S)-2-((4- fluorophenoxy)methyl)cyclopentan-1 -amine hydrochloride (Intermediate 2, 107 mg, 0.44 mmol) and DIPEA (0.25 mL, 1.45 mmol) in DCM (2 mL) and the reaction stirred at rt overnight. The reaction was diluted with DCM (10 mL) and water and the phases were separated. The organic phase was concentrated in vacuo and purification by gradient flash column chromatography eluting with 0-100% EtOAc in isohexane, followed by re-purification by gradient flash column chromatography eluting with 0-30% DCM in MeCN, yielded the title compound (65 mg, 0.16 mmol) as a sticky tan glassy solid.

Data in Table 3.

Procedure 3:

Example 9, W-[( 1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6- pyrimidin-2-yl-benzamide

F

2-Methoxy-6-(pyrimidin-2-yl)benzoic acid (Intermediate 17, 177 mg, 0.41 mmol) was dissolved in DCM (2 ml_) and oxalyl chloride (0.02 ml_, 0.22 mmol) was added dropwise followed by DMF (0.01 ml_). After stirring at rt for 1 hr the reaction mixture was concentrated in vacuo. (1 S,2S)-2-((4-Fluorophenoxy)methyl)cyclopentan-1- amine hydrochloride (Intermediate 2, 50 mg, 0.20 mmol) in DCM (2 ml_) was added to the residue followed by dropwise addition of Et 3 N (0.14 ml_, 1.02 mmol) and the reaction mixture stirred at rt for 3.5 h. DCM (5 ml_) and saturated aqueous NaHCOs solution (5 ml_) were added and the phases were separated. The aqueous phase was extracted with DCM (2 x 5 ml_) and the combined organic phases were concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-7% MeOH in DCM, followed by preparative reversed phase HPLC (Phenomenex Gemini-NX 5pm C18 column, 100 x 30 mm, eluting with 40 to 70% MeCN/Solvent B over 12.5 min at 30 mL/min [where solvent B is 0.2% of (28% NH3/H2O) in H2O], collecting fractions by monitoring at 205 nm) yielded the title compound (9 mg, 0.02 mmol) as a pale yellow glassy solid.

Data in Table 3.

Example 29, N-[( 1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-4- (triazol-2-yl)pyridine-3-carboxamide

1-Chloro-/\/,/\/,2-trimethyl-1-propenylamine (97 pl_, 0.73 mmol) was added to a solution of 2-methoxy-4-(2/-/-1 ,2,3-triazol-2-yl)nicotinic acid (Intermediate 33, 80 mg, 0.36 mmol) in DCM (2 ml_) and stirred at rt for 2 h before concentration in vacuo to yield a colourless gum which solidified upon standing. The gum was redissolved in DCM (3 ml_) and added dropwise to a stirred solution of (1 S,2S)-2-((4- fluorophenoxy)methyl)cyclopentan-1 -amine hydrochloride (Intermediate 2, 107 mg, 0.44 mmol) and DIPEA (193 mI_, 1.1 1 mmol) in DCM (5 ml_). After stirring for 1 h at rt the mixture was concentrated in vacuo and purification by gradient flash column chromatography eluting with 0-70% EtOAc in isohexane yielded the title compound (90 mg, 0.22 mmol) as a white solid.

Data in Table 3.

Typical procedure for the preparation of examples via Stille coupling, as exemplified by the preparation of the below examples in Procedure 4.

Procedure 4: Example 11 , N-[( 1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-methoxy-6- pyrazin-2-yl-benzamide

A stirred solution of 2-bromo-/\/-((1 S,2S)-2-((4-fluorophenoxy)methyl)cyclopentyl)-6- methoxybenzamide (Intermediate 39, 100 mg, 0.23 mmol), 2-

(tributylstannyl)pyrazine (Intermediate 40, 102 mg, 0.28 mmol) and CsF (70 mg, 0.46 mmol) in DMF (5 mL) was degassed with Ar, followed by the addition of Pd(PPh 3 ) 4 (26 mg, 0.02 mmol) and heating at 120°C for 16 h in a sealed tube. EtOAc and H 2 0 (20 mL each) were added and the phases were separated. The organic phase was washed with brine (10 mL), dried (Na 2 S04) and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-50% EtOAc in petroleum ether yielded the target compound (55 mg, 0.13 mmol) as an off white solid.

Data in Table 3.

Example 13, N-[( 1 S,2S)-2-[(4-fluorophenoxy)methyl]cyclopentyl]-2-(5- fluoropyrimidin-2-yl)-6-methoxy-benzamide

Step 1. Pd(PPh3)2Cl2 (35 mg, 0.042 mmol) was added to a solution of 2-bromo-5- fluoropyrimidine (Intermediate 42, 150 mg, 0.84 mmol) and hexamethylditin (415 mg, 1.27 mmol) in dry THF (3 ml_) which had been degassed with Ar. After heating at 100°C for 16 h in a sealed tube the reaction mixture was filtered through celite and concentrated in vacuo to yield 5-fluoro-2-(trimethylstannyl)pyrimidine as a black gum which was used without analysis or purification in Step 2.

Step 2. A stirred solution of 2-bromo-/\/-((1 S,2S)-2-((4- fluorophenoxy)methyl)cyclopentyl)-6-methoxybenzamide (Intermediate 39, 100 mg, 0.23 mmol) and crude 5-fluoro-2-(trimethylstannyl)pyrimidine (280 mg) in 1 ,4- dioxane (3 ml_) was degassed with Ar. Pd(P‘Bu 3 ) 2 (26 mg, 0.02 mmol) was added and the reaction mixture was heated at 120°C for 16 h in a sealed tube. EtOAc (25 mL) and H 2 0 (20 mL) were added and the phases were separated. The organic phase was washed with brine (15 mL), dried (Na 2 SC>4) and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-50% EtOAc in petroleum ether yielded the title compound (34 mg, 0.07 mmol) as a white solid.

Data in Table 3.

Typical procedure for the preparation of examples via S N Ar, as exemplified by the preparation of the below example in Procedure 5.

Procedure 5:

Example 31 , N-[( 1 S,2S)-2-[(5-fluoro-2-pyridyl)oxymethyl]cyclopentyl]-2- methoxy-6-(triazol-2-yl)benzamide Step 1. Et 3 N (0.16 ml_, 1.14 mmol) was added to a suspension of 2-methoxy-6-(2/-/- 1 ,2,3-triazol-2-yl)benzoic acid (Intermediate 1 1 , 100 mg, 0.46 mmol), methyl (1 S,2S)- 2-aminocyclopentane-1-carboxylate (Intermediate 7, 90.1 mg, 0.50 mmol), EDCI (109 mg, 0.57 mmol) and HOBt (87.3 mg, 0.57 mmol) in DCM (2 ml_) at rt under N 2 . After stirring for 5 h at rt DCM (3 ml_) was added and the phases were separated. The organic phase was washed with 1 M aqueous HCI (5 ml_) and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-100% EtOAc in isohexane yielded methyl (1 S,2S)-2-[[2-methoxy-6-(triazol-2- yl)benzoyl]amino]cyclopentanecarboxylate (1 17 mg, 0.34 mmol) as a colourless glass.

LCMS (Method 5): m/z 345.4 (ES+), at 1.04 min.

1 H NMR: (400 MHz, CDCI 3 ) d: 7.78 (s, 2H), 7.56-7.37 (m, 2H), 6.96 (dd, J = 8.1 , 1.2 Hz, 1 H), 5.82 (d, J = 7.2 Hz, 1 H), 4.51 (quin, J = 6.9 Hz, 1 H), 3.88 (s, 3H), 3.68 (s, 3H), 2.83 (dt, J = 8.8, 7.0 Hz, 1 H), 2.26-2.12 (m, 1 H), 2.07-1.55 (m, 5H).

Step 2. LiBH 4 (4M in THF, 0.17 ml_, 0.68 mmol) was added slowly to a solution of methyl (1 S,2S)-2-[[2-methoxy-6-(triazol-2- yl)benzoyl]amino]cyclopentanecarboxylate (1 17 mg, 0.34 mmol) in THF (2 ml_) under N 2 at 0°C and the reaction mixture was stirred at 0°C for 5 min then rt overnight. Saturated aqueous NH 4 CI solution (5 ml_) was added cautiously, and the mixture was extracted with EtOAc (3 x 10 ml_). The combined organic phases were washed with brine (10 ml_) and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-100% EtOAc in isohexane, then 0-5% MeOH in EtOAc yielded N-[( 1 S,2S)-2-(hydroxymethyl)cyclopentyl]-2-methoxy-6-(triazol-2- yl)benzamide (88.0 mg, 0.28 mmol) as a colourless glassy solid.

LCMS (Method 5): m/z 317.4 (ES+), at 0.87 min.

1 H NMR: (400 MHz, CDCI 3 ) d: 7.80 (d, J = 0.6 Hz, 2H), 7.51-7.43 (m, 2H), 6.98 (dd, J = 7.9, 1.4 Hz, 1 H), 5.91 (s, 1 H), 4.18-4.06 (m, 1 H), 3.89 (s, 3H), 3.75 (dd, J = 1 1.3, 4.1 Hz, 1 H), 3.57 (dd, J = 1 1.4, 7.4 Hz, 1 H), 2.87 (br. s, 1 H), 2.15-1.91 (m, 2H), 1.80 (ddd, J = 12.7, 8.3, 6.3 Hz, 1 H), 1.72-1.57 (m, 2H), 1.42 (ddq, J = 25.5, 12.6, 8.3 Hz,

2H).

Step 3. NaH (60% dispersion in mineral oil, 12.2 mg, 0.31 mmol) was added to a solution of N-[( 1 S,2S)-2-(hydroxymethyl)cyclopentyl]-2-methoxy-6-(triazol-2- yl)benzamide (88.0 mg, 0.28 mmol) in DMF (2 mL) under N 2 . After stirring for 10 min at rt 2,5-difluoropyridine (Intermediate 43, 0.03 mL, 0.29 mmol) was added and the reaction stirred at rt overnight before addition of further 2,5-difluoropyridine (0.03 mL, 0.29 mmol) and heating at 50°C for 2 h. Further NaH (60% dispersion in mineral oil, 12.2 mg, 0.31 mmol) was added and the reaction heated for 1 h before partitioning between EtOAc (10 mL) and H 2 0 (10 mL). The aqueous phase was extracted with EtOAc (2 x 10 mL) and the combined organic phases were washed with brine (2 x 20 mL) and concentrated in vacuo. Purification by gradient flash column chromatography eluting with 0-100% EtOAc in isohexane yielded the title compound (79 mg, 0.19 mmol) as a white solid.

Data in Table 3.

Further examples prepared by the above procedures are detailed in Table 3 Table 2 - Intermediates

Table 3 - Example compounds

BIOLOGICAL ACTIVITY

Cloning, Baculovirus generation, large-scale infection of SF21 cells and membrane preparation. Human OX1 or OX2 receptor were cloned into Invitrogen’s (ThermoFisher Scientific, UK) Bac-to-Bac Baculovirus Expression System. P0 baculovirus was generated by transfecting SF9 cells with bacmid DNA using Cellfectin® II transfection reagent (ThermoFisher Scientific, UK, Catalog number 10362-100). Following P0 generation P1 virus was then generated ready for large scale infection and membrane preparation. SF21 cells were grown in expression medium ESF921 (Expression Systems, USA, catalog number 96-001-01) supplemented with 10% heat-inactivated FBS and 1 % Pen/Strep and were infected at a cell density of 2.5x10 6 cells/mL and a multiplicity of infection of 1.0 for both human OX1R and OX2R. Incubation was carried out at over 48 h in a shaking incubator set at 27°C. The cell culture was then centrifuged at 2,500 ref for 10 min at 4°C. The pellets were resuspended in cold PBS supplemented with Roche’s Complete EDTA-free protease inhibitor cocktail tablets (Roche Applied Sciences, catalog number 05056489001 ), 1 mM PMSF and 1 mM EDTA. The resuspended cell paste was then centrifuged at 3,273 ref for 12 min at 4°C. The supernatant was discarded and the pellet frozen at -80°C. The cell pellet from a 4 L culture was resuspended in buffer containing 50 mM HEPES pH 7.5, 150 mM NaCI, 8 Roche EDTA-Free protease inhibitor cocktail tablets and 1 mM PMSF. The suspension was left stirring at rt for 1 h and then homogenised for 90 s at 9,500 rpm using a VDI 25 (VWR, USA) homogeniser. The cells were then lysed using a Microfluidiser processor M-1 10L Pneumatic (Microfluidics, USA) at 60 PSI and membranes collected by ultracentrifugation at 204.7k x g for 1 h. The supernatant was discarded and the pellet was resuspended and homogenised in 90 s at 9,500 rpm in 50 mL (25 mL for each 2 L culture) of buffer containing 50 mM Hepes pH 7.5, 150 mM NaCI, 3 Roche EDTA-free protease inhibitor cocktail tablets and 1 mM PMSF. The resulting membranes were then stored at -80°C.

EXAMPLE A

[ 3 H]-radioligand binding assay. After thawing, membrane homogenates were re suspended in the binding buffer (8.5 mM HEPES, pH 7.4, 1.3 mM CaCh, 1.2 mM MgS0 4 , 1 18 mM NaCI, 4.7 mM KCI, 4 mM NaHCOs, 1.2 mM KH 2 P0 4 , 1 1 mM glucose) to a final assay concentration of 6.4 pg (OX1) or 1.4 pg (OX2) protein per well. Saturation isotherms were determined by the addition of various concentrations (0-30 nM) of [ 3 H]-4-(2,6-difluoro-4-methoxybenzyl)-2-(5,6-dimethoxypy ridin-3-yl)-2/-/- 1 ,2,4-benzothiadiazin-3(4/-/)-one 1 , 1 -dioxide (Christopher et at, MedChemComm., 2015, 6, 947-955) in a total reaction volume of 250 pl_ for 90 min at rt. At the end of the incubation, membranes were filtered onto a 96-well GF/B filter pre-incubated with 0.5% polyethylenimine, with a Tomtec cell harvester and washed 5 times with 0.5 ml_ distilled water. Non-specific binding (NSB) was measured in the presence of 3.33 mM [4-(5-chloro-1 ,3-benzoxazol-2-yl)-7-methyl-1 ,4-diazepan-1-yl][5- methyl-2-(2/-/-1 ,2,3-triazol-2-yl)phenyl]methanone (Wertz et at, Angew. Chem. !nt. Ed., 201 1 , 50, 1 151 1-11515). Radioactivity on the filter was counted (1 min) on a Microbeta radiometric plate counter (Perkin Elmer) after addition of 50 mI_ of scintillation fluid (LabLogic: Part# SG-BXX-14). For competition binding experiments, membranes were incubated with [ 3 H]-4-(2,6-difluoro-4-methoxybenzyl)-2-(5,6- dimethoxypyridin-3-yl)-2/-/-1 ,2,4-benzothiadiazin-3(4/-/)-one 1 ,1 -dioxide at a concentration equal to the K D value of the radioligand (1.5 nM for OXi and 0.75 nM for OX2 receptors respectively) and 10 concentrations of the inhibitory compound (between the ranges of 10 mM - 0.94 pmol). IC50 values were derived from the inhibition curve and the equilibrium dissociation constant (K,) values were calculated using the Cheng-Prusoff equation. The pK, values (where pK, = -logi 0 K,) of compounds of the invention are shown in Table 4.

Generation of CHO stable cell lines expressing the human OX 1 R and OX 2 R.

Stable cell lines for the human OX1R and OX2R receptors were generated through the transfection of CHO cells using the transfection reagent Genejuice (Novagen number 70967) and cDNA coding for either the human OX1R or OX2R. Cells were cultured in Sigma Nutrient Mixture F-12 Ham media (catalog number N6658) supplemented with 10% heat-inactivated FBS. Forty-eight hours after transfection, cells were harvested and placed under geneticin selection by culturing in Sigma Nutrient Mixture F-12 Ham media supplemented with 10% heat-inactivated FBS and geneticin (ThermoFisher Catalog number: 10131035). Selection of the final stable clone for each receptor was made on the basis of receptor expression, assay signal to noise and robustness though increasing passage. Selected clones for the human OX1 R and OX2R were then frozen down into liquid nitrogen using Cell Freezing Medium-DMSO (Sigma-Aldrich catalog number: C6164). EXAMPLE B

IPone accumulation assay. A CHO cell line stably expressing either human OX1 or OX2 receptor was used with the IPone HTRF assay kit (CisBio: Part# 62IPAPEJ) to measure receptor activation. The assay was optimised to measure the ability (potency; fpKb) of antagonists to reduce agonist (orexin A)-induced inositol phosphate turnover. Briefly, cells were plated onto half area 96-well white walled plates at a density of 12,500 cells/well. Sixteen hours post-plating cell growth media was replaced with antagonist concentration-response curve diluted in stimulation buffer (supplied with the kit). Cells were incubated in a humidified incubator (37 °C) for 30 min before an ECso challenge concentration (~20 nM (OX1) and 40 nM (OX2)) of orexin A (Tocris catalogue number 1455). After 30 min stimulation in a humidified incubator (37 °C) the assay was terminated by the addition of detection mixture as per manufacturer’s instructions. The concentration of compound which reduced orexin-A stimulated turnover of inositol phosphates by 50% (IC50) was calculated by fitting to a four parameter sigmoidal dose response curve. The IC50 values were converted to fpK B values using the orexin A pECso value estimated on each plate as well as the challenge concentration interpolated from the orexin A control curve. The fpK B values of compounds of the invention are shown in Table 4. Table 4 - pKi and fpK h Data

nd = not determined.




 
Previous Patent: AUTOMATED BED OR BABY COT

Next Patent: SAFETY BANKING IN STABLES