WO2018146466A1 - Antagonistes des récepteurs de l'orexine - Google Patents

Antagonistes des récepteurs de l'orexine Download PDF

Info

Publication number
WO2018146466A1
WO2018146466A1 PCT/GB2018/050340 GB2018050340W WO2018146466A1 WO 2018146466 A1 WO2018146466 A1 WO 2018146466A1 GB 2018050340 W GB2018050340 W GB 2018050340W WO 2018146466 A1 WO2018146466 A1 WO 2018146466A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
disorder
disease
sleep
Prior art date
Application number
PCT/GB2018/050340
Other languages
English (en)
Inventor
David Sheppard
Daniele Andreotti
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 WO2018146466A1 publication Critical patent/WO2018146466A1/fr

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

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.
  • DORAs function predominantly through OX2R antagonism.
  • OX2R knock-out mice exhibit a subtle, barely detectable sleep phenotype, whereas OX2R knock-out 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).
  • 0X1 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 0X1 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 0X2 R 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).
  • the orexin system also interacts with brain dopamine systems. Intracerebroventricular (i.c.v.) injections of orexins in mice increase locomotor activity, grooming and stereotypy.
  • 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 : 21 18-2122), and enteric peristalsis (Satoh et al., Neuropharmacology, 2006, 51 : 466-473), 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, diarrho
  • 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: 1 1-20). 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.c.v., intracisternally or intrathecally increases the mean arterial pressure, heart rate, renal sympathetic nerve activity and plasma catecholamine or vasopressin levels (Samson et al., Brain Res, 1999, 831 : 248-253; Samson et al., 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 etal., 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 al., 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 al., 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 0X1 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 al., 2010, supra). Orexin receptor antagonists may be useful for the treatment of stress, anxiety and panic disorders.
  • 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.
  • a pharmaceutical 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 (V) and a compound of formula (XVI), to produce the compound of Formula I):
  • 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 (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 (V)(a) and a compound of formula (XVI), to produce the compound of Formula (l)(a):
  • LG is a leaving group, such as a chloro, bromo, mesylate, or tosylate group.
  • the compound of Formula (V) or (V)(a) is reacted with the compound of Formula (XVI) in the presence of a base in a solvent.
  • a base is CS2CO3
  • the solvent is DMF
  • the reaction is carried out at an elevated temperature, preferably 100°C.
  • the compound of Formula (XVI) is 2-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):
  • PG2 is an amine-protecting group.
  • 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 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 (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):
  • the compound of Formula (III) or (lll)(a) is reacted with 2-fluoro-4- methoxybenzoic acid in the presence of a coupling reagent and an organic base, in an aprotic solvent, at a temperature from 0-100°C.
  • the coupling reagent is (1-[Bis(dimethylamino)methylene]-1 H- 1 ,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate) (HATU)
  • the organic base is DIPEA
  • the aprotic solvent is DMF
  • the temperature is preferably 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 compound 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.
  • 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 (XI):
  • 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 (XI) 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 (Xl)(a):
  • PGi is an amine-protecting group
  • PG2 is an amine- protecting group
  • the deprotection is acid-mediated deprotection in an aprotic solvent.
  • methods of the invention for producing a compound of Formula (I) further comprise producing a compound of formula (XI) from a compound of formula (X) by a cyclisation reaction:
  • PGi is an amine-protecting group
  • 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) are in a cis position.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise producing a compound of formula (Xl)(a) from a compound of formula (X)(a) by an acid-mediated cyclisation reaction:
  • PGi is an amine-protecting group
  • PG2 is an amine- protecting group
  • the acid-mediated cyclisation reaction is carried out in the presence of an acid in an aprotic solvent.
  • methods of the invention for producing a compound of Formula (l)(a) further comprise preparing a compound of Formula (X)(a) from a compound of Formula (Vl)(a), (Vll)(a), (Vlll)(a), and (IX)(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 (X)(a) from a compound of Formula (Xll)(a), (Xlll)(a), (XIV)(a), and (XV)(a) as illustrated by Scheme 3 below.
  • PGi is an amine protecting group.
  • PGi is a Cbz, MeOZ, ⁇ BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, fe/f-butyl carbamate, 9-Fluorenylmethyl carbamate, benzhydryl, or trityl group, in particular PGi is a carbamate group, more particularly fe/f-butyl carbamate.
  • PG 2 is a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, fe/f-butyl carbamate, 9-Fluorenylmethyl carbamate, benzhydryl, or trityl group.
  • PG2 is a benzyl group.
  • PGi is an amine-protecting group
  • PG2 is an amine- protecting group
  • PGi is a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, te/f-butyl carbamate, 9-Fluorenylmethyl carbamate, benzhydryl, or trityl group, in particular PGi is a carbamate group, more particularly te/f-butyl carbamate.
  • PG 2 is a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, te/f-butyl carbamate, 9-Fluorenylmethyl carbamate, benzhydryl, or trityl group.
  • PG 2 is a benzyl 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 (XI) 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 (XI) further comprises converting the compound of Formula (XI) to a compound of Formula
  • a method of the invention for producing a compound of Formula (Xl)(a) further comprises converting the compound of Formula (Xl)(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.
  • Particular examples of suitable amine protecting groups include the following:
  • fe/f-Butyloxycarbonyl ('BOC) group (common in solid phase peptide synthesis) - removed by concentrated strong acid (such as HCI or CFsCOOH), or by heating to >80 °C;
  • PMB p-Methoxybenzyl
  • DMPM 3,4-Dimethoxybenzyl
  • PMP p-methoxyphenyl
  • CAN ammonium cerium(IV) nitrate
  • PGi and PG2 may each independently be selected from: a Cbz, MeOZ, 'BOC, FMOC, Ac, Bz, Bn, carbamate, PMB, Ts, Troc, sulphonamide, acetamide, benzamide, fe/f-butylcarbamate, 9-Fluorenylmethyl carbamate, benzhydryl, trityl group.
  • PGi is a carbamate group, preferably f-butyl carbamate
  • 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., c/ ' s 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.
  • 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.
  • 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.
  • treat 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). 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).
  • 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).
  • 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 coadministered 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.
  • 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
  • the active agents of this invention may also be administered by non-oral routes.
  • the 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 ⁇ 9/ ⁇ 9/ ⁇ " ⁇ 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 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 substituents are as defined above with reference to the compound of Formula (I), or (l)(a), above.
  • 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.
  • 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), (X)(a) and (Xl)(a) as illustrated by Scheme 1.
  • 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, fe/f-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 (lll)(a) may be prepared from a compound of formula (ll)(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 under reflux.
  • a suitable reducing agent such as UAIH4
  • a suitable aprotic solvent such as 2-MeTHF
  • 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), in the presence of a suitable organic base, such as DIPEA, in an appropriate aprotic solvent, such as DMF, 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) (HATU)
  • HATU (1 -[Bis(dimethylamino)m
  • suitable deprotection conditions such as transfer hydrogenation, using for example ammonium formate and Pd(OH)2 on charcoal in a suitable solvent, such as EtOH, at a suitable temperature, such as between rt and reflux.
  • a compound of formula (l)(a) may be prepared from a compound of formula (V)(a) and a compound of formula (XVI) in the presence of a suitable base, such as CS2CO3, in a suitable solvent, such as DMF, at an elevated temperature, such as 100°C.
  • a suitable base such as CS2CO3
  • a suitable solvent such as DMF
  • a compound of Formula (X)(a) may be prepared from a compound of Formula (Vl)(a), (Vll)(a), (Vlll)(a), and (IX)(a), as illustrated by Scheme 2:
  • PGi is an amine protecting group, as hereinbefore defined.
  • a compound of formula (Vll)(a) may be prepared by condensation of Boc-L-alanine (Vl)(a) and Meldrum's acid in the presence of a suitable condensation reagent, such as EDC.HCI, in the presence of a suitable catalyst, such as DMAP, in a suitable solvent, such as EtOAc, 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
  • EtOAc a suitable solvent, such as EtOAc
  • a compound of formula (Vlll)(a) may be prepared from a compound of formula (Vll)(a) by treatment with heat in a suitable solvent, such as EtOAc, at an appropriate elevated temperature, such as reflux.
  • a compound of formula (IX)(a) may be prepared from a compound of formula (VI I l)(a) using an appropriate hydrogenolysis reaction in the presence of a suitable catalyst, such as PtC>2, in the presence of hydrogen gas at a suitable pressure, such as 5 bar, and in a suitable solvent, such as EtOAc.
  • a compound of formula (X)(a) may be prepared from a compound of formula (IX)(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 ⁇ , 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 ⁇ , 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 ⁇ , in a suitable solvent, such as
  • a compound of Formula (X)(a) may alternatively be prepared from a compound of Formula (Xll)(a), (Xlll)(a), (XIV)(a), and (XV)(a) as illustrated by Scheme 3:
  • PGi is an amine protecting group, as hereinbefore defined.
  • a compound of formula (Xlll)(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 ⁇ .
  • a compound of formula (XIV)(a) may be prepared by the oxidation of a compound of formula (XIII) 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 (XV)(a) may be prepared from a compound of formula (XIV)(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 suitable non-nucleophilic base such as LHMDS
  • a suitable aprotic solvent such as THF
  • a suitable electrophilic source of phenylselenide such as PhSeBr
  • a compound of formula (X)(a) may be prepared from a compound of formula (XV)(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
  • DCM a suitable solvent
  • a suitable low temperature such as -40°C.
  • a compound of Formula (l)(a) may be converted to its corresponding salt using methods known to those skilled in the art.
  • amines of Formula (l)(a) may be treated with trifluoroacetic acid (TFA), HCI, 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.
  • TFA trifluoroacetic acid
  • HCI trifluoroacetic acid
  • maleic acid maleic acid
  • citric acid in a solvent such as diethyl ether (Et20), CH2CI2, tetrahydrofuran (THF), or methanol (MeOH)
  • Et20 diethyl ether
  • CH2CI2 tetrahydrofuran
  • MeOH methanol
  • the acid is HCI and the solvent is isopropanol.
  • 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.
  • 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.
  • the eluant conditions are 1 M methanolic NH3.
  • EDC- HCI 48.63 g, 253.68 mmol was added portion wise over 20 mins to a solution of Boc- L-alanine (40 g, 21 1.4 mmol), Meldrum's acid (33.55 g, 232.54 mmol), and DMAP (38.91 g, 317.10 mmol) in DCM (813 ml_) at 0 °C. The mixture was stirred at rt for 2 hr then concentrated under reduced pressure. The residue was diluted with EtOAc (900 ml_) and washed with brine (800 ml_).
  • N-(Methoxymethyl)-N-(trimethylsilylmethyl) benzylamine (89.5 mL, 349.85 mmol) was added dropwise to an ice-cooled stirred solution of te/f-butyl (2S)-2-methyl-5-oxo-2,5- dihydro-1 H-pyrrole-1-carboxylate (Preparation 4, 57.5 g, 291.54 mmol) in DCM (1 150 mL) and TFA (6.03 mL, 78.72 mmol) and the mixture then stirred at rt for 3 hrs. The reaction mixture was partitioned between DCM and sat. NaHCOs Soln., the layers separated and the aqueous extracted with DCM (2x).
  • the crude product was purified by column chromatography using a silica gel cartridge eluting with cyclohexane: EtOAc (100:0 to 50:50) .
  • EtOAc cyclohexane
  • the product was dissolved in EtOAc and washed with water and brine (1 : 1 , 3x) then the organic phase dried (Na2S0 4 ) and evaporated under vacuum to afford the title compound, 2.86 g, 67.5 %.
  • 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]).
  • IC50 half maximal inhibitory concentration
  • 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.

Landscapes

  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des antagonistes des récepteurs de l'orexine, des compositions pharmaceutiques comprenant les antagonistes, des procédés de fabrication des antagonistes, leur utilisation pour la modulation du récepteur de l'orexine, et l'utilisation des antagonistes en tant que médicaments, en particulier pour le traitement de maladies, de troubles ou d'états médiés par l'activité du récepteur de l'orexine. Plus spécifiquement, l'invention concerne un composé de formule (I) comprenant des sels de celui-ci, des polymorphes et des isomères de celui-ci (y compris des isomères optiques, géométriques et tautomères) tels que définis dans la description et des composés isotopiquement marqués de formule (I).
PCT/GB2018/050340 2017-02-09 2018-02-07 Antagonistes des récepteurs de l'orexine WO2018146466A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1702174.2A GB201702174D0 (en) 2017-02-09 2017-02-09 Orexin receptor antagonists
GB1702174.2 2017-02-09

Publications (1)

Publication Number Publication Date
WO2018146466A1 true WO2018146466A1 (fr) 2018-08-16

Family

ID=58462153

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2018/050340 WO2018146466A1 (fr) 2017-02-09 2018-02-07 Antagonistes des récepteurs de l'orexine

Country Status (2)

Country Link
GB (1) GB201702174D0 (fr)
WO (1) WO2018146466A1 (fr)

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003002561A1 (fr) 2001-06-28 2003-01-09 Smithkline Beecham P.L.C. Derives d'amine cycliques n-aroyle utilises comme antagonistes du recepteur de l'orexine
WO2003051872A1 (fr) 2001-12-19 2003-06-26 Smithkline Beecham P.L.C. Derives d'ethylene diamine et utilisation en tant qu'antagonistes de recepteurs d'orexine
WO2004033418A2 (fr) 2002-10-11 2004-04-22 Actelion Pharmaceuticals Ltd. Derives d'acide sulfonylamino-acetique
WO2004041791A1 (fr) 2002-11-06 2004-05-21 Glaxo Group Limited Derives d'amine cyclique n-aryle acetyle utilises comme antagonistes de l'orexine
WO2007126934A2 (fr) 2006-03-29 2007-11-08 Merck & Co., Inc. Antagonistes des récepteurs de l'orexine sous forme de composés amidoéthylthioéther
WO2007126935A2 (fr) 2006-03-29 2007-11-08 Merck & Co., Inc. Antagonistes des récepteurs de l'orexine sous forme de composés diazépane
WO2008008551A2 (fr) 2006-07-14 2008-01-17 Merck & Co., Inc. Prolines bis-amides substituées en 2 antagonistes du récepteur de l'oréxine
WO2008008517A2 (fr) 2006-07-14 2008-01-17 Merck & Co., Inc. Diazépans pontés antagonistes du récepteur de l'oréxine
WO2008008518A1 (fr) 2006-07-14 2008-01-17 Merck & Co., Inc. Diazépans substitués antagonistes du récepteur de l'oréxine
US20080132490A1 (en) 2006-12-01 2008-06-05 Bergman Jeffrey M Substituted diazepan orexin receptor antagonists
WO2008143856A1 (fr) 2007-05-18 2008-11-27 Merck & Co., Inc. Antagonistes des récepteurs de l'oréxine à diazépam à pont oxo
WO2009022311A2 (fr) 2007-08-15 2009-02-19 Actelion Pharmaceuticals Ltd Dérivés du 1,2-diamido-éthylène
WO2009058238A1 (fr) 2007-10-29 2009-05-07 Merck & Co., Inc. Antagonistes des récepteurs de l'orexine, de type diazépan substitué
WO2009063485A2 (fr) 2007-07-24 2009-05-22 Viridis Biopharma Pvt Ltd. Traitement de conditions et troubles de maladie humaine mettant en œuvre des analogues de la vitamine k et leurs dérivés
WO2009124956A1 (fr) 2008-04-10 2009-10-15 Glaxo Group Limited Dérivés de pyridine utilisés pour traiter des troubles liés aux orexines
WO2010017260A1 (fr) 2008-08-07 2010-02-11 Merck & Co., Inc. Antagonistes de récepteur de tripyridyle carboxamide d'orexine
WO2010048010A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Pipéridines 2,5-disubstituées comme antagonistes du récepteur de l’orexine
WO2010048014A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes de récepteur de l'orexine de type pyrrolidine 2,4-disubstituée
WO2010048012A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes de récepteur d’orexine de pipéridine 2,5-disubstituée
WO2010048013A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes de récepteur d’orexine de morpholine 2,5-disubstituée
WO2010048017A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes des récepteurs de l'orexine de type azépane disubstitué
WO2010051238A1 (fr) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Antagonistes des récepteurs de l’orexine à base de pyridazine-carboxamide
WO2010051236A1 (fr) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Antagonistes d'isonicotinamide des récepteurs de l'orexine
WO2010051237A1 (fr) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Antagonistes carboxamide phényl 2,5-disubstitué du récepteur de l’orexine
WO2010060471A1 (fr) 2008-11-26 2010-06-03 Glaxo Group Limited Dérivés de la pipéridine utiles en tant qu’antagonistes du récepteur de l’orexine
WO2010060472A1 (fr) 2008-11-26 2010-06-03 Glaxo Group Limited Dérivés de l’imidazopyridazine agissant en tant qu’antagonistes de l’orexine
WO2010060470A1 (fr) 2008-11-26 2010-06-03 Glaxo Group Limited Dérivés de la pipéridine utiles en tant qu’antagonistes du récepteur de l’orexine
WO2010063663A1 (fr) 2008-12-02 2010-06-10 Glaxo Group Limited Dérivés de n-{[(1r,4s,6r)-3-(2-pyridinylcarbonyl)-3-azabicyclo[4.1.0]hept-4-yl]methyl}-2-heteroarylamine et leurs utilisations
WO2010063662A1 (fr) 2008-12-02 2010-06-10 Glaxo Group Limited Dérivés de n-{[(1s,4s,6s)-3-(2-pyridinylcarbonyl)-3-azabicyclo[4.1.0]hept-4-yl]methyl}-2-heteroarylamine et leurs utilisations
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 (fr) 2008-12-23 2010-07-01 Glaxo Group Limited Dérivés de pipéridine pouvant être utilisés en tant qu'agonistes de l'orexine
WO2011050198A1 (fr) * 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Octahydropyrrolo[3,4-c]pyrroles disubstitués en tant que modulateurs de récepteur d'orexine
WO2012145581A1 (fr) 2011-04-20 2012-10-26 Janssen Pharmaceutica Nv Octahydropyrrolo [3,4-c] pyrroles disubstitués utilisés comme modulateurs du récepteur de l'orexine

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003002561A1 (fr) 2001-06-28 2003-01-09 Smithkline Beecham P.L.C. Derives d'amine cycliques n-aroyle utilises comme antagonistes du recepteur de l'orexine
WO2003051872A1 (fr) 2001-12-19 2003-06-26 Smithkline Beecham P.L.C. Derives d'ethylene diamine et utilisation en tant qu'antagonistes de recepteurs d'orexine
WO2004033418A2 (fr) 2002-10-11 2004-04-22 Actelion Pharmaceuticals Ltd. Derives d'acide sulfonylamino-acetique
WO2004041791A1 (fr) 2002-11-06 2004-05-21 Glaxo Group Limited Derives d'amine cyclique n-aryle acetyle utilises comme antagonistes de l'orexine
WO2007126934A2 (fr) 2006-03-29 2007-11-08 Merck & Co., Inc. Antagonistes des récepteurs de l'orexine sous forme de composés amidoéthylthioéther
WO2007126935A2 (fr) 2006-03-29 2007-11-08 Merck & Co., Inc. Antagonistes des récepteurs de l'orexine sous forme de composés diazépane
WO2008008518A1 (fr) 2006-07-14 2008-01-17 Merck & Co., Inc. Diazépans substitués antagonistes du récepteur de l'oréxine
WO2008008517A2 (fr) 2006-07-14 2008-01-17 Merck & Co., Inc. Diazépans pontés antagonistes du récepteur de l'oréxine
WO2008008551A2 (fr) 2006-07-14 2008-01-17 Merck & Co., Inc. Prolines bis-amides substituées en 2 antagonistes du récepteur de l'oréxine
US20080132490A1 (en) 2006-12-01 2008-06-05 Bergman Jeffrey M Substituted diazepan orexin receptor antagonists
WO2008143856A1 (fr) 2007-05-18 2008-11-27 Merck & Co., Inc. Antagonistes des récepteurs de l'oréxine à diazépam à pont oxo
WO2009063485A2 (fr) 2007-07-24 2009-05-22 Viridis Biopharma Pvt Ltd. Traitement de conditions et troubles de maladie humaine mettant en œuvre des analogues de la vitamine k et leurs dérivés
WO2009022311A2 (fr) 2007-08-15 2009-02-19 Actelion Pharmaceuticals Ltd Dérivés du 1,2-diamido-éthylène
WO2009058238A1 (fr) 2007-10-29 2009-05-07 Merck & Co., Inc. Antagonistes des récepteurs de l'orexine, de type diazépan substitué
WO2009124956A1 (fr) 2008-04-10 2009-10-15 Glaxo Group Limited Dérivés de pyridine utilisés pour traiter des troubles liés aux orexines
WO2010017260A1 (fr) 2008-08-07 2010-02-11 Merck & Co., Inc. Antagonistes de récepteur de tripyridyle carboxamide d'orexine
US20100160344A1 (en) 2008-10-09 2010-06-24 Giuseppe Alvaro Novel compounds
US20100160345A1 (en) 2008-10-09 2010-06-24 Giuseppe Alvaro Novel compounds
WO2010048010A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Pipéridines 2,5-disubstituées comme antagonistes du récepteur de l’orexine
WO2010048012A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes de récepteur d’orexine de pipéridine 2,5-disubstituée
WO2010048017A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes des récepteurs de l'orexine de type azépane disubstitué
WO2010048014A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes de récepteur de l'orexine de type pyrrolidine 2,4-disubstituée
WO2010048013A1 (fr) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. Antagonistes de récepteur d’orexine de morpholine 2,5-disubstituée
WO2010051238A1 (fr) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Antagonistes des récepteurs de l’orexine à base de pyridazine-carboxamide
WO2010051236A1 (fr) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Antagonistes d'isonicotinamide des récepteurs de l'orexine
WO2010051237A1 (fr) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Antagonistes carboxamide phényl 2,5-disubstitué du récepteur de l’orexine
WO2010060472A1 (fr) 2008-11-26 2010-06-03 Glaxo Group Limited Dérivés de l’imidazopyridazine agissant en tant qu’antagonistes de l’orexine
WO2010060470A1 (fr) 2008-11-26 2010-06-03 Glaxo Group Limited Dérivés de la pipéridine utiles en tant qu’antagonistes du récepteur de l’orexine
WO2010060471A1 (fr) 2008-11-26 2010-06-03 Glaxo Group Limited Dérivés de la pipéridine utiles en tant qu’antagonistes du récepteur de l’orexine
WO2010063663A1 (fr) 2008-12-02 2010-06-10 Glaxo Group Limited Dérivés de n-{[(1r,4s,6r)-3-(2-pyridinylcarbonyl)-3-azabicyclo[4.1.0]hept-4-yl]methyl}-2-heteroarylamine et leurs utilisations
WO2010063662A1 (fr) 2008-12-02 2010-06-10 Glaxo Group Limited Dérivés de n-{[(1s,4s,6s)-3-(2-pyridinylcarbonyl)-3-azabicyclo[4.1.0]hept-4-yl]methyl}-2-heteroarylamine et leurs utilisations
WO2010072722A1 (fr) 2008-12-23 2010-07-01 Glaxo Group Limited Dérivés de pipéridine pouvant être utilisés en tant qu'agonistes de l'orexine
WO2011050198A1 (fr) * 2009-10-23 2011-04-28 Janssen Pharmaceutica Nv Octahydropyrrolo[3,4-c]pyrroles disubstitués en tant que modulateurs de récepteur d'orexine
WO2012145581A1 (fr) 2011-04-20 2012-10-26 Janssen Pharmaceutica Nv Octahydropyrrolo [3,4-c] pyrroles disubstitués utilisés comme modulateurs du récepteur de l'orexine

Non-Patent Citations (69)

* Cited by examiner, † Cited by third party
Title
"Design of Prodrugs", 1985, ELSEVIER
"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 ET AL., 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 ET AL., 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
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, 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, 2009, pages 1 - 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 ET AL., 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 ET AL., BRITISH JOURNAL OF PHARMACOLOGY, vol. 171, 2014, pages 332 - 350
LIN ET AL., 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, HYPERTENSION, vol. 37, 2001, pages 1382 - 1387
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: "Pharmaceutical Salts", J PHARM SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
SAKURAI T. ET AL., 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., BRJ 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
GB201702174D0 (en) 2017-03-29

Similar Documents

Publication Publication Date Title
EP2491031B1 (fr) Composés hétérocycliques condensés en tant que modulateurs de récepteur d'orexine
US9447117B2 (en) Substituted 2-azabicycles and their use as orexin receptor modulators
TW593280B (en) CRF antagonistic quino- and quinazolines
US10150766B2 (en) P2X7 modulators
US10513523B2 (en) Imidazopyrazines and pyrazolopyrimidines and their use as AMPA receptor modulators
US8835633B2 (en) Process for the preparation of benzoimidazol-2-yl pyrimidine derivatives
TW201427957A (zh) 適用於治療對多巴胺d3受體調節有反應的病症之醯胺基環烷基化合物
WO2018227058A1 (fr) Agonistes du récepteur 2 du peptide formylé de pipéridinone
US8575364B2 (en) Modulators of serotonin receptor
EP3634409B1 (fr) Antagonistes des récepteurs de l'orexine
WO2022217232A1 (fr) Chromanes et benzofuranes utilisés en tant qu'agonistes de 5-ht1a et de taar1
WO2018146466A1 (fr) Antagonistes des récepteurs de l'orexine
JP2021500416A (ja) 疼痛及び疼痛関連状態を治療するための新規アルコキシアミノ誘導体
CA2984305C (fr) Composes indolones et utilisation de ces derniers en tant que modulateurs des recepteurs ampa
WO2018206959A1 (fr) Antagonistes des récepteurs de l'orexine
EP2470022A1 (fr) Composés et procédés
CN113166147A (zh) 用于治疗疼痛和疼痛相关病症的新的四氢嘧啶二氮杂卓和四氢吡啶二氮杂卓化合物
CA3215043A1 (fr) Modulateurs de taar1 et de serotonine, et compositions pharmaceutiques et leurs procedes d'utilisation
AU2013204436B2 (en) Process for the preparation of benzoimidazol-2-yl pyrimidine derivatives
WO2022070068A1 (fr) Inhibiteurs de la dihydroorotate déshydrogénase
WO2009102937A1 (fr) Procédés pour la préparation de modulateurs constitués de cathepsine s de tétrahydro-pyrazolo-pyridine à liaison carbone

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: 18711617

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: 18711617

Country of ref document: EP

Kind code of ref document: A1