WO2024115797A2 - Dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and use thereof as orexin receptors agonists for treating or preventing neurological diseases - Google Patents
Dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and use thereof as orexin receptors agonists for treating or preventing neurological diseases Download PDFInfo
- Publication number
- WO2024115797A2 WO2024115797A2 PCT/EP2023/088019 EP2023088019W WO2024115797A2 WO 2024115797 A2 WO2024115797 A2 WO 2024115797A2 EP 2023088019 W EP2023088019 W EP 2023088019W WO 2024115797 A2 WO2024115797 A2 WO 2024115797A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- hydrogen atom
- compound
- orexin
- formula
- alkyl
- Prior art date
Links
- 208000012902 Nervous system disease Diseases 0.000 title claims abstract description 31
- 208000025966 Neurological disease Diseases 0.000 title claims abstract description 30
- 108050000742 Orexin Receptor Proteins 0.000 title claims abstract description 26
- 102000008834 Orexin receptor Human genes 0.000 title claims abstract description 26
- IDUSJBBWEKNWAK-UHFFFAOYSA-N 3,4-dihydro-2h-1,2-benzothiazine Chemical compound C1=CC=C2SNCCC2=C1 IDUSJBBWEKNWAK-UHFFFAOYSA-N 0.000 title abstract description 10
- BGDOLELXXPTPFX-UHFFFAOYSA-N 3,4-dihydro-2h-1,2-benzoxazine Chemical class C1=CC=C2ONCCC2=C1 BGDOLELXXPTPFX-UHFFFAOYSA-N 0.000 title abstract description 8
- NTURVSFTOYPGON-UHFFFAOYSA-N Dihydroquinazoline Chemical compound C1=CC=C2C=NCNC2=C1 NTURVSFTOYPGON-UHFFFAOYSA-N 0.000 title abstract description 8
- 239000000556 agonist Substances 0.000 title description 27
- 150000001875 compounds Chemical class 0.000 claims abstract description 138
- 102000002512 Orexin Human genes 0.000 claims abstract description 76
- 108060005714 orexin Proteins 0.000 claims abstract description 76
- 208000019116 sleep disease Diseases 0.000 claims abstract description 41
- 239000003814 drug Substances 0.000 claims abstract description 30
- 238000011282 treatment Methods 0.000 claims abstract description 28
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 22
- 230000005062 synaptic transmission Effects 0.000 claims abstract description 19
- 230000001010 compromised effect Effects 0.000 claims abstract description 18
- 230000002093 peripheral effect Effects 0.000 claims abstract description 17
- 230000002265 prevention Effects 0.000 claims abstract description 16
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 121
- 239000000126 substance Substances 0.000 claims description 58
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 57
- 125000003118 aryl group Chemical group 0.000 claims description 53
- 208000007590 Disorders of Excessive Somnolence Diseases 0.000 claims description 51
- 208000018737 Parkinson disease Diseases 0.000 claims description 50
- 125000005843 halogen group Chemical group 0.000 claims description 48
- 230000007958 sleep Effects 0.000 claims description 43
- 125000001072 heteroaryl group Chemical group 0.000 claims description 42
- 125000000623 heterocyclic group Chemical group 0.000 claims description 41
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 claims description 38
- 239000000203 mixture Substances 0.000 claims description 36
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 35
- 125000000923 (C1-C30) alkyl group Chemical group 0.000 claims description 32
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 claims description 30
- 208000035959 Narcolepsy type 1 Diseases 0.000 claims description 29
- 208000004461 narcolepsy 1 Diseases 0.000 claims description 29
- 208000033409 Narcolepsy type 2 Diseases 0.000 claims description 28
- 201000010099 disease Diseases 0.000 claims description 28
- 208000035475 disorder Diseases 0.000 claims description 27
- 206010020765 hypersomnia Diseases 0.000 claims description 27
- 208000020016 psychiatric disease Diseases 0.000 claims description 26
- 125000003837 (C1-C20) alkyl group Chemical group 0.000 claims description 25
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 24
- 229940079593 drug Drugs 0.000 claims description 24
- 150000003839 salts Chemical class 0.000 claims description 22
- 125000002252 acyl group Chemical group 0.000 claims description 13
- 208000019901 Anxiety disease Diseases 0.000 claims description 12
- 208000010877 cognitive disease Diseases 0.000 claims description 12
- 208000016588 Idiopathic hypersomnia Diseases 0.000 claims description 11
- 208000032859 Synucleinopathies Diseases 0.000 claims description 11
- 208000015122 neurodegenerative disease Diseases 0.000 claims description 11
- 201000005439 recurrent hypersomnia Diseases 0.000 claims description 11
- 208000019022 Mood disease Diseases 0.000 claims description 10
- 208000028698 Cognitive impairment Diseases 0.000 claims description 9
- 206010019280 Heart failures Diseases 0.000 claims description 9
- 208000011580 syndromic disease Diseases 0.000 claims description 9
- 208000024827 Alzheimer disease Diseases 0.000 claims description 8
- 206010008874 Chronic Fatigue Syndrome Diseases 0.000 claims description 8
- 208000035895 Guillain-Barré syndrome Diseases 0.000 claims description 8
- 206010053712 Hypersomnia-bulimia syndrome Diseases 0.000 claims description 8
- 201000008178 Kleine-Levin syndrome Diseases 0.000 claims description 8
- 206010049567 Miller Fisher syndrome Diseases 0.000 claims description 8
- 208000005793 Restless legs syndrome Diseases 0.000 claims description 8
- 208000012396 long COVID-19 Diseases 0.000 claims description 8
- 208000029766 myalgic encephalomeyelitis/chronic fatigue syndrome Diseases 0.000 claims description 8
- 201000002859 sleep apnea Diseases 0.000 claims description 8
- 208000001640 Fibromyalgia Diseases 0.000 claims description 7
- 230000002060 circadian Effects 0.000 claims description 7
- 239000003937 drug carrier Substances 0.000 claims description 7
- 230000036541 health Effects 0.000 claims description 7
- 206010065687 Bone loss Diseases 0.000 claims description 6
- 208000000112 Myalgia Diseases 0.000 claims description 6
- 206010040047 Sepsis Diseases 0.000 claims description 6
- 230000036506 anxiety Effects 0.000 claims description 6
- 230000003292 diminished effect Effects 0.000 claims description 6
- 230000003340 mental effect Effects 0.000 claims description 6
- 208000013465 muscle pain Diseases 0.000 claims description 6
- 208000024335 physical disease Diseases 0.000 claims description 6
- 230000000241 respiratory effect Effects 0.000 claims description 6
- 208000006096 Attention Deficit Disorder with Hyperactivity Diseases 0.000 claims description 5
- 208000036864 Attention deficit/hyperactivity disease Diseases 0.000 claims description 5
- 208000015802 attention deficit-hyperactivity disease Diseases 0.000 claims description 5
- 208000034799 Tauopathies Diseases 0.000 claims description 4
- 239000002775 capsule Substances 0.000 claims description 3
- 229940102223 injectable solution Drugs 0.000 claims description 3
- 238000007911 parenteral administration Methods 0.000 claims description 3
- 230000037317 transdermal delivery Effects 0.000 claims description 3
- 230000000926 neurological effect Effects 0.000 abstract description 14
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 166
- -1 cycloalkyl urea compounds Chemical class 0.000 description 66
- 239000000047 product Substances 0.000 description 55
- 238000005160 1H NMR spectroscopy Methods 0.000 description 47
- 238000001644 13C nuclear magnetic resonance spectroscopy Methods 0.000 description 35
- 229910052739 hydrogen Inorganic materials 0.000 description 23
- 201000003631 narcolepsy Diseases 0.000 description 23
- 208000002193 Pain Diseases 0.000 description 20
- 230000000694 effects Effects 0.000 description 19
- 238000000034 method Methods 0.000 description 19
- OFNHNCAUVYOTPM-IIIOAANCSA-N orexin-a Chemical compound C([C@@H](C(=O)N[C@@H](C)C(=O)N[C@@H](C)C(=O)NCC(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(C)C)C(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)CNC(=O)[C@H](C)NC(=O)CNC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H]1NC(=O)[C@H](CO)NC(=O)[C@@H]2CSSC[C@@H](C(=O)N[C@H](C(N[C@@H](CCCN=C(N)N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@H](C(=O)N2)[C@@H](C)O)=O)CSSC1)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC(C)C)NC(=O)[C@H]1N(CCC1)C(=O)[C@H]1NC(=O)CC1)C1=CNC=N1 OFNHNCAUVYOTPM-IIIOAANCSA-N 0.000 description 17
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 15
- 239000004480 active ingredient Substances 0.000 description 15
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 15
- 230000036407 pain Effects 0.000 description 15
- 210000002569 neuron Anatomy 0.000 description 14
- 230000000446 orexinergic effect Effects 0.000 description 14
- 230000001965 increasing effect Effects 0.000 description 13
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 12
- 230000001276 controlling effect Effects 0.000 description 11
- 239000000243 solution Substances 0.000 description 11
- 206010041349 Somnolence Diseases 0.000 description 10
- 102000005962 receptors Human genes 0.000 description 10
- 108020003175 receptors Proteins 0.000 description 10
- 208000024891 symptom Diseases 0.000 description 10
- 208000028017 Psychotic disease Diseases 0.000 description 9
- 230000027455 binding Effects 0.000 description 9
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 9
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 8
- 208000014094 Dystonic disease Diseases 0.000 description 8
- 239000005557 antagonist Substances 0.000 description 8
- 239000004615 ingredient Substances 0.000 description 8
- 238000012360 testing method Methods 0.000 description 8
- 230000001225 therapeutic effect Effects 0.000 description 8
- 102000004175 Cathepsin H Human genes 0.000 description 7
- 108090000619 Cathepsin H Proteins 0.000 description 7
- 102000006441 Dopamine Plasma Membrane Transport Proteins Human genes 0.000 description 7
- 108010044266 Dopamine Plasma Membrane Transport Proteins Proteins 0.000 description 7
- 102000008092 Norepinephrine Plasma Membrane Transport Proteins Human genes 0.000 description 7
- 108010049586 Norepinephrine Plasma Membrane Transport Proteins Proteins 0.000 description 7
- 208000025535 REM sleep behavior disease Diseases 0.000 description 7
- 230000004913 activation Effects 0.000 description 7
- 230000015572 biosynthetic process Effects 0.000 description 7
- 210000004027 cell Anatomy 0.000 description 7
- 230000003247 decreasing effect Effects 0.000 description 7
- 230000004064 dysfunction Effects 0.000 description 7
- 208000010118 dystonia Diseases 0.000 description 7
- 238000010304 firing Methods 0.000 description 7
- 239000003446 ligand Substances 0.000 description 7
- 206010012289 Dementia Diseases 0.000 description 6
- 241000699670 Mus sp. Species 0.000 description 6
- 208000036110 Neuroinflammatory disease Diseases 0.000 description 6
- KWYUFKZDYYNOTN-UHFFFAOYSA-M Potassium hydroxide Chemical compound [OH-].[K+] KWYUFKZDYYNOTN-UHFFFAOYSA-M 0.000 description 6
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 6
- 230000037007 arousal Effects 0.000 description 6
- 125000004432 carbon atom Chemical group C* 0.000 description 6
- 230000000875 corresponding effect Effects 0.000 description 6
- 210000005064 dopaminergic neuron Anatomy 0.000 description 6
- 206010015037 epilepsy Diseases 0.000 description 6
- 208000004296 neuralgia Diseases 0.000 description 6
- 230000003959 neuroinflammation Effects 0.000 description 6
- 238000002360 preparation method Methods 0.000 description 6
- 239000002287 radioligand Substances 0.000 description 6
- 230000036385 rapid eye movement (rem) sleep Effects 0.000 description 6
- 208000011117 substance-related disease Diseases 0.000 description 6
- 238000003786 synthesis reaction Methods 0.000 description 6
- 208000001573 Cataplexy Diseases 0.000 description 5
- 206010008748 Chorea Diseases 0.000 description 5
- 206010010904 Convulsion Diseases 0.000 description 5
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 5
- 208000023105 Huntington disease Diseases 0.000 description 5
- 208000028482 Hypothalamic disease Diseases 0.000 description 5
- 208000019430 Motor disease Diseases 0.000 description 5
- 208000002033 Myoclonus Diseases 0.000 description 5
- 102100028656 Sigma non-opioid intracellular receptor 1 Human genes 0.000 description 5
- 206010044565 Tremor Diseases 0.000 description 5
- 238000010171 animal model Methods 0.000 description 5
- 125000004429 atom Chemical group 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 230000002708 enhancing effect Effects 0.000 description 5
- 230000002267 hypothalamic effect Effects 0.000 description 5
- 230000005764 inhibitory process Effects 0.000 description 5
- 230000015654 memory Effects 0.000 description 5
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 description 5
- 230000004770 neurodegeneration Effects 0.000 description 5
- 208000021722 neuropathic pain Diseases 0.000 description 5
- 230000000324 neuroprotective effect Effects 0.000 description 5
- 201000000980 schizophrenia Diseases 0.000 description 5
- 208000022925 sleep disturbance Diseases 0.000 description 5
- 208000012661 Dyskinesia Diseases 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 4
- 208000019695 Migraine disease Diseases 0.000 description 4
- 208000008589 Obesity Diseases 0.000 description 4
- 206010034010 Parkinsonism Diseases 0.000 description 4
- 101710104750 Sigma non-opioid intracellular receptor 1 Proteins 0.000 description 4
- 208000013738 Sleep Initiation and Maintenance disease Diseases 0.000 description 4
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 4
- 230000002159 abnormal effect Effects 0.000 description 4
- 230000001154 acute effect Effects 0.000 description 4
- 125000000217 alkyl group Chemical group 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 4
- 208000012601 choreatic disease Diseases 0.000 description 4
- ZPUCINDJVBIVPJ-LJISPDSOSA-N cocaine Chemical compound O([C@H]1C[C@@H]2CC[C@@H](N2C)[C@H]1C(=O)OC)C(=O)C1=CC=CC=C1 ZPUCINDJVBIVPJ-LJISPDSOSA-N 0.000 description 4
- 230000007423 decrease Effects 0.000 description 4
- 230000006735 deficit Effects 0.000 description 4
- 238000013461 design Methods 0.000 description 4
- VYFYYTLLBUKUHU-UHFFFAOYSA-N dopamine Chemical compound NCCC1=CC=C(O)C(O)=C1 VYFYYTLLBUKUHU-UHFFFAOYSA-N 0.000 description 4
- 206010016256 fatigue Diseases 0.000 description 4
- 230000004634 feeding behavior Effects 0.000 description 4
- 125000005842 heteroatom Chemical group 0.000 description 4
- 239000003112 inhibitor Substances 0.000 description 4
- 230000002401 inhibitory effect Effects 0.000 description 4
- 206010027599 migraine Diseases 0.000 description 4
- 229910052757 nitrogen Inorganic materials 0.000 description 4
- 235000020824 obesity Nutrition 0.000 description 4
- OHOWSYIKESXDMN-WMQZXSDYSA-N orexin-b Chemical compound C([C@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@@H](N)CCSC)[C@@H](C)O)[C@@H](C)CC)C(=O)N[C@@H](CC(N)=O)C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)NCC(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)NCC(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H](CCCN=C(N)N)C(N)=O)C1=CNC=N1 OHOWSYIKESXDMN-WMQZXSDYSA-N 0.000 description 4
- 229910052760 oxygen Inorganic materials 0.000 description 4
- 239000002244 precipitate Substances 0.000 description 4
- 102200036626 rs104893877 Human genes 0.000 description 4
- 210000003523 substantia nigra Anatomy 0.000 description 4
- 229910052717 sulfur Inorganic materials 0.000 description 4
- 239000011593 sulfur Substances 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- PLRACCBDVIHHLZ-UHFFFAOYSA-N 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine Chemical compound C1N(C)CCC(C=2C=CC=CC=2)=C1 PLRACCBDVIHHLZ-UHFFFAOYSA-N 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 208000031091 Amnestic disease Diseases 0.000 description 3
- 206010002653 Anosmia Diseases 0.000 description 3
- 208000020925 Bipolar disease Diseases 0.000 description 3
- 206010007559 Cardiac failure congestive Diseases 0.000 description 3
- 208000027534 Emotional disease Diseases 0.000 description 3
- 201000011240 Frontotemporal dementia Diseases 0.000 description 3
- 208000004454 Hyperalgesia Diseases 0.000 description 3
- 206010020772 Hypertension Diseases 0.000 description 3
- 206010062767 Hypophysitis Diseases 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 101001135571 Mus musculus Tyrosine-protein phosphatase non-receptor type 2 Proteins 0.000 description 3
- 206010028980 Neoplasm Diseases 0.000 description 3
- 208000027089 Parkinsonian disease Diseases 0.000 description 3
- 206010062519 Poor quality sleep Diseases 0.000 description 3
- 208000006011 Stroke Diseases 0.000 description 3
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 3
- 206010047700 Vomiting Diseases 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 150000001299 aldehydes Chemical class 0.000 description 3
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 3
- 210000004556 brain Anatomy 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 239000000460 chlorine Substances 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 238000005859 coupling reaction Methods 0.000 description 3
- 235000014632 disordered eating Nutrition 0.000 description 3
- 229960003638 dopamine Drugs 0.000 description 3
- 208000024732 dysthymic disease Diseases 0.000 description 3
- 230000002996 emotional effect Effects 0.000 description 3
- 238000002474 experimental method Methods 0.000 description 3
- 239000000835 fiber Substances 0.000 description 3
- 238000009472 formulation Methods 0.000 description 3
- 229930195712 glutamate Natural products 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 206010022437 insomnia Diseases 0.000 description 3
- 230000014759 maintenance of location Effects 0.000 description 3
- 208000024714 major depressive disease Diseases 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 239000003176 neuroleptic agent Substances 0.000 description 3
- 230000000701 neuroleptic effect Effects 0.000 description 3
- DIVDFFZHCJEHGG-UHFFFAOYSA-N oxidopamine Chemical compound NCCC1=CC(O)=C(O)C=C1O DIVDFFZHCJEHGG-UHFFFAOYSA-N 0.000 description 3
- 239000001301 oxygen Substances 0.000 description 3
- 208000019906 panic disease Diseases 0.000 description 3
- 230000008506 pathogenesis Effects 0.000 description 3
- 230000001575 pathological effect Effects 0.000 description 3
- 230000035790 physiological processes and functions Effects 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000033764 rhythmic process Effects 0.000 description 3
- 229920006395 saturated elastomer Polymers 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 201000009032 substance abuse Diseases 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- 230000002618 waking effect Effects 0.000 description 3
- SNICXCGAKADSCV-JTQLQIEISA-N (-)-Nicotine Chemical compound CN1CCC[C@H]1C1=CC=CN=C1 SNICXCGAKADSCV-JTQLQIEISA-N 0.000 description 2
- IWYDHOAUDWTVEP-SSDOTTSWSA-N (R)-mandelic acid Chemical compound OC(=O)[C@H](O)C1=CC=CC=C1 IWYDHOAUDWTVEP-SSDOTTSWSA-N 0.000 description 2
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 2
- FHIFCKSBNKKCJM-UHFFFAOYSA-N 2-amino-4-nitrobenzamide Chemical compound NC(=O)C1=CC=C([N+]([O-])=O)C=C1N FHIFCKSBNKKCJM-UHFFFAOYSA-N 0.000 description 2
- SOBQOVZAFJDEJI-UHFFFAOYSA-N 2-amino-5-nitrobenzamide Chemical compound NC(=O)C1=CC([N+]([O-])=O)=CC=C1N SOBQOVZAFJDEJI-UHFFFAOYSA-N 0.000 description 2
- SJZRECIVHVDYJC-UHFFFAOYSA-N 4-hydroxybutyric acid Chemical compound OCCCC(O)=O SJZRECIVHVDYJC-UHFFFAOYSA-N 0.000 description 2
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical group [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 2
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 2
- 208000003200 Adenoma Diseases 0.000 description 2
- 206010001233 Adenoma benign Diseases 0.000 description 2
- 206010002091 Anaesthesia Diseases 0.000 description 2
- 206010002383 Angina Pectoris Diseases 0.000 description 2
- KXDAEFPNCMNJSK-UHFFFAOYSA-N Benzamide Chemical compound NC(=O)C1=CC=CC=C1 KXDAEFPNCMNJSK-UHFFFAOYSA-N 0.000 description 2
- 201000006474 Brain Ischemia Diseases 0.000 description 2
- UXZAJSZFFARTEI-YRPNKDGESA-N CS(=O)(=O)N[C@@H]1[C@@H](N(CCC1)C(=O)OC)CO[C@@H]1CC[C@@H](CC1)C1=CC=CC=C1 Chemical compound CS(=O)(=O)N[C@@H]1[C@@H](N(CCC1)C(=O)OC)CO[C@@H]1CC[C@@H](CC1)C1=CC=CC=C1 UXZAJSZFFARTEI-YRPNKDGESA-N 0.000 description 2
- 206010008120 Cerebral ischaemia Diseases 0.000 description 2
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 208000000094 Chronic Pain Diseases 0.000 description 2
- 208000019888 Circadian rhythm sleep disease Diseases 0.000 description 2
- RGHNJXZEOKUKBD-SQOUGZDYSA-N D-gluconic acid Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C(O)=O RGHNJXZEOKUKBD-SQOUGZDYSA-N 0.000 description 2
- 206010012218 Delirium Diseases 0.000 description 2
- 206010067889 Dementia with Lewy bodies Diseases 0.000 description 2
- 206010012335 Dependence Diseases 0.000 description 2
- 208000020401 Depressive disease Diseases 0.000 description 2
- 206010013980 Dyssomnias Diseases 0.000 description 2
- 208000030814 Eating disease Diseases 0.000 description 2
- 208000019454 Feeding and Eating disease Diseases 0.000 description 2
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 2
- 201000001498 Froelich syndrome Diseases 0.000 description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 2
- 102000003688 G-Protein-Coupled Receptors Human genes 0.000 description 2
- 108090000045 G-Protein-Coupled Receptors Proteins 0.000 description 2
- 208000011688 Generalised anxiety disease Diseases 0.000 description 2
- AEMRFAOFKBGASW-UHFFFAOYSA-N Glycolic acid Chemical compound OCC(O)=O AEMRFAOFKBGASW-UHFFFAOYSA-N 0.000 description 2
- 101000986786 Homo sapiens Orexin/Hypocretin receptor type 1 Proteins 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 2
- 208000035154 Hyperesthesia Diseases 0.000 description 2
- 206010058359 Hypogonadism Diseases 0.000 description 2
- 208000031773 Insulin resistance syndrome Diseases 0.000 description 2
- 208000032382 Ischaemic stroke Diseases 0.000 description 2
- 208000001456 Jet Lag Syndrome Diseases 0.000 description 2
- 201000007493 Kallmann syndrome Diseases 0.000 description 2
- 201000002832 Lewy body dementia Diseases 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- 208000001089 Multiple system atrophy Diseases 0.000 description 2
- 208000037212 Neonatal hypoxic and ischemic brain injury Diseases 0.000 description 2
- 208000021384 Obsessive-Compulsive disease Diseases 0.000 description 2
- 206010053142 Olfacto genital dysplasia Diseases 0.000 description 2
- KDLHZDBZIXYQEI-UHFFFAOYSA-N Palladium Chemical compound [Pd] KDLHZDBZIXYQEI-UHFFFAOYSA-N 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- 206010035004 Pickwickian syndrome Diseases 0.000 description 2
- 206010065016 Post-traumatic pain Diseases 0.000 description 2
- 206010073211 Postural tremor Diseases 0.000 description 2
- 208000017442 Retinal disease Diseases 0.000 description 2
- 206010038923 Retinopathy Diseases 0.000 description 2
- 208000010340 Sleep Deprivation Diseases 0.000 description 2
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 2
- 208000007107 Stomach Ulcer Diseases 0.000 description 2
- 208000000323 Tourette Syndrome Diseases 0.000 description 2
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 208000005298 acute pain Diseases 0.000 description 2
- 230000032683 aging Effects 0.000 description 2
- 230000001668 ameliorated effect Effects 0.000 description 2
- 230000006933 amyloid-beta aggregation Effects 0.000 description 2
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 description 2
- 230000037005 anaesthesia Effects 0.000 description 2
- 235000019558 anosmia Nutrition 0.000 description 2
- 230000003110 anti-inflammatory effect Effects 0.000 description 2
- 230000036528 appetite Effects 0.000 description 2
- 235000019789 appetite Nutrition 0.000 description 2
- 206010003119 arrhythmia Diseases 0.000 description 2
- 210000004227 basal ganglia Anatomy 0.000 description 2
- 230000006399 behavior Effects 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 239000000090 biomarker Substances 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical compound CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- 238000004364 calculation method Methods 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 125000003917 carbamoyl group Chemical group [H]N([H])C(*)=O 0.000 description 2
- 230000000747 cardiac effect Effects 0.000 description 2
- 230000000768 catecholaminergic effect Effects 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 206010008118 cerebral infarction Diseases 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 229910052801 chlorine Inorganic materials 0.000 description 2
- 201000001883 cholelithiasis Diseases 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 229960003920 cocaine Drugs 0.000 description 2
- 208000014439 complex regional pain syndrome type 2 Diseases 0.000 description 2
- 230000008878 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 125000004093 cyano group Chemical group *C#N 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 229940072719 danavorexton Drugs 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- XBDQKXXYIPTUBI-UHFFFAOYSA-N dimethylselenoniopropionate Natural products CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 231100000673 dose–response relationship Toxicity 0.000 description 2
- 230000035622 drinking Effects 0.000 description 2
- 235000021271 drinking Nutrition 0.000 description 2
- 235000005686 eating Nutrition 0.000 description 2
- 230000019439 energy homeostasis Effects 0.000 description 2
- 238000013100 final test Methods 0.000 description 2
- 239000011737 fluorine Substances 0.000 description 2
- 229910052731 fluorine Inorganic materials 0.000 description 2
- 230000006870 function Effects 0.000 description 2
- 230000002496 gastric effect Effects 0.000 description 2
- 208000029364 generalized anxiety disease Diseases 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 230000000848 glutamatergic effect Effects 0.000 description 2
- 125000001183 hydrocarbyl group Chemical group 0.000 description 2
- 239000001257 hydrogen Substances 0.000 description 2
- 208000031424 hyperprolactinemia Diseases 0.000 description 2
- 239000003326 hypnotic agent Substances 0.000 description 2
- 230000000147 hypnotic effect Effects 0.000 description 2
- 210000003016 hypothalamus Anatomy 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 208000014674 injury Diseases 0.000 description 2
- 150000007529 inorganic bases Chemical class 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 238000000185 intracerebroventricular administration Methods 0.000 description 2
- SUMDYPCJJOFFON-UHFFFAOYSA-N isethionic acid Chemical compound OCCS(O)(=O)=O SUMDYPCJJOFFON-UHFFFAOYSA-N 0.000 description 2
- 208000033915 jet lag type circadian rhythm sleep disease Diseases 0.000 description 2
- 238000011813 knockout mouse model Methods 0.000 description 2
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 2
- 210000004558 lewy body Anatomy 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 206010027175 memory impairment Diseases 0.000 description 2
- 229910021645 metal ion Inorganic materials 0.000 description 2
- 210000000274 microglia Anatomy 0.000 description 2
- 230000001730 monoaminergic effect Effects 0.000 description 2
- 125000002950 monocyclic group Chemical group 0.000 description 2
- 230000036651 mood Effects 0.000 description 2
- BQJCRHHNABKAKU-KBQPJGBKSA-N morphine Chemical compound O([C@H]1[C@H](C=C[C@H]23)O)C4=C5[C@@]12CCN(C)[C@@H]3CC5=CC=C4O BQJCRHHNABKAKU-KBQPJGBKSA-N 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- TXXHDPDFNKHHGW-UHFFFAOYSA-N muconic acid Chemical compound OC(=O)C=CC=CC(O)=O TXXHDPDFNKHHGW-UHFFFAOYSA-N 0.000 description 2
- 201000006417 multiple sclerosis Diseases 0.000 description 2
- 230000003961 neuronal insult Effects 0.000 description 2
- 208000015238 neurotic disease Diseases 0.000 description 2
- 239000002858 neurotransmitter agent Substances 0.000 description 2
- 229960002715 nicotine Drugs 0.000 description 2
- SNICXCGAKADSCV-UHFFFAOYSA-N nicotine Natural products CN1CCCC1C1=CC=CN=C1 SNICXCGAKADSCV-UHFFFAOYSA-N 0.000 description 2
- 230000000422 nocturnal effect Effects 0.000 description 2
- 150000007530 organic bases Chemical class 0.000 description 2
- 230000010355 oscillation Effects 0.000 description 2
- 201000008482 osteoarthritis Diseases 0.000 description 2
- 208000033300 perinatal asphyxia Diseases 0.000 description 2
- 208000023515 periodic limb movement disease Diseases 0.000 description 2
- 230000002085 persistent effect Effects 0.000 description 2
- 230000000144 pharmacologic effect Effects 0.000 description 2
- 125000000587 piperidin-1-yl group Chemical group [H]C1([H])N(*)C([H])([H])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 2
- 125000003367 polycyclic group Chemical group 0.000 description 2
- 230000000750 progressive effect Effects 0.000 description 2
- 238000011552 rat model Methods 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 230000001850 reproductive effect Effects 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- YGSDEFSMJLZEOE-UHFFFAOYSA-N salicylic acid Chemical compound OC(=O)C1=CC=CC=C1O YGSDEFSMJLZEOE-UHFFFAOYSA-N 0.000 description 2
- 238000003345 scintillation counting Methods 0.000 description 2
- 229940125723 sedative agent Drugs 0.000 description 2
- 239000000932 sedative agent Substances 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 230000011664 signaling Effects 0.000 description 2
- 230000003860 sleep quality Effects 0.000 description 2
- 230000037322 slow-wave sleep Effects 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 125000003003 spiro group Chemical group 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 239000007858 starting material Substances 0.000 description 2
- 231100000736 substance abuse Toxicity 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 235000019640 taste Nutrition 0.000 description 2
- 210000001103 thalamus Anatomy 0.000 description 2
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 230000007704 transition Effects 0.000 description 2
- 230000008733 trauma Effects 0.000 description 2
- 206010044652 trigeminal neuralgia Diseases 0.000 description 2
- SFLSHLFXELFNJZ-QMMMGPOBSA-N (-)-norepinephrine Chemical compound NC[C@H](O)C1=CC=C(O)C(O)=C1 SFLSHLFXELFNJZ-QMMMGPOBSA-N 0.000 description 1
- QBYIENPQHBMVBV-HFEGYEGKSA-N (2R)-2-hydroxy-2-phenylacetic acid Chemical compound O[C@@H](C(O)=O)c1ccccc1.O[C@@H](C(O)=O)c1ccccc1 QBYIENPQHBMVBV-HFEGYEGKSA-N 0.000 description 1
- YWPHCCPCQOJSGZ-LLVKDONJSA-N (2r)-2-[(2-ethoxyphenoxy)methyl]morpholine Chemical compound CCOC1=CC=CC=C1OC[C@@H]1OCCNC1 YWPHCCPCQOJSGZ-LLVKDONJSA-N 0.000 description 1
- YONLFQNRGZXBBF-ZIAGYGMSSA-N (2r,3r)-2,3-dibenzoyloxybutanedioic acid Chemical compound O([C@@H](C(=O)O)[C@@H](OC(=O)C=1C=CC=CC=1)C(O)=O)C(=O)C1=CC=CC=C1 YONLFQNRGZXBBF-ZIAGYGMSSA-N 0.000 description 1
- KWTSXDURSIMDCE-QMMMGPOBSA-N (S)-amphetamine Chemical compound C[C@H](N)CC1=CC=CC=C1 KWTSXDURSIMDCE-QMMMGPOBSA-N 0.000 description 1
- MIOPJNTWMNEORI-GMSGAONNSA-N (S)-camphorsulfonic acid Chemical compound C1C[C@@]2(CS(O)(=O)=O)C(=O)C[C@@H]1C2(C)C MIOPJNTWMNEORI-GMSGAONNSA-N 0.000 description 1
- BJEPYKJPYRNKOW-REOHCLBHSA-N (S)-malic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O BJEPYKJPYRNKOW-REOHCLBHSA-N 0.000 description 1
- RFVFQQWKPSOBED-PSXMRANNSA-N 1-myristoyl-2-palmitoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)O[C@@H](COP([O-])(=O)OCC[N+](C)(C)C)COC(=O)CCCCCCCCCCCCC RFVFQQWKPSOBED-PSXMRANNSA-N 0.000 description 1
- YFGHCGITMMYXAQ-UHFFFAOYSA-N 2-[(diphenylmethyl)sulfinyl]acetamide Chemical compound C=1C=CC=CC=1C(S(=O)CC(=O)N)C1=CC=CC=C1 YFGHCGITMMYXAQ-UHFFFAOYSA-N 0.000 description 1
- MGCGMYPNXAFGFA-UHFFFAOYSA-N 2-amino-5-nitrobenzonitrile Chemical compound NC1=CC=C([N+]([O-])=O)C=C1C#N MGCGMYPNXAFGFA-UHFFFAOYSA-N 0.000 description 1
- SDHXWAPVLOGAJR-UHFFFAOYSA-N 2-chloro-5-nitrobenzamide Chemical compound NC(=O)C1=CC([N+]([O-])=O)=CC=C1Cl SDHXWAPVLOGAJR-UHFFFAOYSA-N 0.000 description 1
- UPHOPMSGKZNELG-UHFFFAOYSA-N 2-hydroxynaphthalene-1-carboxylic acid Chemical compound C1=CC=C2C(C(=O)O)=C(O)C=CC2=C1 UPHOPMSGKZNELG-UHFFFAOYSA-N 0.000 description 1
- UJVBZCCNLAAMOV-UHFFFAOYSA-N 2h-1,2-benzothiazine Chemical compound C1=CC=C2C=CNSC2=C1 UJVBZCCNLAAMOV-UHFFFAOYSA-N 0.000 description 1
- VVVBTTVYHSVMOA-UHFFFAOYSA-N 2h-quinazolin-1-amine Chemical compound C1=CC=C2N(N)CN=CC2=C1 VVVBTTVYHSVMOA-UHFFFAOYSA-N 0.000 description 1
- XROPABQYPLIFDB-UHFFFAOYSA-N 3,4-dihydro-1,2-benzothiazin-2-amine Chemical compound C1=CC=C2SN(N)CCC2=C1 XROPABQYPLIFDB-UHFFFAOYSA-N 0.000 description 1
- DXAJOWOZFPQRQL-UHFFFAOYSA-N 3,4-dihydro-1,2-benzoxazin-2-amine Chemical class C1=CC=C2ON(N)CCC2=C1 DXAJOWOZFPQRQL-UHFFFAOYSA-N 0.000 description 1
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 description 1
- SNJWCDJROJUGJG-UHFFFAOYSA-N 6-nitro-2-phenyl-2,3-dihydro-1h-quinazolin-4-one Chemical compound N1C(=O)C2=CC([N+](=O)[O-])=CC=C2NC1C1=CC=CC=C1 SNJWCDJROJUGJG-UHFFFAOYSA-N 0.000 description 1
- 206010000599 Acromegaly Diseases 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 208000008811 Agoraphobia Diseases 0.000 description 1
- 206010001541 Akinesia Diseases 0.000 description 1
- 201000000736 Amenorrhea Diseases 0.000 description 1
- 206010001928 Amenorrhoea Diseases 0.000 description 1
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 1
- 208000000044 Amnesia Diseases 0.000 description 1
- 208000006820 Arthralgia Diseases 0.000 description 1
- 208000025978 Athletic injury Diseases 0.000 description 1
- 208000008035 Back Pain Diseases 0.000 description 1
- 208000035183 Benign hereditary chorea Diseases 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- 206010006002 Bone pain Diseases 0.000 description 1
- 206010006100 Bradykinesia Diseases 0.000 description 1
- 206010048962 Brain oedema Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- 208000021465 Brief psychotic disease Diseases 0.000 description 1
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 1
- 208000032841 Bulimia Diseases 0.000 description 1
- 206010006550 Bulimia nervosa Diseases 0.000 description 1
- 208000025721 COVID-19 Diseases 0.000 description 1
- VOSAWOSMGPKQEQ-OALUTQOASA-N CS(=O)(=O)N[C@@H]1[C@@H](N(CC1)C(C(C)(C)O)=O)CC=1C(=C(C=CC=1)C1=CC(=CC(=C1)F)F)F Chemical compound CS(=O)(=O)N[C@@H]1[C@@H](N(CC1)C(C(C)(C)O)=O)CC=1C(=C(C=CC=1)C1=CC(=CC(=C1)F)F)F VOSAWOSMGPKQEQ-OALUTQOASA-N 0.000 description 1
- 206010006895 Cachexia Diseases 0.000 description 1
- 206010058019 Cancer Pain Diseases 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 206010007556 Cardiac failure acute Diseases 0.000 description 1
- 208000024172 Cardiovascular disease Diseases 0.000 description 1
- 102000005600 Cathepsins Human genes 0.000 description 1
- 108010084457 Cathepsins Proteins 0.000 description 1
- 208000001387 Causalgia Diseases 0.000 description 1
- 208000003417 Central Sleep Apnea Diseases 0.000 description 1
- 208000017164 Chronobiology disease Diseases 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 241001573498 Compacta Species 0.000 description 1
- 208000023890 Complex Regional Pain Syndromes Diseases 0.000 description 1
- 208000033001 Complex partial seizures Diseases 0.000 description 1
- 208000013586 Complex regional pain syndrome type 1 Diseases 0.000 description 1
- 208000027691 Conduct disease Diseases 0.000 description 1
- 206010066131 Congenital central hypoventilation syndrome Diseases 0.000 description 1
- 208000011990 Corticobasal Degeneration Diseases 0.000 description 1
- 208000009798 Craniopharyngioma Diseases 0.000 description 1
- 208000020406 Creutzfeldt Jacob disease Diseases 0.000 description 1
- 208000003407 Creutzfeldt-Jakob Syndrome Diseases 0.000 description 1
- 208000010859 Creutzfeldt-Jakob disease Diseases 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- 208000014311 Cushing syndrome Diseases 0.000 description 1
- 102000005927 Cysteine Proteases Human genes 0.000 description 1
- 108010005843 Cysteine Proteases Proteins 0.000 description 1
- RGHNJXZEOKUKBD-UHFFFAOYSA-N D-gluconic acid Natural products OCC(O)C(O)C(O)C(O)C(O)=O RGHNJXZEOKUKBD-UHFFFAOYSA-N 0.000 description 1
- 206010011878 Deafness Diseases 0.000 description 1
- 208000024254 Delusional disease Diseases 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- 208000025967 Dissociative Identity disease Diseases 0.000 description 1
- 206010013883 Dwarfism Diseases 0.000 description 1
- 208000032928 Dyslipidaemia Diseases 0.000 description 1
- 231100000491 EC50 Toxicity 0.000 description 1
- 206010014733 Endometrial cancer Diseases 0.000 description 1
- 206010014759 Endometrial neoplasm Diseases 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 208000010235 Food Addiction Diseases 0.000 description 1
- 230000005526 G1 to G0 transition Effects 0.000 description 1
- 208000017228 Gastrointestinal motility disease Diseases 0.000 description 1
- 206010064571 Gene mutation Diseases 0.000 description 1
- 206010018265 Gigantism Diseases 0.000 description 1
- 201000004311 Gilles de la Tourette syndrome Diseases 0.000 description 1
- 208000002705 Glucose Intolerance Diseases 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 206010056438 Growth hormone deficiency Diseases 0.000 description 1
- 208000031886 HIV Infections Diseases 0.000 description 1
- 208000004547 Hallucinations Diseases 0.000 description 1
- 206010019196 Head injury Diseases 0.000 description 1
- 206010019233 Headaches Diseases 0.000 description 1
- 208000010496 Heart Arrest Diseases 0.000 description 1
- 208000016988 Hemorrhagic Stroke Diseases 0.000 description 1
- GVGLGOZIDCSQPN-PVHGPHFFSA-N Heroin Chemical compound O([C@H]1[C@H](C=C[C@H]23)OC(C)=O)C4=C5[C@@]12CCN(C)[C@@H]3CC5=CC=C4OC(C)=O GVGLGOZIDCSQPN-PVHGPHFFSA-N 0.000 description 1
- 206010020112 Hirsutism Diseases 0.000 description 1
- 101000618525 Homo sapiens Membrane transport protein XK Proteins 0.000 description 1
- 101001071429 Homo sapiens Metabotropic glutamate receptor 2 Proteins 0.000 description 1
- 101001098357 Homo sapiens Orexin receptor type 2 Proteins 0.000 description 1
- 208000033830 Hot Flashes Diseases 0.000 description 1
- 206010060800 Hot flush Diseases 0.000 description 1
- 208000031226 Hyperlipidaemia Diseases 0.000 description 1
- 206010020710 Hyperphagia Diseases 0.000 description 1
- 206010020928 Hypnopompic hallucination Diseases 0.000 description 1
- 208000006083 Hypokinesia Diseases 0.000 description 1
- 206010021067 Hypopituitarism Diseases 0.000 description 1
- 206010050515 Hyposmia Diseases 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- 208000025282 Hypothalamo-pituitary disease Diseases 0.000 description 1
- 206010061216 Infarction Diseases 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 206010065390 Inflammatory pain Diseases 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 206010022489 Insulin Resistance Diseases 0.000 description 1
- 206010022520 Intention tremor Diseases 0.000 description 1
- 208000004404 Intractable Pain Diseases 0.000 description 1
- LPHGQDQBBGAPDZ-UHFFFAOYSA-N Isocaffeine Natural products CN1C(=O)N(C)C(=O)C2=C1N(C)C=N2 LPHGQDQBBGAPDZ-UHFFFAOYSA-N 0.000 description 1
- WTDRDQBEARUVNC-LURJTMIESA-N L-DOPA Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C(O)=C1 WTDRDQBEARUVNC-LURJTMIESA-N 0.000 description 1
- WTDRDQBEARUVNC-UHFFFAOYSA-N L-Dopa Natural products OC(=O)C(N)CC1=CC=C(O)C(O)=C1 WTDRDQBEARUVNC-UHFFFAOYSA-N 0.000 description 1
- 201000006792 Lennox-Gastaut syndrome Diseases 0.000 description 1
- 208000019693 Lung disease Diseases 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 206010026749 Mania Diseases 0.000 description 1
- 208000025972 Maternal Obesity Diseases 0.000 description 1
- 102100022472 Membrane transport protein XK Human genes 0.000 description 1
- 208000001145 Metabolic Syndrome Diseases 0.000 description 1
- 102100036837 Metabotropic glutamate receptor 2 Human genes 0.000 description 1
- DUGOZIWVEXMGBE-UHFFFAOYSA-N Methylphenidate Chemical compound C=1C=CC=CC=1C(C(=O)OC)C1CCCCN1 DUGOZIWVEXMGBE-UHFFFAOYSA-N 0.000 description 1
- 208000016285 Movement disease Diseases 0.000 description 1
- TXXHDPDFNKHHGW-CCAGOZQPSA-N Muconic acid Natural products OC(=O)\C=C/C=C\C(O)=O TXXHDPDFNKHHGW-CCAGOZQPSA-N 0.000 description 1
- 208000002740 Muscle Rigidity Diseases 0.000 description 1
- 208000008238 Muscle Spasticity Diseases 0.000 description 1
- 208000029549 Muscle injury Diseases 0.000 description 1
- 208000021642 Muscular disease Diseases 0.000 description 1
- MBBZMMPHUWSWHV-BDVNFPICSA-N N-methylglucamine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 1
- 206010028813 Nausea Diseases 0.000 description 1
- 201000005625 Neuroleptic malignant syndrome Diseases 0.000 description 1
- 208000002537 Neuronal Ceroid-Lipofuscinoses Diseases 0.000 description 1
- 108090000189 Neuropeptides Proteins 0.000 description 1
- 206010029333 Neurosis Diseases 0.000 description 1
- 208000000224 Night Terrors Diseases 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- 208000004166 Obesity Hypoventilation Syndrome Diseases 0.000 description 1
- 102100028141 Orexin/Hypocretin receptor type 1 Human genes 0.000 description 1
- 206010033664 Panic attack Diseases 0.000 description 1
- 208000037273 Pathologic Processes Diseases 0.000 description 1
- RMUCZJUITONUFY-UHFFFAOYSA-N Phenelzine Chemical compound NNCCC1=CC=CC=C1 RMUCZJUITONUFY-UHFFFAOYSA-N 0.000 description 1
- 206010034912 Phobia Diseases 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 208000000609 Pick Disease of the Brain Diseases 0.000 description 1
- 208000010067 Pituitary ACTH Hypersecretion Diseases 0.000 description 1
- 208000020627 Pituitary-dependent Cushing syndrome Diseases 0.000 description 1
- 108010064218 Poly (ADP-Ribose) Polymerase-1 Proteins 0.000 description 1
- 102100023712 Poly [ADP-ribose] polymerase 1 Human genes 0.000 description 1
- 208000036757 Postencephalitic parkinsonism Diseases 0.000 description 1
- 208000004550 Postoperative Pain Diseases 0.000 description 1
- 201000010769 Prader-Willi syndrome Diseases 0.000 description 1
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 1
- 206010036832 Prolactinoma Diseases 0.000 description 1
- OFOBLEOULBTSOW-UHFFFAOYSA-N Propanedioic acid Natural products OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 206010061921 Psychotic disorder due to a general medical condition Diseases 0.000 description 1
- IWYDHOAUDWTVEP-UHFFFAOYSA-N R-2-phenyl-2-hydroxyacetic acid Natural products OC(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-N 0.000 description 1
- 206010071390 Resting tremor Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 208000020186 Schizophreniform disease Diseases 0.000 description 1
- 208000000810 Separation Anxiety Diseases 0.000 description 1
- 208000019568 Shared Paranoid disease Diseases 0.000 description 1
- 208000028810 Shared psychotic disease Diseases 0.000 description 1
- 208000005439 Sleep paralysis Diseases 0.000 description 1
- 206010041010 Sleep terror Diseases 0.000 description 1
- 206010041250 Social phobia Diseases 0.000 description 1
- 208000005392 Spasm Diseases 0.000 description 1
- 206010067672 Spasmodic dysphonia Diseases 0.000 description 1
- 206010041738 Sports injury Diseases 0.000 description 1
- 208000032851 Subarachnoid Hemorrhage Diseases 0.000 description 1
- 208000007271 Substance Withdrawal Syndrome Diseases 0.000 description 1
- 208000011962 Substance-induced mood disease Diseases 0.000 description 1
- 231100000395 Substance-induced mood disorder Toxicity 0.000 description 1
- 208000011963 Substance-induced psychotic disease Diseases 0.000 description 1
- 231100000393 Substance-induced psychotic disorder Toxicity 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- 208000027522 Sydenham chorea Diseases 0.000 description 1
- 102000019355 Synuclein Human genes 0.000 description 1
- 108050006783 Synuclein Proteins 0.000 description 1
- 206010043118 Tardive Dyskinesia Diseases 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 206010068932 Terminal insomnia Diseases 0.000 description 1
- QIOZLISABUUKJY-UHFFFAOYSA-N Thiobenzamide Chemical compound NC(=S)C1=CC=CC=C1 QIOZLISABUUKJY-UHFFFAOYSA-N 0.000 description 1
- 208000009205 Tinnitus Diseases 0.000 description 1
- 206010044074 Torticollis Diseases 0.000 description 1
- 208000016620 Tourette disease Diseases 0.000 description 1
- 208000031674 Traumatic Acute Stress disease Diseases 0.000 description 1
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Triethanolamine Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 1
- 208000026928 Turner syndrome Diseases 0.000 description 1
- 206010046543 Urinary incontinence Diseases 0.000 description 1
- 208000013142 Writer cramp Diseases 0.000 description 1
- 201000000690 abdominal obesity-metabolic syndrome Diseases 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 208000003554 absence epilepsy Diseases 0.000 description 1
- 208000028311 absence seizure Diseases 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 208000026345 acute stress disease Diseases 0.000 description 1
- CGNMLOKEMNBUAI-UHFFFAOYSA-N adrafinil Chemical compound C=1C=CC=CC=1C(S(=O)CC(=O)NO)C1=CC=CC=C1 CGNMLOKEMNBUAI-UHFFFAOYSA-N 0.000 description 1
- 229960002820 adrafinil Drugs 0.000 description 1
- 230000001919 adrenal effect Effects 0.000 description 1
- 230000007000 age related cognitive decline Effects 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 230000008484 agonism Effects 0.000 description 1
- 230000036626 alertness Effects 0.000 description 1
- 229910001420 alkaline earth metal ion Inorganic materials 0.000 description 1
- 206010053552 allodynia Diseases 0.000 description 1
- 230000003281 allosteric effect Effects 0.000 description 1
- VWYANPOOORUCFJ-UHFFFAOYSA-N alpha-Fernenol Chemical compound CC1(C)C(O)CCC2(C)C3=CCC4(C)C5CCC(C(C)C)C5(C)CCC4(C)C3CCC21 VWYANPOOORUCFJ-UHFFFAOYSA-N 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-N alpha-hydroxysuccinic acid Natural products OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 231100000540 amenorrhea Toxicity 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 235000011114 ammonium hydroxide Nutrition 0.000 description 1
- 230000006986 amnesia Effects 0.000 description 1
- 229940025084 amphetamine Drugs 0.000 description 1
- 210000004727 amygdala Anatomy 0.000 description 1
- 229940035674 anesthetics Drugs 0.000 description 1
- 208000022531 anorexia Diseases 0.000 description 1
- 230000008485 antagonism Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 230000007529 anxiety like behavior Effects 0.000 description 1
- 239000002249 anxiolytic agent Substances 0.000 description 1
- 230000000949 anxiolytic effect Effects 0.000 description 1
- 229940005530 anxiolytics Drugs 0.000 description 1
- 235000021407 appetite control Nutrition 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 229960004823 armodafinil Drugs 0.000 description 1
- YFGHCGITMMYXAQ-LJQANCHMSA-N armodafinil Chemical compound C=1C=CC=CC=1C([S@](=O)CC(=O)N)C1=CC=CC=C1 YFGHCGITMMYXAQ-LJQANCHMSA-N 0.000 description 1
- 230000002917 arthritic effect Effects 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 229960002430 atomoxetine Drugs 0.000 description 1
- VHGCDTVCOLNTBX-QGZVFWFLSA-N atomoxetine Chemical compound O([C@H](CCNC)C=1C=CC=CC=1)C1=CC=CC=C1C VHGCDTVCOLNTBX-QGZVFWFLSA-N 0.000 description 1
- 230000003190 augmentative effect Effects 0.000 description 1
- 230000002567 autonomic effect Effects 0.000 description 1
- 210000003050 axon Anatomy 0.000 description 1
- 201000002922 basal ganglia calcification Diseases 0.000 description 1
- 230000003542 behavioural effect Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-N benzenesulfonic acid Chemical compound OS(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-N 0.000 description 1
- 229940092714 benzenesulfonic acid Drugs 0.000 description 1
- 229940049706 benzodiazepine Drugs 0.000 description 1
- 125000003310 benzodiazepinyl group Chemical class N1N=C(C=CC2=C1C=CC=C2)* 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 150000005130 benzoxazines Chemical class 0.000 description 1
- 125000002619 bicyclic group Chemical group 0.000 description 1
- 208000016791 bilateral striopallidodentate calcinosis Diseases 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 208000028683 bipolar I disease Diseases 0.000 description 1
- 208000022257 bipolar II disease Diseases 0.000 description 1
- 206010005159 blepharospasm Diseases 0.000 description 1
- 230000000744 blepharospasm Effects 0.000 description 1
- 230000036772 blood pressure Effects 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 208000006752 brain edema Diseases 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 1
- 229910052794 bromium Inorganic materials 0.000 description 1
- 230000009172 bursting Effects 0.000 description 1
- KDYFGRWQOYBRFD-NUQCWPJISA-N butanedioic acid Chemical compound O[14C](=O)CC[14C](O)=O KDYFGRWQOYBRFD-NUQCWPJISA-N 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- VJEONQKOZGKCAK-UHFFFAOYSA-N caffeine Natural products CN1C(=O)N(C)C(=O)C2=C1C=CN2C VJEONQKOZGKCAK-UHFFFAOYSA-N 0.000 description 1
- AXCZMVOFGPJBDE-UHFFFAOYSA-L calcium dihydroxide Chemical compound [OH-].[OH-].[Ca+2] AXCZMVOFGPJBDE-UHFFFAOYSA-L 0.000 description 1
- 239000000920 calcium hydroxide Substances 0.000 description 1
- 229910001861 calcium hydroxide Inorganic materials 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 210000000748 cardiovascular system Anatomy 0.000 description 1
- 206010007776 catatonia Diseases 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000002490 cerebral effect Effects 0.000 description 1
- 210000004289 cerebral ventricle Anatomy 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 238000002512 chemotherapy Methods 0.000 description 1
- 210000004978 chinese hamster ovary cell Anatomy 0.000 description 1
- 238000013375 chromatographic separation Methods 0.000 description 1
- 230000001684 chronic effect Effects 0.000 description 1
- 208000020832 chronic kidney disease Diseases 0.000 description 1
- 208000022831 chronic renal failure syndrome Diseases 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 229940121657 clinical drug Drugs 0.000 description 1
- 230000019771 cognition Effects 0.000 description 1
- 230000003920 cognitive function Effects 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 238000010668 complexation reaction Methods 0.000 description 1
- 229940124301 concurrent medication Drugs 0.000 description 1
- 208000036970 congenital 1 with or without Hirschsprung disease central hypoventilation syndrome Diseases 0.000 description 1
- 230000036461 convulsion Effects 0.000 description 1
- 208000029078 coronary artery disease Diseases 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 125000000582 cycloheptyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 description 1
- 208000026725 cyclothymic disease Diseases 0.000 description 1
- 206010061428 decreased appetite Diseases 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 230000007850 degeneration Effects 0.000 description 1
- 230000005786 degenerative changes Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 229960002069 diamorphine Drugs 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical compound OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 208000037765 diseases and disorders Diseases 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 206010013461 dissociative amnesia Diseases 0.000 description 1
- 208000018459 dissociative disease Diseases 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 210000004002 dopaminergic cell Anatomy 0.000 description 1
- 230000003291 dopaminomimetic effect Effects 0.000 description 1
- 230000000632 dystonic effect Effects 0.000 description 1
- 208000016570 early-onset generalized limb-onset dystonia Diseases 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 230000002357 endometrial effect Effects 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 230000001667 episodic effect Effects 0.000 description 1
- 201000006517 essential tremor Diseases 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 230000002964 excitative effect Effects 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 230000035558 fertility Effects 0.000 description 1
- 231100000502 fertility decrease Toxicity 0.000 description 1
- 230000001605 fetal effect Effects 0.000 description 1
- 229940073049 firazorexton Drugs 0.000 description 1
- 201000002904 focal dystonia Diseases 0.000 description 1
- 201000002865 focal hand dystonia Diseases 0.000 description 1
- 230000037406 food intake Effects 0.000 description 1
- 235000012631 food intake Nutrition 0.000 description 1
- 238000001640 fractional crystallisation Methods 0.000 description 1
- 238000013467 fragmentation Methods 0.000 description 1
- 238000006062 fragmentation reaction Methods 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- 208000001130 gallstones Diseases 0.000 description 1
- 208000021302 gastroesophageal reflux disease Diseases 0.000 description 1
- 239000003193 general anesthetic agent Substances 0.000 description 1
- 238000002682 general surgery Methods 0.000 description 1
- 201000002886 generalized dystonia Diseases 0.000 description 1
- 210000004392 genitalia Anatomy 0.000 description 1
- 230000007277 glial cell activation Effects 0.000 description 1
- 210000001905 globus pallidus Anatomy 0.000 description 1
- 239000000174 gluconic acid Substances 0.000 description 1
- 235000012208 gluconic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 230000010370 hearing loss Effects 0.000 description 1
- 231100000888 hearing loss Toxicity 0.000 description 1
- 208000016354 hearing loss disease Diseases 0.000 description 1
- 208000019622 heart disease Diseases 0.000 description 1
- 210000001320 hippocampus Anatomy 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 102000044543 human HCRTR1 Human genes 0.000 description 1
- 102000044551 human HCRTR2 Human genes 0.000 description 1
- 150000004677 hydrates Chemical class 0.000 description 1
- 230000006951 hyperphosphorylation Effects 0.000 description 1
- 230000002218 hypoglycaemic effect Effects 0.000 description 1
- 235000019559 hyposmia Nutrition 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 208000003532 hypothyroidism Diseases 0.000 description 1
- 230000002989 hypothyroidism Effects 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000008105 immune reaction Effects 0.000 description 1
- 208000026278 immune system disease Diseases 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 230000007574 infarction Effects 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 208000000509 infertility Diseases 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 231100000535 infertility Toxicity 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 239000000543 intermediate Substances 0.000 description 1
- 230000008991 intestinal motility Effects 0.000 description 1
- 208000020658 intracerebral hemorrhage Diseases 0.000 description 1
- 229910052740 iodine Inorganic materials 0.000 description 1
- 239000011630 iodine Substances 0.000 description 1
- 208000002551 irritable bowel syndrome Diseases 0.000 description 1
- 208000028867 ischemia Diseases 0.000 description 1
- 230000000302 ischemic effect Effects 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 230000000155 isotopic effect Effects 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 208000017169 kidney disease Diseases 0.000 description 1
- 230000003907 kidney function Effects 0.000 description 1
- 239000004310 lactic acid Substances 0.000 description 1
- 235000014655 lactic acid Nutrition 0.000 description 1
- 229960000448 lactic acid Drugs 0.000 description 1
- 210000003796 lateral hypothalamic area Anatomy 0.000 description 1
- 229960004502 levodopa Drugs 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 230000006742 locomotor activity Effects 0.000 description 1
- 210000000627 locus coeruleus Anatomy 0.000 description 1
- 230000002132 lysosomal effect Effects 0.000 description 1
- 208000002780 macular degeneration Diseases 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 229940098895 maleic acid Drugs 0.000 description 1
- 239000001630 malic acid Substances 0.000 description 1
- 235000011090 malic acid Nutrition 0.000 description 1
- 229940099690 malic acid Drugs 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 229960002510 mandelic acid Drugs 0.000 description 1
- 240000004308 marijuana Species 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000006386 memory function Effects 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 229940098779 methanesulfonic acid Drugs 0.000 description 1
- 229960001344 methylphenidate Drugs 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 230000004065 mitochondrial dysfunction Effects 0.000 description 1
- 229960001165 modafinil Drugs 0.000 description 1
- 229960005181 morphine Drugs 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- 230000008450 motivation Effects 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 230000003387 muscular Effects 0.000 description 1
- 230000007886 mutagenicity Effects 0.000 description 1
- 231100000299 mutagenicity Toxicity 0.000 description 1
- 208000010125 myocardial infarction Diseases 0.000 description 1
- LCPQCTBHZPMVFX-UHFFFAOYSA-N n-[4-(1-cyclobutylpiperidin-4-yl)oxyphenyl]-2-morpholin-4-ylacetamide;dihydrochloride Chemical compound Cl.Cl.C=1C=C(OC2CCN(CC2)C2CCC2)C=CC=1NC(=O)CN1CCOCC1 LCPQCTBHZPMVFX-UHFFFAOYSA-N 0.000 description 1
- XGZZHZMWIXFATA-UEZBDDGYSA-N nalfurafine Chemical compound C([C@]12[C@H]3OC=4C(O)=CC=C(C2=4)C[C@@H]2[C@]1(O)CC[C@H]3N(C)C(=O)\C=C\C1=COC=C1)CN2CC1CC1 XGZZHZMWIXFATA-UEZBDDGYSA-N 0.000 description 1
- KVBGVZZKJNLNJU-UHFFFAOYSA-N naphthalene-2-sulfonic acid Chemical compound C1=CC=CC2=CC(S(=O)(=O)O)=CC=C21 KVBGVZZKJNLNJU-UHFFFAOYSA-N 0.000 description 1
- 125000001624 naphthyl group Chemical group 0.000 description 1
- 230000008693 nausea Effects 0.000 description 1
- 210000001577 neostriatum Anatomy 0.000 description 1
- 230000001537 neural effect Effects 0.000 description 1
- 208000007431 neuroacanthocytosis Diseases 0.000 description 1
- 230000000955 neuroendocrine Effects 0.000 description 1
- 208000037860 neuroleptic-induced Akathisia Diseases 0.000 description 1
- 230000016273 neuron death Effects 0.000 description 1
- 201000008051 neuronal ceroid lipofuscinosis Diseases 0.000 description 1
- 230000004112 neuroprotection Effects 0.000 description 1
- 206010029410 night sweats Diseases 0.000 description 1
- 230000036565 night sweats Effects 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- 150000002828 nitro derivatives Chemical class 0.000 description 1
- 230000008452 non REM sleep Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- SFLSHLFXELFNJZ-UHFFFAOYSA-N norepinephrine Natural products NCC(O)C1=CC=C(O)C(O)=C1 SFLSHLFXELFNJZ-UHFFFAOYSA-N 0.000 description 1
- 229960002748 norepinephrine Drugs 0.000 description 1
- 208000001797 obstructive sleep apnea Diseases 0.000 description 1
- 238000012346 open field test Methods 0.000 description 1
- 229940127240 opiate Drugs 0.000 description 1
- 229940005483 opioid analgesics Drugs 0.000 description 1
- 230000001956 orexigenic effect Effects 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 201000002851 oromandibular dystonia Diseases 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 235000020830 overeating Nutrition 0.000 description 1
- 230000036542 oxidative stress Effects 0.000 description 1
- 125000004430 oxygen atom Chemical group O* 0.000 description 1
- 230000001898 pallidal effect Effects 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N papa-hydroxy-benzoic acid Natural products OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- 208000002851 paranoid schizophrenia Diseases 0.000 description 1
- 230000008756 pathogenetic mechanism Effects 0.000 description 1
- 230000009054 pathological process Effects 0.000 description 1
- 230000004963 pathophysiological condition Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 125000001147 pentyl group Chemical group C(CCCC)* 0.000 description 1
- 230000002748 permeability into the brain Effects 0.000 description 1
- 208000030062 persistent idiopathic facial pain Diseases 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 229950010883 phencyclidine Drugs 0.000 description 1
- JTJMJGYZQZDUJJ-UHFFFAOYSA-N phencyclidine Chemical compound C1CCCCN1C1(C=2C=CC=CC=2)CCCCC1 JTJMJGYZQZDUJJ-UHFFFAOYSA-N 0.000 description 1
- 229960000964 phenelzine Drugs 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 125000004193 piperazinyl group Chemical group 0.000 description 1
- 125000003386 piperidinyl group Chemical group 0.000 description 1
- NNACHAUCXXVJSP-UHFFFAOYSA-N pitolisant Chemical compound C1=CC(Cl)=CC=C1CCCOCCCN1CCCCC1 NNACHAUCXXVJSP-UHFFFAOYSA-N 0.000 description 1
- 229960003651 pitolisant Drugs 0.000 description 1
- IUGYQRQAERSCNH-UHFFFAOYSA-N pivalic acid Chemical compound CC(C)(C)C(O)=O IUGYQRQAERSCNH-UHFFFAOYSA-N 0.000 description 1
- 208000028173 post-traumatic stress disease Diseases 0.000 description 1
- 208000000170 postencephalitic Parkinson disease Diseases 0.000 description 1
- 230000001144 postural effect Effects 0.000 description 1
- WSHYKIAQCMIPTB-UHFFFAOYSA-M potassium;2-oxo-3-(3-oxo-1-phenylbutyl)chromen-4-olate Chemical compound [K+].[O-]C=1C2=CC=CC=C2OC(=O)C=1C(CC(=O)C)C1=CC=CC=C1 WSHYKIAQCMIPTB-UHFFFAOYSA-M 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 201000009104 prediabetes syndrome Diseases 0.000 description 1
- 238000009117 preventive therapy Methods 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 201000009395 primary hyperaldosteronism Diseases 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 229940002612 prodrug Drugs 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 201000002212 progressive supranuclear palsy Diseases 0.000 description 1
- 230000000770 proinflammatory effect Effects 0.000 description 1
- 208000030153 prolactin-producing pituitary gland adenoma Diseases 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 235000019260 propionic acid Nutrition 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 230000009979 protective mechanism Effects 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000007111 proteostasis Effects 0.000 description 1
- 229960002601 protriptyline Drugs 0.000 description 1
- BWPIARFWQZKAIA-UHFFFAOYSA-N protriptyline Chemical compound C1=CC2=CC=CC=C2C(CCCNC)C2=CC=CC=C21 BWPIARFWQZKAIA-UHFFFAOYSA-N 0.000 description 1
- 230000001107 psychogenic effect Effects 0.000 description 1
- 238000010926 purge Methods 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 125000003226 pyrazolyl group Chemical group 0.000 description 1
- 125000004076 pyridyl group Chemical group 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- IUVKMZGDUIUOCP-BTNSXGMBSA-N quinbolone Chemical compound O([C@H]1CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)C=C4CC3)C)CC[C@@]21C)C1=CCCC1 IUVKMZGDUIUOCP-BTNSXGMBSA-N 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- CBQGYUDMJHNJBX-RTBURBONSA-N reboxetine Chemical compound CCOC1=CC=CC=C1O[C@H](C=1C=CC=CC=1)[C@@H]1OCCNC1 CBQGYUDMJHNJBX-RTBURBONSA-N 0.000 description 1
- 229960003770 reboxetine Drugs 0.000 description 1
- 230000000306 recurrent effect Effects 0.000 description 1
- 238000000611 regression analysis Methods 0.000 description 1
- 230000022983 regulation of cell cycle Effects 0.000 description 1
- 230000015355 regulation of circadian sleep/wake cycle, sleep Effects 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 208000023504 respiratory system disease Diseases 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 230000003938 response to stress Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 229960004889 salicylic acid Drugs 0.000 description 1
- 229940073599 samelisant Drugs 0.000 description 1
- 208000022610 schizoaffective disease Diseases 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 125000003748 selenium group Chemical group *[Se]* 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 230000001953 sensory effect Effects 0.000 description 1
- 208000025874 separation anxiety disease Diseases 0.000 description 1
- 230000001568 sexual effect Effects 0.000 description 1
- 230000036299 sexual function Effects 0.000 description 1
- 108010080097 sigma-1 receptor Proteins 0.000 description 1
- 230000004617 sleep duration Effects 0.000 description 1
- 230000004620 sleep latency Effects 0.000 description 1
- 230000008667 sleep stage Effects 0.000 description 1
- 208000020685 sleep-wake disease Diseases 0.000 description 1
- 230000004036 social memory Effects 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- 229960003928 sodium oxybate Drugs 0.000 description 1
- 239000012453 solvate Substances 0.000 description 1
- 201000002849 spasmodic dystonia Diseases 0.000 description 1
- 208000018198 spasticity Diseases 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 201000001716 specific phobia Diseases 0.000 description 1
- 210000000278 spinal cord Anatomy 0.000 description 1
- 230000000707 stereoselective effect Effects 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 210000004281 subthalamic nucleus Anatomy 0.000 description 1
- 235000011149 sulphuric acid Nutrition 0.000 description 1
- MQDUVMBBJZLFHF-WOJBJXKFSA-N suntinorexton Chemical compound CCS(=O)(=O)N[C@@H]1CCN([C@@H]1CC1=CC=CC(=C1F)C1=CC(F)=CC=C1)C(=O)C(C)(C)O MQDUVMBBJZLFHF-WOJBJXKFSA-N 0.000 description 1
- 229940073501 suntinorexton Drugs 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 238000011287 therapeutic dose Methods 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 231100000886 tinnitus Toxicity 0.000 description 1
- 235000019505 tobacco product Nutrition 0.000 description 1
- 125000001425 triazolyl group Chemical group 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 229960000281 trometamol Drugs 0.000 description 1
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 1
- 230000002485 urinary effect Effects 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 230000024883 vasodilation Effects 0.000 description 1
- 210000004515 ventral tegmental area Anatomy 0.000 description 1
- 229960001255 viloxazine Drugs 0.000 description 1
- 208000009935 visceral pain Diseases 0.000 description 1
- 230000008673 vomiting Effects 0.000 description 1
- 238000002424 x-ray crystallography Methods 0.000 description 1
- 229910052727 yttrium Inorganic materials 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic 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
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/517—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/535—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
- A61K31/536—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with carbocyclic ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/54—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
- A61K31/5415—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with carbocyclic ring systems, e.g. phenothiazine, chlorpromazine, piroxicam
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/26—Psychostimulants, e.g. nicotine, cocaine
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D239/00—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
- C07D239/70—Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
- C07D239/72—Quinazolines; Hydrogenated quinazolines
- C07D239/86—Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
- C07D239/88—Oxygen atoms
- C07D239/91—Oxygen atoms with aryl or aralkyl radicals attached in position 2 or 3
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D265/00—Heterocyclic compounds containing six-membered rings having one nitrogen atom and one oxygen atom as the only ring hetero atoms
- C07D265/04—1,3-Oxazines; Hydrogenated 1,3-oxazines
- C07D265/12—1,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems
- C07D265/14—1,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring
- C07D265/20—1,3-Oxazines; Hydrogenated 1,3-oxazines condensed with carbocyclic rings or ring systems condensed with one six-membered ring with hetero atoms directly attached in position 4
- C07D265/22—Oxygen atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D279/00—Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
- C07D279/04—1,3-Thiazines; Hydrogenated 1,3-thiazines
- C07D279/08—1,3-Thiazines; Hydrogenated 1,3-thiazines condensed with carbocyclic rings or ring systems
Definitions
- the present invention is directed to dihydro-quinazoline, -benzothiazine and - benzoxazine derivatives for use in the prevention or treatment of neurological, psychiatric, sleep disorders and diseases, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
- the present invention is also directed to pharmaceutical compositions comprising these compounds for use in the prevention and/or treatment of neurological disorders and diseases.
- the present invention is also directed to dihydrobenzothiazine and dihydro-benzoxazine derivatives and the use of these compounds as a medicament.
- Orexin 1 and 2 (0X1 and 0X2, also known as hypocretin 1 and 2 or orexin A and B) are hypothalamic neuropeptides specifically produced in the lateral hypothalamic area (Sakurai et al. 1998). Orexins act on the two G protein-coupled receptors (GPCRs), the orexin 1 receptor (0X1 R) and the orexin 2 receptor (0X2R), participating in a broad range of physiological functions such as sleep/wakefulness (Gao et al. 2021 ; de Lecea et al. 1998; Lin et al. 1999; Ohno et Sakurai 2008), feeding behavior (Sakurai et al. 1998), reward -seeking (Cason et al.
- GPCRs G protein-coupled receptors
- OX1 R is mainly involved in motivation and reward and the OX2R in the modulation of sleep/wake cycle and energy homeostasis (Perrey et Zhang 2020).
- SB-334867 an experimental 0X1 R selective antagonist
- SB-334867 an experimental 0X1 R selective antagonist
- 0X1 R antagonists In contrary to 0X1 R antagonists, a potential therapeutic role of 0X1 R agonists in REM sleep behavior disorder (RBD) is hypothesized due to the interaction between o- synuclein and 0X1 R in pathogenesis. 0X1 R dysfunction can induce the occurrence of RBD and is a potent early sign of Parkinson's disease (PD), but whether the pathogenetic mechanism involved in RBD remains unexplored. In contrast, o-synuclein has been verified to form Lewy bodies in orexin neurons, whose activity and function depend on orexin receptor 1 (0X1 R) (Fan et al. 2023).
- 0X1 R agonists may be a useful strategy to slow or stop the neurodegenerative process of PD and treating RBD.
- NT1 narcolepsy type 1 characterized by excessive daytime sleepiness, cataplexy, hypnagogic/hypnopompic hallucinations, sleep paralysis, and disturbed nighttime sleep (Cao et Guilleminault 2017; Siegel 1999; Thorpy 2020).
- Cerebrospinal fluid (CSF) orexin-1 levels are predictive for narcolepsy (abnormal levels in 89.5% of the overall patient population and 94.7% of HLA DQBI*0602 positive cases (Kanbayashi et al. 2002; Nishino 2007).
- NT1 low or absent CSF hypocretin levels serve as a specific biomarker when in narcolepsy type 2 (NT2) or other variants of central of hypersomnolence disorders or hypersomnias (i.e. idiopathic hypersomnia or secondary or recurrent hypersomnia associated with psychiatric diseases), where this specific biomarker is currently absent (Zhang et al. 2018). Since this finding, orexin agonists have attracted attention of potential treatments development for narcolepsy. In 2008, the first OX2R selective agonists classes were filed to be patented, stimulating the discovery of several compounds, but all acting only as OX2R agonists (Bogen et al. 2021 ; Fujimoto et al. 2022; Yanagisawa 2012; Yukitake et al. 2019; Zhang et al. 2021 ).
- TAK-994 and TAK-861 are active compounds and act as a highly selective agonist of the OX2R.
- TAK-994 is >700- fold selectivity over the OX1 R.
- TAK-994 was the first oral selective OX2R agonist developed.
- the international patent application W02022140317 relates to substituted piperidino compounds, particularly, substituted piperidino compounds having agonist activity, once again only OX2R are targeted.
- Parkinson's Disease is the second most common chronic neurodegenerative disease characterized by progressive loss of dopaminergic neurons in the substantia nigra pars compacta (Hornykiewicz et Kish 1987).
- the incidence of PD in the population over 55 years old is about 1% and the cardinal symptoms of PD include resting tremor, bradykinesia, muscle rigidity, postural instability, and usually associatedd with cognitive impairment, mental disorder, and other nonmotor symptoms (Beitz 2014; Meerwaldt et Hovestadt 1988).
- the cause of PD is not fully understood, but several factors including gene mutation, oxidative stress, mitochondrial dysfunction, neurotransmitter toxicity, failure of protein homeostasis appear to be associated with the development of PD.
- the most common treatment for PD is symptom management.
- the dopamine precursor levodopa is the most widely used clinical drug (Hornykiewicz 1975), which could only attenuate the symptoms, but fails to halt the progressive degeneration of dopaminergic neurons in the substantia nigra.
- Orexinergic receptors are located in many brain structures, such as cortex, hippocampus, amygdala, thalamus, hypothalamus, and basal ganglia (Hervieu et al. 2001 ; Hu et al. 2015). It is known that orexins play important roles in the regulation of sleep, feeding behavior, energy homeostasis, neuroendocrine, and autonomic control and the activity of orexinergic system decreases with aging, which has been implicated in many neurodegenerative disorders (Liu et al. 2018).
- orexinergic systems also play an important role in motor control (Berhe, Gebre, et Assefa 2020; Hu et al. 2015; Song et al. 2015; Wang, Cao, et Wu 2021 ).
- Most of the central motor control structures are innervated by orexinergic fibers (Hu et al. 2015; Liu et al. 2018).
- all the basal ganglia nuclei including the globus pallidus, the subthalamic nucleus, the substantia nigra, and the striatum are innervated by orexigenic fibers (Alrouji et al. 2023; Liu et al. 2018).
- 6-hydroxydopamine (6-OHDA)-induced rat model of PD revealed that the number of orexinergic neurons in the lateral hypothalamus decreases significantly (Long-Biao et al. 2010), when the loss of orexinergic neurons in this animal model of PD seems to resemble the process in parkinsonian patients. Furthermore, the orexin levels in plasma and cerebrospinal fluid decrease dramatically in parkinsonian patients (Drouot et al. 2003; Fronczek et al. 2007). These reports implied the important role of orexinergic systems in PD.
- Orexin-A has neuroprotective effects in cellular models of PD. Orexin-A protects SH-SY5Y cells against 6-OHDA (Esmaeili-Mahani et al. 2013; Pasban-Aliabadi, Esmaeili-Mahani, etAbbasnejad 2017) or MPPC (Feng et al. 2014; Liu et al. 2018) induced toxicity.
- orexin-A exerted neuroprotective effects, which may imply orexin-A as a potential therapeutic target for PD and OX1 R agonists as potential target for treatment of PD.
- orexin-A may act as an immunomodulatory regulator of microglia reducing hypothalamic neuron death in the condition of inflammation.
- orexin-A exerts protective effects by attenuating neuroinflammation in AD and cerebral ischemia (Couvineau et Laburthe 2012; Xiong et al. 2013).
- the anti-inflammatory properties may also be involved in the neuroprotective effects of orexin in PD.
- the potential therapeutic effects of orexins on both motor and non-motor disorders in animal models of PD have also been indicated.
- Cerebroventricular administration of orexin-A alleviates sensory motor deficits in a 6- OHDA-treated rat model of PD (Hadadianpour et al. 2017). Similar results were observed in the MPTP-induced mouse model of PD.
- the application of orexin-A into the cerebral ventricles improves motor performance in both pole and open field tests by attenuating the loss of dopaminergic neurons and fibers (Liu et al. 2018).
- Intrapallidal administration of both orexin-A and orexin-B could also alleviate motor deficits in MPTP-treated parkinsonian mice (Ying Wang et al. 2019).
- chemogenetic activation of orexinergic neurons could reverse the abnormal locomotor activity in the pre-clinical stage in A53T mice (Stanojlovic, Pallais, et Kotz 2019).
- cognitive impairments, one of the common non-motor disorders in PD could also be ameliorated by orexins.
- cognitive impairments, one of the common non- motor disorders in PD could also be ameliorated by orexins.
- orexin-A into CA1 or chemogenetic activation of orexinergic neurons increases the firing activity of CA1 neurons (Chen, Chen, et Du 2017) and ameliorates hippocampal- dependent memory impairment in the A53T mouse model of PD (Stanojlovic, Pallais, et Kotz 2019).
- the first subject-matter of the invention relates to a compound of formula (I): wherein: - X represents –NH-, –S- or –O-; - Y and R 2 , independently of each other represents a hydrogen atom, a halogen atom, –NO 2 or –NH 2 ; - R 1 , R 3 , R 4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R 5 , R 6 , R 7 , R 8 , R 9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR 10 or a (C 1 –C 30 )alkyl chain, especially (C 1 –C 20 )alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C ⁇ C–, –C(R 11
- the present invention also relates to a pharmaceutical composition
- a pharmaceutical composition comprising at least one compound of formula (I) as described above and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of neurological diseases, preferably associated with psychiatric and/or sleep disorders and diseases, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
- the first subject-matter of the invention relates to a compound of formula (I): wherein: - X represents –NH-, –S- or –O-; - Y and R 2 , independently of each other represents a hydrogen atom, a halogen atom, –NO 2 or –NH 2 ,; - R 1 , R 3 , R 4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R 5 , R 6 , R 7 , R 8 , R 9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR 10 or a (C 1 –C 30 )alkyl chain, especially (C 1 –C 20 )alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C ⁇ C–,
- halogen atom means fluorine, chlorine, bromine and iodine atoms.
- alkyl group means a saturated, linear or branched hydrocarbon chain.
- (C 1 –C x )alkyl means an alkyl group such as defined above, containing 1 to X carbon atoms.
- (C 1 –C 6 )alkyl means an alkyl group such as defined above, containing 1 to 6 carbon atoms, such as, for example, methyl, ethyl, isopropyl, tert–butyl, pentyl, etc.
- aryl means an aromatic group, especially a hydrocarbon group, especially containing 6 to 20 carbon atoms, preferably 6 to 10 carbon atoms, and comprising one or more fused rings, such as, for example, a phenyl or naphthyl group.
- fused rings such as, for example, a phenyl or naphthyl group.
- heteroaryl means an aromatic group comprising one or more fused rings and comprising 5 to 10 cyclic atoms, including one or more heteroatoms, advantageously 1 to 4 and even more advantageously 1 or 2, such as, for example, sulfur, nitrogen, oxygen, phosphorus or selenium atoms, and preferably sulfur, nitrogen or oxygen, the other cyclic atoms being carbon atoms.
- acyl means a group of formula -CO-R in which R represents a (C 1 –C 6 ) alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl group, and preferably a (C 1 –C 6 ) alkyl or aryl group, and still more preferably a (C 1 –C 6 ) alkyl group.
- cycloalkyl means a saturated mono– or polycyclic hydrocarbon chain (especially a bicyclic or tricyclic chain). When it is a polycyclic group, the rings can be fused, bridged or joined by a spiro ring junction two by two. Examples include cyclopropyl, cyclopentyl, cyclohexyl and cycloheptyl groups.
- heterocyclic group means a non-aromatic, saturated or unsaturated, monocyclic or polycyclic group (comprising fused, bridged or spiro rings) in which one or several atom(s) of the ring(s) carbon atoms each is replaced with heteroatoms, in particular this term means a 5 to 10–membered ring, saturated or unsaturated, but not aromatic, and containing one or more, advantageously 1 to 4, more advantageously 1 or 2 heteroatoms, the heteroatoms being for example, sulfur, nitrogen or oxygen atoms. It can particularly be a pyrrolidinyl, piperidinyl, piperazinyl or morpholinyl group.
- the aryl, heteroaryl and heterocyclic group when substituted, can be substituted with one or more groups chosen from the group consisting of a halogen atom, a (C 1 –C 6 )alkyl group, an aryl group, -NO 2 , –CN, –OR 19 , –SR 20 , –NR 16 R 17 , –B(OH) 2 , –SO 3 R 17 , and –COOR 18 , in particular chosen from the group consisting of a halogen atom, -NO 2 , –CN, –OR 19 , – SR 20 , –NR 16 R 17 , –B(OH) 2 , –SO 3 R 17 , with R 16 to R 20 representing, independently of each other, a hydrogen atom or a (C 1 –C 6 )alkyl group.
- “pharmaceutically acceptable” means what is used in the preparation of a pharmaceutical composition, which is generally safe, nontoxic and not biologically or otherwise undesirable and which is acceptable for both veterinary and human pharmaceutical use.
- “Pharmaceutically–acceptable salts” of a compound mean of salts that are pharmaceutically acceptable, such as defined here, that have the desired pharmacological activity of the parent compound.
- Such salts include: (1) hydrates and solvates, (2) pharmaceutically–acceptable acid addition salts formed with pharmaceutically– acceptable inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or formed with pharmaceutically–acceptable organic acids such as acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, hydroxynaphthoic acid, 2–hydroxy ethanesulfonic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, muconic acid, 2–naphthalene sulfonic acid, propionic acid, salicylic acid, succinic acid, dibenzoyl–L–tartaric acid, tartaric acid, p–tol
- Acceptable organic bases include diethanolamine, ethanolamine, N–methylglucamine, triethanolamine, tromethamine and the like.
- Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate and sodium hydroxide.
- the compounds according to the invention will be in the form of pharmaceutically–acceptable base addition salts, the base being such as NaOH or KOH, and especially NaOH.
- An “optically pure compound” means an enantiomer in an enantiomeric excess of more than 95%, preferably of more than 96%, more preferably of more than 97%, even more preferably of more than 98%, particularly preferably of more than 99%.
- the present invention also includes all pharmaceutically acceptable isotopic variations of a compound of formula I in which one or more atoms is replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
- Such compounds are identical to those disclosed herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
- isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen such as 2 H and 3 H, carbon such as 11 C, 13 C and 14 C, nitrogen such as 13 N and 15 N, oxygen such as 15 O, 17 O and 18 O, sulfur such as 35 S, fluorine such as 18 F, iodine such as 123 I and 125 I, and chlorine such as 36 Cl.
- isotopically-labelled compounds of formula I for example those incorporating a radioactive isotope, are useful in drug and / or substrate tissue distribution studies.
- a subject administered with a compound of the present invention, or a pharmaceutically acceptable salt thereof is generally a mammal, such as a human being, male or female.
- the amount of compound administered to the subject is an amount sufficient to agonize the orexin receptor in the subject.
- the amount of compound can be an “effective amount”, wherein the subject compound is administered in an amount that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
- An effective amount does not necessarily include considerations of toxicity and safety related to the administration of the compound. It is recognized that one skilled in the art may affect neurological and psychiatric disorders associated with orexin receptor activation by treating a subject presently afflicted with the disorders, or by prophylactically treating a subject likely to be afflicted with the disorders, with an effective amount of a compound of the present invention.
- treatment and “treating” refer to all processes wherein there may be a slowing, interrupting, arresting, controlling, or stopping of the progression of the neurological and psychiatric disorders described herein, but does not necessarily indicate a total elimination of all disorder symptoms, as well as the prophylactic therapy of the mentioned conditions, particularly in a subject that is predisposed to such disease or disorder.
- administration of and or “administering a” compound should be understood to mean providing a compound of the invention or a prodrug of a compound of the invention to the subject.
- compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier, i.e.
- the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
- the orexin receptors (OX1R, OX2R) have been implicated in a wide range of biological functions. This has suggested a potential role for these receptors in a variety of disease processes in humans or other species.
- the compounds of the present invention could therefore have utility in treating, preventing, ameliorating, controlling or reducing the risk of a variety of disorders associated with orexin receptors, including one or more of the following conditions or diseases: narcolepsy with or without cataplexy, narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), Gelineau syndrome (Maladie de Gelineau), narcoleptic syndrome, accompanied by narcolepsy-like symptoms, cataplexy in narcolepsy, excessive daytime sleepiness (EDS) in narcolepsy, hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, and insufficient sleep syndrome and any conditions in accordance with The International Classification of Sleep Disorders, Third Edition (ICSD-3) classifies eight different Central Disorders
- neurological, psychiatric, sleep disorders and diseases refers to psychiatric and/or sleep neurological disorders and diseases, i.e. neurological diseases associated with sleep and/or psychiatric disorders.
- the disorders and diseases in which the central orexin neurotransmission is compromised or central and peripheral orexin receptors are involved.
- the present invention may provide methods for treating or controlling: narcolepsy with or without cataplexy, narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), Gelineau syndrome (Maladie de Gélineau), narcoleptic syndrome, accompanied by narcolepsy-like symptoms, cataplexy in narcolepsy, excessive daytime sleepiness (EDS) in narcolepsy, hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, and insufficient sleep syndrome and any conditions in accordance with The International Classification of Sleep Disorders, Third Edition (ICSD-3) classifies eight different Central Disorders of Hypersomnolence (CDH)(American Academy of Sleep Medicine 2014) ; pathologic daytime sleepiness and/or unappropriated
- the dihydro-quinazoline, -benzothiazine and benzoxazine derivatives as compounds of the present invention may also potentially have utility in treating, preventing, ameliorating, controlling or reducing the risk of a variety of other disorders associated with orexin receptors, including one or more of the following conditions or diseases including enhancing sleep quality, improving sleep quality, increasing sleep efficiency, consolidating sleep maintenance, improving sleep initiation, decreasing sleep latency or onset, decreasing difficulties in falling asleep, increasing sleep continuity, decreasing the number of awakenings during sleep, decreasing intermittent waking during sleep, decreasing nocturnal arousals, decreasing the time spent awake following the initial onset of sleep, increasing the total amount of sleep, reducing the fragmentation of sleep, altering the timing, frequency or duration of sleep stages, or duration of slow-wave sleep and / or REM sleep, promoting slow wave sleep, enhancing EEG - delta activity during sleep, decreasing nocturnal arousals, especially early morning awakenings, increasing daytime alertness, reducing daytime drowsiness, treating or reducing
- a preferred embodiment of the invention is a compound of formula (I) or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, Parkinson's disease and other synucleinopathies, preferably narcolepsy type 1 (NT1), narcolepsy type 2 (NT2) and Parkinson's disease.
- NT1 narcolepsy type 1
- NT2 narcolepsy type 2
- Parkinson's disease preferably narcolepsy type 1 (NT1), narcolepsy type 2 (NT2) and Parkinson's disease
- a preferred embodiment of the invention is a compound of formula (I) or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of REM sleep behavior disorder (RBD).
- RBD REM sleep behavior disorder
- the neurological diseases are selected from the group consisting of narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, Parkinson's disease and other synucleinopathies, preferably narcolepsy type 1 (NT1), narcolepsy type 2 (NT2) and Parkinson's disease.
- NT1 narcolepsy type 1
- NT2 narcolepsy type 2
- Parkinson's disease preferably narcolepsy type 1 (NT1), narcolepsy type 2 (NT2) and Parkinson's disease.
- the neurological diseases preferably the neurological disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved, are selected from the group consisting of narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia and recurrent hypersomnia.
- the neurological disorders targeted are those requiring an agonist of the orexin 1 receptor (OX1R) and/or the orexin 2 receptor (OX2R), preferably both receptors.
- R 2 when one of Y or R 2 represents a halogen atom, –NO 2 or – NH 2 , preferably –NO 2 or –NH 2 , the other represents a hydrogen atom. In some embodiments, when Y represents a halogen atom, –NO 2 or –NH 2 , preferably –NO 2 or – NH 2 , R 2 represents a hydrogen atom. In some embodiments, when R 2 represents a halogen atom, –NO 2 or –NH 2 , preferably –NO 2 or –NH 2 , Y represents a hydrogen atom. R 4 represents preferably a hydrogen atom.
- R 1 , R 2 , R 3 , R 4 each represent a hydrogen atom or R 1 , R 3 , R 4 , Y each represent a hydrogen atom.
- R 1 , R 2 , R 3 , R 4 can each represent a hydrogen atom.
- R 1 , R 3 , R 4 , Y each represent a hydrogen atom.
- R 5 , R 6 , R 7 , R 8 , R 9 each represent, independently of each other, a hydrogen atom or –OR 10 .
- at least four residues out of R 5 , R 6 , R 7 , R 8 and R 9 each represent a hydrogen atom.
- R 5 , R 6 , R 7 , R 8 , R 9 each represent, a hydrogen atom.
- R 5 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group.
- R 5 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 – C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group
- R 6 , R 7 , R 8 , R 9 each represent, a hydrogen atom
- R 6 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group.
- R 6 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 – C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group
- R 5 , R 7 , R 8 , R 9 each represent, a hydrogen atom
- R 7 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group.
- R 7 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 – C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group
- R 5 , R 6 , R 8 , R 9 each represent, a hydrogen atom
- one residue out of R 5 , R 6 , R 7 , R 8 and R 9 represents -OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 – C 20 )alkyl or a phenyl group.
- X can represent -NH-. In some embodiments, X can represent - S-. In some embodiments, X can represent -O-. Preferably, X represents –NH- or –S-.
- the compound of formula (I) is selected from the group consisting of:
- the compound of formula (I) is selected from the group consisting of:
- the compound of formula (I) is selected from the group consisting of: , and mixtures thereof.
- the neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which orexin receptors are involved is selected from the group consisting of narcolepsy type 1, narcolepsy type 2, idiopathic hypersomnia, recurrent hypersomnia, attention- deficit hyperactivity disorder, anxiety and mood disorders, Alzheimer's disease or any other neurodegenerative disorders or cognitive impairment and tauopathies, Parkinson's disease and other synucleinopathies, Guillain-Barre syndrome, chronic fatigue syndrome, long COVID-19 and medical or health conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, restless legs syndrome, fibromyalgia, cardiac failure, diseases related to bone loss, sepsis, syndromes which are manifested by unrefresh
- the dosage of active ingredient in the compositions of this invention may be varied, however, it is necessary that the amount of the active ingredient be such that a suitable dosage form is obtained.
- the active ingredient may be administered to subjects (animals and human) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy.
- the selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment.
- the dose will vary from subject to subject depending upon the nature and severity of disease, the subject's weight, special diets then being followed by a subject, concurrent medication, and other factors which those skilled in the art will recognize.
- dosage levels of between 0.0001 to 100 mg/kg of body weight per day are administered to the subject, e.g., humans, adolescent humans and elderly humans, to obtain effective agonism of orexin receptors.
- a therapeutic dose of a compound of formula (I) comprised between 0.1 mg/kg/day and 100 mg/kg/day is administrated to a patient in need thereof.
- the present invention also relates to a pharmaceutical composition comprising at least one compound of formula (I) as described above and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
- the dosage range will generally be from 0.5 mg to 10.0 g per subject per day which may be administered in single or multiple doses. In one embodiment, the dosage range will be from 0.5 mg to 500 mg per subject per day, preferably from 0.5 mg to 200 mg per subject per day, and more preferably from 5 mg to 50 mg per subject per day.
- Pharmaceutical compositions of the present invention may be provided in a solid dosage formulation such as comprising from 0.5 mg to 500 mg of active ingredient, or comprising from 1 mg to 250 mg of active ingredient.
- the pharmaceutical composition may be provided in a solid dosage formulation comprising 1 mg, 5 mg, 10 mg, 50 mg, 80 mg, 100 mg, 200 mg active ingredient.
- the pharmaceutical composition for use according to the invention comprises between 0.5 mg to 800 mg, preferably between 20 mg to 400 mg of the compound of formula (I).
- the compositions may be provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, such as 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated.
- the compounds may be administered on a regimen of 1 to 4 times per day, such as once or twice per day.
- the compounds may be administered once or multiple times during the day.
- the compounds may be administered upon awakening or otherwise in the morning, or during waking hours.
- the compounds may be administered 25 mg, 30 mg or 250 mg, 1 hour after awakening, 30 minutes after awakening or immediately after awakening.
- the pharmaceutical composition according to the invention is suitable for oral or parenteral administration.
- the pharmaceutical composition according to the invention is in the form of a solution, such as an injectable solution, or a tablet or a capsule or a transdermal delivery system.
- the compounds of the present invention may be used in combination with one or more other drugs in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which compounds of the present invention or the other drugs may have utility, where the combination of the drugs together are safer or more effective than either drug alone.
- Such other drug(s) may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention.
- a pharmaceutical composition in unit dosage form containing such other drugs and the compound of the present invention is contemplated.
- the combination therapy may also include therapies in which the compound of the present invention and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, the compounds of the present invention and the other active ingredients may be used in lower doses than when each is used singly.
- the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to a compound of the present invention.
- the above combinations include combinations of a compound of the present invention not only with one other active compound, but also with two or more other active compounds.
- the weight ratio of the compound of the present invention to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used.
- the weight ratio of the compound of the present invention to the other agent will generally range from 1000:1 to 1:1000, preferably from 200:1 to 1:200.
- Combinations of a compound of the present invention and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. In such combinations the compound of the present invention and other active agents may be administered separately or in conjunction. In addition, the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent.
- the compounds of the present invention may be administered in combination with compounds which are known in the art to be useful for treating or controlling narcolepsy, including e.g., methylphenidate, amphetamine, atomoxetine, reboxetine, viloxazine, phenelzine, protriptyline, gamma–hydroxybutyric acid, sodium oxybate, or other oxybate salts, modafinil, armodafinil, adrafinil, pitolisant, samelisant, nalfurafin, caffeine, and salts thereof, and combinations thereof, and the like.
- compounds which are known in the art to be useful for treating or controlling narcolepsy including e.g., methylphenidate, amphetamine, atomoxetine, reboxetine, viloxazine, phenelzine, protriptyline, gamma–hydroxybutyric acid, sodium oxybate, or other oxybate salt
- the present invention also relates to a compound of formula (II): wherein: - X represents -NH-, –S- or –O-, preferably -NH- or -S-; - Y and R 2 , independently of each other represents a hydrogen atom, a halogen atom, –NO 2 or –NH 2 ,; - R 1 , R 3 , R 4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R 5 , R 6 , R 7 , R 8 , R 9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR 10 or a (C 1 –C 30 )alkyl chain, especially (C 1 –C 20 )alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, hetero
- the compound of formula (II) is not (this compound has been described (Cox et al. 2020) as having an inhibitory activity of amyloid aggregation), (these four compounds has been described as having an inhibitory effect on cell multiplication (Yale et Kalkstein 1967), (this compound has been described as having an anticancer activity (Chinigo et al. 2008), (this compound has been described as having an anti-tumour activity (Mordarski et Chylinska 1971), (this compound has been used for the treatment of PARP-1 related diseases, see WO 2018044136) (Kim et al.
- each of R 1 , R 2 , R 3 and R 4 represents H
- Y represents an halogen atom
- four of R 5 , R 6 , R 7 , R 8 , and R 9 represent H
- Y and R 2 independently of each other represents a hydrogen atom, –NO 2 or –NH 2 .
- one of Y or R 2 represents a halogen atom, –NO 2 or – NH 2 , preferably –NO 2 or –NH 2
- the other represents a hydrogen atom or a halogen atom.
- R 2 represents a hydrogen atom or a halogen atom.
- R 2 when R 2 represents a halogen atom, –NO 2 or –NH 2 , preferably –NO 2 or –NH 2 , Y represents a hydrogen atom or a halogen atom.
- Y or R 2 when one of Y or R 2 represents a halogen atom, –NO 2 or – NH 2 , preferably –NO 2 or –NH 2 , the other represents a hydrogen atom.
- R 2 when Y represents a halogen atom, –NO 2 or –NH 2 , preferably –NO 2 or – NH 2 , R 2 represents a hydrogen atom.
- R 2 represents a halogen atom, –NO 2 or –NH 2 , preferably –NO 2 or –NH 2
- Y represents a hydrogen atom.
- R 4 represents preferably a hydrogen atom.
- R 5 , R 6 or R 7 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or a phenyl group.
- R 1 , R 2 , R 3 , R 4 each represent a hydrogen atom or R 1 , R 3 , R 4 , Y each represent a hydrogen atom.
- R 1 , R 2 , R 3 , R 4 can each represent a hydrogen atom.
- R 1 , R 3 , R 4 , Y each represent a hydrogen atom.
- R 5 , R 6 , R 7 , R 8 , R 9 each represent, independently of each other, a hydrogen atom or –OR 10 .
- at least four residues out of R 5 , R 6 , R 7 , R 8 and R 9 each represent a hydrogen atom.
- R 5 , R 6 , R 7 , R 8 , R 9 each represent, a hydrogen atom.
- R 5 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group.
- R 5 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 – C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group
- R 6 , R 7 , R 8 , R 9 each represent, a hydrogen atom
- R 6 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group.
- R 6 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 – C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group
- R 5 , R 7 , R 8 , R 9 each represent, a hydrogen atom
- R 7 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group.
- R 7 represents –OR 10 with R 10 representing a hydrogen atom, a (C 1 – C 30 )alkyl or phenyl group, preferably a (C 1 –C 20 )alkyl or a phenyl group
- R 5 , R 6 , R 8 , R 9 each represent, a hydrogen atom
- one residue out of R 5 , R 6 , R 7 , R 8 and R 9 represents -OR 10 with R 10 representing a hydrogen atom, a (C 1 –C 30 )alkyl or phenyl group, preferably a (C 1 – C 20 )alkyl or a phenyl group.
- X can represent -NH-. In some embodiments, X can represent - S-. In some embodiments, X can represent -O-. Preferably, X represents –S-.
- the compound of formula (II) is selected from the group consisting of:
- the compound of formula (II) is selected from the group consisting of compounds 99, 100, 145, 146, 148, 149, 151, 152, 153, 154, 155, 157, 158, 161, 167, 182, 185, 191, 194, 195, 196, 243, 244, 292, 293, 301, 302, 304, 305, 308, 310, 311, and pharmaceutically acceptable salts thereof.
- the compound of formula (II) is selected from the group consisting of compounds 99, 148, 152, 153, 154, 155, 157, 161, 167, 191, 194, 195, 196, 301, 302, 304, 305, 308, 310, 311, and pharmaceutically acceptable salts thereof.
- the compound of formula (II) is selected from the group consisting of: , and pharmaceutically acceptable salts thereof.
- the compound of formula (II) is selected from the group consisting of: , , and pharmaceutically acceptable salts thereof.
- the present invention also relates to a pharmaceutical composition comprising a compound of formula (II) and a pharmaceutically acceptable carrier.
- the pharmaceutical composition is as defined above for compound of formula (I).
- the present invention also relates to a compound of formula (II) as described above for use as a medicament.
- the present invention also relates to a compound of formula (II) as described above for use in the manufacture of a medicament.
- the present invention provides a method for treating a subject in need thereof comprising administering to said subject a compound of formula (II) or a composition comprising a compound of formula (II).
- the chromatographic separations of may be achieved as known in the art. Their absolute stereochemistry may be determined by the X-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration.
- racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated.
- the separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.
- the coupling reaction is often the formation of salts using an enantiomerically pure acid or base.
- the diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue.
- the racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art.
- any enantiomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
- the present invention provides a method for treating a subject suffering from neurological diseases, preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved, comprising administering to said subject a compound of formula (I) or a composition comprising a compound of formula (I).
- neurological diseases preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
- said disorders and diseases are as defined above.
- the present invention provides a method for preventing neurological diseases, preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved, comprising administering to a subject an effective amount of a compound of formula (I) or a composition comprising an effective amount of a compound of formula (I).
- neurological diseases preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease
- central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved
- the methods further comprise administration of a second active compound.
- the invention is also related to compounds of formula (I) or pharmaceutical compositions comprising at least one compound of formula (I) for their use in the manufacture of a medicament for the treatment of from neurological diseases, preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
- neurological diseases preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
- said disorders and diseases are as defined above.
- the present invention also relates to the following embodiments: Embodiment 1.
- Embodiment 2 The compound of formula (I) for use according to embodiment 1, characterized in that R 1 , R 2 , R 3 , R 4 each represent a hydrogen atom.
- Embodiment 3 The compound of formula (I) for use according to embodiment 1 or 2, characterized in that R 5 , R 6 , R 7 , R 8 , R 9 , each represent, independently of each other, a hydrogen atom or –OR 10 .
- Embodiment 4. The compound of formula (I) for use according to any one of embodiments 1 to 3, characterized in that at least four residues out of R 5 , R 6 , R 7 , R 8 and R 9 each represent a hydrogen atom.
- Embodiment 9 A pharmaceutical composition comprising at least one compound of formula (I) as defined in any one of embodiments 1 to 8 and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
- Embodiment 10 The pharmaceutical composition for use according to embodiment 9, comprising between 0.5 mg to 800 mg, preferably between 20 mg to 400 mg of the compound of formula (I).
- Embodiment 11 The pharmaceutical composition for use according to embodiment 9 or 10, which is suitable for oral or parenteral administration, preferably in the form of a solution, such as an injectable solution, a tablet, a capsule or a transdermal delivery system.
- Embodiment 13 The compound of formula (II) according to embodiment 12, characterized in that R 1 , R 2 , R 3 , R 4 each represent a hydrogen atom and R 5 , R 6 , R 7 , R 8 , R 9 each represent, independently of each other, a hydrogen atom or –OR 10 .
- Embodiment 14 The compound of formula (II) according to embodiments 12 or 13, characterized in that at least four residues out of R 5 , R 6 , R 7 , R 8 and R 9 each represent a hydrogen atom.
- Embodiment 15 The compound of formula (II) according to embodiment 14, characterized in that R 5 , R 6 , R 7 , R 8 , R 9 , each represent, a hydrogen atom.
- Embodiment 17 The compound of formula (II) according to any one of embodiments 12 to 16, characterized in that it is selected from the group consisting of compounds 99, 148, 152, 153 , 154 , 155, 157, 161 , 167 , 191 , 194, 195 , 196 , and mixtures thereof, preferably 99 , 152 , 155 , 194 , and mixtures thereof.
- a compound of formula (II) according to any of embodiments 12 to 17, for use as a medicament.
- One or more features of any embodiments disclosed herein may be combined and/or rearranged within the scope of the invention to produce further embodiments that are also within the scope of the invention.
- Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be within the scope of the present invention.
- the invention is further described by the following non-limiting Examples. Examples Examples are provided below to facilitate a more complete understanding of the invention. The following examples illustrate the exemplary modes of making and practicing the invention.
- Example 1 Chemistry of dihydro-quinazoline derivatives Design, synthesis and preparations of 6-nitro-2,3-dihydro-1H-quinazolin-4-one Polyphosphoric ester preparation (PPE) P2O4 (150 g) was refluxed in diethyl ether (150 mL) and CHCl3 (300 mL) until a clear solution occurred. Then solvents were removed under reduce pressure to give a clear oil.
- PPE Polyphosphoric ester preparation
- Binding activities on OX1R and OX2R sites These compounds express an activity on OX1R and/or OX2R as agonist which is determined in accordance with the following general experimental method by Eurofins using Chinese hamster ovary (CHO) cells expressing human OX1R, and human embryonic kidney (HEK), 293 cells expressing human OX2R.
- CHO Chinese hamster ovary
- HEK human embryonic kidney
- OX1 (agonist radioligand) human recombinant (CHO cells); Ligand [125I]orexin A; Ligand concentration: 0.1 nM ; Ligand Kd: 0.87 nM; Non-specific: SB 334867 (1 ⁇ M); Incubation: 60 min at RT scintillation counting; Control inhibitor: Orexin-A; Test concentration/dose: IC/EC 50 calculation are provided if 5 or more concentrations are selected; Test sample requirements: Minimum for 1) Screen: 60 ⁇ l of 10 mM stock -OR- 1 mg (pre-weighed) for 10 ⁇ M final testing.
- Results showing an inhibition (or stimulation) between 25% and 50% are indicative of weak to moderate effects.
- Results showing an inhibition (or stimulation) lower than 25% are not considered significant and mostly attributable to variability of the signal around the control level.
- Results showing an inhibition (or stimulation for assays run in basal conditions) higher than 50% are considered to represent significant effects of the test compounds.
- Low to moderate negative values have no real meaning and are attributable to variability of the signal around the control level.
- High negative values ( ⁇ 50%) that are sometimes obtained with high concentrations of test compounds are generally attributable to nonspecific effects of the test compounds in the assays. On rare occasion they could suggest an allosteric effect of the test compound. Table 2.
- Binding activities on dopamine and norepinephrine transporters, metabotropic glutamate 2, cathepsin-H and sigma-1 receptors It has been reported for a long time ago that norepinephrine cells of the locus coeruleus (Hagan et al.1999; Horvath et al.1999) and dopaminergic cells of the Ventral tegmental area (Nakamura et al.2000) all show to increase their firing rates by orexins. The firing rates of these monoaminergic neurons are well known to be associated with sleep/wakefulness states.
- OX2R could act pre- synaptically at the level of both dopaminergic and glutamatergic axons (Bandarabadi et al. 2022).
- Product 152 at 10 -5 M significantly targets on OX2R as agonist (IC 73%) and on OX1R as agonist (IC 63%) in addition acting on metabotropic glutamate 2 (mGlu2) receptors which target for the treatment of psychiatric disorders including schizophrenia, depression, and anxiety, which are characterized by a glutamatergic dysfunction with a significantly binding effect (IC 54%).
- mGlu2 metabotropic glutamate 2
- mGluR 2 antagonism activity is associated with enhanced theta/gamma oscillations and increased transitions from sleep to waking state (Ahnaou, Ver Donck, et Drinkenburg 2014).
- Product 194 at 10 -5 M significantly targets on OX2R as agonist (IC 84%) and weakly on OX1R as agonist (IC ⁇ 50%), acting on dopamine transporter (DAT) and norepinephrine transporter (NET) as a catecholaminergic reuptake inhibitor, with a binding effect on DAT (IC 89%) and NET (IC 58%).
- Therapeutic approaches targeting cathepsins can contribute to prevent or slow down the pathogenesis of neurodegenerative diseases as seen for neuronal ceroid lipofuscinosis, synucleinopathies (Parkinson's disease, Dementia with Lewy Body and Multiple System Atrophy) as well as Alzheimer's and Huntington's disease (Stoka et al. 2023).
- CTSH Cathepsin H
- narcolepsy is a lysosomal cysteine protease that plays a role in various physiological processes, including the immune response.
- CTSH has been implicated in modulating immune reactions within the central nervous system (Wang et al. 2023).
- Neuroinflammation refers to the inflammatory response in the nervous tissue, often involving glial cell activation and the release of pro-inflammatory molecules.
- CTSH can influence neuroinflammation by participating in the processing and presentation of antigens, which is crucial for the activation of immune cells such as microglia.
- Previous studies had also suggested a potential involvement of CTSH in the pathogenesis of narcolepsy (Mogavero et al. 2023).
- Product 96 at 10 -5 M targets on OX1R as agonist (IC 56%) and moreover on CTSH as antagonist (IC 80%), also acting on dopamine transporter (DAT) and norepinephrine transporter (NET) as a catecholaminergic reuptake inhibitor, with a binding effect on DAT (IC 99%) and NET (IC 77%).
- Sigma-1 receptor (S1R) participating in various physiological and pathological processes, such as neurotransmission, neuroprotection and neuroinflammation is considered as a therapeutic target for a range of neurodegenerative diseases, including amnesia and AD and also various synucleinopathies (Wang et Jia 2023).
- S1R agonists find to have multiple mechanisms of action that could be beneficial in AD, such as anti-inflammatory and antioxidant effects, modulation of neurotransmitters, and a neuroprotective effect by inhibiting A ⁇ aggregation and tau hyperphosphorylation is AD (Cummings, Osse, et Kinney 2023; Malar et al. 2023; Shinoda, Nemoto, et Iwamoto 2023).
- Product 90 at 10 -5 M targets on OX2R as agonist (IC 67%) and on S1R as antagonist (IC 75%).
- Orexin-A protects human neuroblastoma SH-SY5Y cells against 6-hydroxydopamine- induced neurotoxicity: involvement of PKC and PI3K signaling pathways (2012) Rejuvenation Research 20(2): 125-33. Perrey, David A, et Yanan Zhang. 2020. « Therapeutics Development for Addiction: Orexin-1 Receptor Antagonists republic Brain research 1731: 145922-145922. Polito, Rita et al. 2018.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- Psychiatry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biomedical Technology (AREA)
- Neurology (AREA)
- Neurosurgery (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention is directed to dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives for use in the prevention or treatment of neurological, psychiatric, sleep disorders and diseases, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. The present invention is also directed to pharmaceutical compositions comprising these compounds for use in the prevention and/or treatment of neurological disorders and diseases. The present invention is also directed to dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and the use of these compounds as a medicament.
Description
Dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and use thereof as orexin receptors agonists for treating or preventing neurological diseases
Field of the invention
The present invention is directed to dihydro-quinazoline, -benzothiazine and - benzoxazine derivatives for use in the prevention or treatment of neurological, psychiatric, sleep disorders and diseases, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. The present invention is also directed to pharmaceutical compositions comprising these compounds for use in the prevention and/or treatment of neurological disorders and diseases. The present invention is also directed to dihydrobenzothiazine and dihydro-benzoxazine derivatives and the use of these compounds as a medicament.
Background of the present invention
Orexin 1 and 2 (0X1 and 0X2, also known as hypocretin 1 and 2 or orexin A and B) are hypothalamic neuropeptides specifically produced in the lateral hypothalamic area (Sakurai et al. 1998). Orexins act on the two G protein-coupled receptors (GPCRs), the orexin 1 receptor (0X1 R) and the orexin 2 receptor (0X2R), participating in a broad range of physiological functions such as sleep/wakefulness (Gao et al. 2021 ; de Lecea et al. 1998; Lin et al. 1999; Ohno et Sakurai 2008), feeding behavior (Sakurai et al. 1998), reward -seeking (Cason et al. 2010; Harris, Wimmer, et Aston -Jones 2005), and stress responses (Johnson et al. 2010). More especially, OX1 R is mainly involved in motivation and reward and the OX2R in the modulation of sleep/wake cycle and energy homeostasis (Perrey et Zhang 2020).
In genetic studies in mice, the lack of orexin function causes narcoleptic symptoms characterized by excessive daytime sleepiness and cataplexy (Lin et al. 1999; Willie et al. 2003), and intracerebroventricular administration of orexins attenuates the narcoleptic symptoms (Mieda 2017). Besides, OX1 R knockout mice exhibit no obvious sleep/wakefulness-related phenotype but OX2R knockout mice show severe narcoleptic phenotype, suggesting that the 0X2R-mediated signaling would be sufficient to prevent the symptoms of narcolepsy/cataplexy (Mieda et al. 2011 ; Saito et al. 2018).
Increasingly for the last decade, a large number of orexin antagonists have been
developed as potential drugs for various pathophysiological conditions involving the orexin system (e.g. for the treatment of insomnia) (Heifetz et al. 2013) but far fewer have been the orexin agonists when they have potential for the treatment of various diseases (Nollet et Leman 2013; Song et al. 2015), including obesity, attention-deficit hyperactivity disorder (ADHD) (Baimel et al. 2015; Cortese, Konofal, et Lecendreux 2008; Heifetz et al. 2013), neurodegenerative diseases (e.g. Alzheimer, Parkinson) (Gao et al. 2021 ; Katsuki et Michinaga 2012), and above all narcolepsy type 1 and 2 (Fujimoto et al. 2022; Mezeiova et al. 2020; Yukitake et al. 2019; Zhang et al. 2021 ).
Using orexin antagonists in animal models, the role of 0X1 R on sleep regulation has been studied. SB-334867 (an experimental 0X1 R selective antagonist) has been reported reversing the REM sleep suppression induced by intracerebroventricular injection of orexin-A (Smith et al. 2003) and above all induces effects on REM sleep and induces Non-REM sleep when administered alone (Morairty et al. 2012).
In contrary to 0X1 R antagonists, a potential therapeutic role of 0X1 R agonists in REM sleep behavior disorder (RBD) is hypothesized due to the interaction between o- synuclein and 0X1 R in pathogenesis. 0X1 R dysfunction can induce the occurrence of RBD and is a potent early sign of Parkinson's disease (PD), but whether the pathogenetic mechanism involved in RBD remains unexplored. In contrast, o-synuclein has been verified to form Lewy bodies in orexin neurons, whose activity and function depend on orexin receptor 1 (0X1 R) (Fan et al. 2023).
0X1 R agonists may be a useful strategy to slow or stop the neurodegenerative process of PD and treating RBD.
Loss of orexinergic neurons in the brain is associated with the cause of narcolepsy type 1 (NT1 ) characterized by excessive daytime sleepiness, cataplexy, hypnagogic/hypnopompic hallucinations, sleep paralysis, and disturbed nighttime sleep (Cao et Guilleminault 2017; Siegel 1999; Thorpy 2020).
Cerebrospinal fluid (CSF) orexin-1 levels are predictive for narcolepsy (abnormal levels in 89.5% of the overall patient population and 94.7% of HLA DQBI*0602 positive cases (Kanbayashi et al. 2002; Nishino 2007).
In NT1 , low or absent CSF hypocretin levels serve as a specific biomarker when in narcolepsy type 2 (NT2) or other variants of central of hypersomnolence disorders or hypersomnias (i.e. idiopathic hypersomnia or secondary or recurrent hypersomnia associated with psychiatric diseases), where this specific biomarker is currently absent (Zhang et al. 2018).
Since this finding, orexin agonists have attracted attention of potential treatments development for narcolepsy. In 2008, the first OX2R selective agonists classes were filed to be patented, stimulating the discovery of several compounds, but all acting only as OX2R agonists (Bogen et al. 2021 ; Fujimoto et al. 2022; Yanagisawa 2012; Yukitake et al. 2019; Zhang et al. 2021 ).
However, it is considered that these compounds are not satisfactory, for example, in terms of activity, pharmacokinetics, permeability into the brain/central nervous system or safety, and the development of an improved compound having OX2R agonist activity is desired.
Sunti norexton, firazorexton and danavorexton are selective OX2R which are under development by Takeda for the treatment of narcolepsy. TAK-994 and TAK-861 are active compounds and act as a highly selective agonist of the OX2R. TAK-994 is >700- fold selectivity over the OX1 R. Related to danavorexton (TAK-925), it reached phase 2 clinical trials for narcolepsy. However, clinical development was discontinued in 2021 for safety reasons (Dauvilliers et al. 2023; Ishikawa et al. 2023). TAK-994, was the first oral selective OX2R agonist developed.
Recently, cycloalkyl urea compounds targeting OX2R as an agonist action have been reported in W02021107023, but these compounds failed to target both OX1 R and OX2R.
The international patent application W02022140317 relates to substituted piperidino compounds, particularly, substituted piperidino compounds having agonist activity, once again only OX2R are targeted.
Parkinson's Disease (PD) is the second most common chronic neurodegenerative disease characterized by progressive loss of dopaminergic neurons in the substantia nigra pars compacta (Hornykiewicz et Kish 1987). The incidence of PD in the population over 55 years old is about 1% and the cardinal symptoms of PD include resting tremor, bradykinesia, muscle rigidity, postural instability, and usually companied with cognitive impairment, mental disorder, and other nonmotor symptoms (Beitz 2014; Meerwaldt et Hovestadt 1988). The cause of PD is not fully understood, but several factors including gene mutation, oxidative stress, mitochondrial dysfunction, neurotransmitter toxicity, failure of protein homeostasis appear to be associated with the development of PD. The most common treatment for PD is symptom management. The dopamine precursor levodopa is the most widely used clinical drug (Hornykiewicz 1975), which could only attenuate the symptoms, but fails to halt the progressive degeneration of dopaminergic neurons in the substantia nigra. In recent years, many efforts were devoted to find endogenous neuroprotective mediators to stop or reverse
the degenerative changes of dopaminergic neurons in the substantia nigra.
Orexinergic receptors are located in many brain structures, such as cortex, hippocampus, amygdala, thalamus, hypothalamus, and basal ganglia (Hervieu et al. 2001 ; Hu et al. 2015). It is known that orexins play important roles in the regulation of sleep, feeding behavior, energy homeostasis, neuroendocrine, and autonomic control and the activity of orexinergic system decreases with aging, which has been implicated in many neurodegenerative disorders (Liu et al. 2018).
In addition, orexinergic systems also play an important role in motor control (Berhe, Gebre, et Assefa 2020; Hu et al. 2015; Song et al. 2015; Wang, Cao, et Wu 2021 ). Most of the central motor control structures are innervated by orexinergic fibers (Hu et al. 2015; Liu et al. 2018). More importantly, all the basal ganglia nuclei, including the globus pallidus, the subthalamic nucleus, the substantia nigra, and the striatum are innervated by orexigenic fibers (Alrouji et al. 2023; Liu et al. 2018).
From approximatively two decades, increasingly numerous studies demonstrated that orexinergic systems are closely correlated with PD (Katsuki et Michinaga 2012; Yasui et al. 2006). It was reported that parkinsonian patients display significant loss of orexinergic neurons in postmortem exams (Fronczek et al. 2007; Thannickal, Lai, et Siegel 2007).
Experiments with 6-hydroxydopamine (6-OHDA)-induced rat model of PD revealed that the number of orexinergic neurons in the lateral hypothalamus decreases significantly (Long-Biao et al. 2010), when the loss of orexinergic neurons in this animal model of PD seems to resemble the process in parkinsonian patients. Furthermore, the orexin levels in plasma and cerebrospinal fluid decrease dramatically in parkinsonian patients (Drouot et al. 2003; Fronczek et al. 2007). These reports implied the important role of orexinergic systems in PD.
Other studies revealed that orexin-A has neuroprotective effects in cellular models of PD. Orexin-A protects SH-SY5Y cells against 6-OHDA (Esmaeili-Mahani et al. 2013; Pasban-Aliabadi, Esmaeili-Mahani, etAbbasnejad 2017) or MPPC (Feng et al. 2014; Liu et al. 2018) induced toxicity.
Finally, the exact role of orexin-A in the animal models of PD, and the protective mechanisms of orexin-A on the nigral dopaminergic neurons seems demonstrated using MPTP parkinsonian mice (Liu et al. 2018). In this animal model, orexin-A exerted neuroprotective effects, which may imply orexin-A as a potential therapeutic target for PD and OX1 R agonists as potential target for treatment of PD.
Another involvement of orexin was also recognized, this in the immune response and neuroinflammation (Duffy et al. 2019; Polito et al. 2018) suggesting that orexin-A may act as an immunomodulatory regulator of microglia reducing hypothalamic neuron
death in the condition of inflammation.
Furthermore, orexin-A exerts protective effects by attenuating neuroinflammation in AD and cerebral ischemia (Couvineau et Laburthe 2012; Xiong et al. 2013). Thus, the anti-inflammatory properties may also be involved in the neuroprotective effects of orexin in PD. The potential therapeutic effects of orexins on both motor and non-motor disorders in animal models of PD have also been indicated.
Cerebroventricular administration of orexin-A alleviates sensory motor deficits in a 6- OHDA-treated rat model of PD (Hadadianpour et al. 2017). Similar results were observed in the MPTP-induced mouse model of PD. The application of orexin-A into the cerebral ventricles improves motor performance in both pole and open field tests by attenuating the loss of dopaminergic neurons and fibers (Liu et al. 2018). Intrapallidal administration of both orexin-A and orexin-B could also alleviate motor deficits in MPTP-treated parkinsonian mice (Ying Wang et al. 2019). Furthermore, chemogenetic activation of orexinergic neurons could reverse the abnormal locomotor activity in the pre-clinical stage in A53T mice (Stanojlovic, Pallais, et Kotz 2019). In addition, cognitive impairments, one of the common non-motor disorders in PD, could also be ameliorated by orexins. In addition, cognitive impairments, one of the common non- motor disorders in PD, could also be ameliorated by orexins. The administration of orexin-A into CA1 or chemogenetic activation of orexinergic neurons increases the firing activity of CA1 neurons (Chen, Chen, et Du 2017) and ameliorates hippocampal- dependent memory impairment in the A53T mouse model of PD (Stanojlovic, Pallais, et Kotz 2019).
Furthermore, sociability and social memory impairments observed in A53T parkinsonian mice are also improved by chemogenetic activation of orexinergic neurons (Stanojlovic et al. 2019). Moreover, the reduced anxiety-like behavior observed in the early stage of PD could be restored by chemogenetic activation of orexinergic neurons in five-month-old A53T mice (Stanojlovic, Pallais, et Kotz 2019). The therapeutic effect of orexin on motor disorders in PD is probably associated with the restoration of dopaminergic neurons and the excitatory effects of orexin on pallidal neurons (Liu et al. 2018; Wang et al. 2019). However, the mechanisms of the therapeutic effects of orexins on non-motor disorders have not been well studied thus far.
Summary of the present invention
The first subject-matter of the invention relates to a compound of formula (I):
wherein: - X represents –NH-, –S- or –O-; - Y and R2, independently of each other represents a hydrogen atom, a halogen atom, –NO2 or –NH2; - R1, R3, R4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C30)alkyl chain, especially (C1–C20)alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, –C(R11)=C(R12)–, -O-, –S–, –NR13–, –C(O)–, –C(S)–, –C=N–, –N=C–, –OC(O)–, – C(O)O–, –SC(O)–, –C(O)S–, –N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R10 represents a hydrogen atom or a (C1–C30)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R11 and R12 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl group; and - R13 to R15 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, and preferably a hydrogen atom or a (C1–C6)alkyl or aryl group, and still more preferably a hydrogen atom or a (C1–C6)alkyl group, or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of neurological diseases, preferably associated with psychiatric and/or sleep disorders and diseases,
advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. The present invention also relates to a pharmaceutical composition comprising at least one compound of formula (I) as described above and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of neurological diseases, preferably associated with psychiatric and/or sleep disorders and diseases, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. Detailed description of the present invention The first subject-matter of the invention relates to a compound of formula (I):
wherein: - X represents –NH-, –S- or –O-; - Y and R2, independently of each other represents a hydrogen atom, a halogen atom, –NO2 or –NH2,; - R1, R3, R4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C30)alkyl chain, especially (C1–C20)alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, –C(R11)=C(R12)–, -O-, –S–, –NR13–, –C(O)–, –C(S)–, –C=N–, –N=C–,–OC(O)–, – C(O)O–, –SC(O)–, –C(O)S–, –N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R10 represents a hydrogen atom or a (C1–C30)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, the aryl, heteroaryl and heterocyclic rings being optionally substituted;
- R11 and R12 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl group; and - R13 to R15 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, and preferably a hydrogen atom or a (C1–C6)alkyl or aryl group, and still more preferably a hydrogen atom or a (C1–C6)alkyl group, or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of neurological diseases, preferably associated with psychiatric and/or sleep disorders and diseases, such as narcolepsy, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. Definitions In the present invention “halogen atom” means fluorine, chlorine, bromine and iodine atoms. In the present invention, “alkyl” group means a saturated, linear or branched hydrocarbon chain. In the present invention, “(C1–Cx)alkyl” means an alkyl group such as defined above, containing 1 to X carbon atoms. For example, “(C1–C6)alkyl” means an alkyl group such as defined above, containing 1 to 6 carbon atoms, such as, for example, methyl, ethyl, isopropyl, tert–butyl, pentyl, etc. In the present invention, “aryl” means an aromatic group, especially a hydrocarbon group, especially containing 6 to 20 carbon atoms, preferably 6 to 10 carbon atoms, and comprising one or more fused rings, such as, for example, a phenyl or naphthyl group. Advantageously, it is a phenyl group. In the present invention, “heteroaryl” means an aromatic group comprising one or more fused rings and comprising 5 to 10 cyclic atoms, including one or more heteroatoms, advantageously 1 to 4 and even more advantageously 1 or 2, such as, for example, sulfur, nitrogen, oxygen, phosphorus or selenium atoms, and preferably sulfur, nitrogen or oxygen, the other cyclic atoms being carbon atoms. Examples of heteroaryl groups are furyl, thienyl, pyrrolyl, pyridinyl, pyrimidinyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, indyl or selenophenyl. In the present invention, “acyl” group means a group of formula -CO-R in which R
represents a (C1–C6) alkyl, cycloalkyl, heterocyclic, aryl, or heteroaryl group, and preferably a (C1–C6) alkyl or aryl group, and still more preferably a (C1–C6) alkyl group. In the present invention, “cycloalkyl” means a saturated mono– or polycyclic hydrocarbon chain (especially a bicyclic or tricyclic chain). When it is a polycyclic group, the rings can be fused, bridged or joined by a spiro ring junction two by two. Examples include cyclopropyl, cyclopentyl, cyclohexyl and cycloheptyl groups. In the present invention, “heterocyclic” group means a non-aromatic, saturated or unsaturated, monocyclic or polycyclic group (comprising fused, bridged or spiro rings) in which one or several atom(s) of the ring(s) carbon atoms each is replaced with heteroatoms, in particular this term means a 5 to 10–membered ring, saturated or unsaturated, but not aromatic, and containing one or more, advantageously 1 to 4, more advantageously 1 or 2 heteroatoms, the heteroatoms being for example, sulfur, nitrogen or oxygen atoms. It can particularly be a pyrrolidinyl, piperidinyl, piperazinyl or morpholinyl group. The aryl, heteroaryl and heterocyclic group, when substituted, can be substituted with one or more groups chosen from the group consisting of a halogen atom, a (C1–C6)alkyl group, an aryl group, -NO2, –CN, –OR19, –SR20, –NR16R17, –B(OH)2, –SO3R17, and –COOR18, in particular chosen from the group consisting of a halogen atom, -NO2, –CN, –OR19, – SR20, –NR16R17, –B(OH)2, –SO3R17, with R16 to R20 representing, independently of each other, a hydrogen atom or a (C1–C6)alkyl group. In the present invention, “pharmaceutically acceptable” means what is used in the preparation of a pharmaceutical composition, which is generally safe, nontoxic and not biologically or otherwise undesirable and which is acceptable for both veterinary and human pharmaceutical use. “Pharmaceutically–acceptable salts” of a compound mean of salts that are pharmaceutically acceptable, such as defined here, that have the desired pharmacological activity of the parent compound. Such salts include: (1) hydrates and solvates, (2) pharmaceutically–acceptable acid addition salts formed with pharmaceutically– acceptable inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or formed with pharmaceutically–acceptable organic acids such as acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, hydroxynaphthoic acid, 2–hydroxy ethanesulfonic acid,
lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, muconic acid, 2–naphthalene sulfonic acid, propionic acid, salicylic acid, succinic acid, dibenzoyl–L–tartaric acid, tartaric acid, p–toluenesulfonic acid, trimethylacetic acid, trifluoroacetic acid and the like, or (3) pharmaceutically–acceptable base addition salts formed when an acid proton present in the parent compound is either replaced by a metal ion, for example an alkaline metal ion, an alkaline–earth metal ion or an aluminum ion, or is coordinated with a pharmaceutically–acceptable organic or inorganic base. Acceptable organic bases include diethanolamine, ethanolamine, N–methylglucamine, triethanolamine, tromethamine and the like. Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate and sodium hydroxide. Preferably, the compounds according to the invention will be in the form of pharmaceutically–acceptable base addition salts, the base being such as NaOH or KOH, and especially NaOH. An “optically pure compound” means an enantiomer in an enantiomeric excess of more than 95%, preferably of more than 96%, more preferably of more than 97%, even more preferably of more than 98%, particularly preferably of more than 99%. The present invention also includes all pharmaceutically acceptable isotopic variations of a compound of formula I in which one or more atoms is replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Such compounds are identical to those disclosed herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen such as 2H and 3H, carbon such as 11C, 13C and 14C, nitrogen such as 13N and 15N, oxygen such as 15O, 17O and 18O, sulfur such as 35S, fluorine such as 18F, iodine such as 123I and 125I, and chlorine such as 36Cl. Certain isotopically-labelled compounds of formula I, for example those incorporating a radioactive isotope, are useful in drug and / or substrate tissue distribution studies. A subject administered with a compound of the present invention, or a pharmaceutically acceptable salt thereof, is generally a mammal, such as a human being, male or female. The amount of compound administered to the subject is an amount sufficient to agonize the orexin receptor in the subject. In an embodiment, the amount of compound can be an “effective amount”, wherein the subject
compound is administered in an amount that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. An effective amount does not necessarily include considerations of toxicity and safety related to the administration of the compound. It is recognized that one skilled in the art may affect neurological and psychiatric disorders associated with orexin receptor activation by treating a subject presently afflicted with the disorders, or by prophylactically treating a subject likely to be afflicted with the disorders, with an effective amount of a compound of the present invention. As used herein, the terms "treatment” and “treating” refer to all processes wherein there may be a slowing, interrupting, arresting, controlling, or stopping of the progression of the neurological and psychiatric disorders described herein, but does not necessarily indicate a total elimination of all disorder symptoms, as well as the prophylactic therapy of the mentioned conditions, particularly in a subject that is predisposed to such disease or disorder. The terms “administration of” and or “administering a” compound should be understood to mean providing a compound of the invention or a prodrug of a compound of the invention to the subject. The term “composition” as used herein is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. Such term is intended to encompass a product comprising the active ingredient(s), and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier, i.e. the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Diseases and disorders The orexin receptors (OX1R, OX2R) have been implicated in a wide range of biological functions. This has suggested a potential role for these receptors in a variety of disease processes in humans or other species. The compounds of the present invention could therefore have utility in treating, preventing, ameliorating, controlling or reducing the risk of a variety of disorders associated with orexin receptors, including one or more
of the following conditions or diseases: narcolepsy with or without cataplexy, narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), Gelineau syndrome (Maladie de Gelineau), narcoleptic syndrome, accompanied by narcolepsy-like symptoms, cataplexy in narcolepsy, excessive daytime sleepiness (EDS) in narcolepsy, hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, and insufficient sleep syndrome and any conditions in accordance with The International Classification of Sleep Disorders, Third Edition (ICSD-3) classifies eight different Central Disorders of Hypersomnolence (CDH)(American Academy of Sleep Medicine 2014) and/ or marked by pathologic daytime sleepiness and/or unappropriated arousal status caused or nor by sleep apnea, nocturnal myoclonus, Rapid Eye Movement (REM) sleep interruptions, jet-lag, shift working, sleep disturbances, dyssomnias, sleep disorders, sleep disturbances, hypersomnia associated with depression, emotional / mood disorders, Alzheimer's disease or neurodegenerative disorders or cognitive impairment and tauopathies, Parkinson's disease and other synucleinopathies, Guillain-Barre syndrome, chronic fatigue syndrome, Long COVID-19 and medical or health conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, restless legs syndrome, fibromyalgia, cardiac failure, diseases related to bone loss, sepsis, syndromes which are manifested by unrefreshing sleep and muscle pain, sleep apnea which is associated with respiratory disturbances during sleep ; conditions which result from a diminished quality of sleep and other diseases related to general orexin system dysfunction. In the present invention, “neurological, psychiatric, sleep disorders and diseases” refers to psychiatric and/or sleep neurological disorders and diseases, i.e. neurological diseases associated with sleep and/or psychiatric disorders. In particular, the disorders and diseases in which the central orexin neurotransmission is compromised or central and peripheral orexin receptors are involved. Thus, in certain embodiments the present invention may provide methods for treating or controlling: narcolepsy with or without cataplexy, narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), Gelineau syndrome (Maladie de Gélineau), narcoleptic syndrome, accompanied by narcolepsy-like symptoms, cataplexy in narcolepsy, excessive daytime sleepiness (EDS) in narcolepsy, hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due
to a medication or substance, and insufficient sleep syndrome and any conditions in accordance with The International Classification of Sleep Disorders, Third Edition (ICSD-3) classifies eight different Central Disorders of Hypersomnolence (CDH)(American Academy of Sleep Medicine 2014) ; pathologic daytime sleepiness and/or unappropriated arousal status caused or nor by sleep apnea, nocturnal myoclonus, REM sleep interruptions, jet-lag, shift working, sleep disturbances, dyssomnias, sleep disorders, sleep disturbances, hypersomnia associated with depression, emotional / mood disorders, Alzheimer's disease or cognitive impairment, Parkinson's disease, dementias, Guillain-Barre syndrome, chronic fatigue syndrome, Long COVID-19 and medical or health conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, restless legs syndrome, fibromyalgia, cardiac failure, diseases related to bone loss, sepsis, syndromes which are manifested by unrefreshing sleep and muscle pain, sleep apnea which is associated with respiratory disturbances during sleep ; conditions which result from a diminished quality of sleep and other diseases related to general orexin system dysfunction ; sleep disturbances associated with diseases such as neurological disorders including neuropathic pain and restless leg syndrome, treating or controlling addiction disorders, treating or controlling psychoactive substance use and abuse, enhancing cognition, increasing memory retention ; diabetes and appetite, taste, eating, or drinking disorders ; insulin resistance syndrome ; hypothalamic diseases ; depression, including major depression and major depression disorder ; ameliorating or reducing the risk of epilepsy, including absence epilepsy, seizures ; pain, including neuropathic pain ; Parkinson's disease, dementia with Lewy’s bodies and synucleinopathies ; Guillain-Barre syndrome ; Long COVID-19 ; Kleine-Levin syndrome ; psychosis, dysthymic, mood, bipolar disorder, psychotic and anxiety disorders ; side effects or complications due to anesthesia and/ or orexin antagonists in a subject which comprises administering to the subject a compound of the present invention. The dihydro-quinazoline, -benzothiazine and benzoxazine derivatives as compounds of the present invention may also potentially have utility in treating, preventing, ameliorating, controlling or reducing the risk of a variety of other disorders associated with orexin receptors, including one or more of the following conditions or diseases including enhancing sleep quality, improving sleep quality, increasing sleep efficiency, consolidating sleep maintenance, improving sleep initiation, decreasing sleep latency or onset, decreasing difficulties in falling asleep, increasing sleep continuity, decreasing the number of awakenings during sleep, decreasing intermittent waking
during sleep, decreasing nocturnal arousals, decreasing the time spent awake following the initial onset of sleep, increasing the total amount of sleep, reducing the fragmentation of sleep, altering the timing, frequency or duration of sleep stages, or duration of slow-wave sleep and / or REM sleep, promoting slow wave sleep, enhancing EEG - delta activity during sleep, decreasing nocturnal arousals, especially early morning awakenings, increasing daytime alertness, reducing daytime drowsiness, treating or reducing excessive daytime sleepiness ; increasing satisfaction with the intensity of sleep, increasing sleep maintenance, idiopathic insomnia, sleep problems, insomnia, night terror, insomnias associated with depression, emotional/mood disorders, Alzheimer's disease or neurodegenerative disorders or cognitive impairment, Parkinson's disease, dementias, Guillain-Barre syndrome, chronic fatigue syndrome, Long COVID-19 and medical or health conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, restless legs syndrome, fibromyalgia, cardiac failure, diseases related to bone loss, sepsis, syndromes which are manifested by unrefreshing sleep and muscle pain, sleep apnea which is associated with respiratory disturbances during sleep ; conditions which result from a diminished quality of sleep ; increasing learning ; augmenting memory ; increasing retention of memory ; eating disorders associated with excessive food intake and complications associated therewith, compulsive eating disorders, obesity (due to any cause, whether genetic or environmental), obesity - related disorders overeating, anorexia, bulimia, cachexia, dysregulated appetite control, hypertension, diabetes, elevated plasma insulin concentrations and insulin resistance, dyslipidemias, hyperlipidemia, endometrial, breast, prostate and colon cancer, osteoarthritis, obstructive sleep apnea, cholelithiasis, gallstones, heart disease, lung disease, abnormal heart rhythms and arrythmias, myocardial infarction, congestive heart failure, coronary heart disease, acute and congestive heart failure, hypotension, hypertension, angina pectoris, infarction, ischemic or hemorrhagic stroke, subarachnoid hemorrhage, chronic renal failure, renal disease, impaired glucose tolerance, craniopharyngioma, Prader-Willi Syndrome, Frohlich's syndrome, GH - deficient subjects, Turner's syndrome, and other pathological conditions showing reduced metabolic activity or a decrease in resting energy expenditure as a percentage of total fat – free mass, e.g, children with acute lymphoblastic leukemia, metabolic syndrome, also known as syndrome X, insulin resistance syndrome, reproductive hormone abnormalities, sexual and reproductive dysfunction, such as impaired fertility, infertility, hypogonadism in males and hirsutism in females, fetal defects associated with maternal obesity, gastrointestinal motility disorders, intestinal motility dyskinesias, obesity - related gastroesophageal reflux,
hypothalamic diseases, hypophysis diseases, respiratory disorders, such as obesity - hypoventilation syndrome (Pickwickian syndrome), Ondine’s syndrome and sleep- related disorders where there is abnormal coupling of activity, particularly through the thalamus ; enhancing cognitive function, including cognitive dysfunctions that comprise deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders ; treating or controlling Guillain-Barre syndrome chronic fatigue syndrome, Long COVID-19 and medical or health conditions associated with circadian rhythmicity ; treating or controlling Kleine-Levin syndrome ; treating or controlling psychosis ; treating or controlling dysthymic, mood, psychotic and anxiety disorders ; treating complications due to anesthesia ; enhancing memory ; increasing memory retention ; increasing immune response ; increasing immune function ; hot flashes ; night sweats ; extending life span ; schizophrenia ; muscle - related disorders that are controlled by the excitation / relaxation rhythms imposed by the neural system such as cardiac rhythm and other disorders of the cardiovascular system ; conditions related to proliferation of cells such as vasodilation or vaso-restriction and blood pressure ; cancer ; cardiac arrhythmia ; hypertension ; congestive heart failure ; conditions of the genital / urinary system ; disorders of sexual function and fertility ; adequacy of renal function ; responsivity to anesthetics ; mood disorders, such as depression or more particularly depressive disorders, for example, single episodic or recurrent major depressive disorders and dysthymic disorders, or bipolar disorders, for example, bipolar I disorder, bipolar II disorder and cyclothymic disorder, mood disorders due to a general medical condition, and substance - induced mood disorders ; affective neurosis, depressive neurosis, anxiety neurosis ; anxiety disorders including acute stress disorder, agoraphobia, generalized anxiety disorder, obsessive - compulsive disorder, panic attack, panic disorder, post - traumatic stress disorder, separation anxiety disorder, social phobia, specific phobia, substance - induced anxiety disorder and anxiety due to a general medical condition ; acute neurological and psychiatric disorders such as cerebral deficits subsequent to cardiac bypass surgery and grafting, stroke, ischemic stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage ; Huntington's Chorea ; Huntington's disease and Tourette syndrome ; Cushing's syndrome / disease ; basophile adenoma ; prolactinoma ; hyperprolactinemia ; hypophysis tumor / adenoma ; hypothalamic diseases ; inflammatory bowel disease ; gastric diskinesia ; gastric ulcers ; Froehlich's syndrome ; adrenohypophysis disease ; hypophysis disease ; adrenohypophysis hypofunction ; adrenohypophysis hyperfunction ; hypothalamic
hypogonadism ; Kallman's syndrome (anosmia, hyposmia) ; functional or psychogenic amenorrhea ; hypopituitarism ; hypothalamic hypothyroidism ; hypothalamic - adrenal dysfunction ; idiopathic hyperprolactinemia ; hypothalamic disorders of growth hormone deficiency ; idiopathic growth deficiency ; dwarfism ; gigantism ; acromegaly ; amyotrophic lateral sclerosis ; multiple sclerosis ; ocular damage ; retinopathy ; cognitive disorders ; idiopathic and drug - induced Parkinson's disease ; muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, seizure disorders, absence seizures, complex partial and generalized seizures ; Lennox - Gastaut syndrome ; cognitive disorders including dementia (associated with Alzheimer's disease, ischemia, trauma, vascular problems or stroke, HIV disease, Parkinson's disease, Dementia with Lewy’s Bodies, Huntington's disease, Pick's disease, Creutzfeldt - Jacob disease, perinatal hypoxia, other general medical conditions or substance abuse) ; delirium, amnestic disorders or age related cognitive decline ; schizophrenia or psychosis including schizophrenia (paranoid, disorganized, catatonic or undifferentiated), schizophreniform disorder, schizoaffective disorder, delusional disorder, brief psychotic disorder, shared psychotic disorder, psychotic disorder due to a general medical condition and substance - induced psychotic disorder ; dissociative disorders including multiple personality syndromes and psychogenic amnesias ; substance - related disorders, substance use, substance abuse, substance seeking, substance reinstatement, all types of psychological and physical addictions and addictive behaviors, reward - related behaviors (including substance – induced delirium, persisting dementia, persisting amnestic disorder, psychotic disorder or anxiety disorder ; tolerance, addictive feeding, addictive feeding behaviors, binge / purge feeding behaviors, dependence, withdrawal or relapse from substances including alcohol, amphetamines, cannabis, cocaine, heroin, morphine, nicotine, opioids, phencyclidine, sedatives, hypnotics or anxiolytics) ; appetite, taste, eating or drinking disorders ; movement disorders, including akinesias and akinetic - rigid syndromes (including Parkinson's disease, drug - induced parkinsonism, postencephalitic parkinsonism, progressive supranuclear palsy, multiple system atrophy, cortico-basal degeneration, parkinsonism - ALS dementia complex and basal ganglia calcification), chronic fatigue syndrome, fatigue, including Parkinson's fatigue, multiple sclerosis fatigue, fatigue caused by a sleep disorder or a circadian rhythm disorder, medication induced parkinsonism (such as neuroleptic - induced parkinsonism, neuroleptic malignant syndrome, neuroleptic - induced acute dystonia, neuroleptic - induced acute akathisia, neuroleptic - induced tardive dyskinesia and medication - induced postural tremor), Gilles de la Tourette's syndrome, epilepsy, and dyskinesias including tremor (such as rest tremor, essential tremor, postural tremor and intention tremor), chorea
(such as Sydenham's chorea, Huntington's disease, benign hereditary chorea, neuro- acanthocytosis, symptomatic chorea, drug - induced chorea and hemi-ballism), myoclonus (including generalized myoclonus and focal myoclonus), tics (including simple tics, complex tics and symptomatic tics), restless leg syndrome and dystonia (including generalized dystonia such as idiopathic dystonia, drug - induced dystonia, symptomatic dystonia and paroxymal dystonia, and focal dystonia such as blepharospasm, oromandibular dystonia, spasmodic dysphonia, spasmodic torticollis, axial dystonia, dystonic writer's cramp and hemiplegic dystonia) ; neurodegenerative disorders including nosological entities such as disinhibition - dementia - parkinsonism - amyotrophy complex ; pallido-ponto-nigral degeneration ; epilepsy ; seizure disorders ; attention deficit/hyperactivity disorder (ADHD) ; conduct disorder ; migraine (including migraine headache) ; headache ; hyperalgesia ; pain ; enhanced or exaggerated sensitivity to pain such as hyperalgesia, causalgia, and allodynia ; acute pain ; burn pain ; atypical facial pain ; neuropathic pain ; back pain ; complex regional pain syndrome I and II ; arthritic pain ; sports injury pain ; pain related to infection e.g. HIV, COVID-19, post chemotherapy pain ; post - stroke pain ; post - operative pain ; neuralgia ; emesis, nausea, vomiting ; gastric dyskinesia ; gastric ulcers ; Kallman's syndrome (anosmia) ; asthma ; cancer ; conditions associated with visceral pain such as irritable bowel syndrome, and angina ; eating disorders ; urinary incontinence ; substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, orexin antagonists etc.) ; psychosis ; schizophrenia ; anxiety (including generalized anxiety disorder, panic disorder, and obsessive compulsive disorder) ; mood disorders (including depression, mania, bipolar disorders) ; trigeminal neuralgia ; hearing loss ; tinnitus ; neuronal damage including ocular damage ; retinopathy ; macular degeneration of the eye ; emesis ; brain edema ; pain, including acute and chronic pain states, severe pain, intractable pain, inflammatory pain, neuropathic pain, post - traumatic pain, bone and joint pain (osteoarthritis), repetitive motion pain, dental pain, cancer pain, myofascial pain (muscular injury, fibromyalgia), perioperative pain (general surgery, gynecological), chronic pain, neuropathic pain, post-traumatic pain, trigeminal neuralgia, migraine and migraine headache and other diseases related to general orexin system dysfunction. A preferred embodiment of the invention is a compound of formula (I) or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric
disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, Parkinson's disease and other synucleinopathies, preferably narcolepsy type 1 (NT1), narcolepsy type 2 (NT2) and Parkinson's disease. A preferred embodiment of the invention is a compound of formula (I) or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of REM sleep behavior disorder (RBD). In some embodiments, the neurological diseases, preferably the neurological disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved, are selected from the group consisting of narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, Parkinson's disease and other synucleinopathies, preferably narcolepsy type 1 (NT1), narcolepsy type 2 (NT2) and Parkinson's disease. In some embodiments, the neurological diseases, preferably the neurological disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved, are selected from the group consisting of narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia and recurrent hypersomnia. In particular, the neurological disorders targeted are those requiring an agonist of the orexin 1 receptor (OX1R) and/or the orexin 2 receptor (OX2R), preferably both receptors. Compounds of formula (I) In a preferred embodiment, when one of Y or R2 represents a halogen atom, –NO2 or – NH2, preferably –NO2 or –NH2, the other represents a hydrogen atom or a halogen atom. In some embodiments, when Y represents a halogen atom, –NO2 or –NH2, preferably – NO2 or –NH2, R2 represents a hydrogen atom or a halogen atom. In some embodiments, when R2 represents a halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2, Y represents a hydrogen atom or a halogen atom. In a preferred embodiment, when one of Y or R2 represents a halogen atom, –NO2 or – NH2, preferably –NO2 or –NH2, the other represents a hydrogen atom. In some embodiments, when Y represents a halogen atom, –NO2 or –NH2, preferably –NO2 or – NH2, R2 represents a hydrogen atom. In some embodiments, when R2 represents a
halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2, Y represents a hydrogen atom. R4 represents preferably a hydrogen atom. In a preferred embodiment of the invention, R1, R2, R3, R4 each represent a hydrogen atom or R1, R3, R4, Y each represent a hydrogen atom. In some embodiments, R1, R2, R3, R4 can each represent a hydrogen atom. In some embodiments, R1, R3, R4, Y each represent a hydrogen atom. R5, R6, R7, R8, R9 can each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C20)alkyl, optionally broken up and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, –C(R11)=C(R12)–,-O-, –S–, –NR13–, –C(O)–,–OC(O)–, –C(O)O–, – N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted. In another preferred embodiment, R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom or –OR10. Preferably, at least four residues out of R5, R6, R7, R8 and R9 each represent a hydrogen atom. In a particular embodiment, R5, R6, R7, R8, R9, each represent, a hydrogen atom. In another embodiment, R5 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group. In some embodiments, R5 represents –OR10 with R10 representing a hydrogen atom, a (C1– C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group, and R6, R7, R8, R9, each represent, a hydrogen atom. In another embodiment, R6 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group. In some embodiments, R6 represents –OR10 with R10 representing a hydrogen atom, a (C1– C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group, and R5, R7, R8, R9, each represent, a hydrogen atom. In another embodiment, R7 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group. In some embodiments, R7 represents –OR10 with R10 representing a hydrogen atom, a (C1– C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group, and R5, R6, R8, R9, each represent, a hydrogen atom. In some embodiments, one residue out of R5, R6, R7, R8 and R9 represents -OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–
C20)alkyl or a phenyl group. In some embodiments, X can represent -NH-. In some embodiments, X can represent - S-. In some embodiments, X can represent -O-. Preferably, X represents –NH- or –S-. Advantageously, the compound of formula (I) is selected from the group consisting of:
and mixtures thereof. Preferably, the compound of formula (I) is selected from the group consisting of:
, , and mixtures thereof.
More preferably, the compound of formula (I) is selected from the group consisting of:
, and
mixtures thereof. In a preferred embodiment of the invention, the neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which orexin receptors are involved is selected from the group consisting of narcolepsy type 1, narcolepsy type 2, idiopathic hypersomnia, recurrent hypersomnia, attention- deficit hyperactivity disorder, anxiety and mood disorders, Alzheimer's disease or any other neurodegenerative disorders or cognitive impairment and tauopathies, Parkinson's disease and other synucleinopathies, Guillain-Barre syndrome, chronic fatigue syndrome, long COVID-19 and medical or health conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, restless legs syndrome, fibromyalgia, cardiac failure, diseases related to bone loss, sepsis, syndromes which are manifested by unrefreshing sleep and muscle pain, sleep apnea which is associated with respiratory disturbances during sleep; conditions which result from a diminished quality of sleep. The dosage of active ingredient in the compositions of this invention may be varied, however, it is necessary that the amount of the active ingredient be such that a suitable dosage form is obtained. The active ingredient may be administered to subjects (animals and human) in need of such treatment in dosages that will provide optimal pharmaceutical efficacy. The selected dosage depends upon the desired therapeutic effect, on the route of administration, and on the duration of the treatment. The dose will vary from subject to subject depending upon the nature and severity of disease, the subject's weight, special diets then being followed by a subject, concurrent medication, and other factors which those skilled in the art will recognize. Generally, dosage levels of between 0.0001 to 100 mg/kg of body weight per day are
administered to the subject, e.g., humans, adolescent humans and elderly humans, to obtain effective agonism of orexin receptors. Advantageously, a therapeutic dose of a compound of formula (I) comprised between 0.1 mg/kg/day and 100 mg/kg/day is administrated to a patient in need thereof. The present invention also relates to a pharmaceutical composition comprising at least one compound of formula (I) as described above and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. The dosage range will generally be from 0.5 mg to 10.0 g per subject per day which may be administered in single or multiple doses. In one embodiment, the dosage range will be from 0.5 mg to 500 mg per subject per day, preferably from 0.5 mg to 200 mg per subject per day, and more preferably from 5 mg to 50 mg per subject per day. Pharmaceutical compositions of the present invention may be provided in a solid dosage formulation such as comprising from 0.5 mg to 500 mg of active ingredient, or comprising from 1 mg to 250 mg of active ingredient. The pharmaceutical composition may be provided in a solid dosage formulation comprising 1 mg, 5 mg, 10 mg, 50 mg, 80 mg, 100 mg, 200 mg active ingredient. In a preferred embodiment, the pharmaceutical composition for use according to the invention comprises between 0.5 mg to 800 mg, preferably between 20 mg to 400 mg of the compound of formula (I). For oral administration, the compositions may be provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, such as 1, 5, 10, 15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, 500, 600, 750, 800, 900, and 1000 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated. The compounds may be administered on a regimen of 1 to 4 times per day, such as once or twice per day. The compounds may be administered once or multiple times during the day. The compounds may be administered upon awakening or otherwise in the morning, or during waking hours. For example, the compounds may be administered 25 mg, 30 mg or 250 mg, 1 hour after awakening, 30 minutes after awakening or immediately after awakening. In a particular embodiment, the pharmaceutical composition according to the invention is suitable for oral or parenteral administration. Preferably, the pharmaceutical composition according to the invention is in the form of a solution,
such as an injectable solution, or a tablet or a capsule or a transdermal delivery system. The compounds of the present invention may be used in combination with one or more other drugs in the treatment, prevention, control, amelioration, or reduction of risk of diseases or conditions for which compounds of the present invention or the other drugs may have utility, where the combination of the drugs together are safer or more effective than either drug alone. Such other drug(s) may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention. When a compound of the present invention is used contemporaneously with one or more other drugs, a pharmaceutical composition in unit dosage form containing such other drugs and the compound of the present invention is contemplated. However, the combination therapy may also include therapies in which the compound of the present invention and one or more other drugs are administered on different overlapping schedules. It is also contemplated that when used in combination with one or more other active ingredients, the compounds of the present invention and the other active ingredients may be used in lower doses than when each is used singly. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to a compound of the present invention. The above combinations include combinations of a compound of the present invention not only with one other active compound, but also with two or more other active compounds. The weight ratio of the compound of the present invention to the second active ingredient may be varied and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when a compound of the present invention is combined with another agent, the weight ratio of the compound of the present invention to the other agent will generally range from 1000:1 to 1:1000, preferably from 200:1 to 1:200. Combinations of a compound of the present invention and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used. In such combinations the compound of the present invention and other active agents may be administered separately or in conjunction. In addition, the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent. The compounds of the present invention may be administered in combination with compounds which are known in the art to be useful for treating or controlling
narcolepsy, including e.g., methylphenidate, amphetamine, atomoxetine, reboxetine, viloxazine, phenelzine, protriptyline, gamma–hydroxybutyric acid, sodium oxybate, or other oxybate salts, modafinil, armodafinil, adrafinil, pitolisant, samelisant, nalfurafin, caffeine, and salts thereof, and combinations thereof, and the like. Compounds of formula (II) The present invention also relates to a compound of formula (II):
wherein: - X represents -NH-, –S- or –O-, preferably -NH- or -S-; - Y and R2, independently of each other represents a hydrogen atom, a halogen atom, –NO2 or –NH2,; - R1, R3, R4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C30)alkyl chain, especially (C1–C20)alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, –C(R11)=C(R12)–, -O-,–S–, –NR13–, –C(O)–, –C(S)–, –C=N–, –N=C–, –OC(O)–, – C(O)O–, –SC(O)–, –C(O)S–, –N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R10 represents a hydrogen atom or a (C1–C30)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R11 and R12 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl group; and - R13 to R15 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, and preferably
a hydrogen atom or a (C1–C6)alkyl or aryl group, and still more preferably a hydrogen atom or a (C1–C6)alkyl group, or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof. Preferably, the compound of formula (II) is not
(this compound has been described (Cox et al. 2020) as having an inhibitory activity of amyloid aggregation),
(these four compounds has been described as having an inhibitory effect on cell multiplication (Yale et Kalkstein 1967),
(this compound has been described as having an anticancer activity (Chinigo et al. 2008),
(this compound has been described as having an anti-tumour activity (Mordarski et
Chylinska 1971),
(this compound has been used for the treatment of PARP-1 related diseases, see WO 2018044136) (Kim et al. 2023), and provided that when X represents O, each of R1, R2, R3 and R4 represents H, Y represents an halogen atom and four of R5, R6, R7, R8, and R9 represent H, then the remaining group of R5, R6, R7, R8, and R9 is not a (C1-C30)alkyl chain preceded by -O- and broken-up by -O- or broken up, followed and/or preceded by –C(R11)=C(R12)–. In some embodiments, when X represents -O-, then R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C30)alkyl chain, especially (C1–C20)alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–,–S–, –NR13–, –C(O)–, –C(S)–, –C=N–, –N=C–, –OC(O)–, – C(O)O–, –SC(O)–, –C(O)S–, –N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted. In a preferred embodiment, when X represents -O-, then Y and R2, independently of each other represents a hydrogen atom, –NO2 or –NH2. In a preferred embodiment, when one of Y or R2 represents a halogen atom, –NO2 or – NH2, preferably –NO2 or –NH2, the other represents a hydrogen atom or a halogen atom. In some embodiments, when Y represents a halogen atom, –NO2 or –NH2, preferably – NO2 or –NH2, R2 represents a hydrogen atom or a halogen atom. In some embodiments, when R2 represents a halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2, Y represents a hydrogen atom or a halogen atom. In a preferred embodiment, when one of Y or R2 represents a halogen atom, –NO2 or – NH2, preferably –NO2 or –NH2, the other represents a hydrogen atom. In some embodiments, when Y represents a halogen atom, –NO2 or –NH2, preferably –NO2 or – NH2, R2 represents a hydrogen atom. In some embodiments, when R2 represents a halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2, Y represents a hydrogen atom. R4 represents preferably a hydrogen atom. In a preferred embodiment of the invention, when X is NH or O, then R5, R6 or R7 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or a phenyl group.
In a preferred embodiment of the invention, R1, R2, R3, R4 each represent a hydrogen atom or R1, R3, R4, Y each represent a hydrogen atom. In some embodiments, R1, R2, R3, R4 can each represent a hydrogen atom. In some embodiments, R1, R3, R4, Y each represent a hydrogen atom. R5, R6, R7, R8, R9 can each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C20)alkyl, optionally broken up and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, –C(R11)=C(R12)–,-O-, –S–, –NR13–, –C(O)–, –OC(O)–, –C(O)O–, – N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted. In another preferred embodiment, R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom or –OR10. Preferably, at least four residues out of R5, R6, R7, R8 and R9 each represent a hydrogen atom. In a particular embodiment, R5, R6, R7, R8, R9, each represent, a hydrogen atom. In another embodiment, R5 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group. In some embodiments, R5 represents –OR10 with R10 representing a hydrogen atom, a (C1– C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group, and R6, R7, R8, R9, each represent, a hydrogen atom. In another embodiment, R6 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group. In some embodiments, R6 represents –OR10 with R10 representing a hydrogen atom, a (C1– C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group, and R5, R7, R8, R9, each represent, a hydrogen atom. In another embodiment, R7 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group. In some embodiments, R7 represents –OR10 with R10 representing a hydrogen atom, a (C1– C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group, and R5, R6, R8, R9, each represent, a hydrogen atom. In some embodiments, one residue out of R5, R6, R7, R8 and R9 represents -OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1– C20)alkyl or a phenyl group. In some embodiments, X can represent -NH-. In some embodiments, X can represent -
S-. In some embodiments, X can represent -O-. Preferably, X represents –S-. Advantageously, the compound of formula (II) is selected from the group consisting of:
and pharmaceutically acceptable salts thereof. In a preferred embodiment of the invention, the compound of formula (II) is selected from the group consisting of compounds 99, 100, 145, 146, 148, 149, 151, 152, 153, 154, 155, 157, 158, 161, 167, 182, 185, 191, 194, 195, 196, 243, 244, 292, 293, 301, 302, 304, 305, 308, 310, 311, and pharmaceutically acceptable salts thereof. More advantageously, the compound of formula (II) is selected from the group consisting of compounds 99, 148, 152, 153, 154, 155, 157, 161, 167, 191, 194, 195, 196, 301, 302, 304, 305, 308, 310, 311, and pharmaceutically acceptable salts thereof. Even more advantageously, the compound of formula (II) is selected from the group consisting of:
,
and pharmaceutically acceptable salts thereof. In a more preferred embodiment, the compound of formula (II) is selected from the group consisting of:
, ,
and pharmaceutically acceptable salts thereof. The present invention also relates to a pharmaceutical composition comprising a compound of formula (II) and a pharmaceutically acceptable carrier. The pharmaceutical composition is as defined above for compound of formula (I).
The present invention also relates to a compound of formula (II) as described above for use as a medicament. The present invention also relates to a compound of formula (II) as described above for use in the manufacture of a medicament. In some embodiments, the present invention provides a method for treating a subject in need thereof comprising administering to said subject a compound of formula (II) or a composition comprising a compound of formula (II). The chromatographic separations of may be achieved as known in the art. Their absolute stereochemistry may be determined by the X-ray crystallography of crystalline products or crystalline intermediates which are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration. If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography. The coupling reaction is often the formation of salts using an enantiomerically pure acid or base. The diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue. The racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art. Alternatively, any enantiomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art. In some embodiments, the present invention provides a method for treating a subject suffering from neurological diseases, preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved, comprising administering to said subject a compound of formula (I) or a composition comprising a compound of formula (I). In particular, said disorders and diseases are as defined above. In some embodiments, the present invention provides a method for preventing neurological diseases, preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors
are involved, comprising administering to a subject an effective amount of a compound of formula (I) or a composition comprising an effective amount of a compound of formula (I). In particular, said disorders and diseases are as defined above. In some embodiments, the methods further comprise administration of a second active compound. In one aspect, the invention is also related to compounds of formula (I) or pharmaceutical compositions comprising at least one compound of formula (I) for their use in the manufacture of a medicament for the treatment of from neurological diseases, preferably neurological, psychiatric, sleep disorders and diseases, such as narcolepsy and Parkinson’s disease, in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. In particular, said disorders and diseases are as defined above. The present invention also relates to the following embodiments: Embodiment 1. A compound of formula (I): wherein: - X represents –NH-, –S- or –O-, preferably -NH- or -S-; - Y represents a halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2; - R1, R2, R3, R4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C30)alkyl chain, especially (C1–C20)alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, –C(R11)=C(R12)–,-O-, –S–, –NR13–, –C(O)–, –C(S)–, –C=N–, –N=C–, –C=C–,– OC(O)–, –C(O)O–, –SC(O)–, –C(O)S–, –N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R10 represents a hydrogen atom or a (C1–C30)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R11 and R12 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl group; and
- R13 to R15 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, and preferably a hydrogen atom or a (C1–C6)alkyl or aryl group, and still more preferably a hydrogen atom or a (C1–C6)alkyl group, or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. Embodiment 2. The compound of formula (I) for use according to embodiment 1, characterized in that R1, R2, R3, R4 each represent a hydrogen atom. Embodiment 3. The compound of formula (I) for use according to embodiment 1 or 2, characterized in that R5, R6, R7, R8, R9, each represent, independently of each other, a hydrogen atom or –OR10. Embodiment 4. The compound of formula (I) for use according to any one of embodiments 1 to 3, characterized in that at least four residues out of R5, R6, R7, R8 and R9 each represent a hydrogen atom. Embodiment 5. The compound of formula (I) for use according to embodiment 4, characterized in that R5, R6, R7, R8, R9, each represent, a hydrogen atom. Embodiment 6. The compound of formula (I) for use according to embodiment 4, characterized in that R5, R6 or R7 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group. Embodiment 7. The compound of formula (I) for use according to any one of embodiments 1 to 6, characterized in that it is selected from the group consisting of compounds 69 , 84 , 87 , 90 , 96 , 97 , 98 , 99 , 148 , 152 , 153 , 154 , 155 , 157 , 161 , 167 , 191 , 194 , 195 , 196 , and mixtures thereof, preferably 84 , 99 , 152 , 155 , 194 , and mixtures thereof. Embodiment 8. The compound of formula (I) for use according to any one of embodiments 1 to 7, characterized in that the neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which orexin receptors are involved is selected from the group consisting of narcolepsy type 1, narcolepsy type 2, idiopathic hypersomnia, recurrent hypersomnia, attention- deficit hyperactivity disorder, anxiety and mood disorders, Alzheimer's disease or any
other neurodegenerative disorders or cognitive impairment and tauopathies, Parkinson's disease and other synucleinopathies, Guillain-Barre syndrome, chronic fatigue syndrome, long COVID-19 and medical or health conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating shift-work schedules, restless legs syndrome, fibromyalgia, cardiac failure, diseases related to bone loss, sepsis, syndromes which are manifested by unrefreshing sleep and muscle pain, sleep apnea which is associated with respiratory disturbances during sleep; conditions which result from a diminished quality of sleep. Embodiment 9. A pharmaceutical composition comprising at least one compound of formula (I) as defined in any one of embodiments 1 to 8 and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of neurological, psychiatric, sleep disorders and diseases in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved. Embodiment 10. The pharmaceutical composition for use according to embodiment 9, comprising between 0.5 mg to 800 mg, preferably between 20 mg to 400 mg of the compound of formula (I). Embodiment 11. The pharmaceutical composition for use according to embodiment 9 or 10, which is suitable for oral or parenteral administration, preferably in the form of a solution, such as an injectable solution, a tablet, a capsule or a transdermal delivery system. Embodiment 12. A compound of formula (II): wherein: - X represents -NH-, –S- or –O-, such as -S- or -O-, preferably -S-; - Y represents a halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2; - R1, R2, R3, R4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C30)alkyl chain, especially (C1–C20)alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic,
–C≡C–, –C(R11)=C(R12)–, -O-, –S–, –NR13–, –C(O)–, –C(S)–, –C=N–, –N=C–, –C=C–,– OC(O)–, –C(O)O–, –SC(O)–, –C(O)S–, –N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R10 represents a hydrogen atom or a (C1–C30)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R11 and R12 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl group; and - R13 to R15 represent, independently of each other, a hydrogen atom or a (C1– C6)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, and preferably a hydrogen atom or a (C1–C6)alkyl or aryl group, and still more preferably a hydrogen atom or a (C1–C6)alkyl group, or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof. Embodiment 13. The compound of formula (II) according to embodiment 12, characterized in that R1, R2, R3, R4 each represent a hydrogen atom and R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom or –OR10. Embodiment 14. The compound of formula (II) according to embodiments 12 or 13, characterized in that at least four residues out of R5, R6, R7, R8 and R9 each represent a hydrogen atom. Embodiment 15. The compound of formula (II) according to embodiment 14, characterized in that R5, R6, R7, R8, R9, each represent, a hydrogen atom. Embodiment 16. The compound of formula (II) according to embodiment 14, characterized in that R5, R6 or R7 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group. Embodiment 17. The compound of formula (II) according to any one of embodiments 12 to 16, characterized in that it is selected from the group consisting of compounds 99, 148, 152, 153 , 154 , 155, 157, 161 , 167 , 191 , 194, 195 , 196 , and mixtures thereof, preferably 99 , 152 , 155 , 194 , and mixtures thereof. Embodiment 18. A compound of formula (II) according to any of embodiments 12 to 17,
for use as a medicament. One or more features of any embodiments disclosed herein may be combined and/or rearranged within the scope of the invention to produce further embodiments that are also within the scope of the invention. Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be within the scope of the present invention. The invention is further described by the following non-limiting Examples. Examples Examples are provided below to facilitate a more complete understanding of the invention. The following examples illustrate the exemplary modes of making and practicing the invention. However, the scope of the invention is not limited to specific embodiments disclosed in these Examples, which are illustrative only, since alternative methods can be utilized to obtain similar results. Example 1. Chemistry of dihydro-quinazoline derivatives Design, synthesis and preparations of 6-nitro-2,3-dihydro-1H-quinazolin-4-one
Polyphosphoric ester preparation (PPE) P2O4 (150 g) was refluxed in diethyl ether (150 mL) and CHCl3 (300 mL) until a clear solution occurred. Then solvents were removed under reduce pressure to give a clear oil. Synthesis of 2-amino-5-nitrobenzamide A mixture of 2-amino-5-nitro-benzonitrile (25.3 mmol, 4 g) and concentrated H2SO4 (24.2 mL) were heated to 65 °C and stirred for 12 hours. The reaction mixture was gradually poured into ice and the pH was brought to 8-9 by 6 M aq. NaOH solution or aq. NH4OH. The precipitate was filtered off and washed with ice cold H2O to give pure benzamide used without further purification. 1H NMR (400 MHz, DMSO-d6): δ ppm 8.54 (d, J= 2.7 Hz, 1H), 8.20 (br.s., 1H), 8.01 (dd, J= 9.0 Hz, 2.8 Hz, 1H), 7.87 (bs, 1H), 7.39 (br.s., 1H), 6.78 (d, J= 9.0 Hz, 1H). General procedure for 6-nitro-2,3-dihydro-1H-quinazolin-4-one:
A mixture of 2-amino-5-nitrobenzamide (1 mmol) and PPE (1 g) was added to the corresponding aldehyde (1 mmol) and the solution was heated at 80°C for 30 min without condenser. The solution thus obtained was poured into cold water. The precipitate was filtered off, washed with water and crystallized in diethyl ether to give pure corresponding DHQ with excellent yield (80-95%).
Product 50: 6-nitro-2-phenyl-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 6.01 (s, 1H) 6.83 (d, J=9.08 Hz, 1H) 7.35-7.50 (m, 5H) 8.10 (dd, J=9.08, 2.75 Hz, 1H) 8.43 (d, J=2.66 Hz, 1H) 8.58 (s, 1H) 8.74 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 66.27 (C-2), 112.63 (C-8), 114.24 (C-10), 124.17 (C- 4), 126.53 (C-2’), 128.65 (C-3’), 128.87 (C-4’), 128.97 (C-7), 137.10 (C-6), 141.10 (C- 1’), 152.13 (C-9), 161.28 (C-4). Chemical Formula: C14H11N3O3: m/z: 269.08 Product 51: 6-nitro-2-(4-hydroxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 5.90 (s, 1H) 6.73-6.85 (m, 3H) 7.28 (d, J=8.53 Hz, 2H) 8.09 (dd, J=9.26, 2.93 Hz, 1H) 8.43 (d, J=2.57 Hz, 2H) 8.57 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 66.30 (C-2), 112.63 (C-10), 114.20 (C-8), 115.26 (C- 3’), 124.25 (C-5), 128.05 (C-2’), 128.93 (C-7), 131.25 (C-1’), 137.01 (C-6), 152.30 (C- 9), 158.03 (C-4’), 161.42 (C-4). Chemical Formula: C14H11N3O4: m/z: 285.07. Product 54: 6-nitro-2-(4-methoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ pm 3.75 (s, 3H), 5.95 (s, 1H), 6.81 (d, J=9.08 Hz, 1H), 6.97 (m, J=8.80 Hz, 2H), 7.38 (m, J=8.71 Hz, 2H), 8.09 (dd, J=9.08, 2.75 Hz, 1H), 8.42 (s, 1H), 8.48 (s, 1H), 8.65 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 55.19 (OCH3), 65.91 (C-2), 112.62 (C-10), 113.90 (C- 2’), 114.20 (C-8), 124.16 (C-5), 127.94 (C-2’), 128.93 (C-7), 132.94 (C-1’), 136.99 (C- 6), 152.19 (C-9), 159.67 (C-4’), 161.36 (C-4). Chemical Formula: C15H13N3O4: m/z: 299.09. Product 55: 6-nitro-2-(3-methoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 3.75 (s, 3H) 5.98 (s, 1H) 6.84 (d, J=9.17 Hz, 1H) 6.90-6.99 (m, 1H) 6.99-7.08 (m, 2H) 7.33 (t, J=8.16 Hz, 1H) 8.05-8.17 (m, 1H) 8.43 (d, J=2.75 Hz, 1H) 8.57 (s, 1H) 8.74 (s, 1H) Chemical Formula: C15H13N3O4: m/z: 299.09. Product 56: 6-nitro-2-(2-methoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 3.91 (s, 3H, OCH3), 5.95 (s, 1H, C-H2), 6.88 (d, J=9.17 Hz, 1H, C-H8), 7.16-7.09 (m, 2H, C-H3’,5’), 7.46 (dt, 1H, J=7.1 Hz, C-H4’), 8.10 (m, 1H, C-H7), 8.20-8.18 (dd, 1H, J=1.2;7.8 Hz, C-H6’), 8.44 (d, J=2.75 Hz, 1H, C-H5) 8.57 (s, 1H), 8.75 (s, 1H). 13C NMR (75 MHz, DMSO-d6): δ 55.84(OCH3), 65.78 (C-2), 112.12 (C-10), 113.31(C-3'), 114.22 (C-8), 120.68(C-5'), 124.18 (C-5), 128.45 (C-7), 131.11(C-6'), 131.87 (C-1’), 131.99(C-4'), 134.56 (C-6), 151.31 (C-9), 158.32(C-2'), 161.44 (C-4). Chemical Formula: C15H13N3O4: m/z: 299.09. Product 60: 6-nitro-2-(4-propoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 0.96 (t, J=7.38 Hz, 3H), 1.63-1.80 (m, 2H), 3.92 (t, J=6.46 Hz, 2H), 5.95 (s, 1H), 6.82 (d, J=9.08 Hz, 1H), 6.96 (m, J=8.71 Hz, 2H), 7.37 (m, J=8.71 Hz, 2H), 8.10 (dd, J=9.08, 2.75 Hz, 1H), 8.43 (d, J=2.66 Hz, 1H), 8.48 (s, 1H), 8.64 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 10.34 (CH3), 21.95 (CH2), 65.98 (O-CH2), 69.06 (C- 2), 112.66 (C-10), 114.24 (C-8), 114.46 (C-3’), 124.20 (C-5), 127.93 (C-2’), 128.95 (C- 7), 132.89 (C-1’), 137.06 (C-6), 152.21 (C-9), 159.13 (C-4’), 161.38 (C-4). Chemical Formula: C17H17N3O4: m/z: 327.12. Product 63: 6-nitro-2-(4-butoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 0.92 (t, J=7.40 Hz, 3H), 1.35-1.48 (m, 2H), 1.62- 1.73 (m, 2H), 3.95 (t, J=6.48 Hz, 2H), 5.95 (s, 1H), 6.82 (d, J=9.08 Hz, 1H), 6.96 (m, J=8.71 Hz, 2H), 7.37 (m, J=8.71 Hz, 2H), 8.10 (dd, J=9.08, 2.75 Hz, 1H), 8.42 (d, J=2.66 Hz, 1H), 8.48 (s, 1H), 8.64 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 13.60 (CH3), 18.62 (CH2), 30.59 (CH2), 57.34 (C-2), 67.20 (OCH2), 69.06 (C-2), 112.68 (C-10), 114.22 (C-8), 114.46 (C-3’), 124.21 (C-5), 127.95 (C-2’), 128.90 (C-7), 132.90 (C-1’), 137.08 (C-6), 152.21 (C-9), 159.14 (C-4’), 161.40 (C-4). Chemical Formula: C18H19N3O4 :m/z: 341.14. Product 69: 6-nitro-2-[4-(hexyloxy)phenyl]-2,3-dihydro-1H-quinazolin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 0.81-0.94 (m, 3H), 1.30 (d, J=7.34 Hz, 4H), 1.41 (br. s., 2H), 1.69 (d, J=14.79 Hz, 2H), 3.96 (t, J=6.48 Hz, 2H), 5.95 (s, 1H), 6.82 (d, J=9.05 Hz, 1H), 6.96 (m, J=8.80 Hz, 2H), 7.36 (m, J=8.68 Hz, 2H), 8.10 (dd, J=9.11, 2.75 Hz, 1H), 8.43 (d, J=2.81 Hz, 1H), 8.47 (s, 1H), 8.63 (s, 1H). Chemical Formula: C20H23N3O4: m/z: 369.17. Product 84: 6-nitro-2-[4-(undecyloxy)phenyl]-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 0.84 (t, J=6.56 Hz, 3H), 1.24 (br. s., 14H), 1.32-1.51 (m, 2H), 1.64-1.83 (m, 2H), 4.05 (t, J=6.46 Hz, 2H), 7.06 (m, J=8.89 Hz, 2H), 7.83 (d, J=8.99 Hz, 1H), 8.16-8.32 (m, 2H), 8.50 (dd, J=8.99, 2.75 Hz, 1H), 8.81 (d, J=2.66 Hz, 1H), 12.78 (br. s., 1H, NH). Chemical Formula: C25H31N3O4: m/z: 437.23. Product 87: 6-nitro-2-(4-dodecyloxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (400 MHz, CDCl3) δ ppm 0.85 (t, J=6.72 Hz, 3H), 1.15-1.34 (m, 16 H), 1.38 (br. s., 2H), 1.61-1.75 (m, 2H), 3.95 (t, J=6.48 Hz, 2H), 5.95 (s, 1H), 6.83 (d, J=9.05 Hz, 1H), 6.95 (m, J=8.68 Hz, 2H), 7.36 (m, J=8.68 Hz, 2 H), 8.04-8.14 (m, 1H), 8.43 (d, J=2.69 Hz, 1H), 8.53 (s, 1H), 8.64 (s, 1H). 13C NMR (101 MHz, DMSO-d6) δ ppm 13.89 (CH3), 22.03 (CH2), 25.43 (CH2), 28.56 (CH2), 28.65 (CH2), 28.70 (CH2), 28.94 (3xCH2), 28.97 (CH2), 31.24 (CH2), 65.89 (OCH2), 67.53 (C-2), 112.62 (C-10), 114.21 (C-8), 114.40 (C-3’), 124.15 (C-5), 127.84 (C-2’), 128.86 (C-7), 132.93 (C-1’), 137.01 (C-6), 152.17 (C-9), 159.08 (C-4’), 161.32 (C-4). Chemical Formula: C26H35N3O4: m/z: 453.26. Product 90: 6-nitro-2-(4-tridecyloxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, CDCl3) δ ppm 0.77-0.99 (m, 3H), 1.28 (br. s., 14H), 1.40-1.55 (m, 2H), 1.68-1.92 (m, 2H), 3.96 (s, 2H), 4.71 (br. s., 1H), 5.33 (br. s., 1H), 6.24 (s, 1 H), 6.68 (d, J=8.99 Hz, 1H), 6.93 (m, J=8.71 Hz, 2H), 7.45 (m, J=8.62 Hz, 2H), 8.16 (d, J=11.55 Hz, 1H), 8.77 (d, J=2.57 Hz, 1H). 13C NMR (75 MHz, CDCl3) δ ppm 14.07 (CH3), 22.64 (CH2), 25.98 (CH2), 29.13 (CH2), 29.30 (CH2), 29.36 (CH2), 29.53 (CH2), 29.58 (2xCH2), 31.87 (CH2), 68.11 (OCH2), 68.27 (C-2), 113.84 (C-10), 114.23 (C-8), 115.10 (C-3’), 125.63 (C-5), 128.49 (C-2’), 129.35 (C-7), 129.44 (C-1’), 139.86 (C-6), 151.38 (C-9), 160.76 (C-4’), 162.66 (C-4).
Chemical Formula: C25H33N3O4: m/z: 439.25. Product 93: 6-nitro-2-(4-tetradecyloxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 0.84 (s, 3H), 1.23 (s, 22H), 1.38 (br. s., 3H), 1.61- 1.75 (m, 2H), 3.94 (t, J=6.51 Hz, 2H), 5.95 (s, 1H), 6.81 (d, J=9.08 Hz, H), 6.94 (d, J=8.71 Hz, 2H), 7.36 (d, J=8.71 Hz, 2 H), 8.09 (d, J=11.83 Hz, 1H), 8.43 (d, J=2.66 Hz, 1H), 8.48 (s, 1H), 8.64 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 13.92 (CH3), 22.08 (CH2), 25.47 (CH2), 28.60 (CH2), 28.70 (CH2), 28.74 (CH2), 29.00 (3xCH2), 29.03 (3x CH2), 31.28 (CH2), 65.95 (OCH2), 67.54 (C-2), 112.65 (C-10), 114.22 (C-8), 114.41 (C-3’), 124.19 (C-5), 127.88 (C-2’), 128.91 (C-7), 132.89 (C-1’), 137.04 (C-6), 152.19 (C-9), 159.12 (C-4’), 161.36 (C-4). Chemical Formula: C28H39N3O4: m/z: 481.29. Product 96: 6-nitro-2-(4-phenoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 6.02 (s, 1H) 6.84 (d, J=9.17 Hz, 1H) 7.05 (d, J=8.62 Hz, 2H) 7.00 (d, J=8.80 Hz, 2H) 7.12-7.21 (m, 1H) 7.35-7.45 (m, 2H) 7.49 (d, J=8.62 Hz, 2H) 8.11 (dd, J=9.08, 2.75 Hz, 1H) 8.44 (d, J=2.75 Hz, 1H) 8.53 (s, 1H) 8.71 (s, 1H) Chemical Formula: C20H15N3O4: m/z: 361.11. Product 98: 6-nitro-2-(2-phenoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 6.28 (s, 1H), 6.74 (d, J=9.08 Hz, 1H), 6.84 (d, J=7.89 Hz, 1H), 6.97 (d, J=7.70 Hz, 2H), 7.07-7.24 (m, 2H), 7.29-7.43 (m, 3H), 7.54 (d, J=8.99 Hz, 1H), 7.96-8.08 (m, 1H), 8.40 (d, J=2.66 Hz, 1H), 8.47 (s, 1H), 8.63 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 62.50 (C-2), 112.42 (C-10), 114.00 (C-8), 118.31 (C- 5’), 118.87 (C-8’), 123.59 (C-3’), 123.72 (C-5), 124.05 (C-2’), 128.42 (C-4’), 128.76 (C- 7), 129.90 (C-9’), 130.52 (C-10’), 131.28 (C-1’), 136.85 (C-6), 152.10 (C-9), 154.46 (C- 7’), 156.22 (C-6’), 161.24 (C-4). Chemical Formula: C20H15N3O4: m/z: 361.11.
General procedure for 7-nitro-2,3-dihydro-1H-quinazolin-4-one:
A mixture of 2-amino-4-nitrobenzamide (CAS: 31930-18-4, 1 mmol) and PPE (1 g) was added to the corresponding aldehyde (1 mmol) and the solution was heated at 80°C for 30 min without condenser. The solution thus obtained was poured into cold water. The precipitate was filtered off, washed with water and crystallized in diethyl ether to give pure corresponding DHQ with excellent yield (80-95%). Product 300: 7-nitro-2-[4-(dodecyloxy)phenyl]-2,3-dihydroquinazolin-4(1H)-one
1H NMR (300 MHz, DMSO-d6) δ ppm : 0.85 (t, J=6.24 Hz, 3H, CH3), 1.24 (br. s., 16 H, CH2), 1.38 (br. s., 2H, CH2), 1.61 - 1.81 (m, 2H, CH2), 3.94 (t, J=6.28 Hz, 2H, OCH2), 5.85 (s, 1H, C-H2), 6.94 (d, J=8.44 Hz, 2H, C-H3',5'), 7.35 - 7.46 (m, 3H, C-H2',6', C-H6), 7.57 (d, 1H, C-H8), 7.65 (s, 1H, N-H), 7.83 (d, J=8.53 Hz, 1H, C-H5), 8.61 (s, 1H, N-H). 13C NMR (75 MHz, DMSO-d6) δ ppm: 13.91 (CH3), 22.06 (CH2), 25.45 (CH2), 28.59 (CH2), 28.67 (CH2), 28.72 (CH2), 28.96 (3xCH2), 29.00 (CH2), 31.26 (CH2), 66.04 (C-2), 67.50 (OCH2), 108.79 (C-8), 110.89 (C-6), 114.29 (C-3',5'), 119.25 (C-10), 128.02 (C-2',6'), 129.07 (C-5), 132.75 (C-1'), 148.24 (C-9), 150.72 (C-7), 159.01 (C-4'), 161.86 (C-4). Chemical Formula: C26H35N3O4: m/z: 453,26 Product 313: 7-nitro-2-[4-(tetradecyloxy)phenyl]-2,3-dihydroquinazolin-4(1H)-one
1H NMR (300 MHz, DMSO-d6) δ ppm: 0.78 - 0.91 (t, 3H, CH3), 1.23-1.38 (m, 22H), 1.70 (br. s., 2H, CH2), 3.94 (t, J=6.46 Hz, 2H, OCH2), 5.84 (s, 1 H, C-H2), 6.94 (d, J=8.71 Hz, 2H, C-H3',5'), 7.38 (d, J=8.71 Hz, 2H, C-H2',6'), 7.40 - 7.45 (dd, J=2.2, 8.53 Hz,1H, C-H6), 7.57 (d, J=2.20 Hz, 1H, C-H8), 7.68 (s, 1H, NH), 7.83 (d, J=8.53 Hz, 1H, C-H5), 8.61 (s,
1H, NH). 13C NMR (DMSO-d6) δ ppm: 13.91 (CH3), 22.06 (C-n), 25.46 (C-g), 28.60 (1C), 28.67 (2C), 28.73 (1C), 28.98 (4C), 29.00 (C-b), 31.26 (C-m), 66.03 (C-2), 67.51 (C-a), 108.81 (C- 8), 110.88 (C-6), 114.29 (C-3',5'), 119.26 (C-10), 128.02 (C-2',6'), 129.07 (C-5), 132.78 (C-1'), 148.24 (C-9), 150.73 (C-7), 159.02 (C-4'), 161.87 (C-4). Chemical Formula: C28H39N3O4: m/z: 481,29 Example 2. Chemistry of dihydro-benzothiazine derivatives A) Designs, synthesis and preparations of 6-nitro-2H-benzo[e][1,3]thiazin- 4(3H)-one The design, synthesis and preparation of 6-nitro-2-(phenyl)-2H-benzo[e][1,3]thiazin- 4(3H)-one has never been described. The design, synthesis and preparation of 2-aryl- 2,3-dihydro-4H-1,3-benzothiazin-4-one and 2-aryl-2,3-dihydro-4-one derivatives has been multiple times cited (Boudet 1959; Geng et al. 2012; Ingram et McClelland 1947; Moreau et Delacoux 1962). Our synthetic route begins as below to obtain thiobenzamide:
2-chloro-5-nitrobenzamide RMN 1H (300 MHz, DMSO-d6) δ ppm 7.78-7.81 (m, 1H, C-H3), 7.86 (br.s., 1H, NH), 8.12 (brs, 1H, NH), 8.23-8.25 (m, 2H, C-H4,6). RMN 13C (75 MHz, DMSO-d6) δ ppm 123.61(C-3), 125.41(C-6), 131.51(C-4), 137.01(C-2), 138.17(C-1), 146.17(C-5), 166.25(CONH2). Chemical Formula: C7H5ClN2O3: m/z: 200.00. 2-mercapto-5-nitrobenzamide 1H (300 MHz, DMSO-d6) δ ppm 7.37-7.40 (d, 1H, J= 8.88 Hz, C-H3), 7.43 (br. s., 1H, NH), 7.50-7.54 (dd, 1H, J= 2.83, 8.88 Hz, C-H4), 8.77 (d, 1H, J= 3.02 Hz, C-H6), 11.41 (brs, 1H, NH). 13C (300 MHz, DMSO-d6) δ ppm, 120.61(C-3), 125.92(C-6), 132.14(C-4), 138.22(C-2), 138.47(C-1), 168.12(CONH2), n.o. (C-5). Chemical Formula: C7H6N2O3S: m/z: 198.01. General procedure
A mixture of 2-mercapto-5-nitrobenzamide (1mmol) and PPE (1 g) was added to the corresponding aldehyde (1 mmol) and the solution was heated to 100°C for 60 min without condenser. The solution thus obtained was poured into cold water. The precipitate was filtered off, washed with water and crystalized in diethyl ether to give pure corresponding dihydro-benzothiazine with excellent yield (80-95%).
Product 148: 6-nitro-2-phenyl-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm: 6.31 (d, J=3.76 Hz, 1 H, C-H2), 7.29 - 7.43 (m, 3 H, C-H3',4',5'), 7.43 - 7.53 (m, 2 H, C-H2',6'), 7.63 (d, J=8.71 Hz, 1 H, C-H8), 8.24 (dd, J=8.67, 2.61 Hz, 1 H, C-H7), 8.70 (d, J=2.66 Hz, 1 H, C-H5), 9.43 (d, J=3.67 Hz, 1 H, N-H). 13C NMR (75 MHz, DMSO-d6) δ ppm: 57.36 (C-2), 123.80 (C-5), 126.38 (C-8), 126.86 (C- 10), 128.62 (C-2',6'), 128.67 (C-7), 128.70 (C-4'), 128.86 (C-3',5'), 138.04 (C-1'), 144.63 (C-6), 145.32 (C-9), 162.77 (C-4). Product 149: 6-nitro-2-(4-hydroxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 6.21 (d, J=3.18 Hz, 1H), 6.76 (m, J=8.68 Hz, 2H), 7.31 (m, J=8.68 Hz, 2H), 7.62 (d, J=8.56 Hz, 1H), 8.18-8.27 (m, 1H), 8.69 (d, J=2.69 Hz, 1H), 9.27 (d, J=3.18 Hz, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 57.71 (C-2), 115.29 (C-3’), 123.85 (C-5), 126.25 (C- 10), 127.39 (C-8), 128.46 (C-2’), 128.50 (C-1’), 128.75 (C-7), 145.21 (C-9), 145.28 (C- 6), 158.01 (C-4’), 162.92 (C-4). Chemical Formula: C14H10N2O4S: m/z: 302.04. Product 151: 6-nitro-2-(2-hydroxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 6.25 (d, J=3.91 Hz, 1H), 6.78 (t, J=7.52 Hz, 1H), 6.87 (d, J=7.46 Hz, 1H), 7.17 (t, J=8.50 Hz, 1H), 7.22 (d, J=7.58 Hz, 1H), 7.58 (d, J=8.56 Hz, 1H), 8.22 (dd, J=8.68, 2.69 Hz, 1H), 8.74 (d, J=2.57 Hz, 1H), 9.18 (d, J=3.91 Hz, 1H), 10.16 (s, 1 H, OH). 13C NMR (101 MHz, DMSO-d6) δ ppm 52.11 (C-2), 54.85 (C-3’), 115.53 (C-5’), 119.01 (C- 5’), 123.87 (C-5), 124.01 (C-10), 126.28 (C-8), 126.80 (C-6’), 128.43 (C-1’), 128.62 (C- 7), 129.90 (C-4’), 145.11 (C-9), 153.89 (C-2’), 163.01 (C-4). Chemical Formula: C14H10N2O4S: m/z: 302.04. Product 152: 6-nitro-2-(4-methoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 3.75 (s, 3H), 6.27 (d, J=3.55 Hz, 1H), 6.95 (m, J=8.80 Hz, 2H), 7.42 (m, J=8.68 Hz, 2H), 7.64 (d, J=8.68 Hz, 1H), 8.25 (d, J=11.37 Hz, 1H), 8.70 (s, 1 H), 9.36 (br. s., 1H). Chemical Formula: C15H12N2O4S: m/z: 316.05. Product 153: 6-nitro-2-(3-methoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 3.74 (s, 3H), 6.27 (d, J=3.85 Hz, 1H), 6.94 (s, 1H), 7.04 (s, 2H), 7.25-7.36 (m, 1H), 7.64 (d, J=8.71 Hz, 1H), 8.24 (dd, J=8.71, 2.66 Hz, 1H), 8.70 (d, J=2.57 Hz, 1H), 9.42 (d, J=3.85 Hz, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 55.13 (OCH3), 57.26 (C-2), 112.68 (C-2’), 114.22 (C- 4’), 119.01 (C-6’), 123.77 (C-5), 125.16 (C-10), 126.41 (C-8), 128.67 (C-7), 129.77 (C- 5’), 139.54 (C-1’), 144.65 (C-9), 145.34 (C-6), 159.21 (C-3’), 162.74 (C-4). Chemical Formula: C15H12N2O4S: m/z: 316.05. Product 154: 6-nitro-2-(2-methoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 3.85 (s, 3H), 6.27 (d, J=4.16 Hz, 1H), 6.93 (t, J=7.52 Hz, 1H), 7.07 (d, J=7.95 Hz, 1H), 7.28 (d, J=9.05 Hz, 1H), 7.34 (t, J=8.62 Hz, 1H), 7.57 (d, J=8.68 Hz, 1H), 8.16-8.26 (m, 1H), 8.74 (d, J=2.69 Hz, 1H), 9.21 (d, J=4.28 Hz, 1H). 13C NMR (101 MHz, DMSO-d6) δ ppm 51.85 (C-2), 55.73 (OCH3), 111.49 (C-3’), 120.37 (C-5’), 123.86 (C-5), 125.79 (C-1’), 126.31 (C-8), 126.70 (C-6’), 128.33 (C-10), 128.62 (C-7), 130.26 (C-4’), 144.77 (C-6), 145.20 (C-9), 155.53 (C-2’), 162.90 (C-4). Chemical Formula: C15H12N2O4S: m/z: 316.05. Product 155: 6-nitro-2-(4-ethoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 1.30 (t, J=6.97 Hz, 3H), 4.01 (q, J=6.97 Hz, 2H), 6.26 (d, J=3.42 Hz, 1H), 6.93 (d, J=8.68 Hz, 2H), 7.40 (d, J=8.68 Hz, 2H), 7.63 (d, J=8.68 Hz, 1H), 8.24 (dd, J=8.68, 2.69 Hz, 1H), 8.70 (d, J=2.57 Hz, 1H), 9.35 (d, J=3.30 Hz, 1H). Chemical Formula: C16H14N2O4S: m/z: 330.07. Product 157: 6-nitro-2-(2-ethoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 1.36 (t, J=6.97 Hz, 3H), 4.07-4.16 (m, 2H), 6.26 (d, J=4.40 Hz, 1H), 6.91 (t, J=7.52 Hz, 1H), 7.06 (d, J=7.83 Hz, 1H), 7.24 (s, 1H), 7.27- 7.36 (m, 1H), 7.57 (d, J=8.68 Hz, 1H), 8.21 (dd, J=8.68, 2.69 Hz, 1H), 8.75 (d, J=2.57 Hz, 1H), 9.20 (d, J=4.28 Hz, 1H). 13C NMR (101 MHz, DMSO-d6) δ ppm 14.52 (CH3), 51.73 (C-2), 63.79 (OCH2), 112.30 (C- 3’), 120.25 (C-5’), 123.87 (C-5), 126.17 (C-1’), 126.36 (C-8), 126.58 (C-6’), 128.21 (C- 10), 128.65 (C-7), 130.16 (C-4’), 144.76 (C-6), 145.19 (C-9), 154.71 (C-2’), 162.86 (C- 4). Chemical Formula: C16H14N2O4S: m/z: 330.07.
Product 158: 6-nitro-2-(4-propoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 0.95 (t, J=7.38 Hz, 3H), 1.60-1.82 (m, 2H), 3.91 (t, J=6.51 Hz, 2H), 6.26 (d, J=3.48 Hz, 1H), 6.93 (m, J=8.71 Hz, 2H), 7.40 (m, J=8.71 Hz, 2H), 7.63 (d, J=8.71 Hz, 1H), 8.24 (dd, J=8.62, 2.66 Hz, 1H), 8.70 (d, J=2.57 Hz, 1H), 9.34 (d, J=3.48 Hz, 1H). Chemical Formula: C17H16N2O4S: m/z: 344.08. Product 161: 6-nitro-2-(4-butoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 0.92 (t, J=7.40 Hz, 3H), 1.35-1.48 (m, 2H), 1.62- 1.73 (m, 2H), 3.95 (t, J=6.48 Hz, 2H), 6.26 (d, J=3.55 Hz, 1H), 6.93 (m, J=8.68 Hz, 2H), 7.40 (m, J=8.56 Hz, 2H), 7.63 (d, J=8.68 Hz, 1H), 8.19-8.28 (m, 1H), 8.70 (d, J=2.57 Hz, 1H), 9.34 (d, J=3.42 Hz, 1H). 13C NMR (101 MHz, DMSO-d6) δ ppm 13.59 (CH3), 18.64 (CH2), 30.61 (CH2), 57.34 (C- 2), 67.19 (OCH2), 114.41 (C-3’), 123.80 (C-5), 126.28 (C-8), 128.33 (C-2’), 128.58 (C- 10), 128.73 (C-7), 129.26 (C-1’), 145.01 (C-6), 145.25 (C-9), 159.07 (C-4’), 162.84 (C- 4). Chemical Formula: C18H18N2O4S: m/z: 358.10. Product 167: 6-nitro-2-[4-(hexyloxy)phenyl]-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 0.80-0.92 (m, 3H), 1.30 (d, J=7.21 Hz, 4H), 1.40 (d, J=6.72 Hz, 2H), 1.62-1.74 (m, 2H), 3.94 (t, J=6.54 Hz, 2H), 6.26 (d, J=3.42 Hz, 1H), 6.93 (m, J=8.68 Hz, 2H), 7.39 (m, J=8.68 Hz, 2H), 7.63 (d, J=8.68 Hz, 1H), 8.24 (dd, J=8.68, 2.69 Hz, 1H), 8.70 (d, J=2.57 Hz, 1H), 9.34 (d, J=3.42 Hz, 1H). 13C NMR (101 MHz, DMSO-d6) δ ppm 13.84 (CH3), 22.00 (CH2), 25.11 (CH2), 28.54 (CH2), 30.91 (CH2), 57.35 (C-2), 67.52 (OCH2), 114.43 (C-3’), 123.82 (C-5), 126.31 (C-8), 128.35 (C-2’), 128.60 (C-10), 128.74 (C-7), 129.27 (C-1’), 145.03 (C-6), 145.26 (C-9), 159.08 (C-4’), 162.87 (C-4).
Chemical Formula: C20H22N2O4S: m/z: 386.13. Product 182: 6-nitro-2-[4-(undecyloxy)phenyl]-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 0.75-0.98 (m, 3H), 1.24 (s, 14H), 1.38 (br. s., 2H), 1.57-1.80 (m, 2H), 3.93 (t, J=6.46 Hz, 2H), 6.25 (d, J=3.48 Hz, 1H), 6.92 (m, J=8.80 Hz, 2H), 7.39 (m, J=8.71 Hz, 2H), 7.63 (d, J=8.71 Hz, 1H), 8.24 (d, J=11.37 Hz, 1H), 8.69 (d, J=2.66 Hz, 1H), 9.34 (d, J=3.39 Hz, 1H). Chemical Formula: C25H32N2O4S: m/z: 456.21. Product 185: 6-nitro-2-(4-dodecyloxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 0.85 (t, J=6.72 Hz, 3H), 1.24 (br. s., 16H), 1.38 (br. s., 2H), 1.62-1.75 (m, 2H), 3.94 (t, J=6.48 Hz, 2H), 6.26 (d, J=3.42 Hz, 1H), 6.92 (m, J=8.80 Hz, 2H), 7.39 (m, J=8.80 Hz, 2H), 7.63 (d, J=8.68 Hz, 1H), 8.24 (dd, J=8.68, 2.57 Hz, 1H), 8.70 (s, 1H), 9.34 (d, J=3.30 Hz, 1H). Chemical Formula: C26H34N2O4S: m/z: 470.22. Product 191: 6-nitro-2-(4-tetradecyloxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 0.85 (t, J=6.72 Hz, 3H), 1.24 (br. s., 16H), 1.38 (br. s., 2H), 1.62-1.75 (m, 2H), 3.94 (t, J=6.48 Hz, 2H), 6.26 (d, J=3.42 Hz, 1H), 6.92 (m, J=8.80 Hz, 2H), 7.39 (m, J=8.80 Hz, 2H), 7.63 (d, J=8.68 Hz, 1H), 8.24 (dd, J=8.68, 2.57 Hz, 1H), 8.70 (s, 1H), 9.34 (d, J=3.30 Hz, 1H). 13C NMR (101 MHz, DMSO-d6) δ ppm 13.81 (CH3), 22.03 (CH2), 25.11 (CH2), 25.43 (CH2), 28.56 (CH2), 28.65 (CH2), 28.70 (2xCH2), 28.94 (3xCH2), 28.97 (CH2), 31.21 (CH2), 57.35 (C-2), 67.52 (OCH2), 114.43 (C-3’), 123.82 (C-5), 126.31 (C-8), 128.35 (C-2’), 128.60 (C-10), 128.74 (C-7), 129.27 (C-1’), 145.03 (C-6), 145.26 (C-9), 159.08 (C-4’), 162.87 (C-4). Chemical Formula: C26H34N2O4S: m/z: 470.22.
Product 194: 6-nitro-2-(4-phenoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 6.32 (d, J=3.55 Hz, 1H), 6.96-7.06 (m, 4H), 7.13- 7.23 (m, 1H), 7.35-7.45 (m, 2H), 7.50 (d, J=8.56 Hz, 2H), 7.65 (d, J=8.56 Hz, 1H), 8.26 (d, J=2.69 Hz, 1H), 8.70 (d, J=2.57 Hz, 1H), 9.40 (d, J=3.67 Hz, 1H). 13C NMR (101 MHz, DMSO-d6) δ ppm 57.06 (C-2), 118.12 (C-8’), 119.13 (C-3’), 123.85 (C-5), 123.95 (C-10’), 126.37 (C-8), 128.63 (C-1’), 128.66 (C-10), 128.79 (C-9’), 130.12 (C-2’), 132.44 (C-7), 144.74 (C-6), 145.32 (C-9), 155.90 (C-4’), 157.34 (C-7’), 162.74 (C-4). Chemical Formula: C20H14N2O4S: m/z: 378.07. Product 195: 6-nitro-2-(3-phenoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 6.28 (d, J=4.16 Hz, 1H), 6.91-7.01 (m, 3H), 7.01- 7.05 (m, 1H), 7.10-7.18 (m, 1H), 7.22 (d, J=7.83 Hz, 1H), 7.33-7.44 (m, 3H), 7.62 (d, J=8.68 Hz, 1H), 8.24 (d, J=2.69 Hz, 1H), 8.63 (d, J=2.57 Hz, 1H), 9.44 (d, J=4.16 Hz, 1H). Chemical Formula: C20H14N2O4S: m/z: 378.07. Product 196: 6-nitro-2-(2-phenoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 6.42 (d, J=3.85 Hz, 1 H), 6.84 (d, J=8.16 Hz, 1 H), 7.04 (d, J=8.71 Hz, 2 H), 7.16 (q, J=7.67 Hz, 2 H), 7.30-7.45 (m, 3 H), 7.50 (d, J=9.26 Hz, 1 H), 7.59 (d, J=8.71 Hz, 1 H), 8.18-8.25 (m, 1 H), 8.72 (d, J=2.66 Hz, 1 H), 9.34 (d, J=3.85 Hz, 1 H). 13C NMR (75 MHz, DMSO-d6) δ ppm 52.19 (C-2), 118.22 (C-3’), 118.93 (C-8’), 123.54 (C- 10’), 123.89 (C-5), 123.98 (C-5’), 126.36 (C-8), 127.86 (C-6’), 128.36 (C-1’), 128.38 (C- 10), 128.61 (C-7), 130.09 (C-9’), 130.57 (C-4’), 144.45 (C-6), 145.27 (C-9), 153.58 (C-
2’), 156.13 (C-7’), 162.88 (C-4). Chemical Formula: C20H14N2O4S: m/z: 378.07. General procedure Reduction of nitro group to amino is done in a hydrogenator apparatus. Nitro derivatives (1 mmol) were introduced in the flask with Pd/C (10% in weight) and ethanol (20 ml). The flask was shacked under 50 psi of hydrogen at room temperature until no more starting material appeared in TLC. Then the solution was filtered off and the solvent was removed under reduce pressure. The desired amino derivatives were then crystallized in diethyl ether. Product 96: 6-nitro-2-(4-phenoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 6.02 (s, 1H) 6.84 (d, J=9.17 Hz, 1H) 7.05 (d, J=8.62 Hz, 2H) 7.00 (d, J=8.80 Hz, 2H) 7.12-7.21 (m, 1H) 7.35-7.45 (m, 2H) 7.49 (d, J=8.62 Hz, 2H) 8.11 (dd, J=9.08, 2.75 Hz, 1H) 8.44 (d, J=2.75 Hz, 1H) 8.53 (s, 1H) 8.71 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm: 66.17 (C-2), 114.40 (C-8), 114.89 (C-10), 118.26 (C-8'',C-12'), 118.71 (C-3',C-5'), 123.61 (C-10'), 124.35 (C-5), 128.30 (C-7), 128.68 (C- 2',C-6'), 130.07 (C-9',C-11'), 136.51 (C-1'), 137.15 (C-6), 147.85 (C-9), 156.45 (C-4'), 156.86 (C-7'), 163.56 (C-4). Chemical Formula: C20H15N3O4: m/z: 361.11. Product 97: 6-nitro-2-(3-phenoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm: 6.01 (br. s., 1 H, C-H2), 6.82 (t., 1 H, C-H10'), 6.99 (d, J=7.61 Hz, 3 H, C-H8,9',11'), 7.07 - 7.19 (m, 2 H, C-H8',12'), 7.23 (d, J=7.24 Hz, 1 H, C- H2'), 7.31 - 7.51 (m, 3 H, C-H4',5',6'), 8.09 (d, J=8.89 Hz, 1 H, C-H7), 8.41 (br. s., 1 H, C- H5), 8.59 (br. s., 1 H, N-H), 8.78 (br. s., 1 H, N-H). 13C NMR (75 MHz, DMSO-d6) δ ppm: 65.73 (C-2), 112.74 (C-10), 114.32 (C-8), 116.46 (C- 2'), 118.67 (C-4'), 118.78 (C-8',12'), 121.32 (C-10'), 123.72 (C-6'), 124.15 (C-5), 128.98
(C-7), 130.06 (C-9',11'), 130.41 (C-5'), 137.23 (C-6), 143.41 (C-1'), 152.05 (C-9), 156.15 (C-3'), 156.88 (C-7'), 161.26 (C-4). Product 98: 6-nitro-2-(2-phenoxyphenyl)-2,3-dihydro-1H-quinazolin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 6.28 (s, 1H), 6.74 (d, J=9.08 Hz, 1H), 6.84 (d, J=7.89 Hz, 1H), 6.97 (d, J=7.70 Hz, 2H), 7.07-7.24 (m, 2H), 7.29-7.43 (m, 3H), 7.54 (d, J=8.99 Hz, 1H), 7.96-8.08 (m, 1H), 8.40 (d, J=2.66 Hz, 1H), 8.47 (s, 1H), 8.63 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 62.50 (C-2), 112.42 (C-10), 114.00 (C-8), 118.31 (C- 5’), 118.87 (C-8’), 123.59 (C-3’), 123.72 (C-5), 124.05 (C-2’), 128.42 (C-4’), 128.76 (C- 7), 129.90 (C-9’), 130.52 (C-10’), 131.28 (C-1’), 136.85 (C-6), 152.10 (C-9), 154.46 (C- 7’), 156.22 (C-6’), 161.24 (C-4). Chemical Formula: C20H15N3O4: m/z: 361.11. Product 99: 6-amino-2-phenyl-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 5.28 (br. s, 2H, NH2), 5.84 (s, 1H, C-H2), 6.61 - 6.69 (m, 1 H), 6.92 (d, J=8.31 Hz, 1H), 7.30 - 7.43 (m, 3 H, C-H3',4',5'), 7.41 - 7.523 (m, 2 H, C-H2',6'), 8.61 (d, J=3.18 Hz, 1H), 9.54 (s, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 57.85 (C-2), 114.48 (C-5), 114.99 (C-3’), 118.17 (C- 7), 120.03 (C-10), 127.64 (C-8), 128.62 (C-2',6'), 128.70 (C-4'), 128.86 (C-3',5'), 128.59 (C-9), 129.29 (C-1’), 147.11 (C-6), 164.98 (C-4). Product 100: 6-amino-2-(4-hydroxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 5.28 (br. s, 2H, NH2), 5.86 (s, 1H), 6.61 - 6.69 (m, 1 H), 6.73 (d, 2H), 6.92 (d, J=8.31 Hz, 1H), 7.21-7.31 (m, 3H), 8.61 (d, J=3.18 Hz, 1H), 9.54 (s, 1H).
13C NMR (75 MHz, DMSO-d6) δ ppm 57.85 (C-2), 114.48 (C-5), 114.99 (C-3’), 118.17 (C- 7), 120.03 (C-10), 127.64 (C-8), 128.34 (C-2’), 128.59 (C-9), 129.29 (C-1’), 147.15 (C- 6), 157.55 (C-4’), 164.98 (C-4). Chemical Formula: C14H12N2O2S: m/z: 272.06. Product 145: 6-amino-2-(4-phenoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 5.30 (br. s., 2H), 5.96 (d, J=3.55 Hz, 1H), 6.67 (dd, J=8.31, 2.57 Hz, 1H), 6.91-6.99 (m, 3H), 6.99-7.05 (m, 2H), 7.13-7.19 (m, 1H), 7.25 (d, J=2.57 Hz, 1H), 7.36-7.43 (m, 2H), 7.46 (d, J=8.56 Hz, 2H), 8.76 (d, J=3.67 Hz, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 57.15 (C-2), 114.44 (C-5), 117.98 (C-6’), 118.22 (C- 7), 118.95 (C-3’), 119.37 (C-10), 123.76 (C-8’), 127.75 (C-8), 128.70 (C-7’), 129.26 (C- 9), 130.09 (C-2’), 133.72 (C-1’), 147.26 (C-6), 156.15 (C-5’), 156.79 (C-4’), 164.79 (C- 4). Chemical Formula: C20H16N2O2S: m/z: 348.09. Product 146: 6-amino-2-(3-phenoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 5.10 (br. s., 2H, NH2), 6.03 (br. s., 1H), 6.84-6.97 (m, 3H), 7.04-7.14 (m, 2H), 7.18 (d, J=7.58 Hz, 1H), 7.25-7.39 (m, 4H), 7.47 (d, J=7.95 Hz, 1H), 7.99 (s, 1H), 9.13 (br. s., 1H). Chemical Formula: C20H16N2O2S: m/z: 348.09. Product 158: 6-nitro-2-(4-propoxyphenyl)-2,3-dihydro-1,3-benzothiazin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 0.95 (t, J=7.38 Hz, 3H), 1.60-1.82 (m, 2H), 3.91 (t, J=6.51 Hz, 2H), 6.26 (d, J=3.48 Hz, 1H), 6.93 (m, J=8.71 Hz, 2H), 7.40 (m, J=8.71 Hz, 2H), 7.63 (d, J=8.71 Hz, 1H), 8.24 (dd, J=8.62, 2.66 Hz, 1H), 8.70 (d, J=2.57 Hz, 1H), 9.34 (d, J=3.48 Hz, 1H). 13C NMR (75 MHz, DMSO-d6) δ ppm 10.34 (CH3), 21.95 (CH2), 65.54 (O-CH2), 114.42 (C-
3’), 123.80 (C-5), 126.28 (C-8), 128.33 (C-2’), 128.58 (C-10), 128.73 (C-7), 129.26 (C- 1’), 145.01 (C-6), 145.25 (C-9), 159.10 (C-4’), 162.83 (C-4). Chemical Formula: C17H16N2O4S: m/z: 344.08. Example 3. Chemistry of dihydro-benzoxazine derivatives General procedure: Using the same method as for benzothiazine:
Product 243: 6-amino-2-(4-phenoxyphenyl)-2,3-dihydro-1,3-benzoxazin-4-one 1H NMR (400 MHz, DMSO-d6) δ ppm 6.17 (d, 1H, J= 1.33 Hz, C-H2), 6.75 (m, 2H, C-H5,7), 6.01-6.04 (d, 4H, J= 8.5 Hz, C-H3’,6’), 7.17 (dt, 1H, J= 7.55 Hz, C-H8’), 7.41 (dt, 2H, J= 7.56 Hz, C-H7’), 7.54-7.57 (d, 2H, J= 8.5 Hz, C-H2’), 8.72 (d, 1H, J= 1.3 Hz, N-H3). 13C NMR (75 MHz, DMSO-d6) δ ppm 84.2(2), 111.44(5), 117.12(8), 118.23(3'), 118.87(4a), 119.28(6'), 120.84(7), 124.11(8'), 129.46(2'), 130.37(7'), 132.31(1'), 143.59(6), 148.32(4'), 156.33(5'), 157.93(8a), 163.37(4). Chemical Formula: C20H16N2O3: m/z: 332.12. Product 244: 6-amino-2-(3-phenoxyphenyl)-2,3-dihydro-1,3-benzoxazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 5.30 (bs, 2H, NH2), 6.17 (d, 1H, J= 1.70 Hz, C-H2), 6.73 (m, 2H, C-H5,7), 6.99-7.04 (m, 4H, C-H8’,9’), 7.14-7.16 (m, 2H, C-H10’,8), 7.29-7.31 (d, 1H, J= 7.74 Hz, C-H6’), 7.36-7.44 (m, 3H, C-H2’,4’,5’), 8.76 (d, 1H, N-H3). 13C NMR (75 MHz, DMSO-d6) δ ppm 83.83(2), 111.39(5), 117.1(2'), 117.36(8), 118.88(4a), 118.91(8'), 119.51(7), 120.78(4'), 122.38(6'), 123.79(10'), 130.19(9'), 130.22(5'), 139.72(1'), 143.57(6), 148.02(8a), 156.46(3'), 156.81(7'), 163.08(4). Chemical Formula: C20H16N2O3: m/z: 332.12. Product 246: 6-nitro-2-phenyl-2,3-dihydro-1,3-benzoxazin-4-one
1H NMR (300 MHz, DMSO-d6) δ ppm 6.59 (d, 1H, J= 1.13 Hz, C-H2), 7.29-7.32 (d, 1H, J= 9.07 Hz, C-H8), 7.42-7.48 (m, 3H, C-H3’,4’,5’), 7.58-7.60 (d, 2H, J= 8.68 Hz, C-H2’,6’), 8.34-8.38 (dd, 1H, J= 2.83, 9.07 Hz, C-H7), 8.54 (d, 1H, J= 2.84 Hz, C-H5), 9.41 (bs, 1H, NH). 13C NMR (75 MHz, DMSO-d6) δ ppm 85.56(C-2), 118.61 (C-10), 118.74 (C-8), 123.41 (C- 5), 127.65 (C-2’,6’), 128.89(C-3’,5’), 129.79 (C-4’), 130.27 (C-7), 136.16 (C-1’), 142.37 (C-6), 161.14 (C-9), 161.64 (C-4). Chemical Formula: C14H10N2O4 :m/z: 270.06 Product 292: 6-nitro-2-(4-phenoxyphenyl)-2,3-dihydro-1,3-benzoxazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 6.58 (d, 1H, J= 1.13 Hz, C-H2), 7.03-7.08 (m, 4H, C-H3’,6’), 7.19 (t, 1H, J= 7.55 Hz, C-H8’), 7.29-7.32 (d, 1H, J= 9.07 Hz, C-H8), 7.42 (dt, 2H, J= 7.37 Hz, C-H7’), 7.59-7.62 (d, 2H, J= 8.68 Hz, C-H2’), 8.35-8.39 (dd, 1H, J= 2.83, 9.07 Hz, C-H7), 8.54 (d, 1H, J= 2.84 Hz, C-H5), 9.37 (bs, 1H, NH). 13C NMR (75 MHz, DMSO-d6) δ ppm 85.35(2), 118.27(3'), 118.53(4a), 118.67(8), 119.44(6'), 123.4(5), 124.28(8'), 129.68(2'), 129.74(7), 130.38(7'), 130.73(1'), 142.33(6), 156.05(4'), 158.5(5'), 161.18(8a), 161.68(4). Chemical Formula: C20H14N2O5: m/z: 362.09. Product 293: 6-nitro-2-(3-phenoxyphenyl)-2,3-dihydro-1,3-benzoxazin-4-one
1H NMR (400 MHz, DMSO-d6) δ ppm 6.58 (d, 1H, J= 1.32 Hz, C-H2), 7.00-7.03 (d, 2H, J= 7.75 Hz, C-H8’), 7.07-7.19 (m, 3H, C-H9’,10’), 7.28-7.31 (d, 1H, J= 9.07 Hz, C-H8), 7.33- 7.48 (m, 4H, C-H2’,4’,5’,6’), 8.33-8.37 (dd, 1H, J= 2.83, 9.07 Hz, C-H7), 8.50-8.51(d, 1H, J= 2.84 Hz, C-H5), 9.43 (bs, 1H, NH). 13C NMR (75 MHz, DMSO-d6) δ ppm 85.05(2), 117.39(2'), 118.59(4ª), 118.77(8),
119.13(8'), 120.16(4'), 122.51(6'), 123.39(5), 124.1(10'), 129.81(7), 130.37(9'), 130.73(5'), 138.33(1'), 142.43(6), 156.33(3'), 157.12(7'), 160.99(8a), 161.48(4). Chemical Formula: C20H14N2O5: m/z: 362.09. Example 4. Binding activities 4.1. Binding activities on OX1R and OX2R sites These compounds express an activity on OX1R and/or OX2R as agonist which is determined in accordance with the following general experimental method by Eurofins using Chinese hamster ovary (CHO) cells expressing human OX1R, and human embryonic kidney (HEK), 293 cells expressing human OX2R. Material and methods OX1 (h) (agonist radioligand) human recombinant (CHO cells); Ligand [125I]orexin A; Ligand concentration: 0.1 nM ; Ligand Kd: 0.87 nM; Non-specific: SB 334867 (1 μM); Incubation: 60 min at RT scintillation counting; Control inhibitor: Orexin-A; Test concentration/dose: IC/EC50 calculation are provided if 5 or more concentrations are selected; Test sample requirements: Minimum for 1) Screen: 60 μl of 10 mM stock -OR- 1 mg (pre-weighed) for 10µM final testing. 2) Dose Response: 90 μl of 10 mM stock - OR- 1 mg (pre-weighed) for a top concentration at 10 µM. OX2 (h) (agonist radioligand) human recombinant (HEK-293 cells); Ligands [125I]orexin A; Ligand concentration: 0.04 nM ; Ligand Kd: 0.2 nM; Non-specific: Orexin-B (1 μM); Incubation: 180 min at RT scintillation counting; Control inhibitor: Orexin-B; Test concentration/dose: IC/EC50 calculation are provided if 5 or more concentrations are selected; Test sample requirements: Minimum for 1) Screen: 60 μl of 10 mM stock -OR- 1 mg (pre-weighed) for 10µM final testing. 2) Dose Response: 90 μl of 10 mM stock - OR- 1 mg (pre-weighed) for a top concentration at 10 µM. The compounds recited in Table 1 were tested at 10-5 M, calculated as a % inhibition of control (IC) specific binding of a radioactively labeled ligand specific for OX1R and/ or OX2R target. This binding profile panel was broadly defined with roughly an equal number of selective, central and peripheral therapeutically relevant targets, including native animal tissues, radioligands and specific enzymes involved in cell cycle regulation in accordance with Eurofins Standard Operating Procedure. For radioligand binding experiments, the half maximal inhibitory concentration (IC50) and the half maximal effective concentration (EC50) values were determined (via
computer software) by nonlinear regression analysis of the competition curves using Hill equation curve fitting. The inhibition constants (Ki) were calculated using the Cheng–Prusoff equation (Ki = IC50/(1+ (L/KD)), where L is the concentration of radioligand in the assay, and KD is the affinity of the radioligand for the receptor (Cheng et Prusoff 1973). Results The results obtained according to these binding assays are presented in Table 1. Table 1. Binding activity tested at 10-5 M on OX1R and OX2R as agonist for these selected nitro- or amino-dihydro-quinazoline, nitro- or amino-dihydro-benzothiazine and nitro- or amino-dihydro-benzoxazine derivatives
Results showing an inhibition (or stimulation) between 25% and 50% are indicative of weak to moderate effects.
Results showing an inhibition (or stimulation) lower than 25% are not considered significant and mostly attributable to variability of the signal around the control level. Results showing an inhibition (or stimulation for assays run in basal conditions) higher than 50% are considered to represent significant effects of the test compounds. Low to moderate negative values have no real meaning and are attributable to variability of the signal around the control level. High negative values (≥ 50%) that are sometimes obtained with high concentrations of test compounds are generally attributable to nonspecific effects of the test compounds in the assays. On rare occasion they could suggest an allosteric effect of the test compound. Table 2. Pharmacological activity on OX1R and OX2R as agonist for these selected dihydro-quinazoline, dihydro-benzothiazine and dihydro-benzoxazine derivatives
*: Compounds for which the absence of mutagenicity has been demonstrated by the in vitro micronucleus assay according to the OECD guideline N°487 4.2. Binding activities on dopamine and norepinephrine transporters, metabotropic glutamate 2, cathepsin-H and sigma-1 receptors It has been reported for a long time ago that norepinephrine cells of the locus coeruleus (Hagan et al.1999; Horvath et al.1999) and dopaminergic cells of the Ventral tegmental area (Nakamura et al.2000) all show to increase their firing rates by orexins. The firing rates of these monoaminergic neurons are well known to be associated with sleep/wakefulness states. They fire tonically during awake period, less during NREM
sleep, and cease firing during REM sleep (Vanni-Mercier, Sakai, et Jouvet 1984), displaying similar firing patterns with orexin neurons. Previous studies have suggested that firing of these wake-active monoaminergic neurons mediated arousal was supported by orexins. More recently, a prominent role of orexin-to-dopamine circuits mediated by OX2R in the regulation of theta oscillations across vigilance states has been reported (Bandarabadi et al. 2022). In addition to orexin ability to regulate firing rate and induce burst firing though activation of receptors on the somato-dendritic compartment, OX2R could act pre- synaptically at the level of both dopaminergic and glutamatergic axons (Bandarabadi et al. 2022). Product 152 at 10-5 M significantly targets on OX2R as agonist (IC 73%) and on OX1R as agonist (IC 63%) in addition acting on metabotropic glutamate 2 (mGlu2) receptors which target for the treatment of psychiatric disorders including schizophrenia, depression, and anxiety, which are characterized by a glutamatergic dysfunction with a significantly binding effect (IC 54%). Using a sleep–wake electroencephalogram model in rat to study the central functional activity and target engagement following inactivation of the mGluR2 signaling, it is demonstrated that mGluR2 antagonism activity is associated with enhanced theta/gamma oscillations and increased transitions from sleep to waking state (Ahnaou, Ver Donck, et Drinkenburg 2014). Product 194 at 10-5 M significantly targets on OX2R as agonist (IC 84%) and weakly on OX1R as agonist (IC<50%), acting on dopamine transporter (DAT) and norepinephrine transporter (NET) as a catecholaminergic reuptake inhibitor, with a binding effect on DAT (IC 89%) and NET (IC 58%). Therapeutic approaches targeting cathepsins can contribute to prevent or slow down the pathogenesis of neurodegenerative diseases as seen for neuronal ceroid lipofuscinosis, synucleinopathies (Parkinson's disease, Dementia with Lewy Body and Multiple System Atrophy) as well as Alzheimer's and Huntington's disease (Stoka et al. 2023). Cathepsin H (CTSH) is a lysosomal cysteine protease that plays a role in various physiological processes, including the immune response. In the context of neuroinflammation, CTSH has been implicated in modulating immune reactions within the central nervous system (Wang et al. 2023). Neuroinflammation refers to the inflammatory response in the nervous tissue, often involving glial cell activation and
the release of pro-inflammatory molecules. CTSH can influence neuroinflammation by participating in the processing and presentation of antigens, which is crucial for the activation of immune cells such as microglia. Previous studies had also suggested a potential involvement of CTSH in the pathogenesis of narcolepsy (Mogavero et al. 2023). Product 96 at 10-5 M targets on OX1R as agonist (IC 56%) and moreover on CTSH as antagonist (IC 80%), also acting on dopamine transporter (DAT) and norepinephrine transporter (NET) as a catecholaminergic reuptake inhibitor, with a binding effect on DAT (IC 99%) and NET (IC 77%). Sigma-1 receptor (S1R) participating in various physiological and pathological processes, such as neurotransmission, neuroprotection and neuroinflammation is considered as a therapeutic target for a range of neurodegenerative diseases, including amnesia and AD and also various synucleinopathies (Wang et Jia 2023). S1R agonists find to have multiple mechanisms of action that could be beneficial in AD, such as anti-inflammatory and antioxidant effects, modulation of neurotransmitters, and a neuroprotective effect by inhibiting Aβ aggregation and tau hyperphosphorylation is AD (Cummings, Osse, et Kinney 2023; Malar et al. 2023; Shinoda, Nemoto, et Iwamoto 2023). Product 90 at 10-5 M targets on OX2R as agonist (IC 67%) and on S1R as antagonist (IC 75%).
References Ahnaou, A., L. Ver Donck, et W. H. I. M. Drinkenburg. 2014. « Blockade of the Metabotropic Glutamate (mGluR2) Modulates Arousal through Vigilance States Transitions: Evidence from Sleep-Wake EEG in Rodents. » Behavioural brain research 270: 56-67. Alrouji, Mohammed et al. 2023. « Orexin pathway in Parkinson’s disease: a review ». Molecular Biology Reports: 1-14. American Academy of Sleep Medicine. 2014. International Classification of Sleep Disorders. Darien, Ill.: American Acad. of Sleep Medicine. Baimel, Corey et al. 2015. « Orexin/Hypocretin Role in Reward: Implications for Opioid and Other Addictions ». British Journal of Pharmacology 172(2): 334-48. Bandarabadi, Mojtaba et al. 2022. « Orexin action on the dopaminergic system modulates theta during REM sleep and wakefulness ». bioRxiv: 2022.01.30.478401. Beitz, Janice M. 2014. « Parkinson’s disease: a review ». Front Biosci (Schol Ed) 6(1): 65-74. Berhe, Derbew Fikadu, Abadi Kahsu Gebre, et Brhane Teklebrhan Assefa. 2020. « Orexins role in neurodegenerative diseases: From pathogenesis to treatment ». Pharmacology Biochemistry and Behavior 194: 172929. Bogen, Stephane L et al. 2021. « 5-alkyl pyrrolidine orexin receptor agonists ». Boudet, R. 1959. « Sur un nouvel ensemble hétérocyclique du type benzo-meta- thiazine. 2. » Bulletin de la Société Chimique de France (11-1): 1791-93. Cao, Michelle T., et Christian Guilleminault.2017. « Chapter 90 - Narcolepsy: Diagnosis and Management ». In Principles and Practice of Sleep Medicine (Sixth Edition), éd. Meir Kryger, Thomas Roth, et William C. Dement. Elsevier, 873-882.e5. https://www.sciencedirect.com/science/article/pii/B9780323242882000908. Cason, Angie M. et al. 2010. « Role of Orexin/Hypocretin in Reward-Seeking and Addiction: Implications for Obesity. » Physiology & behavior 100(5): 419-28. Chen, Xin-Yi, Lei Chen, et Yi-Feng Du. 2017. « Orexin-A increases the firing activity of hippocampal CA1 neurons through orexin-1 receptors ». Journal of Neuroscience Research 95(7): 1415-26. Cheng, Y., et W. H. Prusoff. 1973. « Relationship between the Inhibition Constant (K1) and the Concentration of Inhibitor Which Causes 50 per Cent Inhibition (I50) of an Enzymatic Reaction. » Biochemical pharmacology 22(23): 3099-3108. Chinigo, Gary M. et al. 2008. « Asymmetric synthesis of 2, 3-dihydro-2-arylquinazolin- 4-ones: methodology and application to a potent fluorescent tubulin inhibitor with anticancer activity ». Journal of medicinal chemistry 51(15): 4620-31.
Cortese, Samuele, Eric Konofal, et Michel Lecendreux. 2008. « Alertness and Feeding Behaviors in ADHD: Does the Hypocretin/Orexin System Play a Role? » Medical hypotheses 71(5): 770-75. Couvineau, Alain, et Marc Laburthe. 2012. « VPAC Receptors: Structure, Molecular Pharmacology and Interaction with Accessory Proteins ». British journal of pharmacology 166(1): 42-50. Cox, Sarah J. et al. 2020. « High-throughput screening at the membrane interface reveals inhibitors of amyloid-β ». Biochemistry 59(24): 2249-58. Cummings, Jeffrey L., Amanda M. Leisgang Osse, et Jefferson W. Kinney. 2023. « Alzheimer’s Disease: Novel Targets and Investigational Drugs for Disease Modification ». Drugs: 1-22. Dauvilliers, Yves et al. 2023. « Oral Orexin Receptor 2 Agonist in Narcolepsy Type 1 ». New England Journal of Medicine 389(4): 309-21. Drouot, X. et al. 2003. « Low levels of ventricular CSF orexin/hypocretin in advanced PD ». Neurology 61(4): 540-43. Duffy, C.M., J.J. Hofmeister, J.P. Nixon, et T.A. Butterick. 2019. « High fat diet increases cognitive decline and neuroinflammation in a model of orexin loss ». Neurobiology of Learning and Memory 157: 41-47. Esmaeili-Mahani, Saeed et al. 2013. « Protective effect of orexin-A on 6- hydroxydopamine-induced neurotoxicity in SH-SY5Y human dopaminergic neuroblastoma cells ». Neurochemistry International 63(8): 719-25. Fan, Jing Kai et al. 2023. « α-Synuclein Induced the Occurrence of RBD via Interaction with OX1R and Modulated Its Degradation ». NeuroMolecular Medicine. https://doi.org/10.1007/s12017-023-08735-4. Feng, Ya et al. 2014. « Neuroprotection by Orexin-A via HIF-1α induction in a cellular model of Parkinson’s disease ». Neuroscience letters 579: 35-40. Fronczek, Rolf et al. 2007. « Hypocretin (orexin) loss in Parkinson’s disease ». Brain 130(6): 1577-85. Fujimoto, Tatsuhiko et al. 2022. « Discovery of TAK-925 as a Potent, Selective, and Brain-Penetrant Orexin 2 Receptor Agonist. » ACS medicinal chemistry letters 13(3): 457-62. Gao, Fan, Tao Liu, Miao Tuo, et Song Chi. 2021. « The role of orexin in Alzheimer disease: From sleep-wake disturbance to therapeutic target ». Neuroscience Letters 765: 136247. Geng, H. J. et al.2012. « Design,synthesis and antifungal activity of 2-aryl-2,3-dihydro- 4H-1,3-benzothiazin-4-one derivatives ». Journal of Shenyang Pharmaceutical
University 29(11): 834-315. Hadadianpour, Zahra et al. 2017. « The effect of orexin-A on motor and cognitive functions in a rat model of Parkinson’s disease ». Neurological Research 39(9): 845-51. Hagan, J. J. et al. 1999. « Orexin A Activates Locus Coeruleus Cell Firing and Increases Arousal in the Rat. » Proceedings of the National Academy of Sciences of the United States of America 96(19): 10911-16. Harris, Glenda C., Mathieu Wimmer, et Gary Aston-Jones. 2005. « A Role for Lateral Hypothalamic Orexin Neurons in Reward Seeking. » Nature 437(7058): 556-59. Heifetz, Alexander et al.2013. « Toward an Understanding of Agonist Binding to Human Orexin-1 and Orexin-2 Receptors with G-Protein-Coupled Receptor Modeling and Site- Directed Mutagenesis ». Biochemistry 52(46): 8246-60. Hervieu, G. J. et al. 2001. « Gene expression and protein distribution of the orexin-1 receptor in the rat brain and spinal cord ». Neuroscience 103(3): 777-97. Hornykiewicz, O., et S. J. Kish. 1987. « Advances in Neurology, Parkinson’s Disease ». Hornykiewicz, Oleh. 1975. « Brain monoamines and parkinsonism ». Natl Inst Drug Abuse Res Monogr Ser 3: 13-21. Horvath, T. L. et al. 1999. « Hypocretin (Orexin) Activation and Synaptic Innervation of the Locus Coeruleus Noradrenergic System. » The Journal of comparative neurology 415(2): 145-59. Hu, Bo et al. 2015. « Roles of the orexin system in central motor control ». Neuroscience & Biobehavioral Reviews 49: 43-54. Ingram, Jack S., et Ernest W. McClelland. 1947. « 146. The formation of some benz-1: 3-thiazine derivatives by the reduction of benz iso thiazolones ». Journal of the Chemical Society (Resumed): 763-64. Ishikawa, Takashi et al. 2023. « TAK-994, a novel orally available brain-penetrant orexin 2 receptor-selective agonist, suppresses fragmentation of wakefulness and cataplexy-like episodes in mouse models of narcolepsy ». Journal of Pharmacology and Experimental Therapeutics 385(3): 193-204. Johnson, Philip L et al.2010. « A key role for orexin in panic anxiety ». Nature Medicine 16(1): 111-15. Kanbayashi, Takashi et al. 2002. « CSF hypocretin‐1 (orexin‐A) concentrations in narcolepsy with and without cataplexy and idiopathic hypersomnia ». Journal of sleep research 11(1): 91-93. Katsuki, Hiroshi, et Shotaro Michinaga. 2012. « Anti-Parkinson Drugs and Orexin Neurons. » Vitamins and hormones 89: 279-90. Kim, Eunhee et al. 2023. Isoquinolinone derivatives, method for preparing the same,
and pharmaceutical composition for preventing or treating poly (adp-ribose) polymerase-1-related diseases, comprising the same as active ingredient. Google Patents. de Lecea, L. et al. 1998. « The Hypocretins: Hypothalamus-Specific Peptides with Neuroexcitatory Activity ». Proceedings of the National Academy of Sciences of the United States of America 95(1): 322-27. Lin, L. et al. 1999. « The Sleep Disorder Canine Narcolepsy Is Caused by a Mutation in the Hypocretin (Orexin) Receptor 2 Gene. » Cell 98(3): 365-76. Liu, Mei-Fang et al. 2018. « Orexin-A exerts neuroprotective effects via OX1R in Parkinson’s disease ». Frontiers in neuroscience 12: 835. Long-Biao, Cui et al. 2010. « 6-羟多巴诱致帕金森病大鼠模型中 orexin系统的进行性变 化 ». Neuroscience Bulletin 26: 381-87. Malar, Dicson S. et al. 2023. « Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders ». CNS drugs: 1-42. Meerwaldt, JD, et A Hovestadt.1988. « Cognitioin in parkinsonism ». Neurology 38(11): 1814-1814. Mezeiova, Eva et al. 2020. « From Orexin Receptor Agonist YNT-185 to Novel Antagonists with Drug-like Properties for the Treatment of Insomnia. » Bioorganic chemistry 103: 104179. Mieda, Michihiro et al. 2011. « Differential Roles of Orexin Receptor-1 and -2 in the Regulation of Non-REM and REM Sleep. » The Journal of neuroscience : the official journal of the Society for Neuroscience 31(17): 6518-26. ———.2017. « The Roles of Orexins in Sleep/Wake Regulation. » Neuroscience research 118: 56-65. Mogavero, Maria Paola et al. 2023. « Genetics and epigenetics of rare hypersomnia ». Trends in Genetics. Morairty, Stephen R. et al. 2012. « Dual Hypocretin Receptor Antagonism Is More Effective for Sleep Promotion than Antagonism of Either Receptor Alone. » PloS one 7(7): e39131. Mordarski, M., et J. B. Chylinska. 1971. « Antitumor properties of 1, 3-oxazine derivatives, derivatives of dihydro-1, 3-oxazine condensed with an aromatic ring in position 5, 6. » Moreau, R. C., et E. Delacoux. 1962. « PREPARATION DE QUELQUES ALCOYL-3 ARYL-2 OXO-4 DIHYDROBENZOTHIAZINES-1, 3 ». BULLETIN DE LA SOCIETE CHIMIQUE DE FRANCE (3): 502-. Nakamura, T. et al. 2000. « Orexin-Induced Hyperlocomotion and Stereotypy Are
Mediated by the Dopaminergic System. » Brain research 873(1): 181-87. Nishino, Seiji. 2007. « Clinical and Neurobiological Aspects of Narcolepsy ». Sleep Medicine 8(4): 373-99. Nollet, Mathieu, et Samuel Leman. 2013. « Role of Orexin in the Pathophysiology of Depression: Potential for Pharmacological Intervention. » CNS drugs 27(6): 411-22. Ohno, Kousaku, et Takeshi Sakurai. 2008. « Orexin neuronal circuitry: Role in the regulation of sleep and wakefulness ». Frontiers in Neuroendocrinology 29(1): 70-87. Pasban-Aliabadi, Hamzeh, Saeed Esmaeili-Mahani, et Mehdi Abbasnejad. 2017. « Orexin-A protects human neuroblastoma SH-SY5Y cells against 6-hydroxydopamine- induced neurotoxicity: involvement of PKC and PI3K signaling pathways ». Rejuvenation Research 20(2): 125-33. Perrey, David A, et Yanan Zhang. 2020. « Therapeutics Development for Addiction: Orexin-1 Receptor Antagonists ». Brain research 1731: 145922-145922. Polito, Rita et al. 2018. « Adiponectin and Orexin-A as a Potential Immunity Link Between Adipose Tissue and Central Nervous System. » Frontiers in physiology 9: 982. Saito, Yuki C. et al. 2018. « Serotonergic Input to Orexin Neurons Plays a Role in Maintaining Wakefulness and REM Sleep Architecture. » Frontiers in neuroscience 12: 892. Sakurai, T. et al. 1998. « Orexins and Orexin Receptors: A Family of Hypothalamic Neuropeptides and G Protein-Coupled Receptors That Regulate Feeding Behavior ». Cell 92(5): 573-75. Shinoda, Yasuharu, Takayuki Nemoto, et Takahiro Iwamoto. 2023. « The Sigma-1 Receptor: Pathophysiological Roles and Therapeutic Potential in Neurodegenerative Diseases ».福岡大学医学紀要= Medical Bulletin of Fukuoka University 50(1): 43-48. Siegel, J. M. 1999. « Narcolepsy: A Key Role for Hypocretins (Orexins). » Cell 98(4): 409-12. Smith, M. I., D. C. Piper, M. S. Duxon, et N. Upton.2003. « Evidence Implicating a Role for Orexin-1 Receptor Modulation of Paradoxical Sleep in the Rat. » Neuroscience letters 341(3): 256-58. Song, Juhyun et al. 2015. « The Role of Orexin in Post-Stroke Inflammation, Cognitive Decline, and Depression. » Molecular brain 8: 16. Stanojlovic, Milos, Jean Pierre Pallais, et Catherine M. Kotz. 2019. « Chemogenetic Modulation of Orexin Neurons Reverses Changes in Anxiety and Locomotor Activity in the A53T Mouse Model of Parkinson’s Disease. » Frontiers in neuroscience 13: 702. Stanojlovic, Milos, Jean Pierre Pallais Yllescas, Aarthi Vijayakumar, et Catherine Kotz. 2019. « Early Sociability and Social Memory Impairment in the A53T Mouse Model of
Parkinson’s Disease Are Ameliorated by Chemogenetic Modulation of Orexin Neuron Activity ». Molecular Neurobiology 56(12): 8435-50. Stoka, Veronika, Olga Vasiljeva, Hiroshi Nakanishi, et Vito Turk. 2023. « The Role of Cysteine Peptidases Cathepsins B, H, C, and X/Z in Neurodegeneration and Cancer ». Thannickal, Thomas C., Yuan-Yang Lai, et Jerome M. Siegel.2007. « Hypocretin (orexin) cell loss in Parkinson’s disease ». Brain 130(6): 1586-95. Thorpy, Michael J. 2020. « Recently Approved and Upcoming Treatments for Narcolepsy ». CNS drugs 34(1): 9-27. Vanni-Mercier, G., K. Sakai, et M. Jouvet. 1984. « [Specific neurons for wakefulness in the posterior hypothalamus in the cat]. » Comptes rendus de l’Academie des sciences. Serie III, Sciences de la vie 298(7): 195-200. Wang, Qinqin, Fei Cao, et Yili Wu. 2021. « Orexinergic system in neurodegenerative diseases ». Frontiers in Aging Neuroscience 13: 713201. Wang, Tao, et Hongmei Jia. 2023. « The Sigma Receptors in Alzheimer’s Disease: New Potential Targets for Diagnosis and Therapy ». International Journal of Molecular Sciences 24(15): 12025. Wang, Yanfeng et al. 2023. « Cathepsin H: molecular characteristics and clues to function and mechanism ». Biochemical Pharmacology: 115585. Wang, Ying et al. 2019. « Orexins alleviate motor deficits via increasing firing activity of pallidal neurons in a mouse model of Parkinson’s disease ». American Journal of Physiology-Cell Physiology 317(4): C800-812. Willie, Jon T. et al. 2003. « Distinct Narcolepsy Syndromes in Orexin Receptor-2 and Orexin Null Mice: Molecular Genetic Dissection of Non-REM and REM Sleep Regulatory Processes ». Neuron 38(5): 715-30. Xiong, Xiaoxing et al. 2013. « Mitigation of Murine Focal Cerebral Ischemia by the Hypocretin/Orexin System is Associated With Reduced Inflammation ». Stroke 44(3): 764-70. Yale, Harry L., et Marion Kalkstein. 1967. « Substituted 2, 3-dihydro-4 (1H)- quinazolinones. A new class of inhibitors of cell multiplication ». Journal of medicinal chemistry 10(3): 334-36. Yanagisawa, Masashi. 2012. « Small-molecule agonists for type-2 orexin receptor ». Yasui, Kenichi et al. 2006. « CSF orexin levels of Parkinson’s disease, dementia with Lewy bodies, progressive supranuclear palsy and corticobasal degeneration ». Journal of the neurological sciences 250(1-2): 120-23. Yukitake, Hiroshi et al. 2019. « TAK-925, an orexin 2 receptor-selective agonist, shows robust wake-promoting effects in mice ». Pharmacology Biochemistry and Behavior
187: 172794. Zhang, Dehui et al. 2021. « Discovery of Arylsulfonamides as Dual Orexin Receptor Agonists. » Journal of medicinal chemistry 64(12): 8806-25. Zhang, Zhongxing et al. 2018. « Exploring the Clinical Features of Narcolepsy Type 1 versus Narcolepsy Type 2 from European Narcolepsy Network Database with Machine Learning ». Scientific reports 8(1): 10628-10628.
Claims
Claims Claim 1. A compound of formula (I):
wherein: - X represents –NH-, –S- or –O-; - Y and R2, independently of each other represents a hydrogen atom, a halogen atom, –NO2 or –NH2; - R1, R3, R4 each represent, independently of each other, a hydrogen atom or a halogen atom; - R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C30)alkyl chain, especially (C1–C20)alkyl, optionally broken up and/or followed and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, – C(R11)=C(R12)–, -O-, –S–, –NR13–, –C(O)–, –C(S)–, –C=N–, –N=C–, –OC(O)–, –C(O)O–, –SC(O)–, –C(O)S–, –N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R10 represents a hydrogen atom or a (C1–C30)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, the aryl, heteroaryl and heterocyclic rings being optionally substituted; - R11 and R12 represent, independently of each other, a hydrogen atom or a (C1–C6)alkyl group; and - R13 to R15 represent, independently of each other, a hydrogen atom or a (C1–C6)alkyl, cycloalkyl, heterocyclic, aryl, heteroaryl or acyl group, and preferably a hydrogen atom or a (C1–C6)alkyl or aryl group, and still more preferably a hydrogen atom or a (C1–C6)alkyl group, or a pharmaceutically acceptable salt thereof, a tautomer, a stereoisomer or mixture of stereoisomers thereof, for use in the prevention and/or treatment of neurological diseases,
preferably associated with psychiatric and/or sleep disorders and diseases, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
Claim 2. The compound of formula (I) for use according to claim 1, characterized in that: - Y represents a halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2, and R2 represents a hydrogen atom or a halogen atom, preferably a hydrogen atom, or - R2 represents a halogen atom, –NO2 or –NH2, preferably –NO2 or –NH2, and Y represents a hydrogen atom or a halogen atom, preferably a hydrogen atom.
Claim 3. The compound of formula (I) for use according to claim 1 or 2, characterized in that R1, R2, R3, R4 each represent a hydrogen atom or R1, R3, R4, Y each represent a hydrogen atom.
Claim 4. The compound of formula (I) for use according to any one of claims 1 to 3, characterized in that R5, R6, R7, R8, R9 each represent, independently of each other, a hydrogen atom, a halogen atom, –OR10 or a (C1–C20)alkyl, optionally broken up and/or preceded by one or more moieties chosen from the group consisting of aryl, heteroaryl, cycloalkyl, heterocyclic, –C≡C–, –C(R11)=C(R12)–,-O-, –S–, –NR13–, –C(O)–, –OC(O)–, –C(O)O–, – N(R14)C(O)– and –C(O)N(R15)– groups, the aryl, heteroaryl and heterocyclic rings being optionally substituted.
Claim 5. The compound of formula (I) for use according to any one of claims 1 to 4, characterized in that R5, R6, R7, R8, R9, each represent, independently of each other, a hydrogen atom or –OR10.
Claim 6. The compound of formula (I) for use according to any one of claims 1 to 5, characterized in that at least four residues out of R5, R6, R7, R8 and R9 each represent a hydrogen atom.
Claim 7. The compound of formula (I) for use according to claim 6, characterized in that R5, R6, R7, R8, R9, each represent, a hydrogen atom.
Claim 8. The compound of formula (I) for use according to claim 6, characterized in that R5, R6 or R7 represents –OR10 with R10 representing a hydrogen atom, a (C1–C30)alkyl or phenyl group, preferably a (C1–C20)alkyl or a phenyl group.
Claim 9. The compound of formula (I) for use according to any one of claims 1 to 8, characterized in that it is selected from the group consisting of:
5
10 attention-deficit hyperactivity disorder, anxiety and mood disorders, Alzheimer's disease or any other neurodegenerative disorders or cognitive impairment and tauopathies, Parkinson's disease and other synucleinopathies, Guillain-Barre syndrome, chronic fatigue syndrome, long COVID-19 and medical or health conditions associated with circadian rhythmicity as well as mental and physical disorders associated with travel across time zones and with rotating 15 shift-work schedules, restless legs syndrome, fibromyalgia, cardiac failure, diseases related to bone loss, sepsis, syndromes which are manifested by unrefreshing sleep and muscle pain,
sleep apnea which is associated with respiratory disturbances during sleep; conditions which result from a diminished quality of sleep.
Claim 11. The compound of formula (I) for use according to any one of claims 1 to 10, characterized in that the neurological diseases are selected from the group consisting of narcolepsy type 1 (NT1 ), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, Parkinson's disease and other synucleinopathies, preferably narcolepsy type 1 (NT1 ), narcolepsy type 2 (NT2) and Parkinson's disease.
Claim 12. A pharmaceutical composition comprising at least one compound of formula (I) as defined in any one of claims 1 to 11 and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of neurological diseases, preferably associated with psychiatric and/or sleep disorders and diseases, advantageously in which central orexin neurotransmission is compromised or in which central and peripheral orexin receptors are involved.
Claim 13. A pharmaceutical composition comprising at least one compound of formula (I) as defined in any one of claims 1 to 11 and a pharmaceutically acceptable carrier for use in the prevention and/or treatment of narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), hypersomnia, idiopathic hypersomnia, recurrent hypersomnia, Kleine-Levin syndrome, hypersomnia associated with a psychiatric disorder, hypersomnia due to a medical disorder, hypersomnia due to a medication or substance, Parkinson's disease and other synucleinopathies, preferably narcolepsy type 1 (NT1), narcolepsy type 2 (NT2) and Parkinson's disease.
Claim 14. The pharmaceutical composition for use according to claim 12 or 13, comprising between 0.5 mg to 800 mg, preferably between 20 mg to 400 mg of the compound of formula (I).
Claim 15. The pharmaceutical composition for use according to any one of claims 12 to 14, which is suitable for oral or parenteral administration, preferably in the form of a solution, such as an injectable solution, a tablet, a capsule or a transdermal delivery system.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP22307075 | 2022-12-30 | ||
EP22307075.6 | 2022-12-30 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2024115797A2 true WO2024115797A2 (en) | 2024-06-06 |
WO2024115797A3 WO2024115797A3 (en) | 2024-09-06 |
Family
ID=85018905
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2023/088020 WO2024141660A2 (en) | 2022-12-30 | 2023-12-29 | Dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and use thereof as orexin receptors agonists for treating or preventing neurological diseases |
PCT/EP2023/088019 WO2024115797A2 (en) | 2022-12-30 | 2023-12-29 | Dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and use thereof as orexin receptors agonists for treating or preventing neurological diseases |
Family Applications Before (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2023/088020 WO2024141660A2 (en) | 2022-12-30 | 2023-12-29 | Dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and use thereof as orexin receptors agonists for treating or preventing neurological diseases |
Country Status (1)
Country | Link |
---|---|
WO (2) | WO2024141660A2 (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2018044136A1 (en) | 2016-09-05 | 2018-03-08 | 충남대학교 산학협력단 | Novel compound, preparation method therefor and pharmaceutical composition containing same as active ingredient for preventing or treating diseases associated with poly(adp-ribose) polymerase-1 (parp-1) |
WO2021107023A1 (en) | 2019-11-27 | 2021-06-03 | 大日本住友製薬株式会社 | Cycloalkyl urea derivative |
WO2022140317A1 (en) | 2020-12-21 | 2022-06-30 | Alkermes, Inc. | Substituted piperidino compounds and related methods of treatment |
Family Cites Families (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US2943087A (en) * | 1960-06-28 | n naoh | ||
ZA991301B (en) * | 1998-02-18 | 1999-09-13 | Neurosearch As | Glutamate receptor modulators. |
BRPI0511748A (en) * | 2004-06-01 | 2008-01-02 | Univ Virginia | pharmaceutical composition, methods for treating cancer and a disease or disorder associated with angiogenesis in an individual in need, to inhibit proliferation of vascular and cancer endothelial cells, and to prepare sc-2-71 and analogs thereof, sc photoaffinity label -2-71, method for disrupting microtubule polymerization in a cell, and for identifying an analog, analog, and kit for administering a pharmaceutical composition to an individual |
US9200046B2 (en) * | 2011-06-29 | 2015-12-01 | Cornell University | Reporter system for high throughput screening of compounds and uses thereof |
WO2015154047A1 (en) * | 2014-04-03 | 2015-10-08 | Restorgenex Corporation | Novel methods |
WO2016201257A2 (en) * | 2015-06-10 | 2016-12-15 | The Johns Hopkins University | Compositions and methods for identifying adp-ribosylated sites by mass spectrometry |
EP3436601B1 (en) * | 2016-03-31 | 2023-03-29 | Centre National De La Recherche Scientifique | Cytidine deaminase expression level in cancer as a new therapeutic target |
CN111635369A (en) * | 2020-06-19 | 2020-09-08 | 宁夏大学 | A kind of preparation method of dihydroquinazolinone derivative |
CN116178283B (en) * | 2023-01-18 | 2024-07-02 | 武汉科技大学 | 2-Phenyl-2,3-dihydroquinazolin-4(1H)-one derivatives and their preparation and application |
-
2023
- 2023-12-29 WO PCT/EP2023/088020 patent/WO2024141660A2/en unknown
- 2023-12-29 WO PCT/EP2023/088019 patent/WO2024115797A2/en unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2018044136A1 (en) | 2016-09-05 | 2018-03-08 | 충남대학교 산학협력단 | Novel compound, preparation method therefor and pharmaceutical composition containing same as active ingredient for preventing or treating diseases associated with poly(adp-ribose) polymerase-1 (parp-1) |
WO2021107023A1 (en) | 2019-11-27 | 2021-06-03 | 大日本住友製薬株式会社 | Cycloalkyl urea derivative |
WO2022140317A1 (en) | 2020-12-21 | 2022-06-30 | Alkermes, Inc. | Substituted piperidino compounds and related methods of treatment |
Non-Patent Citations (82)
Title |
---|
"The Roles of Orexins in Sleep/Wake Regulation", NEUROSCIENCE RESEARCH, vol. 118, pages 56 - 65 |
AHNAOU, A.L. VER DONCKW. H. I. M. DRINKENBURG: "Blockade of the Metabotropic Glutamate (mGluR2) Modulates Arousal through Vigilance States Transitions: Evidence from Sleep-Wake EEG in Rodents", BEHAVIOURAL BRAIN RESEARCH, vol. 270, 2014, pages 56 - 67, XP028860588, DOI: 10.1016/j.bbr.2014.05.003 |
ALROUJI, MOHAMMED ET AL.: "Orexin pathway in Parkinson's disease: a review", MOLECULAR BIOLOGY REPORTS, 2023, pages 1 - 14 |
BAIMEL, COREY ET AL.: "Orexin/Hypocretin Role in Reward: Implications for Opioid and Other Addictions", BRITISH JOURNAL OF PHARMACOLOGY, vol. 172, no. 2, 2015, pages 334 - 48, XP071171719, DOI: 10.1111/bph.12639 |
BANDARABADI, MOJTABA ET AL.: "Orexin action on the dopaminergic system modulates theta during REM sleep and wakefulness", BIORXIV: 2022.01.30.478401, 2022 |
BEITZ, JANICE M: "Parkinson's disease: a review", FRONT BIOSCI, vol. 6, no. 1, 2014, pages 65 - 74 |
BERHE, DERBEW FIKADU, ABADI KAHSU GEBRE, ET BRHANE TEKLEBRHAN ASSEFA: "Orexins role in neurodegenerative diseases: From pathogenesis to treatment", PHARMACOLOGY BIOCHEMISTRY AND BEHAVIOR, vol. 194, 2020, pages 172929, XP086161978, DOI: 10.1016/j.pbb.2020.172929 |
BOGEN, STEPHANE L ET AL., 5-ALKYL PYRROLIDINE OREXIN RECEPTOR AGONISTS, 2021 |
BOUDET, R.: "Sur un nouvel ensemble heterocyclique du type benzo-meta-thiazine. 2", BULLETIN DE (A SOCIETE CHIMIQUE DE FRANCE, vol. 11, no. 1, 1959, pages 1791 - 93 |
CAO, MICHELLE T.CHRISTIAN GUILLEMINAULT: "Principles and Practice of Sleep Medicine", 2017, ELSEVIER, article "Chapter 90 - Narcolepsy: Diagnosis and Management", pages: 873 - 882 |
CASON, ANGIE M. ET AL.: "Role of Orexin/Hypocretin in Reward-Seeking and Addiction: Implications for Obesity", PHYSIOLOGY ΣT BEHAVIOR, vol. 100, no. 5, 2010, pages 419 - 28, XP027085470 |
CHEN, XIN-YILEI CHENYI-FENG DU: "Orexin-A increases the firing activity of hippocampal CA1 neurons through orexin-1 receptors", JOURNAL OF NEUROSCIENCE RESEARCH, vol. 95, no. 7, 2017, pages 1415 - 26 |
CHENG, Y.W. H. PRUSOFF: "Relationship between the Inhibition Constant (K1) and the Concentration of Inhibitor Which Causes 50 per Cent Inhibition (I50) of an Enzymatic Reaction", BIOCHEMICAL PHARMACOLOGY, vol. 22, no. 23, 1973, pages 3099 - 3108, XP000942582, DOI: 10.1016/0006-2952(73)90196-2 |
CHINIGO, GARY M. ET AL.: "Asymmetric synthesis of 2, 3-dihydro-2-arylquinazolin-4-ones: methodology and application to a potent fluorescent tubulin inhibitor with anticancer activity", JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, no. 15, 2008, pages 4620 - 31, XP055658845, DOI: 10.1021/jm800271c |
CORTESE, SAMUELEERIC KONOFALMICHEL LECENDREUX: "Alertness and Feeding Behaviors in ADHD: Does the Hypocretin/Orexin System Play a Role?", MEDICAL HYPOTHESES, vol. 71, no. 5, 2008, pages 770 - 75, XP025470085, DOI: 10.1016/j.mehy.2008.06.017 |
COUVINEAU, ALAINMARC LABURTHE: "VPAC Receptors: Structure, Molecular Pharmacology and Interaction with Accessory Proteins", BRITISH JOURNAL OF PHARMACOLOGY, vol. 166, no. 1, 2012, pages 42 - 50, XP071171096, DOI: 10.1111/j.1476-5381.2011.01676.x |
COX, SARAH J ET AL.: "High-throughput screening at the membrane interface reveals inhibitors of amyloid-B", BIOCHEMISTRY, vol. 59, no. 24, 2020, pages 2249 - 58, XP093050043, DOI: 10.1021/acs.biochem.0c00328 |
CUMMINGS, JEFFREY L.AMANDA M. LEISGANG OSSEJEFFERSON W. KINNEY: "Alzheimer's Disease: Novel Targets and Investigational Drugs for Disease Modification", DRUGS, 2023, pages 1 - 22 |
DAUVILLIERS, YVES ET AL.: "Oral Orexin Receptor 2 Agonist in Narcolepsy Type 1", NEW ENGLAND JOURNAL OF MEDICINE, vol. 389, no. 4, 2023, pages 309 - 21 |
DROUOT, X. ET AL.: "Low levels of ventricular CSF orexin/hypocretin in advanced PD", NEUROLOGY, vol. 61, no. 4, 2003, pages 540 - 43 |
DUFFY, C.M.J.J. HOFMEISTERJ.P. NIXONT.A. BUTTERICK: "High fat diet increases cognitive decline and neuroinflammation in a model of orexin loss", NEUROBIOLOGY OF LEARNING AND MEMORY, vol. 157, 2019, pages 41 - 47, XP085611213, DOI: 10.1016/j.nlm.2018.11.008 |
ESMAEILI-MAHANI, SAEED ET AL.: "Protective effect of orexin-A on 6-hydroxydopamine-induced neurotoxicity in SH-SY5Y human dopaminergic neuroblastoma cells", NEUROCHEMISTRY INTERNATIONAL, vol. 63, no. 8, 2013, pages 719 - 25, XP028784510, DOI: 10.1016/j.neuint.2013.09.022 |
FAN, JING KAI ET AL.: "α-Synuclein Induced the Occurrence of RBD via Interaction with OX1 R and Modulated Its Degradation", NEUROMOLECULAR MEDICINE, 2023, Retrieved from the Internet <URL:https://doi.org/10.1007/s12017-023-08735-4> |
FENG, YA ET AL.: "Neuroprotection by Orexin-A via HIF-1α induction in a cellular model of Parkinson's disease", NEUROSCIENCE LETTERS, vol. 579, 2014, pages 35 - 40, XP029046801, DOI: 10.1016/j.neulet.2014.07.014 |
FRONCZEK, ROLF ET AL.: "Hypocretin (orexin) loss in Parkinson's disease", BRAIN, vol. 130, no. 6, 2007, pages 1577 - 85 |
FUJIMOTO, TATSUHIKO ET AL.: "Discovery of TAK-925 as a Potent, Selective, and Brain-Penetrant Orexin 2 Receptor Agonist.", ACS MEDICINAL CHEMISTRY LETTERS, vol. 13, no. 3, 2022, pages 457 - 62, XP055923040, DOI: 10.1021/acsmedchemlett.1c00626 |
GAO, FANTAO LIUMIAO TUOSONG CHI: "The role of orexin in Alzheimer disease: From sleep-wake disturbance to therapeutic target", NEUROSCIENCE LETTERS, vol. 765, 2021, pages 136247, XP086847164, DOI: 10.1016/j.neulet.2021.136247 |
GENG, H. J ET AL.: "Design,synthesis and antifungal activity of 2-aryl-2,3-dihydro-4H-1,3-benzothiazin-4-one derivatives", JOURNAL OF SHENYANG PHARMACEUTICAL UNIVERSITY, vol. 29, no. 11, 2012, pages 834 - 315 |
HADADIANPOUR, ZAHRA ET AL.: "The effect of orexin-A on motor and cognitive functions in a rat model of Parkinson's disease", NEUROLOGICAL RESEARCH, vol. 39, no. 9, 2017, pages 845 - 51 |
HAGAN, J. J. ET AL.: "Orexin A Activates Locus Coeruleus Cell Firing and Increases Arousal in the Rat.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 96, no. 19, 1999, pages 10911 - 16 |
HARRIS, GLENDA C.MATHIEU WIMMERGARY ASTON-JONES: "A Role for Lateral Hypothalamic Orexin Neurons in Reward Seeking", NATURE, vol. 437, no. 7058, 2005, pages 556 - 59 |
HEIFETZ, ALEXANDER ET AL.: "Toward an Understanding of Agonist Binding to Human Orexin-1 and Orexin-2 Receptors with G-Protein-Coupled Receptor Modeling and Site-Directed Mutagenesis", BIOCHEMISTRY, vol. 52, no. 46, 2013, pages 8246 - 60, XP055209152, DOI: 10.1021/bi401119m |
HERVIEU, G. J ET AL.: "Gene expression and protein distribution of the orexin-1 receptor in the rat brain and spinal cord", NEUROSCIENCE, vol. 103, no. 3, 2001, pages 777 - 97, XP085030643, DOI: 10.1016/S0306-4522(01)00033-1 |
HORNYKIEWICZ, 0.S. J. KISH., ADVANCES IN NEUROLOGY, PARKINSON'S DISEASE, 1987 |
HORNYKIEWICZ, OLEH.: "Brain monoamines and parkinsonism", NATL INST DRUG ABUSE RES MONOGR SER, vol. 3, 1975, pages 13 - 21 |
HORVATH, T. L. ET AL.: "Hypocretin (Orexin) Activation and Synaptic Innervation of the Locus Coeruleus Noradrenergic System", THE JOURNAL OF COMPARATIVE NEUROLOGY, vol. 415, no. 2, 1999, pages 145 - 59 |
HU, BO ET AL.: "Roles of the orexin system in central motor control", NEUROSCIENCE ΣT BIOBEHAVIORAL REVIEWS, vol. 49, 2015, pages 43 - 54 |
INGRAM, JACK S.ERNEST W. MCCLELLAND: "146. The formation of some benz-1: 3-thiazine derivatives by the reduction of benz iso thiazolones", JOURNAL OF THE CHEMICAL SOCIETY (RESUMED), 1947, pages 763 - 64 |
INTERNATIONAL CLASSIFICATION OF SLEEP DISORDERS, 2014 |
ISHIKAWA, TAKASHI ET AL.: "TAK-994, a novel orally available brain-penetrant orexin 2 receptor-selective agonist, suppresses fragmentation of wakefulness and cataplexy-like episodes in mouse models of narcolepsy", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 385, no. 3, 2023, pages 193 - 204, XP093120935, DOI: 10.1124/jpet.122.001449 |
JOHNSON, PHILIP L ET AL.: "A key role for orexin in panic anxiety", NATURE MEDICINE, vol. 16, no. 1, 2010, pages 111 - 15, XP055066615, DOI: 10.1038/nm.2075 |
KANBAYASHI, TAKASHI ET AL.: "CSF hypocretin - 1 (orexin - A) concentrations in narcolepsy with and without cataplexy and idiopathic hypersomnia", JOURNAL OF SLEEP RESEARCH, vol. 11, no. 1, 2002, pages 91 - 93 |
KATSUKI, HIROSHISHOTARO MICHINAGA: "Anti-Parkinson Drugs and Orexin Neurons", VITAMINS AND HORMONES, vol. 89, 2012, pages 279 - 90 |
KIM, EUNHEE ET AL.: "Isoquinolinone derivatives, method for preparing the same, and pharmaceutical composition for preventing or treating poly (adp-ribose) polymerase-1-related diseases, comprising the same as active ingredient", GOOGLE, 2023 |
LECEA, L. ET AL.: "The Hypocretins: Hypothalamus-Specific Peptides with Neuroexcitatory Activity", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 95, no. 1, 1998, pages 322 - 27, XP002105411, DOI: 10.1073/pnas.95.1.322 |
LIN, L. ET AL.: "The Sleep Disorder Canine Narcolepsy Is Caused by a Mutation in the Hypocretin (Orexin) Receptor 2 Gene", CELL, vol. 98, no. 3, 1999, pages 365 - 76, XP002153571, DOI: 10.1016/S0092-8674(00)81965-0 |
LIU, MEI-FANG ET AL.: "Orexin-A exerts neuroprotective effects via OX1 R in Parkinson's disease", FRONTIERS IN NEUROSCIENCE, vol. 12, 2018, pages 835 |
MALAR, DICSON S. ET AL.: "Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders", CNS DRUGS, 2023, pages 1 - 42 |
MEERWALDT, JDA HOVESTADT: "Cognitioin in parkinsonism", NEUROLOGY, vol. 38, no. 11, 1988, pages 1814 - 1814 |
MEZEIOVA, EVA ET AL.: "From Orexin Receptor Agonist YNT-185 to Novel Antagonists with Drug-like Properties for the Treatment of Insomnia.", BIOORGANIC CHEMISTRY, vol. 103, 2020, pages 104179, XP055964326, DOI: 10.1016/j.bioorg.2020.104179 |
MIEDA, MICHIHIRO ET AL.: "Differential Roles of Orexin Receptor-1 and -2 in the Regulation of Non-REM and REM Sleep", THE JOURNAL OF NEUROSCIENCE: THE OFFICIAL JOURNAL OF THE SOCIETY FOR NEUROSCIENCE, vol. 31, no. 17, 2011, pages 6518 - 26 |
MOGAVERO, MARIA PAOLA ET AL.: "Genetics and epigenetics of rare hypersomnia", TRENDS IN GENETICS, 2023 |
MORAIRTY, STEPHEN R. ET AL.: "Dual Hypocretin Receptor Antagonism Is More Effective for Sleep Promotion than Antagonism of Either Receptor Alone", PLOS ONE, vol. 7, no. 7, 2012, pages 39131 |
NAKAMURA, T. ET AL.: "Orexin-Induced Hyperlocomotion and Stereotypy Are Mediated by the Dopaminergic System", BRAIN RESEARCH, vol. 873, no. 1, 2000, pages 181 - 87, XP002425801, DOI: 10.1016/S0006-8993(00)02555-5 |
NISHINO, SEIJI.: "Clinical and Neurobiological Aspects of Narcolepsy", SLEEP MEDICINE, vol. 8, no. 4, 2007, pages 373 - 99, XP022070273, DOI: 10.1016/j.sleep.2007.03.008 |
NOLLET, MATHIEUSAMUEL LEMAN: "Role of Orexin in the Pathophysiology of Depression: Potential for Pharmacological Intervention", CNS DRUGS, vol. 27, no. 6, 2013, pages 411 - 22 |
OHNO, KOUSAKUTAKESHI SAKURAI: "Orexin neuronal circuitry: Role in the regulation of sleep and wakefulness", FRONTIERS IN NEUROENDOCRINOLOGY, vol. 29, no. 1, 2008, pages 70 - 87, XP022409381, DOI: 10.1016/j.yfrne.2007.08.001 |
PASBAN-ALIABADI, HAMZEHSAEED ESMAEILI-MAHANIMEHDI ABBASNEJAD: "Orexin-A protects human neuroblastoma SH-SY5Y cells against 6-hydroxydopamine-induced neurotoxicity: involvement of PKC and PI3K signaling pathways", REJUVENATION RESEARCH, vol. 20, no. 2, 2017, pages 125 - 33 |
PERREY, DAVID AYANAN ZHANG: "Therapeutics Development for Addiction: Orexin-1 Receptor Antagonists", BRAIN RESEARCH, vol. 1731, 2020, pages 145922 - 145922 |
POLITO, RITA ET AL.: "Adiponectin and Orexin-A as a Potential Immunity Link Between Adipose Tissue and Central Nervous System", FRONTIERS IN PHYSIOLOGY, vol. 9, 2018, pages 982 |
SAITO, YUKI C. ET AL.: "Serotonergic Input to Orexin Neurons Plays a Role in Maintaining Wakefulness and REM Sleep Architecture", FRONTIERS IN NEUROSCIENCE, vol. 12, 2018, pages 892 |
SAKURAI, T. ET AL.: "Orexins and Orexin Receptors: A Family of Hypothalamic Neuropeptides and G Protein-Coupled Receptors That Regulate Feeding Behavior", CELL, vol. 92, no. 5, 1998, pages 573 - 75 |
SIEGEL, J. M.: "Narcolepsy: A Key Role for Hypocretins (Orexins", CELL, vol. 98, no. 4, 1999, pages 409 - 12, XP000941943, DOI: 10.1016/S0092-8674(00)81969-8 |
SMITH, M. I.D. C. PIPERM. S. DUXONN. UPTON: "Evidence Implicating a Role for Orexin-1 Receptor Modulation of Paradoxical Sleep in the Rat", NEUROSCIENCE LETTERS, vol. 341, no. 3, 2003, pages 256 - 58 |
SONG, JUHYUN ET AL.: "The Role of Orexin in Post-Stroke Inflammation, Cognitive Decline, and Depression", MOLECULAR BRAIN, vol. 8, 2015, pages 16, XP021218349, DOI: 10.1186/s13041-015-0106-1 |
STANOJLOVIC, MILOSJEAN PIERRE PALLAIS YLLESCASAARTHI VIJAYAKUMARCATHERINE KOTZ: "Early Sociability and Social Memory Impairment in the A53T Mouse Model of Parkinson's Disease Are Ameliorated by Chemogenetic Modulation of Orexin Neuron Activity", MOLECULAR NEUROBIOLOGY, vol. 56, no. 12, 2019, pages 8435 - 50, XP036926963, DOI: 10.1007/s12035-019-01682-x |
STANOJLOVIC, MILOSJEAN PIERRE PALLAISCATHERINE M. KOTZ: "Chemogenetic Modulation of Orexin Neurons Reverses Changes in Anxiety and Locomotor Activity in the A53T Mouse Model of Parkinson's Disease", FRONTIERS IN NEUROSCIENCE, vol. 13, 2019, pages 702 |
STOKA, VERONIKAOLGA VASILJEVAHIROSHI NAKANISHIVITO TURK, THE ROLE OF CYSTEINE PEPTIDASES CATHEPSINS B, H, C, AND X/Z IN NEURODEGENERATION AND CANCER, 2023 |
THANNICKAL, THOMAS C.YUAN-YANG LAIJEROME M. SIEGEL: "Hypocretin (orexin) cell loss in Parkinson's disease", BRAIN, vol. 130, no. 6, 2007, pages 1586 - 95 |
THORPY, MICHAEL J.: "Recently Approved and Upcoming Treatments for Narcolepsy", CNS DRUGS, vol. 34, no. 1, 2020, pages 9 - 27, XP055791029, DOI: 10.1007/s40263-019-00689-1 |
VANNI-MERCIER, G.K. SAKAIM. JOUVET: "Specific neurons for wakefulness in the posterior hypothalamus in the cat].", COMPTES RENDUS DE I'ACADEMIE DES SCIENCES, vol. 298, no. 7, 1984, pages 195 - 200 |
WANG, QINQINFEI CAOYILI WU: "Orexinergic system in neurodegenerative diseases", FRONTIERS IN AGING NEUROSCIENCE, vol. 13, 2021, pages 713201 |
WANG, TAOHONGMEI JIA: "The Sigma Receptors in Alzheimer's Disease: New Potential Targets for Diagnosis and Therapy", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 24, no. 15, 2023, pages 12025 |
WANG, YANFENG ET AL.: "Cathepsin H: molecular characteristics and clues to function and mechanism", BIOCHEMICAL PHARMACOLOGY, 2023, pages 115585 |
WANG, YING ET AL.: "Orexins alleviate motor deficits via increasing firing activity of pallidal neurons in a mouse model of Parkinson's disease", AMERICAN JOURNAL OF PHYSIOLOGY-CELL PHYSIOLOGY, vol. 317, no. 4, 2019, pages 800 - 812 |
WILLIE, JON T. ET AL.: "Distinct Narcolepsy Syndromes in Orexin Receptor-2 and Orexin Null Mice: Molecular Genetic Dissection of Non-REM and REM Sleep Regulatory Processes", NEURON, vol. 38, no. 5, 2003, pages 715 - 30 |
XIONG, XIAOXING ET AL.: "Mitigation of Murine Focal Cerebral Ischemia by the Hypocretin/Orexin System is Associated With Reduced Inflammation", STROKE, vol. 44, no. 3, 2013, pages 764 - 70 |
YALE, HARRY L.MARION KALKSTEIN: "Substituted 2, 3-dihydro-4 (1H)-quinazolinones. A new class of inhibitors of cell multiplication", JOURNAL OF MEDICINAL CHEMISTRY, vol. 10, no. 3, 1967, pages 334 - 36 |
YASUI, KENICHI ET AL.: "CSF orexin levels of Parkinson's disease, dementia with Lewy bodies, progressive supranuclear palsy and corticobasal degeneration", JOURNAL OF THE NEUROLOGICAL SCIENCES, vol. 250, 2006, pages 120 - 23 |
YUKITAKE, HIROSHI ET AL.: "TAK-925, an orexin 2 receptor-selective agonist, shows robust wake-promoting effects in mice", PHARMACOLOGY BIOCHEMISTRY AND BEHAVIOR, vol. 187, 2019, pages 172794, XP085903575, DOI: 10.1016/j.pbb.2019.172794 |
ZHANG, DEHUI ET AL.: "Discovery of Arylsulfonamides as Dual Orexin Receptor Agonists", JOURNAL OF MEDICINAL CHEMISTRY, vol. 64, no. 12, 2021, pages 8806 - 25 |
ZHANG, ZHONGXING ET AL.: "Exploring the Clinical Features of Narcolepsy Type 1 versus Narcolepsy Type 2 from European Narcolepsy Network Database with Machine Learning", SCIENTIFIC REPORTS, vol. 8, no. 1, 2018, pages 10628 - 10628 |
Also Published As
Publication number | Publication date |
---|---|
WO2024141660A2 (en) | 2024-07-04 |
WO2024141660A3 (en) | 2024-08-29 |
WO2024115797A3 (en) | 2024-09-06 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2021216324B2 (en) | Heterocyclic GLP-1 agonists | |
EP4141001B1 (en) | Kappa opioid receptor antagonists and products and methods related thereto | |
KR101805754B1 (en) | Compositions of protein receptor tyrosine kinase inhibitors | |
EP4103563A1 (en) | Heterocyclic glp-1 agonists | |
WO2021219019A1 (en) | Heterocyclic glp-1 agonists | |
EP3617195B1 (en) | Novel tetrahydronaphthyl urea derivatives as inhibitors of tropomyosin receptor kinase a for the treatment of pain | |
JP6402115B2 (en) | Spiro-quinazolinone derivatives useful for the treatment of neurological diseases and conditions | |
WO2020015343A1 (en) | Novel compounds and their uses as acc inhibitors | |
JP2015143255A (en) | Method for treating pain syndrome and other disorders | |
JP2022036285A (en) | Piperidinyl nociceptin receptor compounds | |
WO2024115797A2 (en) | Dihydro-quinazoline, -benzothiazine and -benzoxazine derivatives and use thereof as orexin receptors agonists for treating or preventing neurological diseases | |
EP3894414B1 (en) | 1-(2,6-diazaspiro[3.3]heptan-6-yl)-5,6-dihydro-4h-benzo[f][1,2,4]triazolo[4,3-a][1,4]diazepine derivatives as vasopressin antagonists for the treatment of neuro-psychological disorders | |
JP7711074B2 (en) | Heterocyclic GLP-1 agonists | |
HK40023306A (en) | Kappa opioid receptor antagonists and products and methods related thereto | |
HK40023306B (en) | Kappa opioid receptor antagonists and products and methods related thereto |