WO2018206959A1 - Orexin receptor antagonists - Google Patents

Orexin receptor antagonists Download PDF

Info

Publication number
WO2018206959A1
WO2018206959A1 PCT/GB2018/051259 GB2018051259W WO2018206959A1 WO 2018206959 A1 WO2018206959 A1 WO 2018206959A1 GB 2018051259 W GB2018051259 W GB 2018051259W WO 2018206959 A1 WO2018206959 A1 WO 2018206959A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
disorder
disease
sleep
Prior art date
Application number
PCT/GB2018/051259
Other languages
French (fr)
Inventor
David Sheppard
Daniele Andreotti
Fabio Maria Sabbatini
Original Assignee
Benevolentai Bio Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Benevolentai Bio Limited filed Critical Benevolentai Bio Limited
Publication of WO2018206959A1 publication Critical patent/WO2018206959A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system

Definitions

  • This invention relates to orexin receptor antagonists, pharmaceutical compositions comprising the antagonists, methods of making the antagonists, their use for the modulation of the orexin receptor, and to use of the antagonists as medicaments, in particular for treating diseases, disorders, or conditions mediated by orexin receptor activity.
  • Orexin (or hypocretin) signalling is mediated by two receptors and two peptide agonists.
  • the two orexin peptides (orexin A and orexin B), bind to two high affinity G protein-coupled receptors (GPCRs), the orexin-1 and orexin-2 receptors (Sakurai T. et al., Cell, 1998, 92, 573-585).
  • GPCRs G protein-coupled receptors
  • Orexin B binds preferentially to orexin-2 receptors
  • orexin A binds with equal affinity to orexin-1 and orexin-2 receptors.
  • Orexin A and orexin B (the orexins) are cleavage products of the same gene, prepro orexin.
  • neurotrophic factor the precursor from which orexin is produced, are found in the perifornical nucleus, the dorsal hypothalamus and the lateral hypothalamus. Orexinergic cells in these nuclei project to many areas of the brain, extending rostrally to the olfactory bulbs and caudally to the spinal cord.
  • the orexin system acts as an integrator of numerous physiological functions, including sleep/arousal states, sensory, locomotion, cognition, energy homeostasis, endocrine and visceral functions (reviewed by Li et al., British Journal of Pharmacology (2014) 171 , 332- 350).
  • Orexin signalling is essential for the stability of the arousal state. It is thought that the orexin system acts as a regulator of behaviour states by modulating the arousal threshold so that appropriate wakefulness can be maintained to cope with fluctuations in the external and internal environments.
  • a lack of orexin signalling is associated with narcolepsy, a disorder characterised by uncontrollable sleep episodes and hypersomnolence (Nishino et al., Lancet: 355, 39-40. 2000)).
  • Dual Orexin 1 and 2 receptor antagonists (DORAs) have proven efficacy in human patients suffering from insomnia, with Suvorexant (Belsomra) the first Orexin dual antagonist approved as an insomnia treatment.
  • OX1 R knock-out mice exhibit a subtle, barely detectable sleep phenotype, whereas OX2R knockout mice show profound hyper somnolence (Kisanuki et al., Sleep 23, A91. 2000).
  • OX2R mutations alone are responsible for the narcoleptic phenotype (Lin et al., Cell 98:365-376. 1999).
  • OX1 R antagonism may actually have detrimental effects in humans, causing an imbalance in the normal REM/NREM sleep architecture and given the brainstem localisation of OX1 R, particularly in the locus coeruleus, 0X1 R antagonists may act to attenuate vigilance state boundaries, effectively diminishing the threshold for sleep-state transitions (Dugovic et al., Front Neurosci. 8:28. 2014).
  • the evidence suggests that a selective OX2R antagonist may provide safer, more efficacious, treatments than DORAs in humans.
  • disorders of the sleep-wake cycle are, therefore, likely targets for orexin receptor (especially orexin-2 receptor) antagonist therapy.
  • disorders include sleep-wake transition disorders, insomnia, restless legs syndrome, jet-lag, disturbed sleep, and sleep disorders secondary to neurological disorders (for example, manias, depressions, manic depression, schizophrenia, and pain syndromes, such as fibromyalgia, neuropathic pain).
  • orexin system also interacts with brain dopamine systems.
  • Intracerebroventricular (i.c.v.) injections of orexins in mice increase locomotor activity, grooming and stereotypy. These effects are reversed by administration of D 2 dopamine receptor antagonists (Nakamura et al., Brain Research, 2000, 873(1), 181-7). Therefore, orexin-receptor antagonists may be useful to treat various neurological disorders, for example, Parkinson's disease, Tourette's syndrome, anxiety, delirium and dementias.
  • Brain interstitial fluid levels of amyloid-beta were demonstrated to fluctuate diurnally in both humans and rodents with sleep deprivation in rodents leading to significant increases in brain interstitial fluid levels of amyloid-beta.
  • Infusion of a dual orexin antagonist in rodents suppressed interstitial levels of amyloid-beta and abolished the natural diurnal variation of amyloid-beta.
  • Orexin neurons project to many regions of the brain associated with reward function (T. Sakurai, supra).
  • Orexin receptor antagonists are likely to be useful for the treatment of drug addiction, and alcoholism.
  • Intracisternal or intraventromedial hypothalamus injections of orexin-A stimulate gastric acid secretion by activating the vagal system through orexin-1 receptors (Takahashi et al., Biochem Biophys Res Commun, 1999, 254: 623-627; Yamada et al., Neurosci Lett, 2005, 376: 137-142; Eliassi et al., J Neuroendocrinol, 2009, 21 : 177-182; Kermani and Eliassi, Neuroscience, 2012, 226: 81-88).
  • Orexin-A can modify gastrointestinal motility, including gastric emptying, gastric interdigestive motility (Naslund et al., Am J Physiol Gastrointest Liver Physiol, 2002, 282: G470-G479; Ehrstrom et al., J Clin Endocrinol Metab, 2005, 90: 2370-2377; Ehrstrom et al., Regul Pept, 2005, 132: 9-16; Bulbul et al., Peptides, 2010, 31 : 2118-2122), and enteric peristalsis (Satoh et al., Neuropharmacology, 2006, 51 : 466 ⁇ 173), as well as colonic motility (Kirchgessner and Liu, Neuron, 1999, 24: 941-951 ; Nozu et al., Neurosci Lett, 201 1 , 498: 143-146). Orexin receptor antagonists are therefore potential treatments for ulcers, irritable bowel syndrome, diarr
  • Body weight may also be affected by orexin-mediated regulation of appetite and metabolism (T. Sakurai et al., Cell, 1998, 92(4), 573-585; T. Sakurai, Reg. Pept, 1999, 85(1), 25-30).
  • Orexins also regulate energy intake or storage and expenditure by modulating the feeding and energy homeostatis-related circuits.
  • Orexinergic neurons in the LH have a prominent role in sending the steady-state levels of physiologically relevant metabolites and integrate this information into a coherent value that is conveyed to arousal centres (Adamantidis and de Lecea, Trends Endocrinol Metab., 2008, 19: 362-370; 2009; Carter et al., Current Opin Pharmacol 2009, 9: 39-45).
  • orexinergic neurons In addition to a direct interaction with feeding circuits and with humoral signals affecting feeding, orexinergic neurons have a significant regulatory role in the brain reward system, and particularly for palatable food.
  • Orexin-1 receptor has been shown to mediate food motivation and reward-based feeding behaviour in mice and rats (Sharf et al., Brain Res, 2010, 1314: 130-138; Choi et al., Neuroscience, 2010, 167: 11-20). Orexin receptor antagonists, therefore, are likely to be useful in treatment of overweight or obesity, and conditions related to overweight or obesity, such as insulin resistance, type II diabetes, hyperlipidemia, gallstones, angina, hypertension, breathlessness, tachycardia, infertility, sleep apnoea, back and joint pain, varicose veins and osteoarthritis.
  • Orexin-deficient mice exhibit lower arterial blood pressure, heart rate and sympathetic tone (Kayaba et al., 2003, Am J Physiol Regul Integr Comp Physiol 285: R581-R593).
  • Orexin administered i.e. v., intracisternally or intrathecally increases the mean arterial pressure, heart rate, renal sympathetic nerve activity and plasma catecholamine or vasopressin levels (Samson et ai, Brain Res, 1999, 831 : 248-253; Samson et ai, Sleep Med Rev, 2005, 9: 243-252; Shirasaka et al., Am J Physiol, 1999, 277 (6 Pt 2): R1780-R1785; Shirasaka et al., Regul Pept, 2002, 104: 91-95; Matsumura et al., Hypertension, 2001 , 37: 1382-1387; Hirota et ai, Brain Res, 2003, 981 : 143-150
  • orexin receptor antagonists may be useful for treatment of hypertension, tachycardia and other arrhythmias, angina pectoris and acute heart failure. It has been suggested that orexinergic neurons are involved in sleep apnoea syndrome, as patients show increased plasma levels of orexin-A (Igarashi et ai, Chest, 2003, 124: 1381— 1385; Busquets et al., Respiration, 2004, 71 : 575-579).
  • Hypercapnia and associated reflexes may increase the activity of orexinergic neurons during sleep, facilitating microarousals and a cascade of sympathetic activity that results in elevated blood pressure during the night.
  • orexin receptor antagonists could be used to prevent these peaks of blood pressure in mild sleep apnoea.
  • the role of the orexinergic system in stress responses has been well established.
  • the term 'stress' means a subjective state perceiving or anticipating the adverse disturbances in surroundings, which further activates various stress mediators to elicit proper responses (Joels, Epilepsia, 2009, 50: 586-597). Activation of orexinergic neurons results in some stress-like effects.
  • Orexin can activate the hypothalamic-pituitary-adrenal (HPA) axis, including corticotrophin releasing factor (CRF), adrenocorticotropic hormone (ACTH) and corticosterone, stimulate stress-related behaviours, like grooming and chewing of inedible material, and enhance the activation of the monoamine system in a stress-like manner (Berridge et ai, Brain Res, 2010, 1314: 91-102).
  • CRF corticotrophin releasing factor
  • ACTH adrenocorticotropic hormone
  • corticosterone corticosterone
  • Panic attacks involve activation of the HPA axis and the autonomic system. Intrahypothalamic administration of an RNAi to orexin or an OX1 receptor antagonist has been shown to block these panic responses in rats injected with sodium lactate, and elevated levels of orexin-A have been detected in humans with panic disorder (Johnson et ai, 2010, supra). Orexin receptor antagonists may be useful for the treatment of stress, anxiety and panic disorders. Identification of orexin receptor antagonists is of great significance in the development of therapeutic agents for the treatment of a wide variety of disorders that are mediated through orexin receptors.
  • N-aroyl cyclic amine derivatives (WO 2003/002561), ethylene diamine derivatives (W02003/051872), sulfonylamino-acetic acid derivatives (W0 2004/033418), N-aryl acetyl cyclic amine derivatives (W0 2004/041791), diazepan derivatives (WO 2007/126935), amidoethylthioether derivatives (WO 2007/126934), 2-substituted proline bis-amide derivatives (WO 2008/008551), bridged diazepan derivatives (WO 2008/008517), substituted diazepan derivatives (WO 2008/008518; US 2008/0132490, WO 2009/058238), oxo bridged diazepan derivatives (WO 2008/143856), 1 ,2-diamido ethylene derivatives (WO 2009/022311), heteroaryl derivatives (WO 2009/0163485),
  • Compounds of the invention include compounds of Formula (I) and salts thereof, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labelled compounds of Formula (I).
  • the central hydrogen atoms (shown in the above structural Formula) on the methylated octahydropyrrolo[3,4-c]pyrrole group are in a cis position.
  • the compound of Formula (I) is a compound of Formula (l)(a):
  • Compounds of the invention include compounds of Formula (l)(a) and salts thereof, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labelled compounds of Formula (l)(a).
  • Compounds of the invention have been found to have surprisingly high potency as orexin receptor antagonists, in particular as selective 0X2 receptor antagonists. We have found that incorporation of the methyl substituent at position 4 of the octahydropyrrolo[3,4-c]pyrrole group provides compounds of the invention with an unexpectedly high increase in potency as an orexin receptor antagonist compared with a corresponding desmethylated compound.
  • compounds of the invention are expected to be particularly effective in the treatment of diseases, disorders or conditions mediated by orexin receptor activity, especially those associated with 0X2 receptor activity.
  • Compounds of the invention may be more effective than conventional compounds in the treatment of such diseases, disorders or conditions at similar doses to the conventional compounds.
  • compounds of the invention may be as effective as conventional compounds at lower doses than the conventional compounds but have fewer, or reduced side effects.
  • composition comprising a compound of Formula (I) or (l)(a), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.
  • a compound or composition of the invention for use as a medicament.
  • the invention also provides a compound or composition of the invention for use in treating a disease, disorder or condition mediated by orexin receptor activity.
  • a compound or composition of the invention in the manufacture of a medicament for treating a disease, disorder or condition mediated by orexin receptor activity.
  • a method of treating a disease, disorder or condition mediated by orexin receptor activity which comprises administering an effective amount of a compound of Formula (I) or (l)(a), or a pharmaceutically acceptable salt thereof, to a subject in need thereof.
  • the invention also provides a method of producing a compound of Formula (I), which comprises reacting a compound of Formula (VII) and a cyanating agent, to produce the compound of Formula (I):
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (VII) are in a cis position, thereby producing a compound of Formula (I) with corresponding hydrogen atoms in a cis position.
  • a method of producing a compound of Formula (l)(a) which comprises reacting a compound of Formula (Vll)(a) with a cyanating agent to produce the compound of Formula (l)(a):
  • the compound of Formula (I) or (l)(a) is prepared from the compound of Formula (VII) or (Vll)(a) by a copper catalysed cyanation reaction, for example, using potassium hexacyanoferrate(ll) trihydrate in the presence of a suitable ligand, such as 1-butylimidazole, and a copper catalyst, such as Cul, in a suitable solvent, such as toluene, at elevated temperature, typically 150°C.
  • a suitable ligand such as 1-butylimidazole
  • a copper catalyst such as Cul
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (VII) by reacting a compound of Formula (VI) and a brominating agent, to produce the compound of Formula (VII):
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (VI) are in a cis position, thereby producing a compound of Formula (VII) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (Vll)(a) by reacting a compound of Formula (Vl)(a) with a brominating agent to produce the compound of Formula (Vll)(a):
  • the compound of Formula (VII) is prepared from the compound of Formula (VI), or the compound of Formula (Vll)(a) is prepared from the compound of Formula (Vl)(a), by bromination, for example, using a suitable brominating agent, such as N-bromo succinimide (NBS), in a suitable solvent, such as MeCN, at between room temperature and 100°C, typically at 50°C.
  • a suitable brominating agent such as N-bromo succinimide (NBS)
  • NBS N-bromo succinimide
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (VI) by reacting a compound of Formula V) and a compound of Formula (XVIII), to produce the compound of Formula (VI):
  • LG is a leaving group.
  • suitable leaving groups include: a chloro, bromo, mesylate or tosylate group.
  • LG is chloro.
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (V) are in a cis position, thereby producing a compound of Formula (VI) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (Vl)(a) by reacting a compound of Formula (V)(a) and a compound of Formula (XVIII), to produce the compound of Formula (Vl)(a):
  • the compound of Formula (V) or (V)(a) is reacted with the compound of Formula (XVII I) in the presence of a suitable base, such as CS2CO3 or K2CO3, in a polar solvent, such as DMF or EtOH.
  • a suitable base such as CS2CO3 or K2CO3
  • a polar solvent such as DMF or EtOH.
  • the base is CS2CO3
  • the solvent is DMF
  • the reaction is carried out at an elevated temperature, preferably 100°C.
  • the compound of Formula (XVIII) is 6-Chloro-4,6-dimethylpyrimidine (i.e. LG is chloro).
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (V) by deprotection of a compound of Formula (IV):
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (IV) are in a cis position, thereby producing a compound of Formula (V) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (V)(a) by deprotection of a compound of Formula (IV)(a):
  • the deprotection is carried out by transfer or palladium-catalyzed hydrogenation.
  • palladium catalyzed hydrogenation may be carried out using 10% palladium on charcoal, in EtOH under an atmosphere of hydrogen at room temperature.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (IV) by reacting a compound of Formula (III) with 2-fluoro-4-methoxybenzoic acid to form the compound of Formula (IV):
  • PG2 is an amine-protecting group.
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (III) are in a cis position, thereby producing a compound of Formula (IV) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (IV)(a) by reacting a compound of Formula (lll)(a) with 2-fluoro-4-methoxybenzoic acid to form the compound of Formula (IV)(a):
  • PG2 is an amine-protecting group.
  • the compound of Formula (III) or (lll)(a) is reacted with 2-fluoro-4- methoxybenzoic acid in the presence of a coupling reagent, such as HATU or T3P, and an organic base, such as ⁇ or DIPEA, in an aprotic solvent, such as MeCN or DMF, at a temperature from 0-100°C.
  • a coupling reagent such as HATU or T3P
  • an organic base such as ⁇ or DIPEA
  • an aprotic solvent such as MeCN or DMF
  • 2-fluoro-4-methoxybenzoic acid is converted in situ to 2-fluoro-4-methoxybenzoyl chloride, by treatment with a suitable reagent, such as SOCI2 or PCI3, in a non polar solvent, such as toluene, at elevated
  • the compound of Formula (III) or (lll)(a) is reacted with 2-fluoro-4- methoxybenzoyl chloride, prepared in situ from 2-fluoro-4-methoxybenzoic acid, in the presence of ⁇ in MeCN, preferably at room temperature.
  • 2-fluoro-4-methoxybenzoic acid is treated with SOCI2, in toluene at 80°C to prepare 2-fluoro-4-methoxybenzoyl chloride in situ, followed by reaction with a compound of Formula (III) or (lll)(a) in the presence of ⁇ in DCM at room temperature.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (III) by reduction of a compound of Formula (II) to produce the compound of Formula (III):
  • PG2 is an amine-protecting group.
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (II) are in a cis position, thereby producing a compound of Formula (III) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (lll)(a) by reduction of a compound of Formula (ll)(a) to produce the com ound of Formula (lll)(a):
  • PG2 is an amine-protecting group.
  • the reduction is carried out using a reducing agent in an aprotic solvent at a temperature from 0°C to reflux.
  • the reducing agent is UAIH4
  • the aprotic solvent is 2-MeTHF
  • the reduction is preferably carried out at room temperature.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (II) by deprotection of a compound of Formula (XIII):
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XIII) are in a cis position, thereby producing a compound of Formula (II) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (ll)(a) by deprotection of a compound of Formula (Xlll)(a):
  • PGi is an amine-protecting group and PG2 is an amine-protecting group.
  • the deprotection is an acid-mediated deprotection in an aprotic solvent.
  • the acid is TFA
  • the solvent is DCM
  • the deprotection is preferably carried out at a temperature from 0°C to room temperature.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XIII) from compounds of Formulae (XII) and (XIX) by a cyclisation reaction:
  • PGi is an amine-protecting group and PG2 is an amine-protecting group.
  • the choice of reagent(s) for use in the cyclisation will depend on which stereoisomer, or mixture of stereoisomers, is to be produced.
  • the cyclisation reaction is selected such that the central hydrogen atoms on the methylated octahydropyrrolo[3,4-c]pyrrole group of the compound of Formula (XI II) are in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XI I l)(a) from a compound of Formula (Xll)(a) and a compound of Formula (XIX), by an acid-mediated cyclisation reaction:
  • PGi is an amine-protecting group and PG2 is an amine-protecting group.
  • the acid-mediated cyclisation reaction is carried out in the presence of an acid in an aprotic solvent.
  • the acid is TFA
  • the aprotic solvent is DCM
  • the reaction is preferably carried out at a temperature from 0°C to room temperature.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise preparing a compound of Formula (Xll)(a) from a compound of Formula (Vlll)(a), (IX)(a), (X)(a), (Xl)(a), and (Xll)(a), as illustrated by Scheme 2 below.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise preparing a compound of Formula (Xll)(a) from a compound of Formula (XIV)(a), (XV)(a), (XVI)(a), and (XVII)(a) as illustrated by Scheme 3 below.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise preparing a compound of Formula (Vl)(a) from a compound of Formula (lll)(a), (XX)(a), (XXI)(a), (XVIII), (XXII)(a) and (XXIII)(a) as illustrated by Scheme 4 below.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (VI) by reacting a compound of Formula (XXIII) and 2-fluoro-4-methoxybenzoic acid to produce the compound of Formula (VI):
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XXIII) are in a cis position, thereby producing a compound of Formula (VI) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (Vl)(a) by reacting a compound of Formula (XXIII)(a) and 2-fluoro-4-methoxybenzoic acid to produce the compound of Formula
  • the compound of Formula (XXIII) or (XXIII)(a) and 2-fluoro-4- methoxybenzoic acid are reacted in the presence of a coupling reagent and an organic base, in an aprotic solvent, at a temperature from 0-100°C.
  • a coupling reagent is HATU, or T3P
  • the organic base is DIPEA or Et 3 N
  • the aprotic solvent is DMF or MeCN
  • the temperature is preferably room temperature.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XXIII) by deprotection of a compound of Formula (XXII):
  • PGi is an amine-protecting group
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XXII) are in a cis position, thereby producing a compound of Formula (XXIII) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XXIII)(a) by deprotection of a compound of Formula (XXII)(a): CH
  • the deprotection is an acid-mediated deprotection in an aprotic solvent.
  • the acid is TFA
  • the solvent is DCM
  • the deprotection is preferably carried out at a temperature from 0°C to room temperature.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XXII) by reacting a compound of Formula (XXI) and a compound of Formula (XVIII) to produce the compound of Formula (XXII):
  • PGi is an amine-protecting group
  • LG is a leaving group
  • Suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group.
  • LG is chloro.
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XXI) are in a cis position, thereby producing a compound of Formula (XXII) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XXII)(a) by reacting a compound of Formula (XXI)(a) and a compound of Formula (XVIII) to produce the compound of Formula (XXII)(a):
  • the compound of Formula (XXI) or (XXI)(a) is reacted with the compound of Formula (XVI) in the presence of a base in a solvent.
  • the compound of Formula (XXI) or (XXI)(a) is reacted with the compound of Formula (XVIII) in the presence of a base in a solvent.
  • a base is CS2CO3 or K2CO3
  • the solvent is DMF or EtOH, and the reaction is carried out at an elevated temperature, such as 80-1 10°C.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XXI) by deprotection of a compound of Formula (XX) to produce the compound of Formula (XXI):
  • PGi is an amine-protecting group
  • PG2 IS an amine-protecting group
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XX) are in a cis position, thereby producing a compound of Formula (XXI) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XXI)(a) by deprotection of a compound of Formula (XX)(a) to produce the compound of Formula (XXI)(a):
  • the deprotection is carried out by transfer palladium catalysed hydrogenation.
  • transfer hydrogenation may be carried out using ammonium formate and Pd(OH) 2 on charcoal, in ethanol, at a temperature between room temperature and reflux.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XX) by protecting a compound of Formula (III) to produce the compound of Formula (XX):
  • the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (III) are in a cis position, thereby producing a compound of Formula (XX) with corresponding hydrogen atoms in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XX)(a) by protecting a
  • PGi is an amine-protecting group
  • PG2 IS an amine-protecting group.
  • protection is carried out under standard conditions for introducing an amine protecting group.
  • protection is carried out by treatment with B0C2O in a suitable solvent, such as DCM, typically at room temperature.
  • PGi is an amine-protecting group
  • PG2 is an amine- protecting group
  • the cyclisation reaction is selected such that the central hydrogen atoms on the methylated octahydropyrrolo[3,4-c]pyrrole group of the compound of Formula (XIII) are in a cis position.
  • PGi is an amine-protecting group
  • PG2 is an amine- protecting group
  • a method of the invention for producing a compound of Formula (XIII) further comprises converting the compound of Formula (XIII) to a compound of
  • a method of the invention for producing a compound of Formula (XIH)(a) further comprises converting the compound of Formula (XI I l)(a) to a compound of Formula (l)(a).
  • methods of the invention for producing a compound of Formula (I) further comprise reacting the compound of Formula (I) to yield a corresponding
  • methods of the invention for producing a compound of Formula (l)(a) further comprise reacting the compound of Formula (l)(a) to yield a corresponding pharmaceutically acceptable salt of the compound of Formula (l)(a).
  • Suitable amine protecting groups are well-known to the skilled person, and include those described in 'Greene's Protective Groups in Organic Synthesis' by Theodora W Greene and Peter G M Wuts, fifth edition, (John Wiley and Sons, 2014), in particular Chapter 7 ("Protection for the Amino Group”), incorporated herein by reference, which also describes methods for the removal of such groups.
  • Suitable amine protecting groups include the following: Carbobenzyloxy (Cbz) group - removed by hydrogenolysis;
  • DMPM 3,4-Dimethoxybenzyl
  • PMP p-methoxyphenyl
  • CAN ammonium cerium(IV) nitrate
  • Ts group - removed by concentrated acid (HBr, H2SO4) and strong reducing agents (sodium in liquid ammonia or sodium naphthalenide); Trichloroethyl chloroformate (Troc) group - removed by Zn insertion in the presence of acetic acid;
  • PGi and PG2 may each independently be selected from: a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, tert- butylcarbamate, 9-Fluorenylmethyl carbamate, benzhydryl, trityl group.
  • PGi is a carbamate group, preferably f-butyl carbamate
  • PG2 is benzyl
  • PGi is a carbamate group, preferably f-butyl carbamate, and PG2 is benzyl.
  • Suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group.
  • LG is chloro.
  • the compounds of the invention include compounds of Formula (I), or (l)(a), and salts thereof as hereinafter defined, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labelled compounds of Formula (I) or (l)(a).
  • any Formula given herein is intended to represent compounds having structures depicted by the structural Formula as well as certain variations or forms.
  • compounds of any Formula given herein may have asymmetric centres and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general Formula, and mixtures thereof, are considered within the scope of the Formula.
  • any Formula given herein is intended to represent a racemate, one or more enantiomeric forms, one or more diastereomeric forms, one or more atropisomeric forms, and mixtures thereof.
  • certain structures may exist as geometric isomers (i.e., cis and trans isomers), as tautomers, or as atropisomers.
  • tautomeric isomerism ('tautomerism') can occur. It follows that a single compound may exhibit more than one type of isomerism.
  • Examples of types of potential tautomerisms shown by the compounds of the invention include; amide hydroxyl-imine and keto enol tautomersims:
  • Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, by chromatography and fractional crystallisation.
  • Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or other derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
  • Chiral compounds of the invention may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on a resin with an asymmetric stationary phase and with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% ethanol, typically from 2 to 20%. Concentration of the eluate affords the enriched mixture. Mixtures of stereoisomers may be separated by conventional techniques known to those skilled in the art (see, for example, "Stereochemistry of Organic Compounds" by E L Eliel (Wiley, New York, 1994)).
  • any Formula given herein is intended to refer also to hydrates, solvates, and polymorphs of such compounds, and mixtures thereof, even if such forms are not listed explicitly.
  • a compound of Formula (I), or (l)(a), or pharmaceutically acceptable salt of a compound of Formula (I), or (l)(a) may be obtained as a solvate.
  • Solvates include those formed from the interaction or complexation of compounds of the invention with one or more solvents, either in solution or as a solid or crystalline form. In some embodiments, the solvent is water and then the solvates are hydrates.
  • certain crystalline forms of a compound of Formula (I), or (l)(a), or a pharmaceutically acceptable salt of a compound of Formula (I), or (l)(a), may be obtained as co-crystals.
  • a compound of Formula (I), or (l)(a), or a pharmaceutically acceptable salt of a compound of Formula (I), or (l)(a) may be obtained in a crystalline form.
  • a compound of Formula (I), or (l)(a) may be obtained in one of several polymorphic forms, as a mixture of crystalline forms, as a polymorphic form, or as an amorphous form.
  • a compound of Formula (I), or (l)(a) may convert in solution between one or more crystalline forms and/or polymorphic forms.
  • any Formula given herein is also intended to represent unlabelled forms as well as isotopically labelled forms of the compounds.
  • Isotopically labelled compounds have structures depicted by the Formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, and fluorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 18 F, respectively.
  • Such isotopically labelled compounds are useful in metabolic studies (preferably with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques (such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)) including drug or substrate tissue distribution assays, or in radioactive treatment of subjects.
  • positron emission tomography PET
  • SPECT single-photon emission computed tomography
  • Substitution with positron emitting isotopes such as 11 C, 18 F, 15 0 and 13 N, can be useful in PET studies for examining substrate receptor occupancy.
  • an 18 F or 11 C labelled compound may be particularly preferred for PET studies.
  • isotopically-labelled compounds of Formula (I) for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • Isotopically labelled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
  • a compound of Formula (I), or (l)(a), and pharmaceutically acceptable salts thereof may be used alone or in combination with one or more additional active ingredients to Formulate pharmaceutical compositions.
  • a pharmaceutical composition therefore comprises an effective amount of a compound of Formula (I), or (l)(a), or a pharmaceutically acceptable salt thereof.
  • the invention includes also pharmaceutically acceptable salts of a compound of Formula (I) or (l)(a).
  • a "pharmaceutically acceptable salt” is intended to mean a salt of a free acid or base of a compound represented by Formula (I), or (l)(a), that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to a subject. See, generally, G.S. Paulekuhn, et al., "Trends in Active Pharmaceutical Ingredient Salt Selection based on Analysis of the Orange Book Database", J. Med. Chem., 2007, 50:6665-72, S.M.
  • Examples of pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of subjects without undue toxicity, irritation, or allergic response.
  • a compound of Formula (I), or (l)(a) may possess a sufficiently acidic group, a sufficiently basic group, or both types of functional groups, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt, bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • Examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen- phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1 ,4-dioates, hexyne-1 ,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates
  • the invention also relates to pharmaceutically acceptable prodrugs of a compound of Formula (I) or (l)(a), and treatment methods employing such pharmaceutically acceptable prodrugs.
  • prodrug means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodrug on being brought to physiological pH is converted to the compound of Formula (I), or (l)(a)).
  • a “pharmaceutically acceptable prodrug” is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject.
  • the present invention also relates to pharmaceutically active metabolites of a compound of Formula (I), or (l)(a), which may also be used in the methods of the invention.
  • a "pharmaceutically active metabolite” means a pharmacologically active product of metabolism in the body of a compound of Formula (I), or (l)(a), or salt thereof.
  • Prodrugs and active metabolites of a compound may be determined using routine techniques known or available in the art. See, e.g., Bertolini, et a/., J Med Chem.
  • a compound of Formula (I), or (l)(a), and its pharmaceutically acceptable salts, pharmaceutically acceptable prodrugs, and pharmaceutically active metabolites, are useful as antagonists of the orexin receptor.
  • the term “treat”, “treating”, or “treatment” as used herein is intended to refer to administration of a compound or composition of the invention to a subject for the purpose of effecting a therapeutic or prophylactic benefit through modulation (in particular, antagonism) of orexin receptor activity. Treating includes reversing, ameliorating, alleviating, inhibiting the progress of, lessening the severity of, or preventing a disease, disorder, or condition, or one or more symptoms of such disease, disorder or condition mediated through modulation of orexin receptor activity.
  • subject refers to a mammalian patient in need of such treatment, such as a human.
  • the invention relates to methods of using the compounds or compositions described herein to treat subjects diagnosed with or suffering from a disease, disorder, or condition mediated by orexin receptor activity, such as: disorders of the sleep-wake cycle, metabolic disorders, neurological disorders, and other disorders (e.g., feeding, drinking, arousal, stress, addiction, especially drug addiction, metabolism and reproduction).
  • a disease, disorder, or condition mediated by orexin receptor activity such as: disorders of the sleep-wake cycle, metabolic disorders, neurological disorders, and other disorders (e.g., feeding, drinking, arousal, stress, addiction, especially drug addiction, metabolism and reproduction).
  • Sleep disorders include, but are not limited to, disorders of the sleep-wake cycle, sleep-wake transition disorders, insomnia, restless legs syndrome, jet-lag, disturbed sleep, and sleep disorders secondary to neurological disorders (e.g., manias, depressions, manic depression, schizophrenia, and pain syndromes (e.g., fibromyalgia, neuropathic).
  • neurological disorders e.g., manias, depressions, manic depression, schizophrenia, and pain syndromes (e.g., fibromyalgia, neuropathic).
  • Metabolic disorders include, but are not limited to, overweight or obesity and conditions related to overweight or obesity, such as insulin resistance, type II diabetes, hyperlipidemia, gallstones, angina, hypertension, breathlessness, tachycardia, arrhythmias, angina pectoris, acute heart failure, infertility, sleep apnoea, back and joint pain, varicose veins and osteoarthritis.
  • Neurological disorders include, but are not limited to, Parkinson's disease, Alzheimer's disease, Huntington's disease, Tourette's Syndrome, catatonia, anxiety, stress disorder, panic disorder, delirium and dementias.
  • Other disorders include, but are not limited to, ulcers, irritable bowel syndrome, diarrhoea and gastroesophageal reflux.
  • compounds of the invention are particularly advantageous for treatment of various diseases (such as, but not restricted to, sleep disorders) because they exhibit rapid and high levels of intestinal absorption, leading to rapid achievement of maximal concentration in the body and the brain, and a sufficient but relatively short exposure period.
  • an effective amount of a compound of the invention is administered to a subject suffering from or diagnosed as having such a disease, disorder, or condition.
  • An "effective amount” means an amount or dose sufficient to generally bring about the desired therapeutic or prophylactic benefit in a subject in need of such treatment for the designated disease, disorder, or condition.
  • Effective amounts or doses of a compound of the present invention may be ascertained by routine methods such as modelling, dose escalation studies or clinical trials, and by taking into consideration routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the compound, the severity and course of the disease, disorder, or condition, the subject's previous or ongoing therapy, the subject's health status and response to drugs, and the judgment of the treating physician.
  • An example of a dose is in the range of from about 0.001 to about 200 mg of compound per kg of subject's body weight per day, preferably about 0.05 to 100 mg/kg/day, or about 1 to 35 mg/kg/day, in single or divided dosage units (e.g., BID, TID, QID).
  • an illustrative range for a suitable dosage amount is from about 0.05 to about 7 g/day, or about 0.2 to about 2.5 g/day.
  • the dose may be adjusted for preventative or maintenance treatment.
  • the dosage or the frequency of administration, or both may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained.
  • treatment may cease.
  • Subjects may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • the active agents of the invention may be used in combination with additional active ingredients in the treatment of the above conditions.
  • the additional active ingredients may be co-administered separately with an active agent of the invention, or included with such an agent in a pharmaceutical composition according to the invention.
  • additional active ingredients are those that are known or discovered to be effective in the treatment of conditions, disorders, or diseases mediated by orexin receptor activity, such as another orexin modulator or a compound active against another target associated with the particular condition, disorder, or disease.
  • the combination may serve to increase efficacy (e.g., by including in the combination a compound potentiating the potency or effectiveness of an active agent according to the invention), decrease one or more side effects, or decrease the required dose of the active agent according to the invention.
  • the active agents of the invention are used, alone or in combination with one or more additional active ingredients, to Formulate pharmaceutical compositions of the invention.
  • a pharmaceutical composition of the invention comprises: (a) an effective amount of at least one active agent in accordance with the invention; and (b) a pharmaceutically acceptable excipient.
  • a "pharmaceutically acceptable excipient” refers to a substance that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to a subject, such as an inert substance, added to a pharmacological composition or otherwise used as a vehicle, carrier, or diluent to facilitate administration of an agent and that is compatible therewith.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.
  • compositions containing one or more dosage units of the active agents may be prepared using suitable pharmaceutical excipients and compounding techniques known or that become available to those skilled in the art.
  • the compositions may be administered in the inventive methods by a suitable route of delivery, e.g., oral, parenteral, rectal, topical, or ocular routes, or by inhalation.
  • the preparation may be in the form of tablets, capsules, sachets, dragees, powders, granules, lozenges, powders for reconstitution, liquid preparations, or suppositories.
  • the compositions are Formulated for intravenous infusion, topical administration, or oral administration.
  • the compounds of the invention can be provided in the form of tablets or capsules, or as a solution, emulsion, or suspension.
  • the compounds may be Formulated to yield a dosage of, e.g., from about 0.05 to about 100 mg/kg daily, or from about 0.05 to about 35 mg/kg daily, or from about 0.1 to about 10 mg/kg daily.
  • a total daily dosage of about 5 mg to 5 g daily may be accomplished by dosing once, twice, three, or four times per day.
  • Oral tablets may include a compound according to the invention mixed with pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservative agents.
  • suitable inert fillers include sodium and calcium carbonate, sodium and calcium phosphate, lactose, starch, sugar, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, and the like.
  • Exemplary liquid oral excipients include ethanol, glycerol, water, and the like.
  • Starch, polyvinyl-pyrrolidone (PVP), sodium starch glycolate, microcrystalline cellulose, and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin.
  • the lubricating agent if present, may be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract, or may be coated with an enteric coating.
  • Capsules for oral administration include hard and soft gelatin capsules.
  • compounds of the invention may be mixed with a solid, semi-solid, or liquid diluent.
  • Soft gelatin capsules may be prepared by mixing the compound of the invention with water, an oil such as peanut oil or olive oil, liquid paraffin, a mixture of mono and di-glycerides of short chain fatty acids, polyethylene glycol 400, or propylene glycol.
  • Liquids for oral administration may be in the form of suspensions, solutions, emulsions or syrups or may be lyophilized or presented as a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid compositions may optionally contain: pharmaceutically-acceptable excipients such as suspending agents (for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminium stearate gel and the like); non-aqueous vehicles, e.g., oil (for example, almond oil or fractionated coconut oil), propylene glycol, ethyl alcohol, or water; preservatives (for example, methyl or propyl p-hydroxybenzoate or sorbic acid); wetting agents such as lecithin; and, if desired, flavouring or colouring agents.
  • suspending agents for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethyl
  • compositions may be Formulated for rectal administration as a suppository.
  • parenteral use including intravenous, intramuscular, intraperitoneal, or subcutaneous routes, the compounds of the invention may be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity or in parenterally acceptable oil.
  • Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride.
  • Such forms will be presented in unit-dose form such as ampules or disposable injection devices, in multi- dose forms such as vials from which the appropriate dose may be withdrawn, or in a solid form or pre-concentrate that can be used to prepare an injectable Formulation.
  • Illustrative infusion doses may range from about 1 to 1000 ⁇ g/kg/minute of compound, admixed with a pharmaceutical carrier over a period ranging from several minutes to several days.
  • the compounds may be mixed with a pharmaceutical carrier at a concentration of about 0.1 % to about 10% of drug to vehicle.
  • a pharmaceutical carrier for topical administration, may be mixed with a pharmaceutical carrier at a concentration of about 0.1 % to about 10% of drug to vehicle.
  • Another mode of administering the compounds of the invention may utilize a patch Formulation to affect transdermal delivery.
  • Compounds of the invention may alternatively be administered in methods of this invention by inhalation, via the nasal or oral routes, e.g., in a spray Formulation also containing a suitable carrier.
  • HATU (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate)
  • the skilled person will appreciate that it may be necessary or desirable at any stage in the synthesis of compounds of the invention to protect one or more sensitive groups, so as to prevent undesirable side reactions.
  • the protecting groups used in the preparation of the compounds of the invention may be used in a conventional manner. See, for example, those described in 'Greene's Protective Groups in Organic Synthesis' by Theodora W Greene and Peter G M Wuts, fifth edition, (John Wiley and Sons, 2014), in particular Chapter 7 ("Protection for the Amino Group"), incorporated herein by reference, which also describes methods for the removal of such groups.
  • the compounds of the invention may be prepared by any method known in the art for the preparation of compounds of analogous structure.
  • the compound of the invention can be prepared by the procedures described by reference to the Schemes that follow, or by the specific methods described in the Examples, or by similar processes to either.
  • a compound of Formula (l)(a) may be prepared from compounds of Formulae (ll)(a),
  • PGi and PG2 are each independently an amine protecting group; LG is a leaving group.
  • PGi and PG 2 may each independently be selected from: a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, tert- butylcarbamate, 9-Fluorenylmethyl carbamate, benzhydryl, trityl group.
  • PGi is a carbamate group, preferably f-butyl carbamate, and PG2 is benzyl.
  • Suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group.
  • LG is chloro.
  • a suitable acid such as TFA
  • a suitable aprotic solvent such as DCM
  • a compound of Formula (lll)(a) may be prepared from a compound of Formula (l)(a) using a suitable reducing agent, such as UAIH4, in a suitable aprotic solvent, such as 2-MeTHF, at a suitable temperature, such as between 0°C and reflux, preferably at room temperature.
  • a compound of Formula (IV)(a) may be prepared from a compound of Formula (lll)(a) and 2- fluoro-4-methoxybenzoic acid using an amide bond forming reaction in the presence of a suitable coupling reagent, such as (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5-b]pyridinium
  • HATU 3-oxid hexafluorophosphate
  • T3P propylphosphonic anhydride
  • a compound of Formula (IV)(a) may be prepared from a compound of Formula (lll)(a) and 2-fluoro-4- methoxybenzoyl chloride, prepared in situ from 2-fluoro-4-methoxybenzoic acid, by treatment with a suitable reagent such as SOCI2 or PCI3 in a non polar solvent such as toluene at elevated temperature, followed by reaction with a compound of Formula (lll)(a), in the presence of a suitable organic base, such as EhN or DIPEA in a suitable solvent such as DCM.
  • a suitable organic base such as EhN or DIPEA in a suitable solvent such as DCM.
  • suitable deprotection conditions such as transfer or palladium catalysed hydrogenation, using for example palladium on charcoal in a suitable solvent, such as EtOH, at a suitable temperature, such as rt, under an atmosphere of hydrogen.
  • a compound of Formula (Vl)(a) may be prepared from a compound of Formula (V)(a) and a compound of Formula (XVI I I) in the presence of a suitable base, such as Cs 2 C0 3 or K2CO 3 , in a suitable solvent, such as DMF or EtOH, at an elevated temperature, such as 80-100°C.
  • a suitable base such as Cs 2 C0 3 or K2CO 3
  • a suitable solvent such as DMF or EtOH
  • a compound of Formula (Vl l)(a) may be prepared from a compound of Formula (Vl)(a) by a bromination using a suitable brominating agent such as N-bromo succinimide (NBS) in a suitable solvent such as MeCN at between room temperature and 100°C, typically at 50°C.
  • a suitable brominating agent such as N-bromo succinimide (NBS)
  • NBS N-bromo succinimide
  • a compound of Formula (l)(a) may be prepared from a compound of Formula (Vl l)(a) by a cyanation reaction using a suitable cyanating agent such as potassium hexacyanoferrate(l l) trihydrate in the presence of a suitable copper catalyst such as Cul and a suitable ligand such as 1-butylimidazole in a suitable solvent such as toluene at elevated temperature, typically 150°C.
  • a suitable cyanating agent such as potassium hexacyanoferrate(l l) trihydrate
  • a suitable copper catalyst such as Cul
  • a suitable ligand such as 1-butylimidazole
  • a compound of Formula (Xl l)(a) may be prepared from a compound of Formula (VI I l)(a), (IX)(a), (X)(a), and (Xl)(a), as illustrated by Scheme 2:
  • PGi is an amine protecting group, as hereinbefore defined.
  • a compound of Formula (IX)(a) may be prepared by condensation of Boc-L-alanine (VI I l)(a) and Meldrum's acid in the presence of a suitable condensation reagent, such as EDC.HCI, optionally in the presence of a suitable catalyst, such as DMAP, in a suitable solvent, such as DCM, at a suitable temperature, such as between 0°C and rt.
  • a suitable condensation reagent such as EDC.HCI
  • a suitable catalyst such as DMAP
  • DCM suitable solvent
  • suitable temperature such as between 0°C and rt.
  • a compound of Formula (X)(a) may be prepared from a compound of Formula (IX)(a) by treatment with heat in a suitable solvent, such as EtOAc, at an appropriate elevated temperature, between 60°C and reflux, optionally at reduced pressure.
  • a suitable solvent such as EtOAc
  • a compound of Formula (X)(a) may be obtained directly from Boc-L-alanine (VI I l)(a), without isolating the compound of Formula (IX)(a).
  • a compound of Formula (Xl)(a) may be prepared by the reduction of a compound of Formula (X)(a) in the presence of a suitable catalyst such as PtC>2 in the presence of hydrogen gas at a suitable pressure in a solvent such as EtOAc.
  • this transformation may be achieved by treatment of a compound of Formula (X)(a) with a suitable reducing agent such as NaBhU in the presence of an acid such as AcOH is a suitable solvent such as DCM at between 0°C and room temperature.
  • a compound of Formula (Xll)(a) may be prepared from a compound of Formula (Xl)(a) using a suitable base-mediated elimination reaction using, for instance an in situ conversion of the alcohol into a methansulfonyl leaving group in the presence of a suitable non-nucleophilic base, such as Et 3 N, in a suitable solvent, such as DCM, at a suitable temperature, such as 0°C to rt.
  • a suitable base-mediated elimination reaction using, for instance an in situ conversion of the alcohol into a methansulfonyl leaving group in the presence of a suitable non-nucleophilic base, such as Et 3 N, in a suitable solvent, such as DCM, at a suitable temperature, such as 0°C to rt.
  • a suitable base-mediated elimination reaction using, for instance an in situ conversion of the alcohol into a methansulfonyl leaving group in the presence of a suitable non-nucleophilic base, such as Et 3 N, in a suitable
  • a compound of Formula (Xll)(a) may alternatively be prepared from a compound of Formula (XIV)(a), (XV)(a), (XVI)(a), and (XVII)(a) as illustrated by Scheme 3:
  • PGi is an amine protecting group, as hereinbefore defined.
  • a compound of Formula (XV)(a) may be prepared from (2S)-2-methylpyrrolidine hydrochloride and B0C2O in a suitable solvent, such as DCM, in the presence of a suitable base, such as Et 3 N.
  • a compound of Formula (XVI)(a) may be prepared by the oxidation of a compound of Formula (XV) using appropriate oxidising agents such as ruthenium(IV)oxide hydrate and sodium metaperiodate in water at a suitable temperature, such as 15-20°C.
  • a compound of Formula (XVI I)(a) may be prepared from a compound of Formula (XVI)(a) by deprotonation using a suitable non-nucleophilic base, such as LHMDS, in a suitable aprotic solvent, such as THF, at low temperature, such as -78°C, followed by quenching with a suitable electrophilic source of phenylselenide, such as PhSeBr, at a suitable temperature, such as rt.
  • a compound of Formula (Xll)(a) may be prepared from a compound of Formula (XVII)(a) by an oxidative elimination reaction using a suitable oxidising agent, such as MCPBA, in a suitable solvent, such as DCM, at a suitable low temperature, such as -40°C.
  • a suitable oxidising agent such as MCPBA
  • a suitable solvent such as DCM
  • a compound of Formula (Vll)(a) may alternatively be prepared from a compound of Formula (XX)(a), (XXI)(a), (XVIII) and (XXII)(a) as illustrated by Scheme 4:
  • PGi and PG2 are each independently an amine protecting group; LG is a leaving group.
  • PGi and PG 2 may each independently be selected from: a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, tert- butylcarbamate, 9-Fluorenylmethyl carbamate, benzhydryl, trityl group.
  • PGi is a carbamate group, preferably f-butyl carbamate, and PG2 is benzyl.
  • suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group.
  • LG is chloro.
  • a compound of Formula (XX)(a) may be prepared from a compound of Formula (lll)(a) under standard conditions for introducing an amine protecting group.
  • PGi 'BoC
  • a compound of Formula (lll)(a) is treated with Boc 2 0 in a suitable solvent such as DCM, typically at room temperature to provide a compound of Formula (XX)(a).
  • suitable deprotection conditions such as transfer or palladium catalysed hydrogenation, using for example ammonium formate and palladium hydroxide on charcoal in EtOH at reflux.
  • suitable deprotection conditions such as transfer or palladium catalysed hydrogenation, using for example ammonium formate and palladium hydroxide on charcoal in EtOH at reflux.
  • suitable deprotection conditions such as transfer or palladium catalysed hydrogenation, using for example ammonium formate and palladium hydroxide on charcoal in EtOH at reflux.
  • a skilled person will appreciate that alternative hydrogenation conditions are available to effect this transformation.
  • a compound of Formula (XXII)(a) may be prepared
  • a compound of Formula (Vl)(a) may be prepared from a compound of Formula (XXIII)(a) and 2-fluoro-4-methoxybenzoic acid using an amide bond forming reaction in the presence of a suitable coupling reagent, such as (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate) (HATU) or propylphosphonic anhydride (T3P), in the presence of a suitable organic base, such as DIPEA or Et 3 N, in an appropriate aprotic solvent, such as DMF or MeCN, at a suitable temperature, such as 0-100°C, preferably at rt.
  • a suitable coupling reagent such as (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate) (
  • a compound of Formula (I) or (l)(a) may be converted to its corresponding salt using methods known to those skilled in the art.
  • amines of Formula (I) or (l)(a) may be treated with trifluoroacetic acid (TFA), HCI, sulfuric acid (H2SO4), maleic acid, or citric acid in a solvent such as diethyl ether (Et20), CH2CI2, tetrahydrofuran (THF), or methanol (MeOH) to provide the corresponding salt forms.
  • Compounds prepared according to the schemes described above may be obtained as single enantiomers, diastereomers, or regioisomers, by enantio- , diastero-, or regiospecific synthesis, or by resolution.
  • Compounds prepared according to the schemes above may alternately be obtained as racemic (1 : 1) or non-racemic (not 1 : 1) mixtures or as mixtures of diastereomers or regioisomers.
  • single enantiomers may be isolated using conventional separation methods known to one skilled in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation.
  • regioisomeric or diastereomeric mixtures are obtained, single isomers may be separated using conventional methods such as chromatography or crystallization.
  • EtOAc (3.9 L) was added again and the solution maintained at 0°C for 18 hrs.
  • the solution was heated to 80°C with an internal vacuum of 800 mBar in distillation mode (internal temperature 66-67°C) and stirred for 1.5 hr.
  • the solvent was evaporated to minimum volume (1 L) and the solution used in the next step without further purification. A drop of this solution was evaporated to obtain an analytical sample.
  • the organic phase was washed with water (2 L) then passed through a thin layer of Celite® to break the emulsion.
  • the organic solution was washed again with water (2 L) filtered through a thin layer of Celite® then charged into a reactor.
  • the solution was cooled to 0 °C and allowed to stand for 15 hrs. The temperature was increased to 20 °C, the solvent evaporated to dryness, then azeotroped with MeTHF to obtain the title compound as a dark orange oil, 380 g, in quantitative yield.
  • 2-Fluoro-4-methoxybenzoic acid 35g, 204 mmol was suspended in SOCI2 (70 mL) and the mixture stirred at 80 °C for 5 hr. The mixture was evaporated under vacuum and azeotroped with toluene (3x50 mL). The resulting oil was dissolved in DCM (300 mL), cooled to 0 °C and Et 3 N (85 mL, 582 mmol) added.
  • Potassium hexacyanoferrate(ll) trihydrate (9.12 g, 21.6 mmol) was dried in vacuo at 80°C for 12 hrs.
  • Cul (4.9 g, 26.0 mmol) and 2-[(3aS,4S,6aR)-5-(2-fluoro-4-methoxybenzoyl)-4-methyl- octahydropyrrolo[3,4-c]pyrrol-2-yl]-5-bromo-4,6-dimethylpyrimidine Preparation 10, 10 g, 21.6 mmol
  • the reaction mixture was stirred in toluene (30.16 ml_) and 1- butylimidazole (5.8 ml_) at 150°C for 19 hrs.
  • the cooled mixture was partitioned between DCM and water, the emulsion filtered and washed with brine.
  • the organic phase was evaporated under reduced pressure to afford a crude product.
  • the reaction was repeated and the two batches of crude product combined.
  • the crude was purified by column chromatography on silica gel eluting with DCM:MeOH (100:0 to 90: 10). This product was dissolved in MeCN (95 ml_) and water was added dropwise (145 ml_) to obtain a precipitate which was filtered and washed with water to afford a beige solid.
  • a 96-well plate SPA was developed using antagonist radioligands (OX-radioligands); [3H]- SB674042 (Langmead et a/., 2004; Malherbe et a/., 2009a) for hOX1 R and [3H]-EMPA (Malherbe et al., 2009b) for hOX2R.
  • Cell membranes were prepared from recombinant cell lines; a CHO cell line transformed with hOX1 R (GeneBank accession# NM_001525) and a HEK cell line transformed with hOX2R (GeneBank accession* NM_001526.2).
  • Assay plates were read using PerkinElmer Microbeta2 allowing the binding profile of the compound of Example 1 to be assessed at room temperature (19°C) over several time points (15, 60, 90, 120, 150 minutes, and 18 hours) in hOX1 R and hOX2R assay formats.
  • End-point data values were determined as Specific Binding (SB) and expressed as a percentage of control (%SB), with 100% being the SB of OX-radioligand in the absence of competing unlabelled ligand, and 0% being the SB of the relevant OX-radioligand in the presence of an excess of the competing cold/unlabelled ligand (Suvorexant; a dual OXR- antagonist [DORA]).
  • SB Specific Binding
  • %SB percentage of control
  • IC50 half maximal inhibitory concentration
  • Example 1 has high affinity for hOX1 R and hOX2R, with 61 -fold selectivity for hOX2R.
  • Example 1 was prepared and tested by analogous methods to those described above in Example 1 and Example 2 in respect of the compound of Example 1.
  • Binding pKi 60 Binding pKi (60 Equilibration hOX2R/hOX1 R** min) min) Time (minutes)
  • Example 1 has higher affinity for hOX2R than comparative Example A.
  • the compound of Example 1 differs from comparative Example A of by virtue of 4-methyl substitution of octahydropyrrolo[3,4-c]pyrrol-2-yl moiety. The results therefore evidence that this methyl substitution gives rise to the increased affinity at hOX2R when compared to comparative Example A.
  • OX1 Optin 1 Receptor
  • OX2 Orexin 2 Receptor

Abstract

The present invention relates to orexin receptor antagonists, pharmaceutical compositi comprising the antagonists, methods of making the antagonists, their use for the modulation of the orexin receptor, and to use of the antagonists as medicaments, in particular for treating diseases, disorders, or conditions mediated by orexin receptor activity. More specifically, there is provided a compound of Formula (I) including salts thereof, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as defined herein and isotopically-labelled compounds of Formula (I).

Description

Orexin Receptor Antagonists
This invention relates to orexin receptor antagonists, pharmaceutical compositions comprising the antagonists, methods of making the antagonists, their use for the modulation of the orexin receptor, and to use of the antagonists as medicaments, in particular for treating diseases, disorders, or conditions mediated by orexin receptor activity.
Orexin (or hypocretin) signalling is mediated by two receptors and two peptide agonists. The two orexin peptides (orexin A and orexin B), bind to two high affinity G protein-coupled receptors (GPCRs), the orexin-1 and orexin-2 receptors (Sakurai T. et al., Cell, 1998, 92, 573-585). Orexin B binds preferentially to orexin-2 receptors, whereas orexin A binds with equal affinity to orexin-1 and orexin-2 receptors. Orexin A and orexin B (the orexins) are cleavage products of the same gene, prepro orexin. In the central nervous system neurons expressing prepro-orexin, the precursor from which orexin is produced, are found in the perifornical nucleus, the dorsal hypothalamus and the lateral hypothalamus. Orexinergic cells in these nuclei project to many areas of the brain, extending rostrally to the olfactory bulbs and caudally to the spinal cord.
The orexin system acts as an integrator of numerous physiological functions, including sleep/arousal states, sensory, locomotion, cognition, energy homeostasis, endocrine and visceral functions (reviewed by Li et al., British Journal of Pharmacology (2014) 171 , 332- 350).
Orexin signalling is essential for the stability of the arousal state. It is thought that the orexin system acts as a regulator of behaviour states by modulating the arousal threshold so that appropriate wakefulness can be maintained to cope with fluctuations in the external and internal environments. A lack of orexin signalling is associated with narcolepsy, a disorder characterised by uncontrollable sleep episodes and hypersomnolence (Nishino et al., Lancet: 355, 39-40. 2000)). Dual Orexin 1 and 2 receptor antagonists (DORAs) have proven efficacy in human patients suffering from insomnia, with Suvorexant (Belsomra) the first Orexin dual antagonist approved as an insomnia treatment. However evidence is mounting to suggest that these DORAs function predominantly through OX2R antagonism. For example, OX1 R knock-out mice exhibit a subtle, barely detectable sleep phenotype, whereas OX2R knockout mice show profound hyper somnolence (Kisanuki et al., Sleep 23, A91. 2000). Furthermore, in dogs OX2R mutations alone are responsible for the narcoleptic phenotype (Lin et al., Cell 98:365-376. 1999). Questions have been raised in the literature as to whether OX1 R antagonism may actually have detrimental effects in humans, causing an imbalance in the normal REM/NREM sleep architecture and given the brainstem localisation of OX1 R, particularly in the locus coeruleus, 0X1 R antagonists may act to attenuate vigilance state boundaries, effectively diminishing the threshold for sleep-state transitions (Dugovic et al., Front Neurosci. 8:28. 2014). The evidence suggests that a selective OX2R antagonist may provide safer, more efficacious, treatments than DORAs in humans.
Disorders of the sleep-wake cycle are, therefore, likely targets for orexin receptor (especially orexin-2 receptor) antagonist therapy. Examples of such disorders include sleep-wake transition disorders, insomnia, restless legs syndrome, jet-lag, disturbed sleep, and sleep disorders secondary to neurological disorders (for example, manias, depressions, manic depression, schizophrenia, and pain syndromes, such as fibromyalgia, neuropathic pain).
The orexin system also interacts with brain dopamine systems. Intracerebroventricular (i.c.v.) injections of orexins in mice increase locomotor activity, grooming and stereotypy. These effects are reversed by administration of D2 dopamine receptor antagonists (Nakamura et al., Brain Research, 2000, 873(1), 181-7). Therefore, orexin-receptor antagonists may be useful to treat various neurological disorders, for example, Parkinson's disease, Tourette's syndrome, anxiety, delirium and dementias.
There is evidence for a role for orexin in the pathogenesis of Alzheimer's disease (Kang et al, Science Express, 2009, 1-10). Brain interstitial fluid levels of amyloid-beta were demonstrated to fluctuate diurnally in both humans and rodents with sleep deprivation in rodents leading to significant increases in brain interstitial fluid levels of amyloid-beta. Infusion of a dual orexin antagonist in rodents suppressed interstitial levels of amyloid-beta and abolished the natural diurnal variation of amyloid-beta. The reduction of interstitial fluid amyloid-beta levels is correlated with reduced amyloid plaque formation, a hallmark of Alzheimer's disease, and consequently the regulation of sleep time could potentially inhibit amyloid-beta aggregation and slow the progression of Alzheimer's disease.
Orexin neurons project to many regions of the brain associated with reward function (T. Sakurai, supra). Several studies have demonstrated a direct role for orexin in the reinstatement of opioid-, ***e-, alcohol-, and nicotine-seeking behaviour (Harris et al, 2005, Nature 437: 556-559; Aston-Jones et al, 2008, Neuropharmacology 56 (Suppl. 1): 1 12-121 ; Lawrence et al, 2006, Br J Pharmacol 148: 752-759; Plaza-Zabala et al, 2010, J Neurosci 30, 2300-2310). Thus, orexin receptor antagonists are likely to be useful for the treatment of drug addiction, and alcoholism. Intracisternal or intraventromedial hypothalamus injections of orexin-A stimulate gastric acid secretion by activating the vagal system through orexin-1 receptors (Takahashi et al., Biochem Biophys Res Commun, 1999, 254: 623-627; Yamada et al., Neurosci Lett, 2005, 376: 137-142; Eliassi et al., J Neuroendocrinol, 2009, 21 : 177-182; Kermani and Eliassi, Neuroscience, 2012, 226: 81-88). Orexin-A can modify gastrointestinal motility, including gastric emptying, gastric interdigestive motility (Naslund et al., Am J Physiol Gastrointest Liver Physiol, 2002, 282: G470-G479; Ehrstrom et al., J Clin Endocrinol Metab, 2005, 90: 2370-2377; Ehrstrom et al., Regul Pept, 2005, 132: 9-16; Bulbul et al., Peptides, 2010, 31 : 2118-2122), and enteric peristalsis (Satoh et al., Neuropharmacology, 2006, 51 : 466^173), as well as colonic motility (Kirchgessner and Liu, Neuron, 1999, 24: 941-951 ; Nozu et al., Neurosci Lett, 201 1 , 498: 143-146). Orexin receptor antagonists are therefore potential treatments for ulcers, irritable bowel syndrome, diarrhoea and gastroesophageal reflux.
Body weight may also be affected by orexin-mediated regulation of appetite and metabolism (T. Sakurai et al., Cell, 1998, 92(4), 573-585; T. Sakurai, Reg. Pept, 1999, 85(1), 25-30). Various findings suggest that orexinergic neurons maintain a proper balance between energy intake or storage and expenditure initially by monitoring the nutritional state of the body in real time. Orexins also regulate energy intake or storage and expenditure by modulating the feeding and energy homeostatis-related circuits. Orexinergic neurons in the LH have a prominent role in sending the steady-state levels of physiologically relevant metabolites and integrate this information into a coherent value that is conveyed to arousal centres (Adamantidis and de Lecea, Trends Endocrinol Metab., 2008, 19: 362-370; 2009; Carter et al., Current Opin Pharmacol 2009, 9: 39-45). In addition to a direct interaction with feeding circuits and with humoral signals affecting feeding, orexinergic neurons have a significant regulatory role in the brain reward system, and particularly for palatable food. The orexin-1 receptor has been shown to mediate food motivation and reward-based feeding behaviour in mice and rats (Sharf et al., Brain Res, 2010, 1314: 130-138; Choi et al., Neuroscience, 2010, 167: 11-20). Orexin receptor antagonists, therefore, are likely to be useful in treatment of overweight or obesity, and conditions related to overweight or obesity, such as insulin resistance, type II diabetes, hyperlipidemia, gallstones, angina, hypertension, breathlessness, tachycardia, infertility, sleep apnoea, back and joint pain, varicose veins and osteoarthritis.
Orexin-deficient mice exhibit lower arterial blood pressure, heart rate and sympathetic tone (Kayaba et al., 2003, Am J Physiol Regul Integr Comp Physiol 285: R581-R593). Orexin administered i.e. v., intracisternally or intrathecally increases the mean arterial pressure, heart rate, renal sympathetic nerve activity and plasma catecholamine or vasopressin levels (Samson et ai, Brain Res, 1999, 831 : 248-253; Samson et ai, Sleep Med Rev, 2005, 9: 243-252; Shirasaka et al., Am J Physiol, 1999, 277 (6 Pt 2): R1780-R1785; Shirasaka et al., Regul Pept, 2002, 104: 91-95; Matsumura et al., Hypertension, 2001 , 37: 1382-1387; Hirota et ai, Brain Res, 2003, 981 : 143-150). These effects that are blocked or attenuated by the OX1 receptor antagonist SB-334867 (Hirota et ai, 2003, supra; Shahid et ai, Br J Pharmacol, 2011 , 162: 961-973). Thus, orexin receptor antagonists may be useful for treatment of hypertension, tachycardia and other arrhythmias, angina pectoris and acute heart failure. It has been suggested that orexinergic neurons are involved in sleep apnoea syndrome, as patients show increased plasma levels of orexin-A (Igarashi et ai, Chest, 2003, 124: 1381— 1385; Busquets et al., Respiration, 2004, 71 : 575-579). Hypercapnia and associated reflexes may increase the activity of orexinergic neurons during sleep, facilitating microarousals and a cascade of sympathetic activity that results in elevated blood pressure during the night. Thus, orexin receptor antagonists could be used to prevent these peaks of blood pressure in mild sleep apnoea.
The role of the orexinergic system in stress responses has been well established. The term 'stress' means a subjective state perceiving or anticipating the adverse disturbances in surroundings, which further activates various stress mediators to elicit proper responses (Joels, Epilepsia, 2009, 50: 586-597). Activation of orexinergic neurons results in some stress-like effects. Orexin can activate the hypothalamic-pituitary-adrenal (HPA) axis, including corticotrophin releasing factor (CRF), adrenocorticotropic hormone (ACTH) and corticosterone, stimulate stress-related behaviours, like grooming and chewing of inedible material, and enhance the activation of the monoamine system in a stress-like manner (Berridge et ai, Brain Res, 2010, 1314: 91-102). A link between orexin and stress-related pathologies, such as panic disorder, has been established (Johnson et ai, Nat Med, 2010, 16: 111-1 15; Lungwitz et ai, Physiol Behav, 2012, 107: 726-732). Panic attacks involve activation of the HPA axis and the autonomic system. Intrahypothalamic administration of an RNAi to orexin or an OX1 receptor antagonist has been shown to block these panic responses in rats injected with sodium lactate, and elevated levels of orexin-A have been detected in humans with panic disorder (Johnson et ai, 2010, supra). Orexin receptor antagonists may be useful for the treatment of stress, anxiety and panic disorders. Identification of orexin receptor antagonists is of great significance in the development of therapeutic agents for the treatment of a wide variety of disorders that are mediated through orexin receptors. Various small-molecule orexin receptor modulators have been reported, including: N-aroyl cyclic amine derivatives (WO 2003/002561), ethylene diamine derivatives (W02003/051872), sulfonylamino-acetic acid derivatives (W0 2004/033418), N-aryl acetyl cyclic amine derivatives (W0 2004/041791), diazepan derivatives (WO 2007/126935), amidoethylthioether derivatives (WO 2007/126934), 2-substituted proline bis-amide derivatives (WO 2008/008551), bridged diazepan derivatives (WO 2008/008517), substituted diazepan derivatives (WO 2008/008518; US 2008/0132490, WO 2009/058238), oxo bridged diazepan derivatives (WO 2008/143856), 1 ,2-diamido ethylene derivatives (WO 2009/022311), heteroaryl derivatives (WO 2009/0163485), methyl substituted piperidinyl derivatives (WO 2009/124956), N,N-disubstituted-1 ,4-diazepane derivatives (Cox et al., Bioorganic & Medicinal Chemistry Letters, 2009, 19(1 1), 2997-3001), Orexin/Hypocretin receptor ligands (Boss, et al., Journal of Medicinal Chemistry, 2009, 52(4), 891-903) 3,9- diazabicyclo[4.2.1]nonanes (Coleman et al., Bioorganic & Medicinal Chemistry Letters, 2010, 20(14), 4201-4205), the dual orexin receptor antagonist, [(7R)-4-(5-Chloro-1 ,3-benzoxazol- 2-yl)-7 -methyl-1 ,4- diazepan- 1-yl][5-methyl-2-(2H-1 ,2,3-triazol-2-yl)phenyl]methanone, (Cox, et. al., Journal of Medicinal Chemistry, 2010 53(14) 5320-5332), pyridazine carboxamide derivatives (WO 2010/051238), 2,5- disubstituted benzamide derivatives (WO 2010/051237), isonicotinamides (WO 2010/051236), heterocyclylbenzoylpiperazines derivatives (WO 2010/48012), substituted diazepane derivatives (WO 2010/048017), substituted pyrrolidine derivatives (WO 2010/048014), triazolylbenzoylpiperidine derivatives (WO 2010/048010), triazolylbenzoylmorpholine derivatives (WO 2010/048013), conformationally restrained N,N disubstituted 1 ,4-diazapane derivatives (Coleman et al., Bioorganic & Medicinal Chemistry Letters, 2010, 20(7), 231 1- 2315), tripyridyl carboxamide derivatives (WO 2010/017260), imidazopyridylmethyl substituted piperidine derivatives (WO 2010/072722), imidazopyrazine substituted piperidine derivatives (US 2010/160344; US 2010/0160345; WO 2010/060472), N-{[(1 R,4S,6R)-3-(2-pyridinylcarbonyl)-3- azabicyclo[4.1.0]hept-4-yl]methyl}-2-heteroarylamine derivatives (WO 2010/063663), N-{[(1 S,4S,6S)-3-(2-pyridinylcarbonyl)-3-azabicyclo[4.1.0]hept-4-yl]methyl}-2-heteroarylamine derivatives (WO 2010/063662), imidazopyrimidine derivatives (WO 2010/060471), imidazopyrazine derivatives (WO 2010/060470), disubstituted octahydropyrrolo[3,4-c]pyrrole derivatives (WO 2012/145581).
There remains a need, however, for potent orexin receptor modulators, in particular orexin receptor antagonists.
According to the invention, there is provided a compound of Formula (I):
Figure imgf000007_0001
Compounds of the invention include compounds of Formula (I) and salts thereof, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labelled compounds of Formula (I).
In some embodiments, the central hydrogen atoms (shown in the above structural Formula) on the methylated octahydropyrrolo[3,4-c]pyrrole group are in a cis position. In particular embodiments, the compound of Formula (I) is a compound of Formula (l)(a):
Figure imgf000007_0002
(l)(a)
2-[(3aS,4S,6aR)-5-(2-Fluoro-4-methoxybenzoyl)-4-methyl-octahydropyrrolo[3,4-c]pyrrol-2- yl]-4,6-dimethylpyrimidine-5-carbonitrile
Compounds of the invention include compounds of Formula (l)(a) and salts thereof, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labelled compounds of Formula (l)(a). Compounds of the invention have been found to have surprisingly high potency as orexin receptor antagonists, in particular as selective 0X2 receptor antagonists. We have found that incorporation of the methyl substituent at position 4 of the octahydropyrrolo[3,4-c]pyrrole group provides compounds of the invention with an unexpectedly high increase in potency as an orexin receptor antagonist compared with a corresponding desmethylated compound. In view of their surprisingly high potency as orexin receptor antagonists, compounds of the invention are expected to be particularly effective in the treatment of diseases, disorders or conditions mediated by orexin receptor activity, especially those associated with 0X2 receptor activity. Compounds of the invention may be more effective than conventional compounds in the treatment of such diseases, disorders or conditions at similar doses to the conventional compounds. Alternatively or additionally, compounds of the invention may be as effective as conventional compounds at lower doses than the conventional compounds but have fewer, or reduced side effects.
There is also provided according to the invention a pharmaceutical composition comprising a compound of Formula (I) or (l)(a), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. There is further provided according to the invention a compound or composition of the invention for use as a medicament.
The invention also provides a compound or composition of the invention for use in treating a disease, disorder or condition mediated by orexin receptor activity.
According to the invention there is also provided use of a compound or composition of the invention in the manufacture of a medicament for treating a disease, disorder or condition mediated by orexin receptor activity. There is further provided according to the invention a method of treating a disease, disorder or condition mediated by orexin receptor activity, which comprises administering an effective amount of a compound of Formula (I) or (l)(a), or a pharmaceutically acceptable salt thereof, to a subject in need thereof. The invention also provides a method of producing a compound of Formula (I), which comprises reacting a compound of Formula (VII) and a cyanating agent, to produce the compound of Formula (I):
Figure imgf000009_0001
Figure imgf000009_0002
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (VII) are in a cis position, thereby producing a compound of Formula (I) with corresponding hydrogen atoms in a cis position.
In particular embodiments, there is provided a method of producing a compound of Formula (l)(a), which comprises reacting a compound of Formula (Vll)(a) with a cyanating agent to produce the compound of Formula (l)(a):
Figure imgf000009_0003
(VN)(a)
Figure imgf000010_0001
(l)(a)
In some embodiments, the compound of Formula (I) or (l)(a) is prepared from the compound of Formula (VII) or (Vll)(a) by a copper catalysed cyanation reaction, for example, using potassium hexacyanoferrate(ll) trihydrate in the presence of a suitable ligand, such as 1-butylimidazole, and a copper catalyst, such as Cul, in a suitable solvent, such as toluene, at elevated temperature, typically 150°C.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (VII) by reacting a compound of Formula (VI) and a brominating agent, to produce the compound of Formula (VII):
Figure imgf000010_0002
(VI)
Brominating agent
Figure imgf000011_0001
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (VI) are in a cis position, thereby producing a compound of Formula (VII) with corresponding hydrogen atoms in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (Vll)(a) by reacting a compound of Formula (Vl)(a) with a brominating agent to produce the compound of Formula (Vll)(a):
Figure imgf000011_0002
(VN)(a) In some embodiments, the compound of Formula (VII) is prepared from the compound of Formula (VI), or the compound of Formula (Vll)(a) is prepared from the compound of Formula (Vl)(a), by bromination, for example, using a suitable brominating agent, such as N-bromo succinimide (NBS), in a suitable solvent, such as MeCN, at between room temperature and 100°C, typically at 50°C.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (VI) by reacting a compound of Formula V) and a compound of Formula (XVIII), to produce the compound of Formula (VI):
Figure imgf000012_0001
(VI)
where LG is a leaving group. Examples of suitable leaving groups include: a chloro, bromo, mesylate or tosylate group. In particular embodiments, LG is chloro.
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (V) are in a cis position, thereby producing a compound of Formula (VI) with corresponding hydrogen atoms in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (Vl)(a) by reacting a compound of Formula (V)(a) and a compound of Formula (XVIII), to produce the compound of Formula (Vl)(a):
Figure imgf000013_0001
(Vl)(a) where LG is a leaving group.
In some embodiments, the compound of Formula (V) or (V)(a) is reacted with the compound of Formula (XVII I) in the presence of a suitable base, such as CS2CO3 or K2CO3, in a polar solvent, such as DMF or EtOH. In particular embodiments, the base is CS2CO3, and the solvent is DMF, and the reaction is carried out at an elevated temperature, preferably 100°C.
In some embodiments the compound of Formula (XVIII) is 6-Chloro-4,6-dimethylpyrimidine (i.e. LG is chloro).
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (V) by deprotection of a compound of Formula (IV):
Figure imgf000014_0001
(I
Figure imgf000014_0002
(V)
where PG2 is an amine-protecting group
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (IV) are in a cis position, thereby producing a compound of Formula (V) with corresponding hydrogen atoms in a cis position. In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (V)(a) by deprotection of a compound of Formula (IV)(a):
Figure imgf000015_0001
(V)(a) where PG2 is an amine-protecting group.
In some embodiments, (in particular where PG2 is benzyl) the deprotection is carried out by transfer or palladium-catalyzed hydrogenation. In particular embodiments, palladium catalyzed hydrogenation may be carried out using 10% palladium on charcoal, in EtOH under an atmosphere of hydrogen at room temperature.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (IV) by reacting a compound of Formula (III) with 2-fluoro-4-methoxybenzoic acid to form the compound of Formula (IV):
Figure imgf000015_0002
(III)
Figure imgf000016_0001
Amide bond formation
Figure imgf000016_0002
(IV)
where PG2 is an amine-protecting group.
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (III) are in a cis position, thereby producing a compound of Formula (IV) with corresponding hydrogen atoms in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (IV)(a) by reacting a compound of Formula (lll)(a) with 2-fluoro-4-methoxybenzoic acid to form the compound of Formula (IV)(a):
Figure imgf000016_0003
(lll)(a)
+
Figure imgf000017_0001
Amide bond formation
Figure imgf000017_0002
(IV)(a)
where PG2 is an amine-protecting group.
In some embodiments, the compound of Formula (III) or (lll)(a) is reacted with 2-fluoro-4- methoxybenzoic acid in the presence of a coupling reagent, such as HATU or T3P, and an organic base, such as ΕίβΝ or DIPEA, in an aprotic solvent, such as MeCN or DMF, at a temperature from 0-100°C. Alternatively, in some embodiments, 2-fluoro-4-methoxybenzoic acid is converted in situ to 2-fluoro-4-methoxybenzoyl chloride, by treatment with a suitable reagent, such as SOCI2 or PCI3, in a non polar solvent, such as toluene, at elevated
temperature, followed by reaction with a compound of Formula (III) or (lll)(a), in the presence of a suitable organic base, such as ΕίβΝ or DIPEA, in an aprotic solvent, such as DCM or MeCN, typically at a temperature from 0°C to room temperature.
In one embodiment, the compound of Formula (III) or (lll)(a) is reacted with 2-fluoro-4- methoxybenzoyl chloride, prepared in situ from 2-fluoro-4-methoxybenzoic acid, in the presence of ΕίβΝ in MeCN, preferably at room temperature.
In particular embodiments, 2-fluoro-4-methoxybenzoic acid is treated with SOCI2, in toluene at 80°C to prepare 2-fluoro-4-methoxybenzoyl chloride in situ, followed by reaction with a compound of Formula (III) or (lll)(a) in the presence of ΕίβΝ in DCM at room temperature. In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (III) by reduction of a compound of Formula (II) to produce the compound of Formula (III):
Figure imgf000018_0001
where PG2 is an amine-protecting group.
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (II) are in a cis position, thereby producing a compound of Formula (III) with corresponding hydrogen atoms in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (lll)(a) by reduction of a compound of Formula (ll)(a) to produce the com ound of Formula (lll)(a):
Figure imgf000018_0002
(H)(a
Reduction CH
Figure imgf000019_0001
(lll)(a)
where PG2 is an amine-protecting group.
In some embodiments, the reduction is carried out using a reducing agent in an aprotic solvent at a temperature from 0°C to reflux.
In particular embodiments, the reducing agent is UAIH4, the aprotic solvent is 2-MeTHF, and the reduction is preferably carried out at room temperature.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (II) by deprotection of a compound of Formula (XIII):
Figure imgf000019_0002
(XIII)
Deprotection
Figure imgf000019_0003
(II)
where PGi is an amine protecting group, and where PG2 is an amine-protecting group. In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XIII) are in a cis position, thereby producing a compound of Formula (II) with corresponding hydrogen atoms in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (ll)(a) by deprotection of a compound of Formula (Xlll)(a):
Figure imgf000020_0001
(Xlll)(a)
Figure imgf000020_0002
(H)(a)
where PGi is an amine-protecting group and PG2 is an amine-protecting group.
In some embodiments, the deprotection is an acid-mediated deprotection in an aprotic solvent.
In particular embodiments, the acid is TFA, and the solvent is DCM, and the deprotection is preferably carried out at a temperature from 0°C to room temperature.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XIII) from compounds of Formulae (XII) and (XIX) by a cyclisation reaction:
Figure imgf000021_0001
+
PG.
Figure imgf000021_0002
(XIII)
where PGi is an amine-protecting group and PG2 is an amine-protecting group.
It will be appreciated that the choice of reagent(s) for use in the cyclisation will depend on which stereoisomer, or mixture of stereoisomers, is to be produced. In some embodiments, the cyclisation reaction is selected such that the central hydrogen atoms on the methylated octahydropyrrolo[3,4-c]pyrrole group of the compound of Formula (XI II) are in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XI I l)(a) from a compound of Formula (Xll)(a) and a compound of Formula (XIX), by an acid-mediated cyclisation reaction:
Figure imgf000022_0001
(Xl l)(a)
+
PG.
OCH
Figure imgf000022_0002
(XIX)
Cyclisation reaction
CH
Figure imgf000022_0003
(Xlll)(a)
where PGi is an amine-protecting group and PG2 is an amine-protecting group.
In some embodiments, the acid-mediated cyclisation reaction is carried out in the presence of an acid in an aprotic solvent. In particular embodiments, the acid is TFA, the aprotic solvent is DCM, and the reaction is preferably carried out at a temperature from 0°C to room temperature.
In some embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise preparing a compound of Formula (Xll)(a) from a compound of Formula (Vlll)(a), (IX)(a), (X)(a), (Xl)(a), and (Xll)(a), as illustrated by Scheme 2 below.
Figure imgf000023_0001
Figure imgf000023_0002
Scheme 2
where PGi is an amine protecting group. Alternatively, in other embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise preparing a compound of Formula (Xll)(a) from a compound of Formula (XIV)(a), (XV)(a), (XVI)(a), and (XVII)(a) as illustrated by Scheme 3 below.
Figure imgf000023_0003
Scheme 3 where PGi is an amine protecting group.
Alternatively, in other embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise preparing a compound of Formula (Vl)(a) from a compound of Formula (lll)(a), (XX)(a), (XXI)(a), (XVIII), (XXII)(a) and (XXIII)(a) as illustrated by Scheme 4 below.
Figure imgf000024_0001
(Vl)(a)
Scheme 4 where PGi is an amine protecting group, and where PG2 is an amine protecting group. LG is a leaving group.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (VI) by reacting a compound of Formula (XXIII) and 2-fluoro-4-methoxybenzoic acid to produce the compound of Formula (VI):
Figure imgf000025_0001
Figure imgf000025_0002
Amide bond formation
Figure imgf000025_0003
(VI)
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XXIII) are in a cis position, thereby producing a compound of Formula (VI) with corresponding hydrogen atoms in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (Vl)(a) by reacting a compound of Formula (XXIII)(a) and 2-fluoro-4-methoxybenzoic acid to produce the compound of Formula
Figure imgf000026_0001
(XXIII)(a)
+
Figure imgf000026_0002
Amide bond formation _ CH
Figure imgf000026_0003
(Vl)(a)
In some embodiments, the compound of Formula (XXIII) or (XXIII)(a) and 2-fluoro-4- methoxybenzoic acid are reacted in the presence of a coupling reagent and an organic base, in an aprotic solvent, at a temperature from 0-100°C. In particular embodiments, the coupling reagent is HATU, or T3P, the organic base is DIPEA or Et3N, the aprotic solvent is DMF or MeCN, and the temperature is preferably room temperature. A skilled person will appreciate that alternative coupling conditions are available in combination with a range of alternative bases and solvents.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XXIII) by deprotection of a compound of Formula (XXII):
Figure imgf000027_0001
(XXIII)
where PGi is an amine-protecting group.
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XXII) are in a cis position, thereby producing a compound of Formula (XXIII) with corresponding hydrogen atoms in a cis position. In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XXIII)(a) by deprotection of a compound of Formula (XXII)(a): CH
Figure imgf000028_0001
(XXIII)(a) where PGi is an amine-protecting group.
In some embodiments, the deprotection is an acid-mediated deprotection in an aprotic solvent.
In particular embodiments (in particular where PGi = 'BoC), the acid is TFA, and the solvent is DCM, and the deprotection is preferably carried out at a temperature from 0°C to room temperature.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XXII) by reacting a compound of Formula (XXI) and a compound of Formula (XVIII) to produce the compound of Formula (XXII):
Figure imgf000028_0002
(XXI)
+
Figure imgf000029_0001
where PGi is an amine-protecting group, and LG is a leaving group.
Examples of suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group. In particular embodiments, LG is chloro.
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XXI) are in a cis position, thereby producing a compound of Formula (XXII) with corresponding hydrogen atoms in a cis position. In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XXII)(a) by reacting a compound of Formula (XXI)(a) and a compound of Formula (XVIII) to produce the compound of Formula (XXII)(a):
Figure imgf000030_0001
(XXII)(a) where PGi is an amine-protecting group, and LG is a leaving group.
Typically, the compound of Formula (XXI) or (XXI)(a) is reacted with the compound of Formula (XVI) in the presence of a base in a solvent.
In some embodiments, the compound of Formula (XXI) or (XXI)(a) is reacted with the compound of Formula (XVIII) in the presence of a base in a solvent. In particular embodiments, the base is CS2CO3 or K2CO3, and the solvent is DMF or EtOH, and the reaction is carried out at an elevated temperature, such as 80-1 10°C.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XXI) by deprotection of a compound of Formula (XX) to produce the compound of Formula (XXI):
Figure imgf000031_0001
(XX)
Deprotection
Figure imgf000031_0002
(XXI)
where PGi is an amine-protecting group, and PG2 IS an amine-protecting group.
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (XX) are in a cis position, thereby producing a compound of Formula (XXI) with corresponding hydrogen atoms in a cis position.
In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XXI)(a) by deprotection of a compound of Formula (XX)(a) to produce the compound of Formula (XXI)(a):
Figure imgf000031_0003
(XX)(a)
Deprotection
Figure imgf000031_0004
(XXI)(a) where PGi is an amine-protecting group, and PG2 IS an amine-protecting group. In some embodiments (in particular where PG2 = Bn), the deprotection is carried out by transfer palladium catalysed hydrogenation. For example, transfer hydrogenation may be carried out using ammonium formate and Pd(OH)2 on charcoal, in ethanol, at a temperature between room temperature and reflux.
A skilled person will appreciate that alternative hydrogenation conditions are available to effect this transformation.
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise producing a compound of Formula (XX) by protecting a compound of Formula (III) to produce the compound of Formula (XX):
Figure imgf000032_0001
(III)
Protection
Figure imgf000032_0002
(XX) where PGi is an amine-protecting group, and PG2 IS an amine-protecting group.
In some embodiments, the central hydrogen atoms on the methylated octahydropyrrolo[3,4- c]pyrrole group of the compound of Formula (III) are in a cis position, thereby producing a compound of Formula (XX) with corresponding hydrogen atoms in a cis position. In particular embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of Formula (XX)(a) by protecting a
compound of Formula (lll)(a) to produce the compound of Formula (XX)(a):
Figure imgf000033_0001
(lll)(a)
Protection
Figure imgf000033_0002
(XX)(a) where PGi is an amine-protecting group, and PG2 IS an amine-protecting group. In particular embodiments, protection is carried out under standard conditions for introducing an amine protecting group. In some embodiments (in particular where PGi = 'BoC), protection is carried out by treatment with B0C2O in a suitable solvent, such as DCM, typically at room temperature.
In a further aspect, there is provided according to the invention a method of producing a compound of Formula (XIII), which comprises cyclisation of a compound of Formula (XII) with a compound of Formula (XVIII) to produce a compound of Formula (XIII):
Figure imgf000033_0003
+ PG- OCH .
Figure imgf000034_0001
(XVII)
Cyclisation reaction
Figure imgf000034_0002
(XIII)
where PGi is an amine-protecting group, and PG2 is an amine- protecting group.
It will be appreciated that the choice of reagent(s) for use in the cyclisation will depend on which stereoisomer, or mixture of stereoisomers, is to be produced.
In some embodiments, the cyclisation reaction is selected such that the central hydrogen atoms on the methylated octahydropyrrolo[3,4-c]pyrrole group of the compound of Formula (XIII) are in a cis position.
In particular embodiments, there is provided a method of producing a compound of Formula (Xlll)(a), which comprises acid-mediated cyclisation of a compound of Formula (Xll)(a) with a compound of Formula (XIX) to produce a compound of Formula (Xlll)(a):
CH3
Figure imgf000034_0003
(Xl l)(a) OCH 3
Figure imgf000035_0001
(XIX)
Cyclisation reaction
CH
H
Figure imgf000035_0002
(Xlll)(a)
where PGi is an amine-protecting group, and PG2 is an amine- protecting group.
In some embodiments, a method of the invention for producing a compound of Formula (XIII) further comprises converting the compound of Formula (XIII) to a compound of
Formula (I).
In particular embodiments, a method of the invention for producing a compound of Formula (XIH)(a) further comprises converting the compound of Formula (XI I l)(a) to a compound of Formula (l)(a).
There is also provided according to the invention use of a compound of Formula (XII) in a method of producing a compound of Formula (I).
There is also provided according to the invention use of a compound of Formula (Xll)(a) in a method of producing a compound of Formula (l)(a).
In some embodiments, methods of the invention for producing a compound of Formula (I) further comprise reacting the compound of Formula (I) to yield a corresponding
pharmaceutically acceptable salt of the compound of Formula (I). In some embodiments, methods of the invention for producing a compound of Formula (l)(a) further comprise reacting the compound of Formula (l)(a) to yield a corresponding pharmaceutically acceptable salt of the compound of Formula (l)(a).
There is also provided according to the invention, a compound of Formula (II), (ll)(a), (III), (lll)(a), (IV), (IV)(a), (V), (V)(a), (VI), (Vl)(a), (VII), (Vll)(a), (VIII), (Vlll)(a), (IX), (IX)(a), (X), (X)(a), (XI), (Xl)(a), (XII), (Xll)(a), (XIII), (Xlll)(a), (XIV), (XIV)(a), (XV), (XV)(a), (XVI), (XVI)(a), (XVII), (XVII)(a), (XVIII), (XIX), (XX), (XX)(a), (XXI), (XXI)(a), (XXII), or (XXII)(a), or a pharmaceutically acceptable salt, solvate, hydrate, geometrical isomer, tautomer, or optical isomer thereof. Examples of suitable amine protecting groups are well-known to the skilled person, and include those described in 'Greene's Protective Groups in Organic Synthesis' by Theodora W Greene and Peter G M Wuts, fifth edition, (John Wiley and Sons, 2014), in particular Chapter 7 ("Protection for the Amino Group"), incorporated herein by reference, which also describes methods for the removal of such groups.
Particular examples of suitable amine protecting groups include the following: Carbobenzyloxy (Cbz) group - removed by hydrogenolysis;
p-Methoxybenzyl carbonyl (Moz or MeOZ) group - removed by hydrogenolysis, more labile than Cbz;
te/f-Butyloxycarbonyl (lBOC) group (common in solid phase peptide synthesis) - removed by concentrated strong acid (such as HCI or CF3COOH), or by heating to >80 °C;
9-Fluorenylmethyloxycarbonyl (FMOC) group - removed by base, such as piperidine;
Acetyl (Ac) group - removed by treatment with a base, most often, with aqueous or gaseous ammonia or methylamine;
Benzoyl (Bz) group - removed by treatment with a base, most often with aqueous or gaseous ammonia or methylamine;
Benzyl (Bn) group - removed by hydrogenolysis;
Carbamate group - removed by acid and mild heating, for example te/f-butyl carbamate; p-Methoxybenzyl (PMB) - removed by hydrogenolysis, more labile than benzyl;
3,4-Dimethoxybenzyl (DMPM) - removed by hydrogenolysis, more labile than p- methoxybenzyl;
p-methoxyphenyl (PMP) group - removed by ammonium cerium(IV) nitrate (CAN);
Tosyl (Ts) group - removed by concentrated acid (HBr, H2SO4) and strong reducing agents (sodium in liquid ammonia or sodium naphthalenide); Trichloroethyl chloroformate (Troc) group - removed by Zn insertion in the presence of acetic acid;
Other Sulfonamides (Nosyl and Nps) groups - removed by samarium iodide, tributyltin hydride;
Acetamide;
Benzamide;
9-Fluorenylmethyl carbamate;
Benzhydryl;
Trityl.
Thus, PGi and PG2 may each independently be selected from: a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, tert- butylcarbamate, 9-Fluorenylmethyl carbamate, benzhydryl, trityl group. In particular embodiments, PGi is a carbamate group, preferably f-butyl carbamate
In particular embodiments PG2 is benzyl.
In particular embodiments, PGi is a carbamate group, preferably f-butyl carbamate, and PG2 is benzyl.
Examples of suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group. In particular embodiments, LG is chloro.
Examples of methods of producing a compound of Formula (I), or (l)(a), in accordance with the invention are described in detail below.
The compounds of the invention include compounds of Formula (I), or (l)(a), and salts thereof as hereinafter defined, polymorphs, and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labelled compounds of Formula (I) or (l)(a).
Any Formula given herein is intended to represent compounds having structures depicted by the structural Formula as well as certain variations or forms. In particular, compounds of any Formula given herein may have asymmetric centres and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general Formula, and mixtures thereof, are considered within the scope of the Formula. Thus, any Formula given herein is intended to represent a racemate, one or more enantiomeric forms, one or more diastereomeric forms, one or more atropisomeric forms, and mixtures thereof. Furthermore, certain structures may exist as geometric isomers (i.e., cis and trans isomers), as tautomers, or as atropisomers.
Where a compound of Formula (I), or (l)(a), contains for example, a keto or guanidine group or an aromatic moiety, tautomeric isomerism ('tautomerism') can occur. It follows that a single compound may exhibit more than one type of isomerism.
Included within the scope of the claimed compounds of the present invention are all stereoisomers, geometric isomers and tautomeric forms of the compounds of Formula (I), or (l)(a), including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or base addition salts wherein the counter ion is optically active, for example, D-lactate or L-lysine, or racemic, for example, DL-tartrate or DL-arginine.
Examples of types of potential tautomerisms shown by the compounds of the invention include; amide hydroxyl-imine and keto enol tautomersims:
O OH
Figure imgf000038_0001
( CH .
Cis/trans isomers may be separated by conventional techniques well known to those skilled in the art, for example, by chromatography and fractional crystallisation. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or other derivative) using, for example, chiral high pressure liquid chromatography (HPLC). Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on a resin with an asymmetric stationary phase and with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% ethanol, typically from 2 to 20%. Concentration of the eluate affords the enriched mixture. Mixtures of stereoisomers may be separated by conventional techniques known to those skilled in the art (see, for example, "Stereochemistry of Organic Compounds" by E L Eliel (Wiley, New York, 1994)).
Additionally, any Formula given herein is intended to refer also to hydrates, solvates, and polymorphs of such compounds, and mixtures thereof, even if such forms are not listed explicitly. A compound of Formula (I), or (l)(a), or pharmaceutically acceptable salt of a compound of Formula (I), or (l)(a), may be obtained as a solvate. Solvates include those formed from the interaction or complexation of compounds of the invention with one or more solvents, either in solution or as a solid or crystalline form. In some embodiments, the solvent is water and then the solvates are hydrates. In addition, certain crystalline forms of a compound of Formula (I), or (l)(a), or a pharmaceutically acceptable salt of a compound of Formula (I), or (l)(a), may be obtained as co-crystals. In certain embodiments of the invention, a compound of Formula (I), or (l)(a), or a pharmaceutically acceptable salt of a compound of Formula (I), or (l)(a), may be obtained in a crystalline form. In other embodiments, a compound of Formula (I), or (l)(a), may be obtained in one of several polymorphic forms, as a mixture of crystalline forms, as a polymorphic form, or as an amorphous form. In other embodiments, a compound of Formula (I), or (l)(a), may convert in solution between one or more crystalline forms and/or polymorphic forms.
Any Formula given herein is also intended to represent unlabelled forms as well as isotopically labelled forms of the compounds. Isotopically labelled compounds have structures depicted by the Formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, and fluorine, such as 2H, 3H, 11C, 13C, 14C, 13N, 15N, 150, 170, 180, 18F, respectively. Such isotopically labelled compounds are useful in metabolic studies (preferably with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques (such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)) including drug or substrate tissue distribution assays, or in radioactive treatment of subjects. Substitution with positron emitting isotopes, such as 11C, 18F, 150 and 13N, can be useful in PET studies for examining substrate receptor occupancy. In particular, an 18F or 11C labelled compound may be particularly preferred for PET studies. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements. Certain isotopically-labelled compounds of Formula (I) for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
Isotopically labelled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
A compound of Formula (I), or (l)(a), and pharmaceutically acceptable salts thereof, may be used alone or in combination with one or more additional active ingredients to Formulate pharmaceutical compositions. A pharmaceutical composition therefore comprises an effective amount of a compound of Formula (I), or (l)(a), or a pharmaceutically acceptable salt thereof.
The invention includes also pharmaceutically acceptable salts of a compound of Formula (I) or (l)(a). A "pharmaceutically acceptable salt" is intended to mean a salt of a free acid or base of a compound represented by Formula (I), or (l)(a), that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to a subject. See, generally, G.S. Paulekuhn, et al., "Trends in Active Pharmaceutical Ingredient Salt Selection based on Analysis of the Orange Book Database", J. Med. Chem., 2007, 50:6665-72, S.M. Berge, et al., "Pharmaceutical Salts", J Pharm Sci., 1977, 66: 1 -19, and Handbook of Pharmaceutical Salts, Properties, Selection, and Use, Stahl and Wermuth, Eds., Wiley-VCH and VHCA, Zurich, 2002.
Examples of pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of subjects without undue toxicity, irritation, or allergic response. A compound of Formula (I), or (l)(a), may possess a sufficiently acidic group, a sufficiently basic group, or both types of functional groups, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt, bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt. Examples of pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen- phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1 ,4-dioates, hexyne-1 ,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ- hydroxybutyrates, glycolates, tartrates, methane-sulfonates, propanesulfonates, naphthalene-1 -sulfonates, naphthalene-2-sulfonates, and mandelates.
The invention also relates to pharmaceutically acceptable prodrugs of a compound of Formula (I) or (l)(a), and treatment methods employing such pharmaceutically acceptable prodrugs. The term "prodrug" means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodrug on being brought to physiological pH is converted to the compound of Formula (I), or (l)(a)). A "pharmaceutically acceptable prodrug" is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject. Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985. The present invention also relates to pharmaceutically active metabolites of a compound of Formula (I), or (l)(a), which may also be used in the methods of the invention. A "pharmaceutically active metabolite" means a pharmacologically active product of metabolism in the body of a compound of Formula (I), or (l)(a), or salt thereof. Prodrugs and active metabolites of a compound may be determined using routine techniques known or available in the art. See, e.g., Bertolini, et a/., J Med Chem. 1997, 40, 201 1 -2016; Shan, et a/., JPharm Sci. 1997, 86 (7), 765-767; Bagshawe, Drug Dev Res. 1995, 34, 220-230; Bodor, Adv Drug Res. 1984, 13, 224-331 ; Bundgaard, Design of Prodrugs (Elsevier Press, 1985); and Larsen, Design and Application of Prodrugs, Drug Design and Development (Krogsgaard-Larsen, et ai, eds., Harwood Academic Publishers, 1991).
A compound of Formula (I), or (l)(a), and its pharmaceutically acceptable salts, pharmaceutically acceptable prodrugs, and pharmaceutically active metabolites, are useful as antagonists of the orexin receptor. The term "treat", "treating", or "treatment" as used herein is intended to refer to administration of a compound or composition of the invention to a subject for the purpose of effecting a therapeutic or prophylactic benefit through modulation (in particular, antagonism) of orexin receptor activity. Treating includes reversing, ameliorating, alleviating, inhibiting the progress of, lessening the severity of, or preventing a disease, disorder, or condition, or one or more symptoms of such disease, disorder or condition mediated through modulation of orexin receptor activity. The term "subject" refers to a mammalian patient in need of such treatment, such as a human.
Accordingly, the invention relates to methods of using the compounds or compositions described herein to treat subjects diagnosed with or suffering from a disease, disorder, or condition mediated by orexin receptor activity, such as: disorders of the sleep-wake cycle, metabolic disorders, neurological disorders, and other disorders (e.g., feeding, drinking, arousal, stress, addiction, especially drug addiction, metabolism and reproduction). Symptoms or disease states are intended to be included within the scope of "conditions, disorders, or diseases." Sleep disorders include, but are not limited to, disorders of the sleep-wake cycle, sleep-wake transition disorders, insomnia, restless legs syndrome, jet-lag, disturbed sleep, and sleep disorders secondary to neurological disorders (e.g., manias, depressions, manic depression, schizophrenia, and pain syndromes (e.g., fibromyalgia, neuropathic). Metabolic disorders include, but are not limited to, overweight or obesity and conditions related to overweight or obesity, such as insulin resistance, type II diabetes, hyperlipidemia, gallstones, angina, hypertension, breathlessness, tachycardia, arrhythmias, angina pectoris, acute heart failure, infertility, sleep apnoea, back and joint pain, varicose veins and osteoarthritis.
Neurological disorders include, but are not limited to, Parkinson's disease, Alzheimer's disease, Huntington's disease, Tourette's Syndrome, catatonia, anxiety, stress disorder, panic disorder, delirium and dementias. Other disorders include, but are not limited to, ulcers, irritable bowel syndrome, diarrhoea and gastroesophageal reflux.
It will be appreciated that compounds of the invention are particularly advantageous for treatment of various diseases (such as, but not restricted to, sleep disorders) because they exhibit rapid and high levels of intestinal absorption, leading to rapid achievement of maximal concentration in the body and the brain, and a sufficient but relatively short exposure period. In treatment methods according to the invention, an effective amount of a compound of the invention is administered to a subject suffering from or diagnosed as having such a disease, disorder, or condition. An "effective amount" means an amount or dose sufficient to generally bring about the desired therapeutic or prophylactic benefit in a subject in need of such treatment for the designated disease, disorder, or condition. Effective amounts or doses of a compound of the present invention may be ascertained by routine methods such as modelling, dose escalation studies or clinical trials, and by taking into consideration routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the compound, the severity and course of the disease, disorder, or condition, the subject's previous or ongoing therapy, the subject's health status and response to drugs, and the judgment of the treating physician. An example of a dose is in the range of from about 0.001 to about 200 mg of compound per kg of subject's body weight per day, preferably about 0.05 to 100 mg/kg/day, or about 1 to 35 mg/kg/day, in single or divided dosage units (e.g., BID, TID, QID). For a 70kg human, an illustrative range for a suitable dosage amount is from about 0.05 to about 7 g/day, or about 0.2 to about 2.5 g/day.
Once improvement of the subject's disease, disorder, or condition has occurred, the dose may be adjusted for preventative or maintenance treatment. For example, the dosage or the frequency of administration, or both, may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained. Of course, if symptoms have been alleviated to an appropriate level, treatment may cease. Subjects may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms. In addition, the active agents of the invention may be used in combination with additional active ingredients in the treatment of the above conditions. The additional active ingredients may be co-administered separately with an active agent of the invention, or included with such an agent in a pharmaceutical composition according to the invention. In an exemplary embodiment, additional active ingredients are those that are known or discovered to be effective in the treatment of conditions, disorders, or diseases mediated by orexin receptor activity, such as another orexin modulator or a compound active against another target associated with the particular condition, disorder, or disease. The combination may serve to increase efficacy (e.g., by including in the combination a compound potentiating the potency or effectiveness of an active agent according to the invention), decrease one or more side effects, or decrease the required dose of the active agent according to the invention. The active agents of the invention are used, alone or in combination with one or more additional active ingredients, to Formulate pharmaceutical compositions of the invention. A pharmaceutical composition of the invention comprises: (a) an effective amount of at least one active agent in accordance with the invention; and (b) a pharmaceutically acceptable excipient.
A "pharmaceutically acceptable excipient" refers to a substance that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to a subject, such as an inert substance, added to a pharmacological composition or otherwise used as a vehicle, carrier, or diluent to facilitate administration of an agent and that is compatible therewith. Examples of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.
Delivery forms of the pharmaceutical compositions containing one or more dosage units of the active agents may be prepared using suitable pharmaceutical excipients and compounding techniques known or that become available to those skilled in the art. The compositions may be administered in the inventive methods by a suitable route of delivery, e.g., oral, parenteral, rectal, topical, or ocular routes, or by inhalation. The preparation may be in the form of tablets, capsules, sachets, dragees, powders, granules, lozenges, powders for reconstitution, liquid preparations, or suppositories. Preferably, the compositions are Formulated for intravenous infusion, topical administration, or oral administration. For oral administration, the compounds of the invention can be provided in the form of tablets or capsules, or as a solution, emulsion, or suspension. To prepare the oral compositions, the compounds may be Formulated to yield a dosage of, e.g., from about 0.05 to about 100 mg/kg daily, or from about 0.05 to about 35 mg/kg daily, or from about 0.1 to about 10 mg/kg daily. For example, a total daily dosage of about 5 mg to 5 g daily may be accomplished by dosing once, twice, three, or four times per day.
Oral tablets may include a compound according to the invention mixed with pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservative agents. Suitable inert fillers include sodium and calcium carbonate, sodium and calcium phosphate, lactose, starch, sugar, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, and the like. Exemplary liquid oral excipients include ethanol, glycerol, water, and the like. Starch, polyvinyl-pyrrolidone (PVP), sodium starch glycolate, microcrystalline cellulose, and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin. The lubricating agent, if present, may be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract, or may be coated with an enteric coating.
Capsules for oral administration include hard and soft gelatin capsules. To prepare hard gelatin capsules, compounds of the invention may be mixed with a solid, semi-solid, or liquid diluent. Soft gelatin capsules may be prepared by mixing the compound of the invention with water, an oil such as peanut oil or olive oil, liquid paraffin, a mixture of mono and di-glycerides of short chain fatty acids, polyethylene glycol 400, or propylene glycol.
Liquids for oral administration may be in the form of suspensions, solutions, emulsions or syrups or may be lyophilized or presented as a dry product for reconstitution with water or other suitable vehicle before use. Such liquid compositions may optionally contain: pharmaceutically-acceptable excipients such as suspending agents (for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminium stearate gel and the like); non-aqueous vehicles, e.g., oil (for example, almond oil or fractionated coconut oil), propylene glycol, ethyl alcohol, or water; preservatives (for example, methyl or propyl p-hydroxybenzoate or sorbic acid); wetting agents such as lecithin; and, if desired, flavouring or colouring agents.
The active agents of this invention may also be administered by non-oral routes. For example, the compositions may be Formulated for rectal administration as a suppository. For parenteral use, including intravenous, intramuscular, intraperitoneal, or subcutaneous routes, the compounds of the invention may be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity or in parenterally acceptable oil. Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride. Such forms will be presented in unit-dose form such as ampules or disposable injection devices, in multi- dose forms such as vials from which the appropriate dose may be withdrawn, or in a solid form or pre-concentrate that can be used to prepare an injectable Formulation. Illustrative infusion doses may range from about 1 to 1000 μg/kg/minute of compound, admixed with a pharmaceutical carrier over a period ranging from several minutes to several days.
For topical administration, the compounds may be mixed with a pharmaceutical carrier at a concentration of about 0.1 % to about 10% of drug to vehicle. Another mode of administering the compounds of the invention may utilize a patch Formulation to affect transdermal delivery. Compounds of the invention may alternatively be administered in methods of this invention by inhalation, via the nasal or oral routes, e.g., in a spray Formulation also containing a suitable carrier.
Exemplary compounds of the invention, and exemplary compounds useful in methods of the invention will now be described by reference to the illustrative synthetic schemes for their general preparation below and the specific examples that follow. Artisans will recognize that, to obtain the various compounds herein, starting materials may be suitably selected so that the ultimately desired substituents will be carried through the reaction scheme with or without protection as appropriate to yield the desired product. Alternatively, it may be necessary or desirable to employ, in the place of the ultimately desired substituent, a suitable group that may be carried through the reaction scheme and replaced as appropriate with the desired substituent. Unless otherwise specified, the variables are as defined above in reference to Formula (I). Reactions may be performed between the melting point and the reflux temperature of the solvent, and preferably between 0 °C and the reflux temperature of the solvent. Reactions may be heated employing conventional heating or microwave heating. Reactions may also be conducted in sealed pressure vessels above the normal reflux temperature of the solvent.
Abbreviations and acronyms used herein include the following:
Figure imgf000046_0001
ddd Doublet of doublets of doublets
DCM dichloromethane
DIPEA N-ethyldiisopropylamine or
N,N-diisopropylethylamine
DMAP 4-(dimethylamino)pyridine
DMF N,N-dimethylformamide
DMMN Dimethylmalonitrile
DMSO-de hexadeuterodimethyl sulfoxide
EDC.HCI N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride
Et ethyl
Et3N triethylamine
EtOH ethanol
EtOAc ethyl acetate
g gram
HCI hydrochloric acid
HATU (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate)
H20 water
HPLC high pressure liquid chromatography
Hr hour
IPA Isopropyl alcohol
KHSO4 potassium bisulfate
K2CO3 Potassium carbonate
L litre
LCMS liquid chromatography mass spectrometry
LHMDS lithium bis(trimethylsilyl)amide
L1AIH4 lithium aluminium hydride
m multiplet
M molar
mBar millibar
MCPBA 3-chloroperbenzoic acid
Me methyl
MeOH methanol
MeOD-d4 deutero-methanol
2-MeTHF 2-methyltetrahydrofuran
mg milligram MHz mega Hertz mins minutes
ml_ millilitres
mmol millimole
MS m/z mass spectrum peak
MsCI methanesulfonyl chloride
MTBE Methyl te/f-butyl ether
M/V Mass volume ratio
N2 nitrogen
NaBH4 sodium borohydride
NaHCOs sodium bicarbonate
NaOH sodium hydroxide
NBS N-bromosuccinimide
NHs ammonia
NH4CI ammonium chloride
Na2S04 sodium sulphate
Pd(OH)2/C palladium hydroxide on carbon
PhSeBr phenylselenyl bromide
Pt02 platinum (IV) oxide
q quartet
rt room temperature
RT retention time
s singlet
sat. saturated
sex strong cation exchange
SOCI2 Thionyl chloride
soln. solution
t triplet
TBAF terf-butyl ammonium fluoride
TFA trifluoroacetic acid
THF tetrahydrofuran
TIPS triisopropylsilyl chloride
TMS trimethylsilyl
T3P Propylphosphonic anhydride μΙ_ micro litres
v/v volume volume percent w/w Weight/weight percent
All of the derivatives of Formula (I) can be prepared by the procedures described in the general methods presented below or by routine modifications thereof. The present invention also encompasses any one or more of these processes for preparing the derivatives of Formula (I), in addition to any novel intermediates used therein.
The routes below, including those mentioned in the Examples and Preparations, illustrate methods of synthesising the compound of Formula (I). The skilled person will appreciate that the compound of the invention, and intermediates thereto, could be made by methods other than those specifically described herein, for example by adaptation of the methods described herein, for example by methods known in the art. Suitable guides to synthesis, functional group interconversions, use of protecting groups, etc., are for example: "Comprehensive Organic Transformations" by RC Larock, VCH Publishers Inc. (1989); "Advanced Organic Chemistry' by J. March, Wiley Interscience (1985); "Designing Organic Synthesis" by S Warren, Wiley Interscience (1978); "Organic Synthesis - The Disconnection Approach" by S Warren, Wiley Interscience (1982); "Guidebook to Organic Synthesis" by RK Mackie and DM Smith, Longman (1982); "Protective Groups in Organic Synthesis" by TW Greene and PGM Wuts, Fifth Ed, John Wiley and Sons, Inc. (2014); and "Protecting Groups" by PJ, Kocienski, Georg Thieme Verlag (1994); and any updated versions of these standard works.
In addition, the skilled person will appreciate that it may be necessary or desirable at any stage in the synthesis of compounds of the invention to protect one or more sensitive groups, so as to prevent undesirable side reactions. In particular, it may be necessary or desirable to protect amino or carboxylic acid groups. The protecting groups used in the preparation of the compounds of the invention may be used in a conventional manner. See, for example, those described in 'Greene's Protective Groups in Organic Synthesis' by Theodora W Greene and Peter G M Wuts, fifth edition, (John Wiley and Sons, 2014), in particular Chapter 7 ("Protection for the Amino Group"), incorporated herein by reference, which also describes methods for the removal of such groups.
In the general synthetic methods below, unless otherwise specified, the substituents are as defined above with reference to the compound of Formula (I), or (l)(a), above.
Where ratios of solvents are given, the ratios are by volume. The compounds of the invention may be prepared by any method known in the art for the preparation of compounds of analogous structure. In particular, the compound of the invention can be prepared by the procedures described by reference to the Schemes that follow, or by the specific methods described in the Examples, or by similar processes to either.
The skilled person will appreciate that the experimental conditions set forth in the schemes that follow are illustrative of suitable conditions for effecting the transformations shown, and that it may be necessary or desirable to vary the precise conditions employed for the preparation of the compound of Formula (I). It will be further appreciated that it may be necessary or desirable to carry out the transformations in a different order from that described in the schemes, or to modify one or more of the transformations, to provide the desired compound of the invention. A compound of Formula (l)(a) may be prepared from compounds of Formulae (ll)(a),
(lll)(a), (IV)(a), (V)(a), (Vl)(a), (Vll)(a), (Xll)(a), (Xlll)(a) and (XIX) as illustrated by Scheme 1.
Figure imgf000051_0001
where:
PGi and PG2 are each independently an amine protecting group; LG is a leaving group.
PGi and PG2 may each independently be selected from: a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, tert- butylcarbamate, 9-Fluorenylmethyl carbamate, benzhydryl, trityl group.
In particular embodiments, PGi is a carbamate group, preferably f-butyl carbamate, and PG2 is benzyl.
Examples of suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group.
In particular embodiments, LG is chloro. A compound of Formula (Xlll)(a) (where PGi = 'BoC and PG2 = Bn) may be prepared from a compound of Formula (Xll)(a), using an acid-mediated cyclisation reaction with N- (methoxymethyl)-N-(trimethylsilylmethyl)benzylamine (XIX) in the presence of a suitable acid, such as TFA, in a suitable aprotic solvent, such as DCM, at a suitable temperature such as 0°C to rt.
A compound of Formula (ll)(a) may be prepared by acid mediated deprotection of a compound of Formula (Xlll)(a) (where PGi = 'BoC) using an appropriate acid, such as TFA, in a suitable aprotic solvent, such as DCM, at a suitable temperature such as 0°C to rt. A compound of Formula (lll)(a) may be prepared from a compound of Formula (l)(a) using a suitable reducing agent, such as UAIH4, in a suitable aprotic solvent, such as 2-MeTHF, at a suitable temperature, such as between 0°C and reflux, preferably at room temperature.
A compound of Formula (IV)(a) may be prepared from a compound of Formula (lll)(a) and 2- fluoro-4-methoxybenzoic acid using an amide bond forming reaction in the presence of a suitable coupling reagent, such as (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5-b]pyridinium
3-oxid hexafluorophosphate) (HATU) or propylphosphonic anhydride (T3P), in the presence of a suitable organic base, such as DIPEA or ΕίβΝ, in an appropriate aprotic solvent, such as DMF or
MeCN, at a suitable temperature, such as 0-100°C, preferably at rt. Alternatively, a compound of Formula (IV)(a) may be prepared from a compound of Formula (lll)(a) and 2-fluoro-4- methoxybenzoyl chloride, prepared in situ from 2-fluoro-4-methoxybenzoic acid, by treatment with a suitable reagent such as SOCI2 or PCI3 in a non polar solvent such as toluene at elevated temperature, followed by reaction with a compound of Formula (lll)(a), in the presence of a suitable organic base, such as EhN or DIPEA in a suitable solvent such as DCM. A skilled person will appreciate that alternative coupling conditions are available in combination with a range of alternative bases and solvents. A compound of Formula (V)(a) may be prepared from a compound of Formula (IV)(a) (where PG2 = Bn) using suitable deprotection conditions, such as transfer or palladium catalysed hydrogenation, using for example palladium on charcoal in a suitable solvent, such as EtOH, at a suitable temperature, such as rt, under an atmosphere of hydrogen. A skilled person will appreciate that alternative hydrogenation conditions are available to effect this transformation.
A compound of Formula (Vl)(a) may be prepared from a compound of Formula (V)(a) and a compound of Formula (XVI I I) in the presence of a suitable base, such as Cs2C03 or K2CO3, in a suitable solvent, such as DMF or EtOH, at an elevated temperature, such as 80-100°C.
A compound of Formula (Vl l)(a) may be prepared from a compound of Formula (Vl)(a) by a bromination using a suitable brominating agent such as N-bromo succinimide (NBS) in a suitable solvent such as MeCN at between room temperature and 100°C, typically at 50°C. A compound of Formula (l)(a) may be prepared from a compound of Formula (Vl l)(a) by a cyanation reaction using a suitable cyanating agent such as potassium hexacyanoferrate(l l) trihydrate in the presence of a suitable copper catalyst such as Cul and a suitable ligand such as 1-butylimidazole in a suitable solvent such as toluene at elevated temperature, typically 150°C.
A compound of Formula (Xl l)(a) may be prepared from a compound of Formula (VI I l)(a), (IX)(a), (X)(a), and (Xl)(a), as illustrated by Scheme 2:
Figure imgf000054_0001
Figure imgf000054_0002
Scheme 2
where:
PGi is an amine protecting group, as hereinbefore defined.
A compound of Formula (IX)(a) may be prepared by condensation of Boc-L-alanine (VI I l)(a) and Meldrum's acid in the presence of a suitable condensation reagent, such as EDC.HCI, optionally in the presence of a suitable catalyst, such as DMAP, in a suitable solvent, such as DCM, at a suitable temperature, such as between 0°C and rt.
A compound of Formula (X)(a) may be prepared from a compound of Formula (IX)(a) by treatment with heat in a suitable solvent, such as EtOAc, at an appropriate elevated temperature, between 60°C and reflux, optionally at reduced pressure.
A compound of Formula (X)(a) may be obtained directly from Boc-L-alanine (VI I l)(a), without isolating the compound of Formula (IX)(a).
A compound of Formula (Xl)(a) may be prepared by the reduction of a compound of Formula (X)(a) in the presence of a suitable catalyst such as PtC>2 in the presence of hydrogen gas at a suitable pressure in a solvent such as EtOAc. Alternatively, this transformation may be achieved by treatment of a compound of Formula (X)(a) with a suitable reducing agent such as NaBhU in the presence of an acid such as AcOH is a suitable solvent such as DCM at between 0°C and room temperature. A compound of Formula (Xll)(a) may be prepared from a compound of Formula (Xl)(a) using a suitable base-mediated elimination reaction using, for instance an in situ conversion of the alcohol into a methansulfonyl leaving group in the presence of a suitable non-nucleophilic base, such as Et3N, in a suitable solvent, such as DCM, at a suitable temperature, such as 0°C to rt. A skilled person will appreciate that alternative leaving group and elimination conditions are available in combination with a range of alternative bases and solvents.
A compound of Formula (Xll)(a) may alternatively be prepared from a compound of Formula (XIV)(a), (XV)(a), (XVI)(a), and (XVII)(a) as illustrated by Scheme 3:
Figure imgf000055_0001
Scheme 3 where:
PGi is an amine protecting group, as hereinbefore defined.
A compound of Formula (XV)(a) may be prepared from (2S)-2-methylpyrrolidine hydrochloride and B0C2O in a suitable solvent, such as DCM, in the presence of a suitable base, such as Et3N.
A compound of Formula (XVI)(a) may be prepared by the oxidation of a compound of Formula (XV) using appropriate oxidising agents such as ruthenium(IV)oxide hydrate and sodium metaperiodate in water at a suitable temperature, such as 15-20°C. A compound of Formula (XVI I)(a) may be prepared from a compound of Formula (XVI)(a) by deprotonation using a suitable non-nucleophilic base, such as LHMDS, in a suitable aprotic solvent, such as THF, at low temperature, such as -78°C, followed by quenching with a suitable electrophilic source of phenylselenide, such as PhSeBr, at a suitable temperature, such as rt.
A compound of Formula (Xll)(a) may be prepared from a compound of Formula (XVII)(a) by an oxidative elimination reaction using a suitable oxidising agent, such as MCPBA, in a suitable solvent, such as DCM, at a suitable low temperature, such as -40°C.
A compound of Formula (Vll)(a) may alternatively be prepared from a compound of Formula (XX)(a), (XXI)(a), (XVIII) and (XXII)(a) as illustrated by Scheme 4:
Figure imgf000056_0001
(Vl)(a)
Scheme 4
where: PGi and PG2 are each independently an amine protecting group; LG is a leaving group.
PGi and PG2 may each independently be selected from: a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, tert- butylcarbamate, 9-Fluorenylmethyl carbamate, benzhydryl, trityl group.
In particular embodiments, PGi is a carbamate group, preferably f-butyl carbamate, and PG2 is benzyl. Examples of suitable leaving groups include: a chloro, bromo, mesylate, or tosylate group. In particular embodiments, LG is chloro.
A compound of Formula (XX)(a) may be prepared from a compound of Formula (lll)(a) under standard conditions for introducing an amine protecting group. Where PGi = 'BoC, a compound of Formula (lll)(a) is treated with Boc20 in a suitable solvent such as DCM, typically at room temperature to provide a compound of Formula (XX)(a).
A compound of Formula (XXI)(a) may be prepared from a compound of Formula (XX)(a) (where PG2 = Bn) using suitable deprotection conditions, such as transfer or palladium catalysed hydrogenation, using for example ammonium formate and palladium hydroxide on charcoal in EtOH at reflux. A skilled person will appreciate that alternative hydrogenation conditions are available to effect this transformation. A compound of Formula (XXII)(a) may be prepared from a compound of Formula (XXI)(a) and a compound of Formula (XVIII) in the presence of a suitable base, such as CsaCC or K2CO3, in a suitable solvent, such as DMF or EtOH, at an elevated temperature, such as 80- 110°C. A compound of Formula (XXIII)(a) may be prepared by acid mediated deprotection of a compound of Formula (XXII)(a) (where PGi = 'BoC) using an appropriate acid, such as TFA, in a suitable aprotic solvent, such as DCM, at a suitable temperature such as 0°C to rt.
A compound of Formula (Vl)(a) may be prepared from a compound of Formula (XXIII)(a) and 2-fluoro-4-methoxybenzoic acid using an amide bond forming reaction in the presence of a suitable coupling reagent, such as (1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5- b]pyridinium 3-oxid hexafluorophosphate) (HATU) or propylphosphonic anhydride (T3P), in the presence of a suitable organic base, such as DIPEA or Et3N, in an appropriate aprotic solvent, such as DMF or MeCN, at a suitable temperature, such as 0-100°C, preferably at rt. A skilled person will appreciate that alternative coupling conditions are available in combination with a range of alternative bases and solvents.
A compound of Formula (I) or (l)(a) may be converted to its corresponding salt using methods known to those skilled in the art. For example, amines of Formula (I) or (l)(a) may be treated with trifluoroacetic acid (TFA), HCI, sulfuric acid (H2SO4), maleic acid, or citric acid in a solvent such as diethyl ether (Et20), CH2CI2, tetrahydrofuran (THF), or methanol (MeOH) to provide the corresponding salt forms.
The skilled person will appreciate that the experimental conditions set forth in the schemes above are illustrative of suitable conditions for effecting the transformations shown, and that they may also be carried out on compounds of the opposite enantiomer to that described or indeed to a racemic mixture. In the latter case a skilled person will also appreciate that individual isomers may be isolated by means of suitable chromatographic conditions.
Compounds prepared according to the schemes described above may be obtained as single enantiomers, diastereomers, or regioisomers, by enantio- , diastero-, or regiospecific synthesis, or by resolution. Compounds prepared according to the schemes above may alternately be obtained as racemic (1 : 1) or non-racemic (not 1 : 1) mixtures or as mixtures of diastereomers or regioisomers. Where racemic and non-racemic mixtures of enantiomers are obtained, single enantiomers may be isolated using conventional separation methods known to one skilled in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation. Where regioisomeric or diastereomeric mixtures are obtained, single isomers may be separated using conventional methods such as chromatography or crystallization. The following examples are provided to further illustrate the invention and various preferred embodiments.
Examples
General methods
1 H nuclear magnetic resonance (NMR) spectra were in all cases consistent with the proposed structures. Characteristic chemical shifts (δ) are given in parts-per-million downfield from tetramethylsilane (for 1 H-NMR) using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad. The following abbreviations have been used for common solvents: CDCI3, deuterochloroform; DMSO-d6, hexadeuterodimethyl sulfoxide; and MeOD-d4, deuteron-methanol. Where appropriate, tautomers may be recorded within the NMR data; and some exchangeable protons may not be visible.
Mass spectra, MS (m/z), were recorded using either electrospray ionisation (ESI) or atmospheric pressure chemical ionisation (APCI). Where relevant and unless otherwise stated the m/z data provided are for isotopes 19F, 35CI, 79Br and 127l.
Where preparative TLC or silica gel chromatography have been used, one skilled in the art may choose any combination of solvents to purify the desired compound.
Preparation 1
tert-Butyl (2S)-5-hvdroxy-2-methyl-3-oxo-2,3-dihvdro-1 H-pyrrole-1-carboxylate
Figure imgf000059_0001
In to a 20 L reactor was charged DCM (5.2 L) followed by Boc-L-alanine (650 g, 3.43 mol), Meldrum's acid (545 g, 3.77 mol) and DMAP (630 g, 5.15 mol) and the line washed with further DCM (1.3 L). The solution was cooled to 0 °C, then EDC HCI (790 g, 4.122 mol) was added portion wise over 20 mins and the reaction stirred at rt for 3.5 hr. The solution was washed with 1 M citric acid soln. (4x 3.9 L) and water (3.9 L). The organic solution was concentrated to ca. 2.6 L, then EtOAc (6.5 L) was added and the mixture further concentrated to ca. 2.6 L. EtOAc (3.9 L) was added again and the solution maintained at 0°C for 18 hrs. The solution was heated to 80°C with an internal vacuum of 800 mBar in distillation mode (internal temperature 66-67°C) and stirred for 1.5 hr. The solvent was evaporated to minimum volume (1 L) and the solution used in the next step without further purification. A drop of this solution was evaporated to obtain an analytical sample.
LCMS m/z = 314.4 [M-H]- Preparation 2
fe/f-Butyl (2S)-3-hvdroxy-2-methyl-5-oxopyrrolidine-1-carboxylate
Figure imgf000060_0001
The solution of te/f-butyl (2S)-5-hydroxy-2-methyl-3-oxo-2,3-dihydro-1 H-pyrrole-1- carboxylate (Preparation 1 , 732 g, 3.43 mol) was diluted with DCM (5.1 L) and AcOH (2.2 L), and the mixture cooled to 0 °C. NaBH4 (195 g, 5.15 mol) was added portion wise over 6 hrs, water (7.3 L) was carefully added to quench the reaction and the mixture stirred for 15 hrs. The phases were separated, the DCM layer was collected and the aqueous phase washed with DCM (0.7 L x4). The combined organic layers were filtered through a Gooch filter, then AcOH (2.2 L) added and the solution cooled to 0 °C. NaBH4 (97.5 g, 2.57 mol) was added portion wise over 1 hr and the resulting thick suspension stirred for 30 mins. K2CO3 (solution 40% M/V) (7.3 L) was added and the two phases were separated. The water layer was extracted with DCM (1 L x 2) and the combined organic phases were dried (Na2S04) and filtered to obtain a solution which was used for the next step without further purification. 10 mL of this solution was evaporated to dryness to obtain 1.625 g of product (estimated yield 650 g, 88%).
1 HNMR (DMSO-de, 400MHz) δ : 1.18 (d, 3H), 1.45 (s, 9H), 2.35-2.57 (ddd, 2H), 4.05 (quintet, 1 H), 4.24-4.29 (m, 1 H), 5.30 (br s, 1 H). Preparation 3
fe/f-Butyl (2S)-2-methyl-5-oxo-2,5-dihvdro-1 H-pyrrole-1-carboxylate
Figure imgf000060_0002
Et3N (1.06 L, 7.63 mol) was added to a solution of te/f-butyl (2S)-3-hydroxy-2-methyl-5- oxopyrrolidine-1 -carboxylate (Preparation 2, 651 g, 3.05 mol) in DCM (6.5 L) and the mixture cooled to 0 °C. MsCI (283 mL, 3.66 mol) was added dropwise over 2 hrs, and the reaction then allowed to warm to rt, and stirred for 2 hrs. The mixture was washed with water (3 x 2L). The organic solution obtained was used in the following step without any further purification. 10 mL of this solution was evaporated to dryness to obtain 0.843 g of product (estimated yield 590 g, 98%). 1 HNMR (DMSO-d6, 400MHz) δ : 1.35 (d, 3H), 1.48 (s, 9H), 4.58-4.65 (m, 1 H), 6.08 (dd, 1 H), 7.43 (dd, 1 H).
Preparation 4
tert- Butyl (1S,3aR,6aS)-5-benzyl-1-methyl-3-oxo-octahvdropyrrolo[3,4-clpyrrole-2- carboxylate
Figure imgf000061_0001
To an ice-cooled stirred solution of te/f-butyl (2S)-2-methyl-5-oxo-2,5-dihydro-1 H-pyrrole- 1-carboxylate (Preparation 3, 590 g, 2.99 mol) in DCM (5.9 L) and TFA (62 mL, 0.81 mol) was added N-(methoxymethyl)-N-(trimethylsilylmethyl) benzylamine (1721 mL, 6.73 mol) dropwise over 4 hrs and the mixture then stirred at rt for 15 hrs. The reaction mixture was partitioned between DCM and saturated NaHCOs soln. (5.9 L), the layers separated and the aqueous phase extracted twice with DCM (2x 1.2 L). The combined organic layers were evaporated, the residue diluted with EtOAc (3.0 L) and the solution washed with aq. NH4CI soln. (3x 3.0 L at pH ~5) and once with water (3.0 L). The solvent was concentrated to minimum volume (~ 2L) under vacuum to afford an orange oil (1.9 Kg) which was taken up in Cy:EtOAc 95:5 (3.8 L) and stirred for 60 hrs. The resulting solid formed was filtered off to afford the title compound as an off-white solid, (470 g, 49%).
The mother liquors were evaporated to dryness and the orange oil (1.3 Kg) was passed through a Si02 pad (1.2 Kg) eluting with Cy (2.4 L, [discarded]) then Cy:EtOAc 50:50 (6.0 L). The solution collected was evaporated and the residue taken up in EtOAc (590 mL) and charged into a reactor. The solvent was concentrated to a minimum volume, Cy (1.2 L) was added and the mixture stirred at rt for 15 hrs. The slurry was filtered under vacuum, washed with Cy (200 mL) and oven dried to obtain additional title compound as a white solid, (140 g, 14%).
The mother liquors were evaporated to dryness and the orange oil (270 g) taken up in Cy (0.6 L) and stirred for 15 hrs at rt. The solid formed was filtered under vacuum and washed with Cy (4x 25mL portions) to obtain further title compound as a white solid, (60 g, 6%). LCMS m/z = 331.3 [MH]+ Preparation 5
(3S,3aS,6aR)-5-Benzyl-3-methyl-octahvdropyrrolor3,4-clpyrrol-1-one
Figure imgf000062_0001
TFA (1.423 L, 18.58 mol) was added to a solution of te/f-butyl (1 S,3aR,6aS)-5-benzyl-1- methyl-3-oxo-octahydropyrrolo[3,4-c]pyrrole-2-carboxylate (Preparation 4, 470 g, 1.422 mol) in DCM (4.7 L) and the reaction stirred at rt for 15 hrs. The reaction was cooled to -5 °C and freshly prepared 6M NaOH was added dropwise keeping the internal temperature below 15 °C to neutralise the solution. Once addition was complete, EtOAc (300 ml_) was added and the two phases were separated. The organic phase was washed with water (2 L) then passed through a thin layer of Celite® to break the emulsion. The organic solution was washed again with water (2 L) filtered through a thin layer of Celite® then charged into a reactor. The solution was cooled to 0 °C and allowed to stand for 15 hrs. The temperature was increased to 20 °C, the solvent evaporated to dryness, then azeotroped with MeTHF to obtain the title compound as a dark orange oil, 380 g, in quantitative yield.
LCMS m/z = 231.0 [MH]+
Preparation 6
(1S,3aS,6aS)-5- Benzyl- 1 -methyl-octahydropyrrolo[3,4-clpyrrole
Figure imgf000062_0002
UAIH4 pellets (133 g, 3.56 mol) was carefully added to 2-MeTHF (2.6 L) and the dark suspension was stirred at rt for 15 hours. A solution of (3S,3aS,6aR)-5-benzyl-3-methyl- octahydropyrrolo[3,4-c]pyrrol-1-one (Preparation 5, 327 g, 1.422 mol) in 2-MeTHF (660 ml_) was added dropwise at 0 °C over 1 hr and the resulting reaction was heated under reflux for 3 hrs. The reaction was cooled to -10°C, then water (135 ml_), NaOH (15% w/w, 135 ml_) and water (405 ml_) were slowly added in this order over 2 hrs. The white precipitate obtained was stirred for 15 hrs at rt, filtered and washed with 2-MeTHF (2 L). The filtrate was evaporated to dryness to afford the title compound as a clear orange oil, 280 g, 91 %. LCMS m/z = 217.3 [MH]+ Preparation 6A
(1S,3aS,6aS)-5-Benzyl-1 -methyl-octahvdropyrrolor3,4-clpyrrole hydrogen sulfate
Figure imgf000063_0001
A solution of H2SO4 (9.06 g, 92.45 mmol) in IPA (60 mL) was added dropwise over 30 min to a solution of (1 S,3aS,6aS)-5-benzyl-1-methyl-octahydropyrrolo[3,4-c]pyrrole (Preparation 6, 20 g, 92.5 mmol) in MeOH (60 mL) and IPA (40 mL) at rt. The obtained suspension was stirred for ca. 1.5 hrs at rt, then cooled to 10°C and stirred for 1.5 hrs. The resulting suspension was filtered and the solid washed with IPA (20 mL) and MTBE (20 mL) and dried overnight under vacuum, to yield the title compound, 25.5g, 87% yield.
Preparation 7
(1S,3aR,6aS)-5-Benzyl-2-(2-fluoro-4-methoxybenzoyl)-1-methyl-octahydropyrrolor3,4- cl pyrrole
Figure imgf000063_0002
H
2-Fluoro-4-methoxybenzoic acid (35g, 204 mmol) was suspended in SOCI2 (70 mL) and the mixture stirred at 80 °C for 5 hr. The mixture was evaporated under vacuum and azeotroped with toluene (3x50 mL). The resulting oil was dissolved in DCM (300 mL), cooled to 0 °C and Et3N (85 mL, 582 mmol) added. (1 S,3aS,6aS)-5-Benzyl-1 -methyl-octahydropyrrolo[3,4- c]pyrrole hydrogen sulfate (Preparation 6a, 49.2 g, 194 mmol) was added portion wise over 5 mins and the reaction then stirred for 30 mins. Sat. NaHCC soln. (300 mL) was added and the mixture was stirred for 30 mins. The organic phase was separated, additional sat. NaHCC>3 soln. (200 mL) added and the mixture stirred for 18 hrs. The organic phase was separated, dried (Na2S04) and evaporated under reduced pressure to afford the title compound.
LCMS m/z = 369.0 [MH]+ Preparation 8
(1S,3aR,6aS)-2-(2-Fluoro-4-methoxybenzoyl)-1 -methyl-octahvdropyrrolor3,4-clpyrrole
Figure imgf000064_0001
(1S,3aR,6aS)-5-Benzyl-2-(2-fluoro-4-methoxybenzoyl)-1-methyl-octahydropyrrolo[3,4- c]pyrrole (Preparation 7, 75.5 g, 202 mmol) was dissolved in EtOH (1 L), Pd/C 10% (1 1 g, 10% w/w) was added and the solution stirred under H2 atmosphere (6 bar) for 20 hrs. The mixture was filtered through a pad of Celite® and the solvent was evaporated in vacuo to obtain the title compound as a sticky oil, 56 g, 99%.
LCMS m/z = 279.0 [MH]+
Preparation 9
2-r(3aS,4S,6aR)-5-(2-fluoro-4-methoxybenzoyl)-4-methyl-octahydropyrrolor3,4-clpyrrol-2- yl1-4,6-dimethylpyrimidine
_ CH
Figure imgf000064_0002
A mixture of (1S,3aR,6aS)-2-(2-fluoro-4-methoxybenzoyl)-1 -methyl-octahydropyrrolo[3,4- c]pyrrole (Preparation 8, 10 g, 35.93 mmol), 2-chloro-4,6-dimethylpyrimidine (5.64 g, 39.52 mmol) and Cs2C03 (14.05 g, 43.12 mmol) in DMF (100 ml_) was heated at 100°C for 18 hrs. The cooled reaction was partitioned between brine and EtOAc, the two phases separated and the organic phase dried (Na2SC>4), filtered and concentred in vacuo. The crude product was dissolved in EtOH (5 V). Water was added drop wise (18 V), the mixture was filtered and washed with additional water and then Et20 to afford the title compound, 10.84 g, 28.19 mmol, 78% yield, as a beige solid. The aqueous phase was evaporated and the residue purified by column chromatography on silica gel eluting with DCM:EtOAc, (95:5 to 50:50) to afford additional product, 2.35 g, 17% yield, as a beige solid.
LCMS m/z = 385.2 [MH]+ Preparation 10
2-r(3aS,4S,6aR)-5-(2-fluoro-4-methoxybenzoyl)-4-rnethyl-octahvdropyrrolor3,4-clpyrrol-2- yll-5-bromo-4,6-dimethylpyrimidine
_ CH
Figure imgf000065_0001
NBS (10.79 g, 60.63 mmol) was added to a solution of 2-[(3aS,4S,6aR)-5-(2-fluoro-4- methoxybenzoyl)-4-methyl-octahydropyrrolo[3,4-c]pyrrol-2-yl]-4,6-dimethylpyrimidine
(Preparation 9, 21.19 g, 55.12 mmol) in MeCN (500 ml_) and the reaction stirred at 50 °C for 1 hr. The cooled mixture was concentrated in vacuo, and the residue partitioned between EtOAc and water. The organic phase was evaporated to afford crude product, which was dissolved in EtOH (53 ml_) and water was added dropwise (158 ml_). The resulting suspension was filtered and the solid washed with water to afford the title compound as a beige solid, 23.27 g, 91 % yield.
1 HNMR (400MHz, DMSO-d6) mixture of rotamers δ: 0.96, 1.30 (2xd, 3H), 2.37, 2.39 (2xs, 6H), 2.60-3.25 (m, 3H), 3.34-4.09 (m, 9H), 6.77-6.96 (m, 2H), 7.26-7.30 (m, 1 H).
Example 1
2-r(3aS,4S,6aR)-5-(2-fluoro-4-methoxybenzoyl)-4-methyl-octahydropyrrolor3,4-clpyrrol-2- yll-4,6-dimethylpyrimidine-5-carbonitrile
Figure imgf000065_0002
(l)(a)
Potassium hexacyanoferrate(ll) trihydrate (9.12 g, 21.6 mmol) was dried in vacuo at 80°C for 12 hrs. Cul (4.9 g, 26.0 mmol) and 2-[(3aS,4S,6aR)-5-(2-fluoro-4-methoxybenzoyl)-4-methyl- octahydropyrrolo[3,4-c]pyrrol-2-yl]-5-bromo-4,6-dimethylpyrimidine (Preparation 10, 10 g, 21.6 mmol) were added. The reaction mixture was stirred in toluene (30.16 ml_) and 1- butylimidazole (5.8 ml_) at 150°C for 19 hrs. The cooled mixture was partitioned between DCM and water, the emulsion filtered and washed with brine. The organic phase was evaporated under reduced pressure to afford a crude product. The reaction was repeated and the two batches of crude product combined. The crude was purified by column chromatography on silica gel eluting with DCM:MeOH (100:0 to 90: 10). This product was dissolved in MeCN (95 ml_) and water was added dropwise (145 ml_) to obtain a precipitate which was filtered and washed with water to afford a beige solid. This was purified by column chromatography on silica gel eluting with Hexane:EtOAc (95:5 to 0: 100) to afford the title compound as a white solid (99% UV purity), 6.31 g, 36%, and a second batch (94% UV purity) as a white solid, 1.9 g, 11 %.
1 HNMR (400MHz, MeOD-d4) (mixture of rotamers) δ: 1.10, 1.42 (2xd, 3H), 2.34-2.50 (m, 6H), 2.70-3.27 (m, 2H), 3.40-4.28 (m, 10H), 6.72-6.92 (m, 2H), 7.22-7.39 (m, 1 H).
LCMS m/z = 410.3 [MH]+
Example 2
In vitro affinity of the compound of Example 1 for human orexin-1 and orexin-2 receptors
The in vitro binding affinity of 2-[(3aS,4S,6aR)-5-(2-fluoro-4-methoxybenzoyl)-4-methyl- octahydropyrrolo[3,4-c]pyrrol-2-yl]-4,6-dimethylpyrimidine-5-carbonitrile
(the compound of Example 1) for human recombinant OX1 (hOX1 R) and OX2 (hOX2R) receptors was determined using a Scintillation Proximity binding Assay (SPA).
A 96-well plate SPA was developed using antagonist radioligands (OX-radioligands); [3H]- SB674042 (Langmead et a/., 2004; Malherbe et a/., 2009a) for hOX1 R and [3H]-EMPA (Malherbe et al., 2009b) for hOX2R. Cell membranes were prepared from recombinant cell lines; a CHO cell line transformed with hOX1 R (GeneBank accession# NM_001525) and a HEK cell line transformed with hOX2R (GeneBank accession* NM_001526.2).
On the day of the assay, membranes containing hOX1 R or hOX2R were captured onto SPA- beads. All test compounds were dissolved in 100% DMSO as 10mM stock solutions, and prepared as 8-point semi-log dilution in 100% DMSO immediately prior to assay (final assay range 5uM to 0.064nM). The compound of Example 1 was co-administered with the appropriate OX-radioligand, and the SPA-membrane preparation added to initiate the assay. During the assay, any compound of Example 1 that bound to the receptors competed with radioligand and reduced the assay signal. Assay plates were read using PerkinElmer Microbeta2 allowing the binding profile of the compound of Example 1 to be assessed at room temperature (19°C) over several time points (15, 60, 90, 120, 150 minutes, and 18 hours) in hOX1 R and hOX2R assay formats.
Data Handling and Analysis.
End-point data values were determined as Specific Binding (SB) and expressed as a percentage of control (%SB), with 100% being the SB of OX-radioligand in the absence of competing unlabelled ligand, and 0% being the SB of the relevant OX-radioligand in the presence of an excess of the competing cold/unlabelled ligand (Suvorexant; a dual OXR- antagonist [DORA]). TB= Total Bound (in presence of 0.5% DMSO);
NSB= Non Specific Bound (in the presence of Suvorexant at 1 μΜ) = 0%;
SB= Specific Bound (TB-NSB) = 100%
Curve fitting and Affinity determination
Assay data were used to determine half maximal inhibitory concentration (IC50) using a four- parameter logistic model in XLfit and/or GraphPad Prism v5. IC50 were derived from plots of %SB against log 10 of the concentration of the compound of Example 1. The pKi were calculated from the IC50 using the Cheng-Prusoff correction: pKi = IC50 /(1+([L]/KD)) where [L] is the radioligand concentration in the displacement assay, and KD is the dissociation constant of the radioligand. The Selectivity value is derived from the Ki [Ki (OX2R)/Ki(OX1 R)].
The results are shown in tables 1 and 2 below:
Table 1
Figure imgf000067_0001
* data at 60min
** selectivity calculated using Ki (Ox2R)/Ki(Ox1 R)
Table 2
Figure imgf000067_0002
The results show that the compound of Example 1 has high affinity for hOX1 R and hOX2R, with 61 -fold selectivity for hOX2R.
Example 3
In vitro affinity of the compound of comparative Example A for human orexin-1 and orexin-2 receptors
2-r(3aS.4S.6aR)-5-(2-fluoro-4-methoxybenzovn-octahvdropyrrolor3.4-clpyrrol-2-yl1-4.6- dimethylpyrimidine-5-carbonitrile (comparative Example A)
Figure imgf000068_0001
was prepared and tested by analogous methods to those described above in Example 1 and Example 2 in respect of the compound of Example 1.
The results for comparative example A are shown in Tables 3 and 4 below
Table 3
Figure imgf000068_0002
* data at 60min
Table 4 Compound hOX1 R hOX2R hOX2R Binding Selectivity
Binding pKi (60 Binding pKi (60 Equilibration hOX2R/hOX1 R** min) min) Time (minutes)
Example A < 5 6.66 nd nd
Data: geomet ric mean, derived 1 "rom 4 duplicate runs
selectivity calculated using Ki(Ox2R)/Ki(Ox1 R)
The results show that the compound of Example 1 has higher affinity for hOX2R than comparative Example A. The compound of Example 1 differs from comparative Example A of by virtue of 4-methyl substitution of octahydropyrrolo[3,4-c]pyrrol-2-yl moiety. The results therefore evidence that this methyl substitution gives rise to the increased affinity at hOX2R when compared to comparative Example A. References
Langmead C.J., Jerman J.C., Brough S.J., Scott C, Porter R.A., Herdon H.J.
(2004). Characterisation of the binding of [3H]-SB-674042, a novel nonpeptide antagonist, to the human orexin-1 receptor. Br. J. Pharmacol. , 141 : 340-346;
Malherbe P., Borroni E., Pinard E., Wettstein j.G., and Knoflach F. (2009a)
Biochemical and Electrophysiological Characterization of Almorexant, a Dual Orexin
Receptor (OX1)/Orexin 2 Receptor (OX2) Antagonist: Comparison with Selective OX1 and OX2 Antagonists. Mol. Pharmacol. 76, 618-631 ;
Malherbe P., Borroni E., Gobbi L, Knust H., Nettekoven M., Pinard E., Roche O., Rogers-Evans M., Wettstein J.G. and Moreauet J-L, (2009b) Biochemical and behavioural characterization of EMPA, a novel high-affinity, selective antagonist for the OX2 receptor. Br. J. Pharmacol. 156, 1326-1341.

Claims

A compound of Formula (I):
Figure imgf000070_0001
(I)
or a pharmaceutically acceptable salt, solvate, hydrate, geometrical isomer, tautomer, or optical isomer thereof.
2. A compound according to claim 1 , wherein the central hydrogen atoms on the methylated octahydropyrrolo[3,4-c]pyrrole group are in a cis position.
3. A compound according to claim 1 or 2, wherein the compound is of Formula (l)(a):
_ CH
Figure imgf000070_0002
(l)(a)
or a pharmaceutically acceptable salt, solvate, hydrate, geometrical isomer, tautomer, or optical isomer thereof.
4. A pharmaceutical composition comprising a compound according to any of claims 1 to 3, and a pharmaceutically acceptable excipient.
5. A compound according to any of claims 1 to 3, or a composition according to claim 4, for use as a medicament.
6. A compound according to any of claims 1 to 3, or a composition according to claim
4, for use in treating a disease, disorder or condition mediated by orexin receptor activity.
7. Use of a compound according to any of claims 1 to 3, or a composition according to claim 4, in the manufacture of a medicament for treating a disease, disorder or condition mediated by orexin receptor activity.
8. A method of treating a disease, disorder or condition mediated by orexin receptor activity, which comprises administering an effective amount of a compound according to any of claims 1 to 3, or a composition according to claim 4, to a subject in need thereof.
9. A method according to claim 8, wherein the subject is a human subject.
10. A compound or composition for use according to claim 6, use according to claim 7, or a method according to claim 8 or 9, wherein the disease, disorder, or condition is selected from the group consisting of: a disorder of the sleep-wake cycle, a metabolic disorder, a neurological disorder, and other disorders, such as, feeding, drinking, arousal, stress, addiction, especially drug addiction, metabolism and reproduction disorders.
1 1. A compound or composition for use according to claim 6, use according to claim 7, or a method according to claim 8 or 9, wherein the disease, disorder, or condition is selected from the group consisting of: disorders of the sleep-wake cycle, sleep-wake transition disorder, insomnia, restless legs syndrome, jet-lag, disturbed sleep, sleep disorders secondary to neurological disorders, manias, depressions, manic depression, schizophrenia, pain syndromes, fibromyalgia, neuropathic pain, catatonia, Parkinson's disease, Tourette's syndrome, anxiety, stress disorder, panic disorder, delirium, dementias, Alzheimer's disease, Huntington's disease, overweight or obesity, conditions related to overweight or obesity, insulin resistance, type II diabetes, hyperlipidemia, gallstones, angina, hypertension, breathlessness, tachycardia, infertility, sleep apnoea, back and joint pain, varicose veins, osteoarthritis, hypertension, tachycardia, arrhythmias, angina pectoris, acute heart failure, ulcers, irritable bowel syndrome, diarrhoea, gastroesophageal reflux, addiction, and alcoholism.
12. A compound or composition for use according to claim 6, use according to claim 7, or a method according to claim 8 or 9, wherein the disease, disorder, or condition is a disorder of the sleep-wake cycle, sleep-wake transition disorder, or insomnia.
13. A method of producing a compound of Formula (I), which comprises reacting a compound of Formula (VII) with a cyanating agent, to produce the compound of Formula (I):
Figure imgf000072_0001
Figure imgf000072_0002
14. A method according to claim 13, wherein the central hydrogen atoms on the methylated octahydropyrrolo[3,4-c]pyrrole group in compound (VII) and compound (I) are in a cis position.
15. A method according to claim 13 or 14, which comprises reacting a compound of Formula (Vll)(a) with a cyanating agent, to produce a compound of Formula (l)(a):
Figure imgf000073_0001
(Vll)(a)
Figure imgf000073_0002
(l)(a)
16. A method according to any of claims 13 to 15, wherein the compound of Formula (VII) or (Vll)(a) undergoes a metal catalysed cyanation reaction, in the presence of a ligand.
17. A method according to claim 16, wherein the cyanating agent is potassium hexacyanoferrate(ll) trihydrate, the ligand is 1-butylimidazole, the catalyst is Cul and the solvent is toluene, and the reaction is carried out at an elevated temperature, preferably 150°C.
PCT/GB2018/051259 2017-05-10 2018-05-10 Orexin receptor antagonists WO2018206959A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1707504.5A GB201707504D0 (en) 2017-05-10 2017-05-10 Orexin receptor antagonists
GB1707504.5 2017-05-10

Publications (1)

Publication Number Publication Date
WO2018206959A1 true WO2018206959A1 (en) 2018-11-15

Family

ID=59065523

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2018/051259 WO2018206959A1 (en) 2017-05-10 2018-05-10 Orexin receptor antagonists

Country Status (2)

Country Link
GB (1) GB201707504D0 (en)
WO (1) WO2018206959A1 (en)

Citations (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003002561A1 (en) 2001-06-28 2003-01-09 Smithkline Beecham P.L.C. N-aroyl cyclic amine derivatives as orexin receptor antagonists
WO2003051872A1 (en) 2001-12-19 2003-06-26 Smithkline Beecham P.L.C. Ethylene diamine derivatives and their use as orexin-receptor antagonists
WO2004033418A2 (en) 2002-10-11 2004-04-22 Actelion Pharmaceuticals Ltd. Sulfonylamino-acetic derivatives and their use as orexin receptor antagonists
WO2004041791A1 (en) 2002-11-06 2004-05-21 Glaxo Group Limited N-aryl acetyl cyclic amine derivatives as orexin antagonists
WO2007126934A2 (en) 2006-03-29 2007-11-08 Merck & Co., Inc. Amidoethylthioether orexin receptor antagonists
WO2007126935A2 (en) 2006-03-29 2007-11-08 Merck & Co., Inc. Diazepan orexin receptor antagonists
WO2008008551A2 (en) 2006-07-14 2008-01-17 Merck & Co., Inc. 2-substituted proline bis-amide orexin receptor antagonists
WO2008008517A2 (en) 2006-07-14 2008-01-17 Merck & Co., Inc. Bridged diazepan orexin receptor antagonists
WO2008008518A1 (en) 2006-07-14 2008-01-17 Merck & Co., Inc. Substituted diazepan orexin receptor antagonists
US20080132490A1 (en) 2006-12-01 2008-06-05 Bergman Jeffrey M Substituted diazepan orexin receptor antagonists
WO2008143856A1 (en) 2007-05-18 2008-11-27 Merck & Co., Inc. Oxo bridged diazepan orexin receptor antagonists
WO2009022311A2 (en) 2007-08-15 2009-02-19 Actelion Pharmaceuticals Ltd 1,2-diamido-ethylene derivatives as orexin antagonists
WO2009058238A1 (en) 2007-10-29 2009-05-07 Merck & Co., Inc. Substituted diazepan orexin receptor antagonists
WO2009063485A2 (en) 2007-07-24 2009-05-22 Viridis Biopharma Pvt Ltd. Treatment of human disease conditions and disorders using vitamin k analogues and derivatives
WO2009124956A1 (en) 2008-04-10 2009-10-15 Glaxo Group Limited Pyridine derivatives used to treat orexin related disorders
WO2010017260A1 (en) 2008-08-07 2010-02-11 Merck & Co., Inc. Tripyridyl carboxamide orexin receptor antagonists
WO2010048014A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,4-disubstituted pyrrolidine orexin receptor antagonists
WO2010048017A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Disubstituted azepan orexin receptor antagonists
WO2010048010A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,5-disubstituted piperidine orexin receptor antagonists
WO2010048012A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,5-disubstituted piperidine orexin receptor antagonists
WO2010048013A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,5-disubstituted morpholine orexin receptor antagonists
WO2010051238A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Pyridazine carboxamide orexin receptor antagonists
WO2010051236A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Isonicotinamide orexin receptor antagonists
WO2010051237A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. 2,5-disubstituted phenyl carboxamide orexin receptor antagonists
WO2010060471A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
WO2010060470A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
WO2010060472A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Imidazopyridazine derivatives acting as orexin antagonists
WO2010063662A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0] hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
WO2010063663A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0]hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
US20100160344A1 (en) 2008-10-09 2010-06-24 Giuseppe Alvaro Novel compounds
US20100160345A1 (en) 2008-10-09 2010-06-24 Giuseppe Alvaro Novel compounds
WO2010072722A1 (en) 2008-12-23 2010-07-01 Glaxo Group Limited Piperidine derivatives useful as orexin antagonists
WO2011050198A1 (en) * 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Disubstituted octahy - dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
WO2012145581A1 (en) 2011-04-20 2012-10-26 Janssen Pharmaceutica Nv Disubstituted octahy-dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
WO2016100157A2 (en) * 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. 6,5-bicyclic octahydropyrrolopyridine orexin receptor antagonists

Patent Citations (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003002561A1 (en) 2001-06-28 2003-01-09 Smithkline Beecham P.L.C. N-aroyl cyclic amine derivatives as orexin receptor antagonists
WO2003051872A1 (en) 2001-12-19 2003-06-26 Smithkline Beecham P.L.C. Ethylene diamine derivatives and their use as orexin-receptor antagonists
WO2004033418A2 (en) 2002-10-11 2004-04-22 Actelion Pharmaceuticals Ltd. Sulfonylamino-acetic derivatives and their use as orexin receptor antagonists
WO2004041791A1 (en) 2002-11-06 2004-05-21 Glaxo Group Limited N-aryl acetyl cyclic amine derivatives as orexin antagonists
WO2007126934A2 (en) 2006-03-29 2007-11-08 Merck & Co., Inc. Amidoethylthioether orexin receptor antagonists
WO2007126935A2 (en) 2006-03-29 2007-11-08 Merck & Co., Inc. Diazepan orexin receptor antagonists
WO2008008518A1 (en) 2006-07-14 2008-01-17 Merck & Co., Inc. Substituted diazepan orexin receptor antagonists
WO2008008517A2 (en) 2006-07-14 2008-01-17 Merck & Co., Inc. Bridged diazepan orexin receptor antagonists
WO2008008551A2 (en) 2006-07-14 2008-01-17 Merck & Co., Inc. 2-substituted proline bis-amide orexin receptor antagonists
US20080132490A1 (en) 2006-12-01 2008-06-05 Bergman Jeffrey M Substituted diazepan orexin receptor antagonists
WO2008143856A1 (en) 2007-05-18 2008-11-27 Merck & Co., Inc. Oxo bridged diazepan orexin receptor antagonists
WO2009063485A2 (en) 2007-07-24 2009-05-22 Viridis Biopharma Pvt Ltd. Treatment of human disease conditions and disorders using vitamin k analogues and derivatives
WO2009022311A2 (en) 2007-08-15 2009-02-19 Actelion Pharmaceuticals Ltd 1,2-diamido-ethylene derivatives as orexin antagonists
WO2009058238A1 (en) 2007-10-29 2009-05-07 Merck & Co., Inc. Substituted diazepan orexin receptor antagonists
WO2009124956A1 (en) 2008-04-10 2009-10-15 Glaxo Group Limited Pyridine derivatives used to treat orexin related disorders
WO2010017260A1 (en) 2008-08-07 2010-02-11 Merck & Co., Inc. Tripyridyl carboxamide orexin receptor antagonists
US20100160344A1 (en) 2008-10-09 2010-06-24 Giuseppe Alvaro Novel compounds
US20100160345A1 (en) 2008-10-09 2010-06-24 Giuseppe Alvaro Novel compounds
WO2010048014A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,4-disubstituted pyrrolidine orexin receptor antagonists
WO2010048010A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,5-disubstituted piperidine orexin receptor antagonists
WO2010048013A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,5-disubstituted morpholine orexin receptor antagonists
WO2010048012A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,5-disubstituted piperidine orexin receptor antagonists
WO2010048017A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Disubstituted azepan orexin receptor antagonists
WO2010051238A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Pyridazine carboxamide orexin receptor antagonists
WO2010051236A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Isonicotinamide orexin receptor antagonists
WO2010051237A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. 2,5-disubstituted phenyl carboxamide orexin receptor antagonists
WO2010060471A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
WO2010060470A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
WO2010060472A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Imidazopyridazine derivatives acting as orexin antagonists
WO2010063662A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0] hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
WO2010063663A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0]hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
WO2010072722A1 (en) 2008-12-23 2010-07-01 Glaxo Group Limited Piperidine derivatives useful as orexin antagonists
WO2011050198A1 (en) * 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Disubstituted octahy - dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
WO2012145581A1 (en) 2011-04-20 2012-10-26 Janssen Pharmaceutica Nv Disubstituted octahy-dropyrrolo [3,4-c] pyrroles as orexin receptor modulators
WO2016100157A2 (en) * 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. 6,5-bicyclic octahydropyrrolopyridine orexin receptor antagonists

Non-Patent Citations (73)

* Cited by examiner, † Cited by third party
Title
"Design of Prodrugs", 1985, ELSEVIER
"GeneBank", Database accession no. NM_001525
"GeneBank", Database accession no. NM_001526.2
"Handbook of Pharmaceutical Salts, Properties, Selection, and Use", 2002, WILEY-VCH AND VHCA
ADAMANTIDIS; DE LECEA, TRENDS ENDOCRINOL METAB., vol. 19, 2008, pages 362 - 370
ASTON-JONES ET AL., NEUROPHARMACOLOGY, vol. 56, no. 1, 2008, pages 112 - 121
BAGSHAWE, DRUG DEV RES, vol. 34, 1995, pages 220 - 230
BERRIDGE ET AL., BRAIN RES, vol. 1314, 2010, pages 91 - 102
BERTOLINI ET AL., J MED CHEM, vol. 40, no. 201, 1997, pages 1 - 2016
BODOR, ADV DRUG RES, vol. 13, 1984, pages 224 - 331
BOSS, JOURNAL OF MEDICINAL CHEMISTRY, vol. 52, no. 4, 2009, pages 891 - 903
BULBUL ET AL., PEPTIDES, vol. 31, 2010, pages 2118 - 2122
BUNDGAARD: "Design of Prodrugs", 1985, ELSEVIER PRESS
BUSQUETS ET AL., RESPIRATION, vol. 71, 2004, pages 575 - 579
CARTER ET AL., CURRENT OPIN PHARMACOL, vol. 9, 2009, pages 39 - 45
CHOI, NEUROSCIENCE, vol. 167, 2010, pages 11 - 20
COLEMAN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 20, no. 14, 2010, pages 4201 - 4205
COLEMAN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 20, no. 7, 2010, pages 2311 - 2315
COX ET AL., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 19, no. 11, 2009, pages 2997 - 3001
COX, JOURNAL OF MEDICINAL CHEMISTRY, vol. 53, no. 14, 2010, pages 5320 - 5332
DUGOVIC ET AL.: "Front Neurosci.", vol. 8, 2014, pages: 28
E L ELIEL: "Stereochemistry of Organic Compounds", 1994, WILEY
EHRSTROM ET AL., J CLIN ENDOCRINOL METAB, vol. 90, 2005, pages 2370 - 2377
EHRSTROM ET AL., REGUL PEPT, vol. 132, 2005, pages 9 - 16
ELIASSI ET AL., J NEUROENDOCRINOL, vol. 21, 2009, pages 177 - 182
G.S. PAULEKUHN ET AL.: "Trends in Active Pharmaceutical Ingredient Salt Selection based on Analysis of the Orange Book Database", J. MED. CHEM., vol. 50, 2007, pages 6665 - 72, XP055536811, DOI: doi:10.1021/jm701032y
HARRIS ET AL., NATURE, vol. 437, 2005, pages 556 - 559
HIROTA ET AL., BRAIN RES, vol. 981, 2003, pages 143 - 150
IGARASHI ET AL., CHEST, vol. 124, 2003, pages 1381 - 1385
J. MARCH: "Advanced Organic Chemistry", 1985, WILEY INTERSCIENCE
JOELS, EPILEPSIA, vol. 50, 2009, pages 586 - 597
JOHNSON ET AL., NAT MED, vol. 16, 2010, pages 111 - 115
KANG ET AL., SCIENCE EXPRESS, 10 January 2009 (2009-01-10)
KAYABA ET AL., AM J PHYSIOL REGUL INTEGR COMP PHYSIOL, vol. 285, 2003, pages R581 - R593
KERMANI; ELIASSI, NEUROSCIENCE, vol. 226, 2012, pages 81 - 88
KIRCHGESSNER; LIU, NEURON, vol. 24, 1999, pages 941 - 951
KISANUKI, SLEEP, vol. 23, 2000, pages A91
LANGMEAD C.J.; JERMAN J.C.; BROUGH S.J.; SCOTT C.; PORTER R.A.; HERDON H.J.: "Characterisation of the binding of [3H]-SB-674042, a novel nonpeptide antagonist, to the human orexin-1 receptor", BR. J. PHARMACOL., vol. 141, 2004, pages 340 - 346, XP002359418, DOI: doi:10.1038/sj.bjp.0705610
LARSEN ET AL.: "Design and Application of Prodrugs, Drug Design and Development", 1991, HARWOOD ACADEMIC PUBLISHERS
LAWRENCE ET AL., BR J PHARMACOL, vol. 148, 2006, pages 752 - 759
LI, BRITISH JOURNAL OF PHARMACOLOGY, vol. 171, 2014, pages 332 - 350
LIN, CELL, vol. 98, 1999, pages 365 - 376
LUNGWITZ ET AL., PHYSIOL BEHAV, vol. 107, 2012, pages 726 - 732
MALHERBE P.; BORRONI E.; GOBBI L.; KNUST H.; NETTEKOVEN M.; PINARD E.; ROCHE O.; ROGERS-EVANS M.; WETTSTEIN J.G.; MOREAUET J-L: "Biochemical and behavioural characterization of EMPA, a novel high-affinity, selective antagonist for the OX2 receptor", BR. J. PHARMACOL., vol. 156, 2009, pages 1326 - 1341
MALHERBE P.; BORRONI E.; PINARD E.; WETTSTEIN J.G.; KNOFLACH F.: "Biochemical and Electrophysiological Characterization of Almorexant, a Dual Orexin Receptor (OX1)/Orexin 2 Receptor (OX2) Antagonist: Comparison with Selective OX1 and OX2 Antagonists", MOL. PHARMACOL., vol. 76, 2009, pages 618 - 631, XP008155137, DOI: doi:10.1124/mol.109.055152
MATSUMURA ET AL., HYPERTENSION, vol. 37, 2001, pages 1382 - 1387
MICHAEL A. LETAVIC ET AL: "Novel Octahydropyrrolo[3,4- c ]pyrroles Are Selective Orexin-2 Antagonists: SAR Leading to a Clinical Candidate", JOURNAL OF MEDICINAL CHEMISTRY, vol. 58, no. 14, 8 July 2015 (2015-07-08), pages 5620 - 5636, XP055493378, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.5b00742 *
NAKAMURA ET AL., BRAIN RESEARCH, vol. 873, no. 1, 2000, pages 181 - 7
NASLUND ET AL., AM J PHYSIOL GASTROINTEST LIVER PHYSIOL, vol. 282, 2002, pages G470 - G479
NISHINO ET AL., LANCET, vol. 355, 2000, pages 39 - 40
NOZU ET AL., NEUROSCI LETT, vol. 498, 2011, pages 143 - 146
PJ, KOCIENSKI: "Protecting Groups", 1994, GEORG THIEME VERLAG
PLAZA-ZABALA ET AL., J NEUROSCI, vol. 30, 2010, pages 2300 - 2310
RC LAROCK: "Comprehensive Organic Transformations", 1989, VCH PUBLISHERS INC.
RK MACKIE; DM SMITH: "Guidebook to Organic Synthesis", 1982, LONGMAN
S WARREN: "Designing Organic Synthesis", 1978, WILEY INTERSCIENCE
S WARREN: "Organic Synthesis - The Disconnection Approach", 1982, WILEY INTERSCIENCE
S.M. BERGE ET AL.: "Pharmaceutical Salts", J PHARM SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
SAKURAI T., CELL, vol. 92, 1998, pages 573 - 585
SAMSON ET AL., BRAIN RES, vol. 831, 1999, pages 248 - 253
SAMSON ET AL., SLEEP MED REV, vol. 9, 2005, pages 243 - 252
SATOH ET AL., NEUROPHARMACOLOGY, vol. 51, 2006, pages 466 - 473
SHAHID ET AL., BR J PHARMACOL, vol. 162, 2011, pages 961 - 973
SHAN ET AL., J PHARM SCI, vol. 86, no. 7, 1997, pages 765 - 767
SHARF ET AL., BRAIN RES, vol. 1314, 2010, pages 130 - 138
SHIRASAKA ET AL., AM J PHYSIOL, vol. 277, 1999, pages R1780 - R1785
SHIRASAKA ET AL., REGUL PEPT, vol. 104, 2002, pages 91 - 95
T. SAKURAI ET AL., CELL, vol. 92, no. 4, 1998, pages 573 - 585
T. SAKURAI, REG. PEPT., vol. 85, no. 1, 1999, pages 25 - 30
TAKAHASHI ET AL., BIOCHEM BIOPHYS RES COMMUN, vol. 254, 1999, pages 623 - 627
THEODORA W GREENE; PETER G M WUTS: "Greene's Protective Groups in Organic Synthesis", 2014, JOHN WILEY AND SONS
TW GREENE; PGM WUTS: "Protective Groups in Organic Synthesis", 2014, JOHN WILEY AND SONS, INC.
YAMADA ET AL., NEUROSCI LETT, vol. 376, 2005, pages 137 - 142

Also Published As

Publication number Publication date
GB201707504D0 (en) 2017-06-21

Similar Documents

Publication Publication Date Title
EP2491031B1 (en) Fused heterocyclic compounds as orexin receptor modulators
US9447117B2 (en) Substituted 2-azabicycles and their use as orexin receptor modulators
TW593280B (en) CRF antagonistic quino- and quinazolines
EP3191492B1 (en) P2x7 modulators
US10513523B2 (en) Imidazopyrazines and pyrazolopyrimidines and their use as AMPA receptor modulators
JP2009507801A (en) Pyridine derivatives and their use in the treatment of mental disorders
US8835633B2 (en) Process for the preparation of benzoimidazol-2-yl pyrimidine derivatives
JP2009510055A (en) Novel azetidine compounds useful in the treatment of functional gastrointestinal disorders, IBS and functional dyspepsia
TW201427957A (en) Acylaminocycloalkyl compounds suitable for treating disorders that respond to modulation of dopamine D3 receptor
WO2018227058A1 (en) Piperidinone formyl peptide 2 receptor agonists
EP3634409B1 (en) Orexin receptor antagonists
WO2022217232A1 (en) Chromans and benzofurans as 5-ht1a and taar1 agonists
CA2984305C (en) Indolone compounds and their use as ampa receptor modulators
WO2018206959A1 (en) Orexin receptor antagonists
WO2018146466A1 (en) Orexin receptor antagonists
JP2010540591A (en) Azabicyclo [3.1.0] hexyl derivatives as modulators of dopamine D3 receptors
AU2013204436B2 (en) Process for the preparation of benzoimidazol-2-yl pyrimidine derivatives
WO2022070068A1 (en) Dihydroorotate dehydrogenase inhibitors
CA3215043A1 (en) Taar1 and serotonin modulators, and pharmaceutical compositions, and methods of use thereof
EA007539B1 (en) Novel pyrazinone derivatives for the treatment of 5-htreceptor-related disorders
WO2009102937A1 (en) Processes for the preparation of carbon-linked tetrahydro-pyrazolo-pyridine modulators of cathepsin s

Legal Events

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

Ref document number: 18731159

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18731159

Country of ref document: EP

Kind code of ref document: A1