NL2035306B1 - 1,3-isoindolinedione derivatives and heteroaromatic analogues for stimulating endothelial cell-pericyte interaction - Google Patents
1,3-isoindolinedione derivatives and heteroaromatic analogues for stimulating endothelial cell-pericyte interaction Download PDFInfo
- Publication number
- NL2035306B1 NL2035306B1 NL2035306A NL2035306A NL2035306B1 NL 2035306 B1 NL2035306 B1 NL 2035306B1 NL 2035306 A NL2035306 A NL 2035306A NL 2035306 A NL2035306 A NL 2035306A NL 2035306 B1 NL2035306 B1 NL 2035306B1
- Authority
- NL
- Netherlands
- Prior art keywords
- compound
- pericyte
- mmol
- disease
- alkyl
- Prior art date
Links
- 125000001072 heteroaryl group Chemical group 0.000 title claims description 11
- 230000003511 endothelial effect Effects 0.000 title description 20
- 230000003993 interaction Effects 0.000 title description 20
- XKJCHHZQLQNZHY-UHFFFAOYSA-N phthalimide Chemical class C1=CC=C2C(=O)NC(=O)C2=C1 XKJCHHZQLQNZHY-UHFFFAOYSA-N 0.000 title description 16
- 230000004936 stimulating effect Effects 0.000 title description 6
- 150000001875 compounds Chemical class 0.000 claims abstract description 180
- 210000003668 pericyte Anatomy 0.000 claims abstract description 86
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 56
- 238000011282 treatment Methods 0.000 claims abstract description 49
- 239000000203 mixture Substances 0.000 claims abstract description 48
- 210000004204 blood vessel Anatomy 0.000 claims abstract description 44
- 201000010099 disease Diseases 0.000 claims abstract description 38
- 238000009472 formulation Methods 0.000 claims abstract description 31
- 239000003814 drug Substances 0.000 claims abstract description 21
- 230000004064 dysfunction Effects 0.000 claims abstract description 21
- 230000001737 promoting effect Effects 0.000 claims abstract description 19
- 230000001225 therapeutic effect Effects 0.000 claims abstract description 17
- 230000002792 vascular Effects 0.000 claims abstract description 17
- 230000006641 stabilisation Effects 0.000 claims abstract description 14
- 238000011105 stabilization Methods 0.000 claims abstract description 13
- 238000000034 method Methods 0.000 claims description 106
- -1 amino, sulfinyl Chemical group 0.000 claims description 80
- 208000032781 Diabetic cardiomyopathy Diseases 0.000 claims description 23
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 21
- 125000000217 alkyl group Chemical group 0.000 claims description 19
- 229910052739 hydrogen Inorganic materials 0.000 claims description 19
- 208000035475 disorder Diseases 0.000 claims description 17
- 125000000896 monocarboxylic acid group Chemical group 0.000 claims description 16
- 239000001257 hydrogen Substances 0.000 claims description 13
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 13
- 229920006395 saturated elastomer Polymers 0.000 claims description 13
- 125000000218 acetic acid group Chemical group C(C)(=O)* 0.000 claims description 12
- 229910052736 halogen Inorganic materials 0.000 claims description 12
- 150000002367 halogens Chemical group 0.000 claims description 12
- 125000001501 propionyl group Chemical group O=C([*])C([H])([H])C([H])([H])[H] 0.000 claims description 12
- 208000030518 CARASIL syndrome Diseases 0.000 claims description 11
- 101000941994 Homo sapiens Protein cereblon Proteins 0.000 claims description 11
- 102100032783 Protein cereblon Human genes 0.000 claims description 11
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 claims description 11
- 208000031464 Cavernous Central Nervous System Hemangioma Diseases 0.000 claims description 10
- 208000005145 Cerebral amyloid angiopathy Diseases 0.000 claims description 10
- 210000003169 central nervous system Anatomy 0.000 claims description 10
- 208000020832 chronic kidney disease Diseases 0.000 claims description 10
- 125000005842 heteroatom Chemical group 0.000 claims description 10
- 125000000623 heterocyclic group Chemical group 0.000 claims description 10
- 229930195733 hydrocarbon Natural products 0.000 claims description 10
- 150000002430 hydrocarbons Chemical class 0.000 claims description 10
- 208000017169 kidney disease Diseases 0.000 claims description 10
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 10
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 10
- 239000004215 Carbon black (E152) Substances 0.000 claims description 9
- 239000003085 diluting agent Substances 0.000 claims description 9
- 208000019745 retinal vasculopathy with cerebral leukodystrophy Diseases 0.000 claims description 9
- 208000029941 retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations Diseases 0.000 claims description 9
- 208000010482 CADASIL Diseases 0.000 claims description 8
- 208000033221 Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy Diseases 0.000 claims description 8
- 208000031953 Hereditary hemorrhagic telangiectasia Diseases 0.000 claims description 8
- 239000002671 adjuvant Substances 0.000 claims description 8
- 230000008499 blood brain barrier function Effects 0.000 claims description 8
- 210000001218 blood-brain barrier Anatomy 0.000 claims description 8
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 8
- 125000006414 CCl Chemical group ClC* 0.000 claims description 6
- 125000001424 substituent group Chemical group 0.000 claims description 6
- 208000024827 Alzheimer disease Diseases 0.000 claims description 5
- 208000033935 Cerebral autosomal dominant arteriopathy-subcortical infarcts-leukoencephalopathy Diseases 0.000 claims description 5
- 208000033909 Cerebral autosomal recessive arteriopathy-subcortical infarcts-leukoencephalopathy Diseases 0.000 claims description 5
- 208000023105 Huntington disease Diseases 0.000 claims description 5
- 206010067362 Radiation necrosis Diseases 0.000 claims description 5
- 208000006011 Stroke Diseases 0.000 claims description 5
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 claims description 5
- 208000016886 cerebral arteriopathy with subcortical infarcts and leukoencephalopathy Diseases 0.000 claims description 5
- 206010015037 epilepsy Diseases 0.000 claims description 5
- 230000000670 limiting effect Effects 0.000 claims description 5
- 201000006417 multiple sclerosis Diseases 0.000 claims description 5
- 210000002220 organoid Anatomy 0.000 claims description 5
- 208000020431 spinal cord injury Diseases 0.000 claims description 5
- 206010010356 Congenital anomaly Diseases 0.000 claims description 4
- 208000002249 Diabetes Complications Diseases 0.000 claims description 4
- 208000007342 Diabetic Nephropathies Diseases 0.000 claims description 4
- 206010012689 Diabetic retinopathy Diseases 0.000 claims description 4
- 125000003118 aryl group Chemical group 0.000 claims description 4
- 230000017531 blood circulation Effects 0.000 claims description 4
- 208000033679 diabetic kidney disease Diseases 0.000 claims description 4
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 claims description 4
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 claims description 4
- 125000004123 n-propyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])* 0.000 claims description 4
- 208000011580 syndromic disease Diseases 0.000 claims description 4
- 208000010159 IgA glomerulonephritis Diseases 0.000 claims description 3
- 206010021263 IgA nephropathy Diseases 0.000 claims description 3
- 206010061218 Inflammation Diseases 0.000 claims description 3
- 208000005777 Lupus Nephritis Diseases 0.000 claims description 3
- 208000018737 Parkinson disease Diseases 0.000 claims description 3
- 208000030886 Traumatic Brain injury Diseases 0.000 claims description 3
- 201000004810 Vascular dementia Diseases 0.000 claims description 3
- 239000003937 drug carrier Substances 0.000 claims description 3
- 230000004054 inflammatory process Effects 0.000 claims description 3
- 210000000056 organ Anatomy 0.000 claims description 3
- 208000030761 polycystic kidney disease Diseases 0.000 claims description 3
- 230000000451 tissue damage Effects 0.000 claims description 3
- 231100000827 tissue damage Toxicity 0.000 claims description 3
- 230000009529 traumatic brain injury Effects 0.000 claims description 3
- 208000032131 Diabetic Neuropathies Diseases 0.000 claims description 2
- 206010020772 Hypertension Diseases 0.000 claims description 2
- 125000004186 cyclopropylmethyl group Chemical group [H]C([H])(*)C1([H])C([H])([H])C1([H])[H] 0.000 claims description 2
- 230000001631 hypertensive effect Effects 0.000 claims description 2
- 125000001972 isopentyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])C([H])([H])* 0.000 claims description 2
- 125000001971 neopentyl group Chemical group [H]C([*])([H])C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 claims description 2
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 2
- 125000005301 thienylmethyl group Chemical group [H]C1=C([H])C([H])=C(S1)C([H])([H])* 0.000 claims description 2
- 125000004453 alkoxycarbonyl group Chemical group 0.000 claims 8
- 125000004448 alkyl carbonyl group Chemical group 0.000 claims 8
- 125000005843 halogen group Chemical group 0.000 claims 5
- 125000004209 (C1-C8) alkyl group Chemical group 0.000 claims 3
- 125000004178 (C1-C4) alkyl group Chemical group 0.000 claims 2
- 125000006519 CCH3 Chemical group 0.000 claims 2
- 125000004432 carbon atom Chemical group C* 0.000 claims 2
- 125000001316 cycloalkyl alkyl group Chemical group 0.000 claims 2
- 125000000753 cycloalkyl group Chemical group 0.000 claims 2
- 125000004475 heteroaralkyl group Chemical group 0.000 claims 2
- 125000000592 heterocycloalkyl group Chemical group 0.000 claims 2
- 125000005885 heterocycloalkylalkyl group Chemical group 0.000 claims 2
- 125000000472 sulfonyl group Chemical group *S(*)(=O)=O 0.000 claims 2
- 206010008190 Cerebrovascular accident Diseases 0.000 claims 1
- 102000036364 Cullin Ring E3 Ligases Human genes 0.000 claims 1
- 206010055171 Hypertensive nephropathy Diseases 0.000 claims 1
- 125000003342 alkenyl group Chemical group 0.000 claims 1
- 125000004183 alkoxy alkyl group Chemical group 0.000 claims 1
- 125000000304 alkynyl group Chemical group 0.000 claims 1
- 208000026106 cerebrovascular disease Diseases 0.000 claims 1
- 238000012423 maintenance Methods 0.000 claims 1
- 230000002265 prevention Effects 0.000 abstract description 4
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 150
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 122
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 94
- 238000005481 NMR spectroscopy Methods 0.000 description 91
- 239000007787 solid Substances 0.000 description 82
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 63
- 235000019439 ethyl acetate Nutrition 0.000 description 58
- 239000011541 reaction mixture Substances 0.000 description 42
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 39
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 36
- 239000000243 solution Substances 0.000 description 35
- 238000004440 column chromatography Methods 0.000 description 32
- 238000004128 high performance liquid chromatography Methods 0.000 description 31
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 29
- 229960003433 thalidomide Drugs 0.000 description 29
- 238000003756 stirring Methods 0.000 description 26
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 24
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 24
- GOTYRUGSSMKFNF-UHFFFAOYSA-N lenalidomide Chemical compound C1C=2C(N)=CC=CC=2C(=O)N1C1CCC(=O)NC1=O GOTYRUGSSMKFNF-UHFFFAOYSA-N 0.000 description 24
- 239000000725 suspension Substances 0.000 description 23
- 239000012043 crude product Substances 0.000 description 21
- 229960004942 lenalidomide Drugs 0.000 description 20
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 19
- 241000699670 Mus sp. Species 0.000 description 19
- NSNHWTBQMQIDCF-UHFFFAOYSA-N dihydrate;hydrochloride Chemical compound O.O.Cl NSNHWTBQMQIDCF-UHFFFAOYSA-N 0.000 description 19
- 238000001644 13C nuclear magnetic resonance spectroscopy Methods 0.000 description 17
- 239000000047 product Substances 0.000 description 15
- 238000010992 reflux Methods 0.000 description 15
- 239000002904 solvent Substances 0.000 description 14
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 13
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 13
- IGAZHQIYONOHQN-UHFFFAOYSA-N Alexa Fluor 555 Chemical compound C=12C=CC(=N)C(S(O)(=O)=O)=C2OC2=C(S(O)(=O)=O)C(N)=CC=C2C=1C1=CC=C(C(O)=O)C=C1C(O)=O IGAZHQIYONOHQN-UHFFFAOYSA-N 0.000 description 12
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 12
- 229910001868 water Inorganic materials 0.000 description 12
- 210000004556 brain Anatomy 0.000 description 11
- 230000000069 prophylactic effect Effects 0.000 description 11
- 238000001959 radiotherapy Methods 0.000 description 11
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 10
- 239000003981 vehicle Substances 0.000 description 10
- ROFZMKDROVBLNY-UHFFFAOYSA-N 4-nitro-2-benzofuran-1,3-dione Chemical compound [O-][N+](=O)C1=CC=CC2=C1C(=O)OC2=O ROFZMKDROVBLNY-UHFFFAOYSA-N 0.000 description 9
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 9
- 239000012267 brine Substances 0.000 description 9
- 238000006243 chemical reaction Methods 0.000 description 9
- 230000000694 effects Effects 0.000 description 9
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 9
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 8
- WETWJCDKMRHUPV-UHFFFAOYSA-N acetyl chloride Chemical compound CC(Cl)=O WETWJCDKMRHUPV-UHFFFAOYSA-N 0.000 description 8
- 239000012346 acetyl chloride Substances 0.000 description 8
- VHRGRCVQAFMJIZ-UHFFFAOYSA-N cadaverine Chemical compound NCCCCCN VHRGRCVQAFMJIZ-UHFFFAOYSA-N 0.000 description 8
- 239000003153 chemical reaction reagent Substances 0.000 description 8
- NUJJHSPYMWDZSJ-UHFFFAOYSA-N ethyl 4-nitro-1,3-dioxoisoindole-2-carboxylate Chemical compound C1=CC([N+]([O-])=O)=C2C(=O)N(C(=O)OCC)C(=O)C2=C1 NUJJHSPYMWDZSJ-UHFFFAOYSA-N 0.000 description 8
- 238000002474 experimental method Methods 0.000 description 8
- 230000000971 hippocampal effect Effects 0.000 description 8
- KNCYXPMJDCCGSJ-UHFFFAOYSA-N piperidine-2,6-dione Chemical group O=C1CCCC(=O)N1 KNCYXPMJDCCGSJ-UHFFFAOYSA-N 0.000 description 8
- XQJHPSIERFKZFW-UHFFFAOYSA-N 1-(3-aminoazetidin-1-yl)ethanone;hydrochloride Chemical compound Cl.CC(=O)N1CC(N)C1 XQJHPSIERFKZFW-UHFFFAOYSA-N 0.000 description 7
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 7
- 150000001412 amines Chemical class 0.000 description 7
- 239000003480 eluent Substances 0.000 description 7
- 238000010172 mouse model Methods 0.000 description 7
- 210000002569 neuron Anatomy 0.000 description 7
- 229960000688 pomalidomide Drugs 0.000 description 7
- UVSMNLNDYGZFPF-UHFFFAOYSA-N pomalidomide Chemical compound O=C1C=2C(N)=CC=CC=2C(=O)N1C1CCC(=O)NC1=O UVSMNLNDYGZFPF-UHFFFAOYSA-N 0.000 description 7
- BDKLKNJTMLIAFE-UHFFFAOYSA-N 2-(3-fluorophenyl)-1,3-oxazole-4-carbaldehyde Chemical compound FC1=CC=CC(C=2OC=C(C=O)N=2)=C1 BDKLKNJTMLIAFE-UHFFFAOYSA-N 0.000 description 6
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 6
- 102100037241 Endoglin Human genes 0.000 description 6
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 6
- 241001465754 Metazoa Species 0.000 description 6
- OFBQJSOFQDEBGM-UHFFFAOYSA-N Pentane Chemical compound CCCCC OFBQJSOFQDEBGM-UHFFFAOYSA-N 0.000 description 6
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 6
- 238000010549 co-Evaporation Methods 0.000 description 6
- 210000002889 endothelial cell Anatomy 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 235000017281 sodium acetate Nutrition 0.000 description 6
- 229940087562 sodium acetate trihydrate Drugs 0.000 description 6
- 229910052717 sulfur Inorganic materials 0.000 description 6
- 208000032028 Microvascular Rarefaction Diseases 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 229960000583 acetic acid Drugs 0.000 description 5
- 239000002253 acid Substances 0.000 description 5
- 230000002490 cerebral effect Effects 0.000 description 5
- 230000001684 chronic effect Effects 0.000 description 5
- 238000009109 curative therapy Methods 0.000 description 5
- 238000003818 flash chromatography Methods 0.000 description 5
- VLKZOEOYAKHREP-UHFFFAOYSA-N n-Hexane Chemical compound CCCCCC VLKZOEOYAKHREP-UHFFFAOYSA-N 0.000 description 5
- 230000004770 neurodegeneration Effects 0.000 description 5
- 238000011321 prophylaxis Methods 0.000 description 5
- 238000011002 quantification Methods 0.000 description 5
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 4
- PAYRUJLWNCNPSJ-UHFFFAOYSA-N Aniline Chemical compound NC1=CC=CC=C1 PAYRUJLWNCNPSJ-UHFFFAOYSA-N 0.000 description 4
- 101001092197 Homo sapiens RNA binding protein fox-1 homolog 3 Proteins 0.000 description 4
- CSNNHWWHGAXBCP-UHFFFAOYSA-L Magnesium sulfate Chemical compound [Mg+2].[O-][S+2]([O-])([O-])[O-] CSNNHWWHGAXBCP-UHFFFAOYSA-L 0.000 description 4
- 102100035530 RNA binding protein fox-1 homolog 3 Human genes 0.000 description 4
- IMOZEMNVLZVGJZ-QGZVFWFLSA-N apremilast Chemical compound C1=C(OC)C(OCC)=CC([C@@H](CS(C)(=O)=O)N2C(C3=C(NC(C)=O)C=CC=C3C2=O)=O)=C1 IMOZEMNVLZVGJZ-QGZVFWFLSA-N 0.000 description 4
- 229960001164 apremilast Drugs 0.000 description 4
- 210000004369 blood Anatomy 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- 229910052799 carbon Inorganic materials 0.000 description 4
- 230000015556 catabolic process Effects 0.000 description 4
- 230000004087 circulation Effects 0.000 description 4
- 238000006482 condensation reaction Methods 0.000 description 4
- 239000012362 glacial acetic acid Substances 0.000 description 4
- 150000004820 halides Chemical group 0.000 description 4
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 4
- XNGIFLGASWRNHJ-UHFFFAOYSA-N phthalic acid Chemical compound OC(=O)C1=CC=CC=C1C(O)=O XNGIFLGASWRNHJ-UHFFFAOYSA-N 0.000 description 4
- 125000005543 phthalimide group Chemical class 0.000 description 4
- 238000001556 precipitation Methods 0.000 description 4
- 238000000746 purification Methods 0.000 description 4
- 239000011780 sodium chloride Substances 0.000 description 4
- 229960001603 tamoxifen Drugs 0.000 description 4
- 108010036395 Endoglin Proteins 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 206010058558 Hypoperfusion Diseases 0.000 description 3
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 3
- LGRFSURHDFAFJT-UHFFFAOYSA-N Phthalic anhydride Natural products C1=CC=C2C(=O)OC(=O)C2=C1 LGRFSURHDFAFJT-UHFFFAOYSA-N 0.000 description 3
- RWRDLPDLKQPQOW-UHFFFAOYSA-N Pyrrolidine Chemical compound C1CCNC1 RWRDLPDLKQPQOW-UHFFFAOYSA-N 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 239000012298 atmosphere Substances 0.000 description 3
- 230000027455 binding Effects 0.000 description 3
- JHIWVOJDXOSYLW-UHFFFAOYSA-N butyl 2,2-difluorocyclopropane-1-carboxylate Chemical compound CCCCOC(=O)C1CC1(F)F JHIWVOJDXOSYLW-UHFFFAOYSA-N 0.000 description 3
- 210000004027 cell Anatomy 0.000 description 3
- 230000008602 contraction Effects 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- RIFGWPKJUGCATF-UHFFFAOYSA-N ethyl chloroformate Chemical compound CCOC(Cl)=O RIFGWPKJUGCATF-UHFFFAOYSA-N 0.000 description 3
- 239000000706 filtrate Substances 0.000 description 3
- 230000002519 immonomodulatory effect Effects 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 229910017604 nitric acid Inorganic materials 0.000 description 3
- 229910052760 oxygen Inorganic materials 0.000 description 3
- 229940002612 prodrug Drugs 0.000 description 3
- 239000000651 prodrug Substances 0.000 description 3
- 239000000700 radioactive tracer Substances 0.000 description 3
- 230000009467 reduction Effects 0.000 description 3
- 230000002207 retinal effect Effects 0.000 description 3
- 150000003839 salts Chemical class 0.000 description 3
- 239000000377 silicon dioxide Substances 0.000 description 3
- 239000000758 substrate Substances 0.000 description 3
- 210000004509 vascular smooth muscle cell Anatomy 0.000 description 3
- KDSNLYIMUZNERS-UHFFFAOYSA-N 2-methylpropanamine Chemical compound CC(C)CN KDSNLYIMUZNERS-UHFFFAOYSA-N 0.000 description 2
- MSYGAHOHLUJIKV-UHFFFAOYSA-N 3,5-dimethyl-1-(3-nitrophenyl)-1h-pyrazole-4-carboxylic acid ethyl ester Chemical compound CC1=C(C(=O)OCC)C(C)=NN1C1=CC=CC([N+]([O-])=O)=C1 MSYGAHOHLUJIKV-UHFFFAOYSA-N 0.000 description 2
- QFUNCNMKEINNGS-UHFFFAOYSA-N 3-chloro-6-nitrophthalic acid Chemical compound [N+](=O)([O-])C1=C(C(C(=O)O)=C(C=C1)Cl)C(=O)O QFUNCNMKEINNGS-UHFFFAOYSA-N 0.000 description 2
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 2
- KQVIOSCWNOSSPK-UHFFFAOYSA-N 4-amino-2-(2-methylpropyl)isoindole-1,3-dione Chemical compound C1=CC(N)=C2C(=O)N(CC(C)C)C(=O)C2=C1 KQVIOSCWNOSSPK-UHFFFAOYSA-N 0.000 description 2
- IYNXMRJUAJBGGM-UHFFFAOYSA-N 4-amino-2-phenylisoindole-1,3-dione Chemical compound O=C1C=2C(N)=CC=CC=2C(=O)N1C1=CC=CC=C1 IYNXMRJUAJBGGM-UHFFFAOYSA-N 0.000 description 2
- FHGYVAFCOQSODD-UHFFFAOYSA-N 4-methyl-3-nitrophthalic acid Chemical compound CC1=CC=C(C(O)=O)C(C(O)=O)=C1[N+]([O-])=O FHGYVAFCOQSODD-UHFFFAOYSA-N 0.000 description 2
- BJPJKBJLPHGODJ-UHFFFAOYSA-N 4-methyl-5-nitrophthalic acid Chemical compound CC1=CC(C(O)=O)=C(C(O)=O)C=C1[N+]([O-])=O BJPJKBJLPHGODJ-UHFFFAOYSA-N 0.000 description 2
- OMOHDUGKQSTKSR-UHFFFAOYSA-N 4-nitro-2-phenylisoindole-1,3-dione Chemical compound O=C1C=2C([N+](=O)[O-])=CC=CC=2C(=O)N1C1=CC=CC=C1 OMOHDUGKQSTKSR-UHFFFAOYSA-N 0.000 description 2
- BONIIQYTWOPUQI-UHFFFAOYSA-N 4-nitroisoindole-1,3-dione Chemical compound [O-][N+](=O)C1=CC=CC2=C1C(=O)NC2=O BONIIQYTWOPUQI-UHFFFAOYSA-N 0.000 description 2
- KSLZRIXXMKXDEO-UHFFFAOYSA-N 5-chloro-2-methyl-3-nitrobenzoic acid Chemical compound CC1=C(C(O)=O)C=C(Cl)C=C1[N+]([O-])=O KSLZRIXXMKXDEO-UHFFFAOYSA-N 0.000 description 2
- 200000000007 Arterial disease Diseases 0.000 description 2
- BVKZGUZCCUSVTD-UHFFFAOYSA-M Bicarbonate Chemical compound OC([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-M 0.000 description 2
- 208000031229 Cardiomyopathies Diseases 0.000 description 2
- 208000017667 Chronic Disease Diseases 0.000 description 2
- 206010012289 Dementia Diseases 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 102000058063 Glucose Transporter Type 1 Human genes 0.000 description 2
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 2
- 208000034800 Leukoencephalopathies Diseases 0.000 description 2
- 102000003960 Ligases Human genes 0.000 description 2
- 108090000364 Ligases Proteins 0.000 description 2
- 208000025205 Mantle-Cell Lymphoma Diseases 0.000 description 2
- YNAVUWVOSKDBBP-UHFFFAOYSA-N Morpholine Chemical compound C1COCCN1 YNAVUWVOSKDBBP-UHFFFAOYSA-N 0.000 description 2
- 208000017442 Retinal disease Diseases 0.000 description 2
- 206010038923 Retinopathy Diseases 0.000 description 2
- 108091006296 SLC2A1 Proteins 0.000 description 2
- ATJFFYVFTNAWJD-UHFFFAOYSA-N Tin Chemical compound [Sn] ATJFFYVFTNAWJD-UHFFFAOYSA-N 0.000 description 2
- 102000011017 Type 4 Cyclic Nucleotide Phosphodiesterases Human genes 0.000 description 2
- 108010037584 Type 4 Cyclic Nucleotide Phosphodiesterases Proteins 0.000 description 2
- 102000044159 Ubiquitin Human genes 0.000 description 2
- 108090000848 Ubiquitin Proteins 0.000 description 2
- 208000009443 Vascular Malformations Diseases 0.000 description 2
- 150000001263 acyl chlorides Chemical class 0.000 description 2
- 238000000540 analysis of variance Methods 0.000 description 2
- 150000008064 anhydrides Chemical class 0.000 description 2
- 230000001772 anti-angiogenic effect Effects 0.000 description 2
- HONIICLYMWZJFZ-UHFFFAOYSA-O azetidin-1-ium Chemical compound C1C[NH2+]C1 HONIICLYMWZJFZ-UHFFFAOYSA-O 0.000 description 2
- 230000004888 barrier function 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
- 230000004378 blood-retinal barrier Effects 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 239000008366 buffered solution Substances 0.000 description 2
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 2
- 230000000052 comparative effect Effects 0.000 description 2
- 230000001054 cortical effect Effects 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 230000007831 electrophysiology Effects 0.000 description 2
- 238000002001 electrophysiology Methods 0.000 description 2
- VRHAQNTWKSVEEC-UHFFFAOYSA-N ethyl 1,3-dioxoisoindole-2-carboxylate Chemical compound C1=CC=C2C(=O)N(C(=O)OCC)C(=O)C2=C1 VRHAQNTWKSVEEC-UHFFFAOYSA-N 0.000 description 2
- 210000003128 head Anatomy 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 239000007928 intraperitoneal injection Substances 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 229910052943 magnesium sulfate Inorganic materials 0.000 description 2
- 235000019341 magnesium sulphate Nutrition 0.000 description 2
- 125000005439 maleimidyl group Chemical group C1(C=CC(N1*)=O)=O 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- SYSQUGFVNFXIIT-UHFFFAOYSA-N n-[4-(1,3-benzoxazol-2-yl)phenyl]-4-nitrobenzenesulfonamide Chemical class C1=CC([N+](=O)[O-])=CC=C1S(=O)(=O)NC1=CC=C(C=2OC3=CC=CC=C3N=2)C=C1 SYSQUGFVNFXIIT-UHFFFAOYSA-N 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 230000010412 perfusion Effects 0.000 description 2
- 230000000144 pharmacologic effect Effects 0.000 description 2
- 238000004321 preservation Methods 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 238000001953 recrystallisation Methods 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 210000001525 retina Anatomy 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- FNIHWOMOGHSTAT-UHFFFAOYSA-N tert-butyl 3-(1,3-dioxoisoindol-2-yl)azetidine-1-carboxylate Chemical compound C1N(C(=O)OC(C)(C)C)CC1N1C(=O)C2=CC=CC=C2C1=O FNIHWOMOGHSTAT-UHFFFAOYSA-N 0.000 description 2
- 210000001685 thyroid gland Anatomy 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 230000008728 vascular permeability Effects 0.000 description 2
- 210000003462 vein Anatomy 0.000 description 2
- LPBSHGLDBQBSPI-YFKPBYRVSA-N (2s)-2-amino-4,4-dimethylpentanoic acid Chemical compound CC(C)(C)C[C@H](N)C(O)=O LPBSHGLDBQBSPI-YFKPBYRVSA-N 0.000 description 1
- XGUXJMWPVJQIHI-YFKPBYRVSA-N (2s)-2-azaniumyl-3-cyclopropylpropanoate Chemical compound [O-]C(=O)[C@@H]([NH3+])CC1CC1 XGUXJMWPVJQIHI-YFKPBYRVSA-N 0.000 description 1
- 238000005160 1H NMR spectroscopy Methods 0.000 description 1
- DWUJDNHHQKEOPR-UHFFFAOYSA-N 2-(2-piperidin-4-ylethyl)pyridine Chemical compound C1CNCCC1CCC1=CC=CC=N1 DWUJDNHHQKEOPR-UHFFFAOYSA-N 0.000 description 1
- LWIXHJOHMBBSOY-UHFFFAOYSA-N 2-(azetidin-3-yl)isoindole-1,3-dione;hydrochloride Chemical compound Cl.O=C1C2=CC=CC=C2C(=O)N1C1CNC1 LWIXHJOHMBBSOY-UHFFFAOYSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- 229960000549 4-dimethylaminophenol Drugs 0.000 description 1
- CWJJAFQCTXFSTA-UHFFFAOYSA-N 4-methylphthalic acid Chemical compound CC1=CC=C(C(O)=O)C(C(O)=O)=C1 CWJJAFQCTXFSTA-UHFFFAOYSA-N 0.000 description 1
- CSAPESWNZDOAFU-UHFFFAOYSA-N 5-chloro-2-methylbenzoic acid Chemical compound CC1=CC=C(Cl)C=C1C(O)=O CSAPESWNZDOAFU-UHFFFAOYSA-N 0.000 description 1
- MMVIDXVHQANYAE-UHFFFAOYSA-N 5-nitro-2-benzofuran-1,3-dione Chemical compound [O-][N+](=O)C1=CC=C2C(=O)OC(=O)C2=C1 MMVIDXVHQANYAE-UHFFFAOYSA-N 0.000 description 1
- 206010002091 Anaesthesia Diseases 0.000 description 1
- 206010059245 Angiopathy Diseases 0.000 description 1
- 208000010839 B-cell chronic lymphocytic leukemia Diseases 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- 208000032170 Congenital Abnormalities Diseases 0.000 description 1
- 101100393884 Drosophila melanogaster Glut1 gene Proteins 0.000 description 1
- 238000001061 Dunnett's test Methods 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 101001019600 Homo sapiens Interleukin-17 receptor B Proteins 0.000 description 1
- 102100035014 Interleukin-17 receptor B Human genes 0.000 description 1
- 239000004395 L-leucine Substances 0.000 description 1
- 235000019454 L-leucine Nutrition 0.000 description 1
- 101150101095 Mmp12 gene Proteins 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 230000006181 N-acylation Effects 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 208000012868 Overgrowth Diseases 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 241000183024 Populus tremula Species 0.000 description 1
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 1
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 1
- XGUXJMWPVJQIHI-UHFFFAOYSA-N S-cyclopropylalanine Natural products OC(=O)C(N)CC1CC1 XGUXJMWPVJQIHI-UHFFFAOYSA-N 0.000 description 1
- 101150058068 SLC2A1 gene Proteins 0.000 description 1
- 206010040914 Skin reaction Diseases 0.000 description 1
- 206010043189 Telangiectasia Diseases 0.000 description 1
- 208000007536 Thrombosis Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- HFBMWMNUJJDEQZ-UHFFFAOYSA-N acryloyl chloride Chemical compound ClC(=O)C=C HFBMWMNUJJDEQZ-UHFFFAOYSA-N 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 210000004504 adult stem cell Anatomy 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 229940024606 amino acid Drugs 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 230000037005 anaesthesia Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 208000007502 anemia Diseases 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000001446 anti-myeloma Effects 0.000 description 1
- 230000001028 anti-proliverative effect Effects 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000012223 aqueous fraction Substances 0.000 description 1
- 210000002565 arteriole Anatomy 0.000 description 1
- 210000001367 artery Anatomy 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- HONIICLYMWZJFZ-UHFFFAOYSA-N azetidine Chemical compound C1CNC1 HONIICLYMWZJFZ-UHFFFAOYSA-N 0.000 description 1
- 150000001539 azetidines Chemical class 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- PASDCCFISLVPSO-UHFFFAOYSA-N benzoyl chloride Chemical compound ClC(=O)C1=CC=CC=C1 PASDCCFISLVPSO-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 230000007698 birth defect Effects 0.000 description 1
- 230000006931 brain damage Effects 0.000 description 1
- 231100000874 brain damage Toxicity 0.000 description 1
- 208000029028 brain injury Diseases 0.000 description 1
- UTKQDSJYJIQJBX-MRVPVSSYSA-N butyl (3r)-3-aminopyrrolidine-1-carboxylate Chemical compound CCCCOC(=O)N1CC[C@@H](N)C1 UTKQDSJYJIQJBX-MRVPVSSYSA-N 0.000 description 1
- UTKQDSJYJIQJBX-QMMMGPOBSA-N butyl (3s)-3-aminopyrrolidine-1-carboxylate Chemical compound CCCCOC(=O)N1CC[C@H](N)C1 UTKQDSJYJIQJBX-QMMMGPOBSA-N 0.000 description 1
- DVECBJCOGJRVPX-UHFFFAOYSA-N butyryl chloride Chemical compound CCCC(Cl)=O DVECBJCOGJRVPX-UHFFFAOYSA-N 0.000 description 1
- 229910002091 carbon monoxide Inorganic materials 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 210000005056 cell body Anatomy 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 210000003710 cerebral cortex Anatomy 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 208000037976 chronic inflammation Diseases 0.000 description 1
- 230000006020 chronic inflammation Effects 0.000 description 1
- 230000006999 cognitive decline Effects 0.000 description 1
- 208000010877 cognitive disease Diseases 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 238000009833 condensation Methods 0.000 description 1
- 230000005494 condensation Effects 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- ZOOSILUVXHVRJE-UHFFFAOYSA-N cyclopropanecarbonyl chloride Chemical compound ClC(=O)C1CC1 ZOOSILUVXHVRJE-UHFFFAOYSA-N 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 230000036425 denaturation Effects 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 229960004132 diethyl ether Drugs 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 238000003255 drug test Methods 0.000 description 1
- 230000005584 early death Effects 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 210000003038 endothelium Anatomy 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000012224 gene deletion Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 230000002489 hematologic effect Effects 0.000 description 1
- VEIWYFRREFUNRC-UHFFFAOYSA-N hydron;piperidine;chloride Chemical compound [Cl-].C1CC[NH2+]CC1 VEIWYFRREFUNRC-UHFFFAOYSA-N 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 150000003949 imides Chemical class 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 239000000677 immunologic agent Substances 0.000 description 1
- 229940124541 immunological agent Drugs 0.000 description 1
- 210000004263 induced pluripotent stem cell Anatomy 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 230000005865 ionizing radiation Effects 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 229960003136 leucine Drugs 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 208000010125 myocardial infarction Diseases 0.000 description 1
- 201000001119 neuropathy Diseases 0.000 description 1
- 230000007823 neuropathy Effects 0.000 description 1
- 208000004235 neutropenia Diseases 0.000 description 1
- 125000000449 nitro group Chemical group [O-][N+](*)=O 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 150000007523 nucleic acids Chemical group 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 208000033808 peripheral neuropathy Diseases 0.000 description 1
- 239000002831 pharmacologic agent Substances 0.000 description 1
- 125000000612 phthaloyl group Chemical group C(C=1C(C(=O)*)=CC=CC1)(=O)* 0.000 description 1
- SIOXPEMLGUPBBT-UHFFFAOYSA-M picolinate Chemical compound [O-]C(=O)C1=CC=CC=N1 SIOXPEMLGUPBBT-UHFFFAOYSA-M 0.000 description 1
- DNUTZBZXLPWRJG-UHFFFAOYSA-M piperidine-1-carboxylate Chemical compound [O-]C(=O)N1CCCCC1 DNUTZBZXLPWRJG-UHFFFAOYSA-M 0.000 description 1
- 125000003386 piperidinyl group Chemical group 0.000 description 1
- 230000004983 pleiotropic effect Effects 0.000 description 1
- 238000010149 post-hoc-test Methods 0.000 description 1
- 239000012286 potassium permanganate Substances 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 238000003825 pressing Methods 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- RZWZRACFZGVKFM-UHFFFAOYSA-N propanoyl chloride Chemical compound CCC(Cl)=O RZWZRACFZGVKFM-UHFFFAOYSA-N 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 230000005810 radionecrosis Effects 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 231100000430 skin reaction Toxicity 0.000 description 1
- 230000035483 skin reaction Effects 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000012453 solvate Substances 0.000 description 1
- 230000003238 somatosensory effect Effects 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 208000009056 telangiectasis Diseases 0.000 description 1
- 231100000378 teratogenic Toxicity 0.000 description 1
- 230000003390 teratogenic effect Effects 0.000 description 1
- WPGLRFGDZJSQGI-UHFFFAOYSA-N tert-butyl 3-aminoazetidine-1-carboxylate Chemical compound CC(C)(C)OC(=O)N1CC(N)C1 WPGLRFGDZJSQGI-UHFFFAOYSA-N 0.000 description 1
- UXAVRNGUMLVNOR-UHFFFAOYSA-N tert-butyl n-(1-acetylazetidin-3-yl)carbamate Chemical compound CC(=O)N1CC(NC(=O)OC(C)(C)C)C1 UXAVRNGUMLVNOR-UHFFFAOYSA-N 0.000 description 1
- VLOLOLUAVFGUTD-UHFFFAOYSA-N tert-butyl n-(azetidin-3-yl)carbamate;hydron;chloride Chemical compound Cl.CC(C)(C)OC(=O)NC1CNC1 VLOLOLUAVFGUTD-UHFFFAOYSA-N 0.000 description 1
- FIDKFEIEZJGDBE-UHFFFAOYSA-N thieno[2,3-c]furan-4,6-dione Chemical compound S1C=CC2=C1C(=O)OC2=O FIDKFEIEZJGDBE-UHFFFAOYSA-N 0.000 description 1
- 206010043554 thrombocytopenia Diseases 0.000 description 1
- GZNAASVAJNXPPW-UHFFFAOYSA-M tin(4+) chloride dihydrate Chemical compound O.O.[Cl-].[Sn+4] GZNAASVAJNXPPW-UHFFFAOYSA-M 0.000 description 1
- FWPIDFUJEMBDLS-UHFFFAOYSA-L tin(II) chloride dihydrate Substances O.O.Cl[Sn]Cl FWPIDFUJEMBDLS-UHFFFAOYSA-L 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000011277 treatment modality Methods 0.000 description 1
- 230000004869 vascular alteration Effects 0.000 description 1
- 208000011733 vascular alteration Diseases 0.000 description 1
- 210000005166 vasculature Anatomy 0.000 description 1
- 210000000264 venule Anatomy 0.000 description 1
- 230000007998 vessel formation Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
- C07D401/04—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
-
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D209/00—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
- C07D209/02—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
- C07D209/44—Iso-indoles; Hydrogenated iso-indoles
- C07D209/48—Iso-indoles; Hydrogenated iso-indoles with oxygen atoms in positions 1 and 3, e.g. phthalimide
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D209/00—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
- C07D209/02—Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
- C07D209/44—Iso-indoles; Hydrogenated iso-indoles
- C07D209/48—Iso-indoles; Hydrogenated iso-indoles with oxygen atoms in positions 1 and 3, e.g. phthalimide
- C07D209/49—Iso-indoles; Hydrogenated iso-indoles with oxygen atoms in positions 1 and 3, e.g. phthalimide and having in the molecule an acyl radical containing a saturated three-membered ring, e.g. chrysanthemumic acid esters
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
- C07D403/04—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D409/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
- C07D409/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
- C07D409/06—Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
- C07D471/04—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D495/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
- C07D495/02—Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
- C07D495/04—Ortho-condensed systems
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Public Health (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- Engineering & Computer Science (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Hematology (AREA)
- Diabetes (AREA)
- Biomedical Technology (AREA)
- Neurology (AREA)
- Neurosurgery (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates to pharmaceutical compound, and formulations comprising the compound, wherein the compound has a structure according to formula I or ||: A4 A3' „@ N-… A \A1 ° (I) A4 A5 / O N—RX \ A 6 ° (II) The compound and formulations are suitable for use in as a medicament in the prevention or treatment of a disease or a disorder characterized by pericyte dysfunction, detachment and/or loss, in a therapeutic treatment for blood vessel stabilization, in a treatment of promoting, restoring or preserving pericyte attachment to blood vessels, and/or in a treatment of a vascular-associated disease in a patient.
Description
P352801NL -1- 1,3-ISOINDOLINEDIONE DERIVATIVES AND HETEROAROMATIC ANALOGUES FOR STIMULATING
ENDOTHELIAL CELL-PERICYTE INTERACTION
The present invention relates to the field of medicine. More particularly, it relates to the use of 1,3- isoindolinedione derivatives and heteroaromatic analogues thereof to stimulate endothelial cell-pericyte interaction and pericyte coverage of blood vessels.
Blood vessels are intricate networks of hollow tubes that continuously adapt structurally and functionally to ensure optimal delivery of nutrients and oxygen to every cell of the body. They are composed of endothelial cells forming the inner lining of the vessel walls and of mural cells referred to as Vascular Smooth Muscle cells (VSMCs) and pericytes (PCs). VSMCs surround arteries and veins whereas PCs are located within the basal membrane of capillaries, venules and terminal arterioles. PCs have important roles in blood vessel formation and stabilization, regulate endothelial cell survival and control blood barrier to solutes and immune cells as well as blood flow. Many serious diseases affecting millions of people worldwide are caused by or associated with PC death or migration away from blood vessels. These pathological responses can lead to chronic inflammation, excessive vessel growth or/and blood vessel rarefaction that ultimately can lead to hypoperfusion and tissue damage.
PC dysfunction has been reported to cause/contribute to the development of a broad range of conditions, such as diabetes (nephropathy and retinopathy), chronic kidney disease (CKD), cardiomyopathy, central nervous system (CNS) disorders such as stroke, epilepsy, spinal cord injury, dementia notably Alzheimer's disease, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis, radiation necrosis and small inherited vessel diseases such as Cerebral autosomal dominant arteriopathy with (Cadasil) and Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (Carasil), Cerebral Amyloid Angiopathy (CAA), Retinal Vasculopathy with Cerebral
Leukoencephalopathy and systemic manifestations (RVCL-S), Hereditary Hemorrhagic Telangiectasia (HHT) and Cerebral Cavernous Malformations (CCM). Most of these conditions currently still lack the availability of effective treatment that is suitable for chronic treatment. The present inventors and others, have previously provided proof of concept data that by promoting PC attachment to blood vessels, they could prevent blood vessel weakness caused by HHT! or by brain irradiation23. Thalidomide was particularly effective in this in both mice and patients suggesting that it may be useful in other diseases with similar underlying pathology.
Thalidomide and its analogs lenalidomide and pomalidomide are immunomodulatory imide drugs (IMiDs), which are highly effective treatment modalities for Multiple Myeloma and del(5q) Myelodysplasic
Syndrome. The class of compounds exerts pleiotropic effects including immunomodulatory, potent anti- inflammatory, anti-angiogenic and direct anti-myeloma activities. Mechanistically, these drugs act as a molecular bridge linking selected neo-substrates to the ubiquitin proteasome system for degradation. All
IMiDs are characterized by a glutarimide ring that binds to Cereblon (CRBN), a substrate of the E3 ubiquitin
P352801NL -2- ligase CRL4 complex to target proteins for degradation. The glutarimide moiety is responsible for most of their pharmacological activities but also for their adverse teratogenic and hematologic effects. Notably, accumulative evidence indicates that the immuno-modulatory, anti-angiogenic and direct anti-proliferative activities of all IMiDs depend only on the glutarimide motive and not on the phthalimide motive.
IMiDs cannot be used to treat chronic diseases. Patients treated with IMiDs can develop serious side effects including possible birth defects, fatigue, weakness, anemia, neuropathy, neutropenia, thrombocytopenia, blood clots causing stroke or heart attack, increase risk of death in people with chronic lymphocytic leukemia (CLL), risk of malighancies, severe liver problems, severe skin reactions, thyroid problems and risk in early death in people who have Mantle Cell Lymphoma (MCL).
Hence, it is an object of the present invention to provide compounds and/or treatments to promote endothelial cells-pericyte interaction and pericyte coverage of blood vessels that, unlike currently known
IMiDs, can be used in chronic treatments.
Accordingly, the present invention is directed to a pharmaceutical compound, wherein the compound has a structure according to formula | or II: 2
JAS
AF ar IV >) {3
AX J
; © (I) wherein
At, A2, A3 and A? each individually represent CR! or N;
A5 and AS each individually represent S, CR’ or N; wherein R' represents hydrogen, halogen, OH, C+-Cs alkyl, NO2, or NR2R3, wherein R2 and R? are each independently selected from H, C1-Cs alkyl, C1-Cs-alkyl-carbonyl, and C+-Cs- alkoxy-carbonyl; wherein Rx represents Y or CHR4R5, wherein Y represents a 4-, 5-, or 6-membered heterocycle comprising 1, 2, or 3 heteroatoms, wherein Y is optionally substituted with one or more substituents selected from the group consisting of halide, saturated or unsaturated C+-Cs hydrocarbon, Ci-Cs-alkoxy-carbonyl, or C+-Cs-alkyl-carbonyl, preferably acetyl or propionyl;
P352801NL -3- wherein R# represents H, Ci-Cs-alkyl, Ci-Ce-cycloalkyl, C1-Ce-cycloalkylalkyl, heteroaryl, C1-Cs- hetercaralkyl, C1-Cs-heterocycloalkyl, or C1-Cs-heterocycloalkylalkyl, wherein R5 represents H, OH, COOH, C+-Cs alkyl, preferably COOH, i-propyl or t-butyl; and wherein
R+ and R5 are not both H.
A reference to compounds of formula (I) and (Il) and sub-references as descried herein also includes pharmaceutically acceptable ionic forms, salts, solvates, isomers, including geometric and stereochemical isomers, tautomers, N- oxides, esters, prodrugs, isotopes and protected forms thereof, preferably pharmaceutically acceptable salts or prodrugs.
In a second aspect, the invention relates to a pharmaceutical compound, wherein the compound has a structure according to formula | or II: 3
AF oS ; is i NN
SE of a (I a so # TN, EE © (I) wherein
A1, A2, A3 and A? each individually represent CR! or N;
A5 and A® each individually represent S, CR! or N; wherein R' represents hydrogen, halogen, OH, C+1-Cs alkyl, NO2, or NR2R3, wherein R2 and R? are each independently selected from H, C1-Cs alkyl, C1-Cs-alkyl-carbonyl, and C4-Cs- alkoxy-carbonyl; wherein Rx represents H, Y or CHR*RS, wherein Y represents a 4-, 5-, or 6-membered heterocycle comprising 1, 2, or 3 heteroatoms, wherein Y is optionally substituted with one or more substituents selected from the group consisting of halide, saturated or unsaturated C:-Cs hydrocarbon, C1-Cs-alkoxy-carbonyl, or Cs-Ce-alkyl-carbonyl, preferably acetyl or propionyl; wherein R+ represents H, Ci-Cs-alkyl, C+-Cs-cycloalkyl, C+-Cs-cycloalkylalkyl, heteroaryl, C1-Cs- heteroaralkyl, C1+-Cs-heterocycloalkyl, or C+-Cs-heterocycloalkylalkyl, wherein R5 represents H, OH, COOH, C+-Cs alkyl, preferably COOH, i-propyl or t-butyl; optionally wherein R+ and R5 are not both H, for use as a medicament in a therapeutic treatment for stimulating endothelial cell-pericyte interaction and promoting PC coverage of blood vessels.
P352801NL -4-
In a third aspect, the invention relates to a pharmaceutical compound, wherein the compound has a structure according to formula | or II: 4
At
II
Fae 3 3 0
G
RE A
Saf (I) 5 wherein
A1, A2, A3 and A+ each individually represent CR! or N;
A5 and AS each individually represent S, CR! or N; wherein Rt represents hydrogen, halogen, OH, C+-Cs alkyl, NO2, or NR2R3, wherein R2 and R3 are each independently selected from H, C1-Cs alkyl, C1+-Cs-alkyl-carbonyl, and C1-Cs- alkoxy-carbonyl; wherein Rx represents H, Y or CHR*RS, wherein Y represents a 4-, 5-, or 6-membered heterocycle comprising 1, 2, or 3 heteroatoms, wherein Y is optionally substituted with one or more substituents selected from the group consisting of halide, saturated or unsaturated C1-Cs hydrocarbon, Ci-Ce-alkoxy-carbonyl, or C+-Cs-alkyl-carbonyl, preferably acetyl or propionyl; wherein R* represents H, Ci-Cs-alkyl, C-Cs-cycloalkyl, C:+-Cs-cycloalkylalkyl, heteroaryl, C1-Cs- hetercaralkyl, C1-Cs-heterocycloalkyl, or C1-Cs-heterocycloalkylalkyl, wherein R5 represents H, OH, COOH, C+-Cs alkyl, preferably COOH, i-propyl or t-butyl; optionally wherein R+ and R® are not both H, for use as a medicament in the prophylactic or curative treatment of a disease or a disorder characterized by pericyte dysfunction, detachment and/or loss.
Figure 1 shows a comparative assessment of prevention of pericyte detachment and vascular leakage induced by radiation therapy by selected phthalimides.
Figure 2 shows a comparative assessment of preventing late vascular rarefaction and neuron loss induced by radiation therapy in the somatosensorial cortex (SS) and in the hippocampal region (Hp) by selected phthalimides.
Figure 3 demonstrate blood vessel stabilization properties by selected phthalimides as illustrated using a mouse model of Hereditary Hemorrhagic Telangiectasia (HHT) type 1.
P352801NL -5-
Figure 4 shows selected analogues of phthalimide that restore endothelial cell-pericyte interaction and blood vessel stability in accordance with an embodiment of the present invention.
The present invention provides new 1,3-isoindolinedione derivatives and heteroaromatic analogs thereof, as well as 1,3-isoindolinedione derivatives and heteroaromatic analogs thereof for use in stimulating endothelial cell-PC interaction and promoting PC coverage of blood vessels, and for use in the prevention or treatment of a disease or a disorder characterized by pericyte dysfunction, pericyte detachment and/or pericyte loss.
It was surprisingly found that these compounds are capable of preventing pericyte (PC) detachment, and promoting endothelial cells-pericyte interaction and pericyte coverage of blood vessels. As such, the compounds of the invention can advantageously be used in prophylactic or curative treatments of diseases or disorders characterized by pericyte dysfunction, detachment and/or loss, including curative therapeutic treatments comprising promotion of endothelial cell-PC interaction and blood vessel stabilization, as well as prophylactic and/or curative therapeutic treatments comprising promotion, restoration or preservation of
PC attachment to blood vessels.
The present inventors found that thalidomide but not its analogue lenalidomide is able to prevent pericyte (PC) detachment (Figure 1A-B) and Blood Brain Barrier (BBB) breakdown (Figure 1C) induced by radiation therapy in mice, as a model of early vessel injury associated with late neuron loss and cognitive decline.
The present inventors have also found that prophylactic treatment with thalidomide but not with its analogue lenalidomide (Figure 2A), by preventing PC detachment and BBB leakage, was able to prevent late capillary rarefaction and to prevent neuron loss in the cortical or hippocampal region induced by radiation therapy in the cortical (Figure 2B-F) or hippocampal region (Figure 2B, 2G-J). It was further observed that thalidomide and pomalidomide, contrary to their analogue lenalidomide, stimulate endothelial cell-PC interaction and blood vessel coverage in a mouse model of HHT using an ex vivo electrophysiology approach to quantify endothelial cell-PC interactions (Figure 3). Consequently, it was inferred that the carbonyl group (C=O) of the phthaloyl ring plays an important role in the ability of thalidomide to promote pericyte recruitment to the endothelium. It was also found that apremilast, an oral small-molecule inhibitor of phosphodiesterase 4 (PDE4) that shares the phthalimide ring with thalidomide but lacks the glutarimide ring and thus fails to bind to cereblon, could also rescue endothelial cell-PC interaction in a mouse model of HHT1 at high concentrations, confirming that the glutarimide ring is not required to promote PC coverage of the vasculature (Figure 3). Hence, it would appear that the isoindolinedione motive, as found in some
IMiDs such as thalidomide, but not in lenalidomide, appears to be the active fragment in compounds promoting PC attachment to blood vessels. It was furthermore observed that the glutarimide motive always present in the IMiDs is not essential to blood vessel stabilization and that major side effects associated with the known IMiDs may advantageously be suppressed by leaving out or replacing this structural motive.
Additionally, it was found that the claimed effects are obtained not only for phthalimide derivatives containing a Ce-aromatic moiety but also by other aromatic or heteroaromatic moieties.
P352801NL -6-
Accordingly, each At, A2, A3 and A* in the 5- or 6-membered aromatic moiety of the compounds as defined herein may individually represent CR or N, and A5 and AS each individually represent S, CR! or N; wherein R' represents hydrogen, halogen, OH, C1-Cs alkyl, NO2, or NR2R3, wherein R2 and R3 are each independently selected from H, C+-Cs alkyl, C+-Cs-alkyl-carbonyl, and C+-Cs-alkoxy-carbonyl.
In an embodiment, At or A2 represents C-NH2 or C-NHC(O)CHs; preferably A! represents C-NHz, and A2, A3 and A+ each individually represent CH, C-CH: or C-Cl, preferably CH. In an embodiment, A2 represents C-NHz, and At, A3 and A+ each individually represent CH, C-CH: or C-Cl, preferably CH.
In one embodiment, one of A5 and AS is S, the remainder of A+, AS and AS being CR! or CH.
The compounds according to the present invention can advantageously be used in treatments, e.g. prophylactic and/or curative therapeutic treatments, of a disease or a disorder characterized by pericyte dysfunction, detachment and/or loss; these involve therapeutic treatments, e.g. curative treatments, comprising promotion of endothelial cell-PC interaction and blood vessel stabilization, and prophylactic and/or curative therapeutic treatments comprising promoting, restoring or preserving pericyte attachment to blood vessels. As used herein, blood vessel stabilization refers inter alia to the protection of BBB integrity, control of blood flow, limitation of inflammation and hypoperfusion, and reduction of tissue damage, or combinations thereof.
Without wishing to be bound to any particular theory, it appears that the major side effects of thalidomide and its analogues such as lenalidomide and pomalidomide are caused at least in part by their aptness and capability to bind to the cereblon protein, which is the sole or primary target protein resulting in the breakdown of multiple neo-substrates mediated by the CRLACRBN ligase complex. Accordingly, the present inventors realized that at least some major side effects of phthalimide derivatives can be suppressed by selecting the Rxgroup such that the compound does essentially not bind to the cereblon protein. This insight allows rapid in vitro screening of toxicity of compounds that exhibit the beneficial effects of the invention.
The compounds as defined herein are N-substituted on the maleimide moiety with an Rx group which is chosen such that the compounds of the present invention do not lead to the binding to cereblon. This effectively excludes glutarimide as present in thalidomide, and hence thalidomide as such. In an embodiment, the lack of binding to cereblon (CRBN) of a compound according to the invention corresponds to the compound exhibiting a half-maximal inhibitory concentration (ICso) higher than 100 uM, preferably higher than 200 pM, more preferably higher than 500 uM or 1000 pM as determined in a competitive binding assay, as compared to the binding of thalidomide to cereblon.
Accordingly, in an embodiment the compound of formula | does not include thalidomide, pomalidomide, lenalidomide or any other compound comprising a glutarimide moiety such as present in these compounds.
The moiety RX is as defined herein and is further selected such that it does not interfere with the pharmaceutical activity.
In an embodiment, R* is Y, wherein Y represents a 4-, 5-, or 6-membered heterocycle comprising 1, 2, or 3 heteroatoms. Preferably, the heteroatoms are independently selected from N, O and S. Preferably,
Y represents a 4-, 5-, or 6-membered heterocycle comprising 1 heteroatom.
P352801NL -7-
Particular good results are obtained with compounds wherein R* represents a substituted or unsubstituted 4-, 5- or 8-membered heterocycle such as a morpholine, azetidine, pyrrolidine or piperidine group.
Accordingly, in an embodiment RX represents a 4-, 5-, or 6-membered heterocycle comprising 1, 2, or 3 heteroatoms, preferably 1 heteroatom, preferably 1 nitrogen atom.
Azetidines as group Rx display particularly good results. Accordingly, in a preferred embodiment of the invention, R* represents
ÔN-R wherein R® represents Ci-Cs-alkyl, C2-Cs-alkenyl, C2-Cs-alkynyl, C1-Cs-alkoxyalkyl, C1-Cs-alkoxy- carbonyl, aryl, or C1-Ce-alkyl-carbonyl, preferably acetyl or propionyl.
Advantageously, these compounds are synthetically relatively easily accessible by a condensation reaction of the corresponding amino acid H2N-C(R3)2-CO:H and the corresponding maleimide derivative.
In an alternative embodiment, R* represents CHR*R?, wherein R+ represents hydrogen, methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, tert-butyl, neopentyl, isopentyl, cyclopropyl-methyl, or thienyl-methyl.
In an embodiment, R5 represents COOH, methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, tert-butyl, preferably COOH, methyl, i-propyl or t-butyl.
In exemplary embodiments, the compound has a structure according to formula (1-2) {2 oo / 0 (8) wherein each Z is independently selected from hydrogen, halogen, OH, C+-Cs alkyl, NO2, and NR2R?, wherein R2 and R3 are each independently selected from H, C+-Cs alkyl, C1-Cs-alkyl-carbonyl, and C1-Cs- alkoxy-carbonyl, preferably wherein each Z is independently selected from H, halogen, NHz, and
NHC (O)CHs; wherein mis 1, 2, 3 or 4, preferably 1 or 2; and wherein R® represents saturated or unsaturated C+-Cs hydrocarbon, C1-Cs-alkoxy-carbonyl, or C1-Cs-alkyl- carbonyl, preferably acetyl or propionyl.
In some exemplary embodiments, A’ or A2 is C-NH2 and Rx is a an N-substituted azetidine group. In a preferred embodiment, the compound has a structure according to formula I-b or formula I-c:
P352801NL -8- 2 §
A
{z 1 | © fl
EY ae {
NH, “ 3 (l-b) 0
Ff ose % = Ay * 3 Ni {z DEN | : ) N Rs 4 ij (lc) wherein each Z is independently selected from hydrogen, halogen, OH, C1-Cs alkyl, and NO:2, preferably wherein each Z is independently selected from H, CHs and CI; wherein m is 1, 2, or 3; and wherein RS represents saturated or unsaturated C+-Cs hydrocarbon, C+-Cs-alkoxy-carbonyl, or C1-Cs-alkyl- carbonyl, preferably acetyl or propionyl.
In another aspect of the invention there is provided a formulation comprising a pharmaceutical compound as defined herein, the formulation further comprising a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier.
Table 1 lists exemplary compounds according to the invention.
Table 1
O 2 Oo in 2 JEN ene A £3
SIT Ne N Fe hy NL FI Ne x A Se
B 1 5, YY 0 No, © 0 @ o Q O et - 0 Ne, SN © JEN 3 <3 es nd . AN or i $i 3 en &F xy NL GN pe
Noo a No Xx Ni Ds hd Ny SN a i N
Se A ò HHT Ty o iH, © ò ®
P352801NL -9-
A
£3 > ad ge .
TO id nt g Ngati N oR \
NH © ety Nei 3 3 : ¥ \ } 3 3
J NH ö G £ © & € 0 Gt
FEIN een =. o we SN oo TN on, EN - ee ANE AY Sy I ) No AY Sy ta No x ; % i ù ds Py
Ni, UO fi, 0 Bn, U (10) (11) (12) 0 Pp 2
SN A 4 © , SEN ol i EE a a NH, Q NH, B (13) (14) (15) 3 Oo i { i ~ i 4 . Ri 5
LAA OT owed MN
TO en pon Te > (16) (17) (18) 0 o} 0 ad . 2 0 0
FT pe Ng
NH, © > NH, © NH, © (19) (20) (21)
P352801NL -10- 0 3 >
N—N LL NC NK yy N= NA /
Se ey a Are 0 wi, O Soa, © )
NH, NH, NB (22) (23) @4) 0 0 7
CF wed] a NC}
A on, eN Aeg ol N
T$ v A |g
Ni © ò ie NH, © 0 26) (27) (NET OTN
N Meg Ned x Sag “3 JON 28) (29) co 0 | 0 7 ed Spe & Fama 1 7 | O
DE NK 5 NONA
Ne pon OH
Ni, © 0 NH, © © NH, © oo) 33) (33) 0 3 = gek ST
N— NH HEI LiL NN © 2 ie X Yond
NH, © Np © (34) (35)
Hence, in an embodiment a compound (1), (2), (3), (4), (5), (6), (7), (8), (9), (10), (11), (12), (13), (14), (15), (18), (17), (18), (19), (20), (21), (22), (23), (24), (25), (26), (27), (28), (29), (30), (31), (32), (33), (34) or (35) as displayed in Table 1 is provided, and/or a compound (1), (2), (3), (4), (5), (6), (7), (8), (9), (10), (11), (12), 5 (13), (14), (15), (16), (17), (18), (19), (20), (21), (22), (23), (24), (25), (26), (27), (28), (29), (30), (31), (32),
P352801NL -11- (33), (34) or {35) for use as a medicament, particularly as a medicament in the prophylactic or curative treatment of a disease or a disorder characterized by pericyte dysfunction, detachment and/or loss, preferably in a treatment of promoting, restoring or preserving pericyte attachment to blood vessels. In an exemplary embodiment, a compound (1), (2), (5), (6), (7), (9), (10), (11), (15), (16), (17), (18), (19), (20), 21), (22), (23), or (24), as displayed in Table 1 is provided, as well as a formulation comprising such compound, for use as medicament, particularly for use as a medicament as defined herein.
Table 2 lists further exemplary compounds according to formula I-a, I-b and I-c.
Table 2 53 BY “ J RY i I; ADNR ner Ff
Comp. NIR IR : 1.005 C-H | C(O)O-t-butyl H : ae [en ow ow oom
Tae [en {ea Goce
P352801NL -12- 1.020 C-NHC(O)CHa ICH [CH CH | C(O)CH=CH: 1.021 C-NHC(O)CHs C-H CH CH C(O)O-t-butyl H
TE [EEE Con 1.023 C-NHC(O)CHs CH | C(O)-n-propyl 1.024 C-NHC(O)CHa CH | C(O)-neopentyl 1.025 C-NH: CHs CH CH C(O)CH: H 7 HCE oa SOR
P352801NL -13-
Hence, in an embodiment compounds 1.001 to 1.088 as disclosed in Table 2 are provided. In another embodiment compounds 1.001 to 1.088 as disclosed in Table 2, as well as a formulation comprising one or more of these compounds and a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier, for use as a medicament are provided, particularly for use as a medicament in the treatment of a disease or a disorder characterized by pericyte dysfunction, detachment and/or loss, preferably in a treatment of promoting, restoring or preserving pericyte attachment to blood vessels.
P352801NL -14-
Table 3 lists exemplary compounds according to formula [-a.
Table 3 3
MW
Co in
Sa Lr
TL N oe CL ER
TOT
2.004 CNH: | CH CH | CH C(O)CH=CH: H 2.006 C-H | C(O)phenyl H 2.008 CNH: | CH CH | CH C(O)-neopentyl H 2.010 C-H | C(O)CH2CHs H 2.012 CH [CNH CH | CH C(O)CH=CH: H 2.014 C-H | C(O)phenyl H 2.022 C-H | C(O)phenyl H ;
P352801NL -15-
CH | GOEREE
CH | EO eagen
TOF
Open)
COT espe
ORT
COT
Open)
Ee {cicero 2.048 C-Cl | C(O)-neopentyl H
COHEN as a {en earn fH 2.052 C-H | C(O)CH=CH2 H
Cowen as [om {en fear os 2.056 C-H | C(O)-neopentyl H
P352801NL -16-
ST | SO
ST | Gowen
SOE
CO
Come i
CORR
CO aes en {a ci [or nea [ee 2.086 C-H | C(O)phenyl CHa
Hence, in an embodiment compounds 2.001 to 2.088 as disclosed in Table 3 are provided. In another embodiment compounds 2.001 to 2.088 as disclosed in Table 3, as well as a formulation comprising one or more of these compounds and a pharmaceutically acceptable excipient, adjuvant, diluent and/or carrier, for use as a medicament are provided, particularly for use as a medicament in the treatment of a disease or a disorder characterized by pericyte dysfunction, detachment and/or loss, preferably in a treatment of promoting, restoring or preserving pericyte attachment to blood vessels.
Excipients are natural or synthetic substances formulated alongside an active ingredient (e.g. a nucleic acid sequence, vector, modified cell or isolated peptide as provided herein), included for the purpose of bulking-up the formulation or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption or solubility. Excipients can also be useful in the manufacturing process, to aid in the handling of the active substance concerned such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation over the expected shelf life. Pharmaceutically acceptable excipients are well known in the art.
A suitable excipient is therefore easily identifiable by one of ordinary skill in the art. By way of example,
P352801NL -17- suitable pharmaceutically acceptable excipients include water, saline, aqueous dextrose, glycerol, ethanol, and the like. Diluents are diluting agents. Pharmaceutically acceptable diluents are well known in the art. A suitable diluent is therefore easily identifiable by one of ordinary skill in the art. Carriers are non-toxic to recipients at the dosages and concentrations employed and are compatible with other ingredients of the formulation. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. Pharmaceutically acceptable carriers are well known in the art. A suitable carrier is therefore easily identifiable by one of ordinary skill in the art. Adjuvants are pharmacological and/or immunological agents that modify the effect of other agents in a formulation.
Pharmaceutically acceptable adjuvants are well known in the art. A suitable adjuvant is therefore easily identifiable by one of ordinary Skill in the art.
The compounds of the invention may be used in therapeutic treatments, including prophylactic and curative therapeutic treatments, of diseases or disorders characterized by pericyte dysfunction or pericyte loss, including curative therapeutic treatments comprising promotion of endothelial cell-pericyte interaction and blood vessel stabilization, as well as prophylactic and/or curative therapeutic treatments comprising restoration, promotion or preservation of pericyte attachment to blood vessels.
In an embodiment, the disease or disorder characterized by pericyte dysfunction, detachment and/or loss is selected from diabetes mellitus complications, chronic kidney disease (CKD), small vessel diseases,
Central Nervous System (CNS) diseases.
In an embodiment, the disease or disorder characterized by pericyte dysfunction, pericyte detachment and/or pericyte loss is selected from diabetic nephropathy (DN), diabetic retinopathy (DR), diabetic cardiomyopathy (DCM), and diabetic neuropathy.
In an embodiment, the disease or disorder characterized by pericyte dysfunction, detachment and/or loss is selected from hypertensive kidney diseases, IgA nephropathy, congenital nephropathy syndrome, lupus nephritis, polycystic kidney disease and allograft nephropathy.
In an embodiment, the disease or disorder characterized by pericyte dysfunction, detachment and/or loss is selected from cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (Cadasil), Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (Carasil), Cerebral Amyloid Angiopathy (CAA), Retinal Vasculopathy with Cerebral
Leukoencephalopathy and systemic manifestations (RVCL-S), Hereditary Hemorrhagic Telangiectasia (HHT), and Cerebral Cavernous Malformations (CCM).
In an embodiment, the disease or disorder characterized by pericyte dysfunction, detachment and/or loss is selected from stroke, epilepsy, spinal cord injury, vascular dementia, Alzheimer's disease,
Huntington's disease, Parkinson's disease, traumatic brain injury, multiple sclerosis, amyotrophic lateral sclerosis, and radiation necrosis.
In a further aspect the invention provides a compound or a formulation as defined herein for use as a medicament for stimulating endothelial cell-pericyte interaction and promoting pericyte coverage of blood vessels in a patient, and in a method for stimulating endothelial cell-PC interaction and promoting and PC
P352801NL -18- coverage of blood vessels in a patient, the method comprising administering compound or a formulation as defined herein.
In another aspect the invention relates to the compounds and formulations as defined herein for use as a medicament in a treatment of promoting endothelial cell-pericyte interaction, particularly of blood vessel stabilization, and/or a treatment of promoting or preserving pericyte attachment to blood vessels, as well as methods for the curative therapeutic treatment of promoting endothelial cell-pericyte interaction and blood vessel stabilization, and/or a treatment of promoting or preserving pericyte attachment to blood vessels.
In one embodiment, the treatment is a prophylactic therapeutic treatment.
In one embodiment, the treatment is curative therapeutic treatment.
The blood vessel stabilization properties and associated medical applicability of the compounds and formulations in accordance with the present invention can be established by an in vivo model using preclinical mouse models of HHT. By coupling electrophysiology and light microscopy, the ability of pericytes to modulate blood vessel constriction in response to an electrical stimulus can be quantified? (See also Figures 1 and 3). These experiments can be performed in whole retinas of control mice and of heterozygous or iKO endoglin mice (HHT1 mouse model) and in iKO endoglin mice treated with selected compounds (cf. Figures 3-4).
By comparing the activity of thalidomide and its analogue lenalidomide in the in vivo model of radio- necrosis and late brain damage, the essential role of both carbonyls groups in the phthalimide could be established (cf. Figures 1 and 2). Figure 1 shows how PC dysfuncticn induced by radiotherapy includes pericyte detachment that leads to BBB leakage, and in figure 2, to late blood capillary rarefaction and neuron loss.
Restoration of endothelial cell-PC interaction can be used to treat chronic diseases with vascular alterations. The compounds ofthe present invention are advantageously capable of sustaining blood vessel integrity limiting capillary rarefaction; promoting/maintaining blood barrier, notably blood-brain barrier (BBB) and blood-retina barrier (BRB), limiting vascular leakage and inflammation; reducing vessel overgrowth and the development of vascular malformations notably telangiectasia, low or high flow vascular malformations; restoring blood flow limiting tissue hypoperfusion and late damage. Thus, the compound according to the present invention, or a pharmaceutically acceptable salt or prodrug thereof, can be used in a medical treatment of vascular-associated diseases.
Accordingly, in one aspect of the invention there is provided the compounds and formulation as defined herein for use as a medicament in a prophylactic or curative treatment of a vascular-associated disease.
In another aspect of the invention there is provided method for the treatment of a vascular-associated disease in a patient, the method comprising administering to the patient a pharmaceutical compound, or a formulation comprising a pharmaceutical compound, wherein the compound has a structure according to formula | or II:
P352801NL -19- {3
AY
ES
> 0 2
IL, Pf eR 0) wherein
A", A2, A3 and A+ each individually represent CR! or N;
A5 and AS each individually represent S, CR! or N; wherein Rt represents hydrogen, halogen, OH, C+-Cs alkyl, NO2, or NR2R3, wherein R2 and R3 are each independently selected from H, C+-Cs alkyl, C+-Ce-alkyl-carbonyl, and C+-Cs- alkoxy-carbonyl; wherein Rx represents H, Y or CHR*RS, wherein Y represents a 4-, 5-, or 6-membered heterocycle comprising 1, 2, or 3 heteroatoms, wherein Y is optionally substituted with one or more substituents selected from the group consisting of halide, saturated or unsaturated C1-Cs hydrocarbon, Ci-Ce-alkoxy-carbonyl, or C+-Cs-alkyl-carbonyl, preferably acetyl or propionyl; wherein R* represents H, Ci-Cs-alkyl, C-Cs-cycloalkyl, C:+-Cs-cycloalkylalkyl, heteroaryl, C1-Cs- heteroaralkyl, C1-Cs-heterocycloalkyl, or C1-Cs-heterocycloalkylalkyl, wherein R5 represents H, OH, COOH, C+-Cs alkyl, preferably COOH, i-propyl or t-butyl; optionally wherein R+ and R5 are not both H.
In a further aspect of the present invention there is provided a method for promoting, restoring or preserving pericyte attachment to blood vessels in a subject, preferably in a method of treatment of a vascular-associated disease, the method comprising administering to the subject the compound or formulation as defined above.
In a further aspect of the present invention there is provided a method for stabilizing blood vessels in a subject, preferably in a method of treatment of a vascular associated disease, the method comprising administering to the subject the compound or formulation as defined above.
Specific examples vascular-associated diseases include diabetes mellitus complications (nephropathy and retinopathy), chronic kidney disease (CKD), cardiomyopathy, central nervous system (CNS) disorders such as stroke, epilepsy, spinal cord injury, dementia notably Alzheimer's disease,
Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis, radiation necrosis and small vessel diseases such as Cerebral autosomal dominant arteriopathy with (Cadasil and Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (Carasil), Cerebral Amyloid
P352801NL -20-
Angiopathy (CAA), Retinal Vasculopathy with Cerebral Leukoencephalopathy and systemic manifestations (RVCL-S), Hereditary Hemorrhagic Telangiectasia (HHT), Cerebral Cavernous Malformations (CCM). Most of these conditions currently still lack the availability of effective treatment that is suitable for chronic treatment.
Figure 1 shows that thalidomide but not its derivative lenalidomide prevents PC detachment and vascular leakage induced by radiation therapy. {A} Diagram of the injection protocol of vehicle, thalidomide or lenalidomide in combination with brain or eye radiation therapy. (B) Retinal PCs stimulated electrically induce localized vascular constriction. Top panels show representative bright field images of one blood capillary before, during and after PC electrical stimulation showing vessel constriction. The graph represents that the mean intensity inducing PC contraction and vascular constriction in control and irradiated mice treated with vehicle alone, thalidomide or lenalidomide. (C} Whole brains photographed after cadaverine Alexa Fluor-555 circulation and quantification of tracer accumulation in control and irradiated mice treated with vehicle alone, thalidomide or lenalidomide after 2 hours of tracer circulation.
Circulation time was 2 hours. All error bars show mean + s.e.m. ‘P<0.05; “P<0.01; *"P<0.001; ““P<0.0001 result from 1-way ANOVA and Dunnett's post hoc tests comparing the mean of each group to the control group. ns: non-significant.
Figure 2 shows that thalidomide but not its derivative lenalidomide prevents late vascular rarefaction and neuron loss induced by radiation therapy in the somatosensorial cortex (SS) and in the hippocampal region (Hp). (A) Diagram of the injection protocol of vehicle, thalidomide or lenalidomide in combination with brain radiation therapy. (B} Confocal image of a brain section stained for NeuN positive neurons showing the somatosensorial (SS) cortex and hippocampal region (Hp). (C-D) Confocal images of the somatosensorial cortex showing endothelial cells stained by GLUT1 (C) and neurons stained by NeuN (D) in control and irradiated mice treated with vehicle alone, thalidomide and lenalidomide. Animals were sacrificed 9 months after receiving a single dose of radiation therapy as indicated in A. (E)} Quantification of the vessel density in the somatosensorial cortex. (F} Number of neurons in the cortical region. (G-H)
Confocal images of the hippocampal region showing endothelial cells stained by GLUT1 (G) and neurons stained by NeuN (H) in control and irradiated mice treated with vehicle alone, thalidomide and lenalidomide. (1) Quantification of the vessel density in the hippocampal region. (F} Number of neurons in the hippocampal region.
Figure 3 reveals that thalidomide and selected FDA-approved analogues exert distinct blood vessel stabilization properties as illustrating using a mouse model of Hereditary Hemorrhagic Telangiectasia (HHT) type 1. (A) Scheme of tamoxifen (Tx) injection for the induction of Eng gene deletion (HHT1) and treatment plan with thalidomide and derivatives. (B) Chemical structures of thalidomide (THA), lenalidomide (LENA), pomalidomide (POMA) and apremilast (APRE). {C} Mean intensity inducing PC contraction in control and in Eng-iKO® mice treated with vehicle alone or that received thalidomide at 4, 40 or 200 mmol kg, lenalidomide at 40, 200 or 400 mmol.kg"t, pomalidomide at 40 mmel.kg’ or apremilast at 40 or 400
P352801NL -21- mmol.kg”’. Figure 4 shows (A) selected analogues of phthalimide that restore endothelial cell-pericyte interaction and blood vessel stability in accordance with the present invention, as demonstrated by (B) the mean intensity inducing PC contraction in control and in Eng-iKO® mice as shown in figure 3C.
The methods and treatment described herein may be a chronic treatment comprising chronic administration of the pharmaceutical compound or formulation. It may also comprise a temporary or acute treatment. In addition, it may be appreciated that the treatment may be curative and/or prophylactic.
Another aspect of the present invention is directed to the use in organoids and organ-on-a-chip (OoC) models. Organoid and OoC are in vitro miniaturized model systems of organs that have gained enormous interest for modeling disease, for personalized medicine, drug testing and cell therapy. They can be established for an increasing variety of organs including brain, kidney, liver, gut, thyroid, prostate, airways either from tissue resident adult stem cells, from biopsies, from embryonic stem cells or induced pluripotent stem cells. Despite considerable success, limitations of current organoid and OoC systems to achieve real life applications remain the limited level of maturity and function and this is also true for the vascular network. Blood vessels of organoids and OoC appear highly immature and leaky with a marked reduction of PC coverage
As used herein, the singular forms "a", "an" and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise. The term "and/or" includes any and all combinations of one or more of the associated listed items. It will be understood that the terms "comprises" and/or "comprising" specify the presence of stated features but do not preclude the presence or addition of one or more other features.
For the purpose of clarity and concise description features are described herein as part of the same or separate embodiments, however, it will be appreciated that the scope of the invention may include embodiments having combinations of all or some of the features described.
If for a compound included herein no absolute stereochemistry has specifically been indicated, the compound may be a racemic mixture, a mixture of diastereoisomers or an enantiomerically pure compound.
Typically, if no absolute stereochemistry has specifically been indicated, the compound is a racemic mixture. Moreover, if no relative stereochemistry has specifically been indicated, the compound is a mixture of diastereoisomers.
The present invention is illustrated by the following non-limiting examples and experiments.
P352801NL -22-
Examples and experiments
Synthesis of compounds lllustrative compounds were synthesized by a condensation of the appropriate anhydride (e.g. with the appropriate phthalic anhydride) with the appropriate amine and the following general procedures.
General procedure A for the condensation reaction using 3-nitrophthalic anhydride and substituted amines.
Equimolar amounts of 3-nitrophthalic anhydride and amine were dissolved in acetic acid and irradiated at 130 °C for 3 hours in a microwave synthesiser. The reaction mixture was subsequently concentrated in vacuo, taken up in dichloromethane and washed with sat. ag. NaHCO: and brine, respectively. The organics were dried over MgSO: and the solvents were evaporated to dryness in vacuo.
The crude product was finally purified by column chromatography.
General procedure B for the condensation reaction using 3-nitrophthalic anhydride and substituted amines.
Equimolar amounts of 3-nitrophthalic anhydride and amine were suspended in THF to which was then added triethylamine (2 equivalents). The suspension was stirred at room temperature until TLC indicated full consumption. The reaction mixture was subsequently concentrated in vacuo, taken up in a 0.6:1 mixture of glacial acetic acid and toluene and irradiated in a MW synthesizer at 130 °C for 0.5 hours.
The resulted solution was then concentrated in vacuo and subsequently purified by column chromatography.
General procedure C for the nitro reduction using Tin (lI) chloride dihydrate.
The corresponding 3-nitrophthalimide was taken up in ethyl acetate, to which was then added an excess amount of Tin (II) chloride dihydrate. The resulting solution was stirred at either 40 °C or at refluxing temperature until completion (TLC). The reaction mixture was then quenched by the addition of saturated aqueous NaHCO:3, layers separated and the aqueous fraction was extracted from ethyl acetate (3x). The combined organics were dried over MgSO:4 and the solvents were evaporated to dryness in vacuo. The crude product was finally purified by column chromatography.
General procedure D for the condensation reaction using ethyl 4-nitro-1,3-dioxoisoindoline-2- carboxylate and substituted amines.
To a stirring solution of ethyl 4-nitro-1,3-dioxoisoindoline-2-carboxylate (1 equivalent) and triethylamine (2 equivalents) in THF was added the appropriate amine in one portion (1 equivalent). The resulting suspension was stirred at room temperature overnight, after which a clear solution was obtained.
The crude reaction mixture was concentrated onto silica and purified by column chromatography.
General procedure E for the N-acylation of 3-(4-nitro-1,3-dioxoiseindelin-2-ylhazetidin-1-ium chloride using various acyl chlorides.
To a stirring suspension of 3-(4-nitro-1,3-dioxoisoindolin-2-yhazetidin-1-ium chloride in dichloromethane were added triethylamine (2.2 equiv.) and the corresponding acyl chloride (1.2 equiv.).
The reaction was stirred at room temperature until a clear solution was obtained, and TLC indicated there
P352801NL -23- was no remaining starting material. The reaction mixture was then concentrated in vacuo and the crude product was purified by column chromatography. 2-Isopropyl-4-nitroiscindoline-1,3-dione: @
Nel E
Ro ~~ % : NO, ©
The compound was synthesized according to general procedure A using the following conditions: 4- nitroiscbenzofuran-1,3-dione (0.10 g, 0.52 mmol), isopropylamine (0.045 mL, 1 equiv.) were taken up in glacial acetic acid (2.5 mL) and irradiated at 130 °C for 3 hours in a microwave synthesiser. Yield: 58% (0.07 g, 0.30 mmol) as a colourless solid. 'H NMR (500 MHz, CDCI) ô 8.12 — 8.05 (m, 2H), 7.91 (dd, J= 8.1, 7.4 Hz, 1H), 4.57 (hept, J= 7.0 Hz, 1H), 1.50 (d, J = 7.0 Hz, 6H). 4-amino-2-isopropylisoindoline-1,3-dione (14): {
Ni, ©
The compound was synthesized according to general procedure C using the following conditions: 2- isopropyl-4-nitroisoindaline-1,3-dione (0.07 g, 0.3 mmol), tin(ll) chloride dihydrate (0.68 g, 10 equiv.) were stirred in ethyl acetate (3 mL) at 40 °C overnight. Yield: 84% (0.05 g, 0.25 mmol) as a bright yellow solid. 'H (400 MHz, CDCls) 6 7.37 (dd, J = 8.3, 7.1 Hz, 1H), 7.10 (dd, J= 7.1, 0.7 Hz, 1H), 6.83 (dd, J = 8.3, 0.7
Hz, 1H), 5.06 (brs, 2H), 4.46 (hept, J = 6.9 Hz, 1H), 1.45 (d, J = 7.0 Hz, 6H). 13C NMR (101 MHz, CDCls) 5170.5, 168.7, 145.0, 135.0, 132.9, 121.1, 112.7, 111.7, 42.6, 20.3. LC-MS (ESI) m/z calcd for
C1H32N202 [M + H]* 204.1, found 205.1. HPLC (method A): 9.181 min, purity 99%. 2-isobutyl-4-nitroisoindoline-1,3-dione: 0 no, 0 7
The compound was synthesized according to general procedure A using the following conditions: 4- nitroisobenzofuran-1,3-dione (0.15 g, 0.78 mmol), isobutylamine (0.068 g, 1.2 equiv.) were taken up in glacial acetic acid (2 mL) and irradiated at 130 °C for 3 hours in a microwave synthesiser. Yield: 89% (0.17 g, 0.69 mmol) as a colourless solid. 'H NMR (400 MHz, CDCl) 6 8.15-8.13 (m, 1H), 8.15 — 8.09
P352801NL -24 - (m, 1H), 7.94 (dd, J =8.2, 7.4 Hz, 1H), 3.55 (d, J = 7.4 Hz, 2H), 2.24 - 2.05 (m, 1H), 0.96 (d, J = 6.8 Hz, 6H). 4-amino-2-isobutylisoindoline-1,3-dione (15): a
Ng, ©
The compound was synthesized according to general procedure C using the following conditions: 2- isobutyl-4-nitroiscindoline-1,3-dione (0.171 g, 0.69 mmol) and tin(ll) chloride dihydrate (1.6 g, 10 equiv.) were stirred in ethyl acetate (3 mL) at 40 °C for 72 hours. Yield: 87% (0.13 g, 0.60 mmol) as a bright yellow solid. ’H NMR (400 MHz, CDCls) 6 7.39 (dd, J= 8.3, 7.1 Hz, 1H), 7.13 (dd, J = 7.1, 0.7 Hz, 1H), 6.85 (dd, J = 8.3, 0.7 Hz, 1H), 5.02 (s, 2H), 3.43 (d, J = 7.4 Hz, 2H), 2.08 (dh, J= 13.7, 6.9 Hz, 1H), 0.92 (d, J=6.7 Hz, 6H). 3C NMR (101 MHz, CDCl) 6 170.62, 169.01, 145.07, 135.16, 132.86, 121.15, 112.87, 111.57, 45.04, 27.99, 20.25. LC-MS (ESI) m/z calcd for C12H14N202 [M + H]* 218.1, found 219.0.
HPLC (method A): 9.737 min, purity 97%. 4-nitro-2-phenylisoindoline-1,3-dione:
Q
YO Ne 3
Say Nn
NO, {3
The compound was synthesized according to general procedure A using the following conditions: 4- nitroisobenzofuran-1,3-dione (0.15 g, 0.78 mmol), aniline (0.045 mL, 1 equiv.) were taken up in glacial acetic acid (2.5 mL) and irradiated at 130 °C for 3 hours in a microwave synthesiser. Yield: 75% (0.16 g, 0.58 mmol) as a colourless crystalline solid. "TH NMR (400 MHz, CDCl) § 8.22 (dd, J = 7.5, 1.0 Hz, 1H), 8.16 (dd, J=8.2,0.9 Hz, 1H), 7.99 (dd, J= 8.1, 7.5 Hz, 1H), 7.57 — 7.48 (m, 2H), 7.47 — 7.40 (m, 3H). 4-amino-2-phenylisoindoline-1,3-dione (16): 9 nf \
Nh, ©
The compound was synthesized according tc general procedure C using the following conditions: 4-nitro- 2-phenylisoindoline-1,3-dione (0.15 g, 0.56 mmol} and tin(ll} chloride dihydrate (0.51 g, 4 equiv.) were stirred in ethyl acetate (15 mL) at 40 °C for 16 hours. Yield: 79% (0.11 g, 0.44 mmol) as a bright yellow solid. TH NMR (400 MHz, CDCl) ò 7.42-7.26 (m, 6H), 7.14 (dd, J=7.1, 0.7 Hz, 1H), 6.80 (dd, J = 8.3, 0.7
Hz, 1H), 5.13 (br s, 2H). 13C NMR (101 MHz, CDCls) ò 169.26, 167.61, 145.84, 135.66, 132.51, 131.93,
P352801NL -25- 129.19, 127.97, 126.68, 121.39, 113.23, 111.00. LC-MS (ESI) m/z calcd for C14H1oN202 [M + H]* 238.1, found 239.0. HPLC (method A): 9.41 min, purity 98%. tert-butyl (S)-3-(4-nitro-1,3-dioxoisoindolin-2-yl}pyrrolidine-1-carboxylate: 3 fo gu VAN 0 : NO, Ù B)
The compound was synthesized according to general procedure B using the following conditions: tert
Butyl (S)-3-aminopyrrolidine-1-carboxylate (0.50 g, 2.68 mmol), 4-nitroisobenzofuran-1,3-dione (0.57 g, 1.1 equiv.), triethylamine (0.7 mL, 2 equiv.). Yield: 93% (0.90 g, 2.50 mmol) as a light brown oil. "H NMR (400 MHz, CDCls) 6 8.22 — 8.08 (m, 2H), 7.96 (t, J = 7.7 Hz, 1H), 4.89 (p, J = 8.2 Hz, 1H), 3.78 — 3.84 (m, 3H), 3.49 - 3.37 (m, 1H), 2.61 (p, J = 9.1 Hz, 1H), 2.16 (d, J = 22.8 Hz, 1H), 1.47 (d, J = 7.3 Hz, 9H). (S)-3-{4-nitro-1,3-dioxoisoindolin-2-yl)pyrrolidin-1-ium chloride:
NN Fa
NG TN
Ng, © tert-Butyl (S)-3-(4-nitro-1,3-dioxoiscindolin-2-yl)pyrrolidine-1-carboxylate (0.90 g, 2.49 mmol) was taken up in methanolic HCI (4N, 25 mL) and stirred at room temperature for 1 hour. After stirring for 1 hour the reaction mixture was filtered and the solids were washed with EtOAc and subsequently dried in vacuo, affording the title compound as a white solid. Yield: 81% (0.60 g, 2.02 mmol). "H NMR (400 MHz, DMSO-
Ds) 8 9.73 (brs, 1H), 9.31 (brs, 1H), 8.30 (dd, J= 8.1, 0.8 Hz, 1H), 8.18 (dd, J= 7.5, 0.8 Hz, 1H}, 8.08 (dd, J = 8.0, 7.6 Hz, 1H), 4.97 — 4.85 (m, 1H), 3.56 — 3.41 (m, 3H), 3.35 — 3.25 (m, 1H), 2.38 — 2.20 (m, 2H). (S)-2-(1-acetylpyrrolidin-3-yl)-4-nitroisoindoline-1,3-dione: 0
To a stirring suspension of (S)-3-(4-nitro-1,3-dioxoiscindolin-2-yl}pyrrolidin-1-ium chloride (0.15 g, 0.50 mmol) in DCM (10 mL) were added triethylamine (0.05 mL, 2 equiv.) and acetyl chloride (0.14 mL, 1.4 equiv.), and the resulted solution was stirred for 2 hours. The reaction mixture was then concentrated in vacuo, affording the crude title compound as a crystalline white solid. Yield: 0.20 g, 0.50 mmol, quant. 'H
NMR (400 MHz, CDCls) 6 8.18 — 8.09 (m, 2H), 7.96 (q, J = 7.6 Hz, 1H), 4.95 (dp, J = 15.6, 8.1 Hz, 1H),
P352801NL -26- 4.02 -3.72 (m, 3H), 3.68 — 3.42 (m, 1H), 2.81 — 2.52 (m, 1H), 2.43 — 2.28 (m, 1H), 2.09 (d, J = 14.0 Hz, 3H). (S)-2-(1-acetylpyrrolidin-3-yl)-4-aminoisoindoline-1,3-dione (25): < Ni, U &
The compound was synthesized according to general procedure C using the following conditions: (S)-2- (1-acetylpyrrolidin-3-yl)-4-nitroisoindcline-1,3-dione (0.15 g, 0.50 mmol) and tin(ll) chloride dihydrate (0.45 g. 4 equiv.) were stirred in ethyl acetate (60 mL) at a reflux for 8 hours. The crude product was purified by column chromatography, eluting with 3% MeOH in DCM as eluent, affording the title compound as a yellow film. Yield: 63% (0.13 g, 0.47 mmol) as a bright yellow solid. 'H NMR (400 MHz, DMSO-De) § 7.42 (ddd, J= 8.5, 7.0, 2.4 Hz, 1H), 7.00 —- 6.91 (m, 2H), 6.48 (d, J = 5.1 Hz, 2H), 4.73 (dp, J = 27.4, 8.0 Hz, 1H), 3.80 — 3.54 (m, 3H), 3.50 (dt, J = 9.9, 7.7 Hz, 0.5H), 3.30 (dt, J = 11.8, 8.1 Hz, 0.5H), 2.47 —- 2.27 (m, 1H), 2.13 (ddtd, J = 32.1, 12.1, 7.6, 4.4 Hz, 1H), 1.94 (d, J = 14.1 Hz, 3H). "*C NMR (101 MHz, DMSO) 5 169.29, 168.15, 167.96, 167.89, 167.85, 146.55, 146.52, 135.20, 135.16, 132.19, 132.15, 121.43, 121.39, 110.67, 110.65, 108.82, 108.75, 48.56, 48.09, 47.37, 46.79, 45.70, 43.96, 29.05, 27.44, 22.32, 21.88. 'H
NMR (500 MHz, DMSO-D, 100 °C) 6 7.43 (dd, J= 8.4, 7.0 Hz, 1H), 7.01 (d, J= 8.4 Hz, 1H),6.97 (d, J = 7.0 Hz, 1H), 6.24 (s, 2H), 4.85 — 4.62 (m, 1H), 4.02 — 3.59 (m, 3H), 3.53 (s, 0.5H), 3.36 (s, 0.5H), 2.48 — 2.32 (m, 1H), 2.30 — 2.05 (m, 1H), 1.96 {s, 3H). LC-MS (ESI) m/z calcd for C14H1sN3O3 [M + H]t 273.1, found 274.0, HPLC (method A}: 6.92 min, purity 99%. tert-butyl (R}-3-(4-nitro-1,3-dioxoisoindolin-2-yl}pyrrolidine-1-carboxylate: o
ESA oo CS
Tu Ven
The compound was synthesized according tc general procedure B using the following conditions: fert-
Butyl (R)-3-aminopyrrolidine-1-carboxylate (0.50 g, 2.68 mmol}, 4-nitroisobenzofuran-1,3-dione (0.57 g, 1.1 equiv.), triethylamine (0.7 mL, 2 equiv.). Yield: 83% (0.81 g, 2.23 mmol) as a light brown oil. "TH NMR (400 MHz, CDCl3) 6 8.13 (dd, J= 7.7, 1.9 Hz, 2H), 7.96 (t, J = 7.8 Hz, 1H), 4.89 (p, J= 8.4 Hz, 1H), 3.83 —3.63 (m, 3H), 3.49 — 3.38 (m, 1H), 2.62 (p, J=9.1 Hz, 1H), 2.19 (brs, 1H), 1.48 (d, J = 7.7 Hz, 9H).
P352801NL -27- (R)-3-(4-nitro-1,3-dioxoisoindolin-2-yl)pyrrolidin-1-ium chloride: 2 at ARE no, © tert-Butyl (R)-3-(4-nitro-1,3-dioxoisoindolin-2-yhpyrrolidine-1-carboxylate (0.80 g, 2.21 mmol) was taken up in methanolic HCI (4N, 25 mL) and stirred at room temperature for 1 hour. After stirring for 1 hour the reaction mixture was filtered and the solids were washed with EtOAc and subsequently dried in vacuo, affording the title compound as a white solid. Yield: 84% (0.55 g, 1.85 mmol). '"H NMR (400 MHz, DMSO-
De) 0 9.64 (s, 1H), 8.24 (s, 1H), 8.30 (dd, J= 8.1, 0.8 Hz, 1H), 8.18 (dd, J= 7.5, 0.9 Hz, 1H), 8.08 (dd, J = 8.0, 7.6 Hz, 1H), 4.92 (tt, J = 8.5, 6.0 Hz, 1H), 3.60 — 3.40 (m, 3H), 3.34 — 3.25 (m, 1H), 2.40 — 2.19 (m, 2Hj). (R)-2-(1-acetylpyrrolidin-3-yl)-4-nitroisoindoline-1,3-dione: £3
NO, 3 a
To a stirring suspension of (R)-3-(4-nitro-1,3-dioxoisoindolin-2-ylypyrrolidin-1-ium chloride (0.15 g, 0.50 mmol) in DCM (10 mL) were added triethylamine (0.05 mL, 2 equiv.) and acetyl chloride (0.14 mL, 1.4 equiv.), and the resulted solution was stirred for 2 hours. The reaction mixture was then concentrated in vacuo, affording the crude title compound as a crystalline white solid. Yield: 0.20 g, 0.50 mmol, quant. 'H
NMR (400 MHz, CDCl) 6 8.13 (ddd, J=7.8, 6.3, 1.7 Hz, 2H), 8.01 —- 7.91 (m, 1H), 4.95 (dp, J= 15.6, 8.2
Hz, 1H), 4.02 — 3.69 (m, 3H), 3.66 — 3.44 (m, 1H), 2.74 — 2.54 (m, 1H), 2.40 — 2.28 (m, 1H), 2.09 (d, J = 14.1 Hz, 3H). (R}-2-(1-acetylpyrrolidin-3-yl}-4-aminoisoindoline-1,3-dione (27): 0
SEN i Ey ni, © &
The compound was synthesized according tc general procedure C using the following conditions: (R)-2- (1-acetylpyrrolidin-3-yl}-4-nitroiscindoline-1,3-dione (0.15 g, 0.50 mmal} and tin(ll) chloride dihydrate (0.45 g, 4 equiv.) were stirred in ethyl acetate (60 mL) at a reflux for 16 hours. The crude product was purified by column chromatography, eluting with 3% MeOH in DCM as eluent, affording the title compound as a yellow film. Yield: 99% (0.13 g, 0.49 mmol) as a bright yellow solid. 'H NMR (400 MHz, DMSO-Ds) 6 7.42 (ddd, J= 8.3, 7.0, 2.5 Hz, 1H), 7.00 — 6.91 (m, 2H), 6.48 (d, J = 5.1 Hz, 2H), 4.73 (dp, J = 27 4, 8.0 Hz, 1H), 3.82 — 3.54 (m, 3H), 3.50 (dt, J = 9.9, 7.8 Hz, 0.5H), 3.30 (dt, J = 11.8, 8.1 Hz, 0.5H), 2.48 — 2.27 (m,
P352801NL -28- 1H), 2.24 — 1.99 (m, 1H), 1.94 (d, J = 14.1 Hz, 3H). "*C NMR (101 MHz, DMSO-Ds) § 169.29, 168.14, 167.96, 167.85, 146.55, 146.52, 135.20, 135.16, 132.19, 132.14, 121.44, 121.39, 110.67, 110.65, 108.80, 108.75, 48.56, 48.09, 47.36, 46.79, 45.70, 43.96, 29.05, 27.44, 22.32, 21.88. "H NMR (500 MHz, DMSO-
De, 100 °C) 6 7.43 (dd, J=8.4, 7.0 Hz, 1H), 7.01 (d, J = 8.4 Hz, 1H), 6.97 (d, 4 = 7.1 Hz, 1H), 6.25 (br s, 2H), 4.85-4.61 (m, 1H), 3.998 — 3.59 (m, 3H), 3.59 — 3.44 (m, 0.5H), 3.44 — 3.25 (m, 0.5H), 2.47 — 2.31 (m, 1H), 2.29 — 2.09 (m, 1H), 1.96 (br s, 3H). LC-MS (ESI) m/z calcd for C14H1sN3Os [M + H]* 273.1, found 274.0. HPLC (method A): 7.18 min, purity 98%. tert-butyl 4-(4-nitro-1,3-dioxoisoindolin-2-yl)piperidine-1-carboxylate: £3
NO, t
The compound was synthesized according to general procedure B using the following conditions: fert- butyl 4-aminopiperidine-1-carboxylate (0.74 g, 3.71 mmol}, 4-nitroisobenzofuran-1,3-dione (0.72 9, 1.0 equiv), triethylamine (0.67 mL, 1.3 equiv.). Yield: 83% (1.16 g, 3.09 mmol) as a colourless oil. 'H NMR (400 MHz, DMSO-Ds) & 8.26 (dd, J = 8.0, 0.9 Hz, 1H), 8.14 (dd, J=7.5, 0.9 Hz, 1H), 8.08 — 8.00 (m, 1H), 4.20 (tt, J=12.2, 4.0 Hz, 1H), 4.13 — 3.93 (m, 2H), 3.00 — 2.68 (m, 2H), 2.08 (qd, J = 12.6, 4.4 Hz, 2H), 1.76 — 1.67 (m, 2H), 1.42 (s, 9H). 4-{4-nitro-1,3-dioxoisoindolin-2-yljpiperidin-1-ium chloride: 0
Neg ee
No, © fert-Butyl 4-(4-nitro-1,3-dioxoiscindolin-2-yl)piperidine-1-carboxylate (0.50 g, 1.33 mmol) was taken up in methanolic HCI (4N, 20 mL) and stirred at room temperature for 1 hour. After stirring for 1 hour the reaction mixture was filtered and the solids were washed with EtOAc and subsequently dried in vacuo, affording the title compound as a white solid. Yield: 56% (0.23 g, 0.75 mmol). 1H NMR (400 MHz, DMSO-
Ds) 8 9.17 (s, 1H), 8.59 (s, 1H), 8.27 (dd, J = 8.1, 0.8 Hz, 1H), 8.16 (dd, J= 7.5, 0.8 Hz, 1H}, 8.10 — 8.01 (m, 1H), 4.34 (tt, J = 12.1, 3.7 Hz, 1H), 3.36 (d, J = 12.5 Hz, 2H), 3.04 (gq, J= 12.4 Hz, 2H), 2.48 - 2.35 {m, 2H), 1.91 (d, J = 12.8 Hz, 2H).
P352801NL -29- 2-(1-acetylpiperidin-4-yl)-4-nitroisoindoline-1,3-dione: aw ld
NG, ©
To a stirring suspension of 4-(4-nitro-1,3-dioxoisoindolin-2-yhpiperidin-1-ium chloride (0.15 g, 0.48 mmol) in DCM (20 mL) were added triethylamine (0.16 mL, 2.4 equiv.) and acetyl chloride (0.08 mL, 2.3 equiv.), and the solution was stirred for 1 hour. The reaction mixture was then washed with brine (2x), the organics dried over MgSOa and evaporated to dryness in vacuo, affording the title compound as a white crystalline solid. Yield: 99% (0.15 g, 0.48 mmol). "H NMR (400 MHz, CDCls) ò 8.14 — 8.07 (m, 2H), 7.96 — 7.88 (m, 1H), 4.84 (ddd, J = 11.1, 4.4, 2.2 Hz, 1H), 4.38 (it, J = 12.2, 4.1 Hz, 1H), 4.02 — 3.92 (m, 1H), 3.16 (td, J=13.4,2.6 Hz, 1H), 2.60 (td, J = 13.2, 2.6 Hz, 1H), 2.52 — 2.28 (m, 2H), 2.15 (s, 3H), 1.85 - 1.71 (m, 2H). 2-(1-acetylpiperidin-4-yl}-4-aminoisoindoline-1,3-dione (29): 3 9! ON =X
The compound was synthesized according to general procedure C using the following conditions: 2-(1- acetylpiperidin-4-yl}-4-nitroisoindoline-1,3-dione (0.15 g, 0.48 mmol) and tin(ll) chloride dihydrate (0.43 g, 4 equiv.) were stirred in ethyl acetate (30 mL) at a reflux for 6 hours. The crude product was purified by column chromatography, eluting with a gradient of 0-2% MeOH in DCM as eluent, affording the title compound as a yellow solid. Yield: 99% (0.14 g, 0.47 mmol) as a bright yellow solid. *H NMR (400 MHz,
CDCl) § 7.41 (dd, $= 8.3, 7.2 Hz, 1H), 7.12 (dd, J=7.1, 0.5 Hz, 1H), 6.85 (dd, J = 8.3, 0.5 Hz, 1H), 5.26 (s,2H), 4.81 (ddd, J=11.1,4.4, 22 Hz, 1H), 4.27 (it, J=12.2,4.1 Hz, 1H), 3.95 (ddd, J=11.4, 4.4, 2.1
Hz, 1H), 3.14 (td, J = 13.4, 2.6 Hz, 1H), 2.59 (td, J = 13.2, 2.6 Hz, 1H), 2.39 (dad, J = 39.9, 12.6, 4.4 Hz, 2H), 2.14 (s, 3H), 1.75 (dd, J = 11.7, 4.2, 2.3 Hz, 2H). 13C NMR (101 MHz, CDCls) § 170.16, 168.97, 168.42, 145.41, 135.35, 132.54, 121.19, 112.81, 111.11, 48.44, 46.23, 41.40, 29.58, 28.98, 21.63.
LC-MS (ESI) m/z calcd for C1sHizN2Os [M + H]* 287.1, found 288.0. HPLC (method A): 7.68 min, purity 99%.
P352801NL -30- tert-butyl 3-methyl-3-(4-nitro-1,3-dioxoisoindolin-2-yl}azetidine-1-carboxylate:
NO, D >
The compound was synthesized according to general procedure B using the following conditions: fert- butyl 3-amino-3-methylazetidine-1-carboxylate (0.50 g, 2.68 mmol), 4-nitroiscobenzofuran-1,3-dione (0.52 g, 1.0 equiv.), triethylamine (0.75 mL, 2 equiv.). Yield: 79% (1.18 g, 3.09 mmol) as a white solid. "H NMR (400 MHz, DMSO-Ds) 6 8.26 (dd, J = 8.0, 0.9 Hz, 1H), 8.13 (dd, J = 7.4, 0.9 Hz, 1H), 8.04 (dd, J = 8.0, 7.5 Hz, 1H), 4.37 (d, J = 8.7 Hz, 2H), 3.82 (d, J = 8.7 Hz, 2H), 1.59 (s, 3H), 1.38 (s, 9H). 3-methyl-3-{4-nitro-1,3-dioxoisoindolin-2-yljazetidin-1-ium chloride:
Q
A NHC
NO,
To a stirring solution of fert-butyl 3-methyl-3-(4-nitro-1,3-dioxoisoindolin-2-ylyazetidine-1-carboxylate (0.75 g, 2.07 mmol} in EtOAc (10 mL) was carefully added 4N HCI in dioxane (10 mL) and the resulted solution was left to stir for 2 hours. The suspension was then filtered and the solids were washed with pet ether and subsequently dried, affording the title compound as a white solid. Yield: 89% (0.66 g, 2.10 mmol). tH
NMR (400 MHz, DMSO-De) ò 10.15 (s, 1H), 9.47 (s, 1H), 8.31 (dd, J= 8.0, 0.9 Hz, 1H), 8.16 (dd, J = 7.5, 0.9 Hz, 1H), 8.07 (dd, J = 8.1, 7.5 Hz, 1H), 4.53 (d, J = 10.4 Hz, 2H), 3.93 (d, J = 11.7 Hz, 1H), 1.73 (s, 3H). 2-(1-acetyl-3-methylazetidin-3-yl}-4-nitroisoindoline-1,3-dione: £3
NJ a N 0 No, ©
To a stirring suspension of 3-methyl-3-(4-nitro-1,3-dioxoiscindolin-2-ylyazetidin-1-ium chloride (0.30 g, 1.01 mmol) in dichloromethane (10 mL) were added subsequently triethylamine (0.34 mL, 2.4 equiv.) and acetyl chloride (0.17 mL, 2.3 equiv.). The resulted solution was stirred for 1 hour, after which TLC showed full conversion. The reaction mixture was diluted with DCM and washed with 0.5N HCI (ag) and brine.
The organics were dried over MgSO: and solvents removed in vacuo, affording the title compound as an off-white solid. Yield: 94% (0.29 g, 0.95 mmol). 'H NMR (400 MHz, DMSO-Ds) § 8.28 (dd, J = 8.0, 0.9 Hz, 1H), 8.15 (dd, J= 7.5, 0.9 Hz, 1H), 8.05 (dd, J= 8.0, 7.5 Hz, 1H), 4.62 (d, J= 8.9 Hz, 1H), 4.34 (d, /= 9.9
Hz, 1H), 4.16 (d, J = 9.1 Hz, 1H), 3.86 (d, J = 9.9 Hz, 1H), 1.78 (s, 3H), 1.60 (s, 3H).
P352801NL -31- 2-(1-acetyl-3-methylazetidin-3-yl}-4-aminoisoindoline-1,3-dione (33): 7 3
Ni, £
The compound was synthesized according to general procedure C using the following conditions: 2-(1- acetyl-3-methylazetidin-3-yD-4-nitroisoindoline-1,3-dione (0.26 g, 0.84 mmol) and tin{ll) chloride dihydrate (0.76 g, 4 equiv.) were stirred in ethyl acetate (25 mL) at a reflux for 16 hours. The crude product was purified by column chromatography, eluting with 100% EtOAc as eluent, affording the title compound as a bright yellow solid. Yield: 85% (0.22 g, 0.80 mmol). "H NMR (400 MHz, CDCl:) 6 7.41 (dd, J = 8.3, 7.1 Hz, 1H), 7.09 (dd, J= 7.1, 0.7 Hz, 1H), 6.89 (dd, J = 8.4, 0.8 Hz, 1H), 5.41 (brs, 2H), 4.71 (d, J = 9.3 Hz, 1H), 4.53 (d, J= 10.6 Hz, 1H), 4.21 (d, J= 9.3 Hz, 1H), 4.15 (d, J = 10.6 Hz, 1H), 1.91 (s, 3H), 1.70 (s, 3H).
BC NMR (101 MHz, CDCls) ò 170.98, 169.45, 167.82, 145.70, 135.46, 132.61, 121.44, 112.60, 110.84, 60.80, 58.46, 49.50, 24.73, 19.02. LC-MS (ESI) m/z calcd for C14H1sN3O3 [M + HJ 273.1, found 274.0.
HPLC (method A): 7.34 min, purity 99%. ethyl 4-nitro-1,3-dioxoisoindoline-2-carboxylate: 0
Ty 3 STN
No, © N
To a stirring suspension of 3-nitrophthalimide (2.0 g, 10.4 mmol) in 17 mL DCM were added triethylamine (6.5 mL, 4.5 equiv.) and DMAP (0.13 g, 0.1 equiv}. This mixture was then cooled to 0 °C using an ice bath before the dropwise addition of ethylchloroformate (5 mL, 5 equiv.). After the addition was completed the solution was maintained at 0 °C for 30 minutes, after which the reaction mixture was diluted with ethyl acetate (250 mL) and washed subsequently with HCI (aq., 3% v/v) and brine, after which the organics were dried over MgSQs and concentrated in vacuo. The crude was purified by recrystallization from a mixture of EtOAc/Hexane. Yield: 70% (1.91 g, 7.23 mmol) as a light brown solid. "TH NMR (400 MHz,
CDCls) ò 8.24 (dd, J= 7.6, 0.9 Hz, 1H), 8.19 (dd, J=8.0, 1.0 Hz, 1H), 8.03 (t, J= 7.8 Hz, 1H), 4.51 (gq, J = 7.1 Hz, 2H), 1.45 (t, J = 7.1 Hz, 3H). ethyl 1,3-dioxo-1,3-dihydro-2H-pyrrelo[3,4-c]pyridine-2-carboxylate: 2
Ge ©
NS oy a
To a stirring suspension of 1H-pyrrolo[3,4-c]pyridine-1,3(2H)-dicne (2.5 g, 16.9 mmol) and triethylamine (3.1 mL, 1.3 equiv.) in DMF (8 mL) was added at 0 °C in dropwise fastion ethyl chloroformate (2.0 mL, 1.2 equiv.). After the addition was completed the solution was maintained at 8 °C for 30 minutes, after which
P352801NL -32- the reaction mixture maintained at room temperature for 1 hour. The reaction mixture was then poured onto ice, extracted with EtOAc (3x 200 mL) and the combined organics washed with brine. The organics were dried over MgSO. and concentrated in vacuo. The crude was purified by column chromatography eluting with 40/60 EtOAc in pentane, affording the title compound as a yellow crystalline solid. Yield: 30% (1.119, 5.04 mmol). "TH NMR (400 MHz, CDCls) & 9.30 (s, 1H), 9.17 (d, J=4.9 Hz, 1H), 7.88 (d, J= 4.8
Hz, 1H), 4.52 (q, J=7.1 Hz, 2H), 1.48 (t, J = 7.1 Hz, 3H). 2-(1-acetylazetidin-3-yl)-1 H-pyrrolo[3,4-c]pyridine-1,3{2H)-dione (4): > o °
The compound was synthesized according to general procedure D using the following reagents: ethyl 1,3- díoxo-1,3-dihydro-2H-pyrrolo[3,4-clpyridine-2-carboxylate (0.15 g, 0.68 mmol), triethylamine (0.19 mL, 2 equiv.), and 1-acetylazetidin-3-aminium chloride (0.1 g, 1 equiv.). The reaction mixture was stirred at room temperature for 2 hours and was then heated to 50 °C for 2 hours. Yield: 30% (0.05 g, 0.21 mmol) as an off-white solid. "H NMR (400 MHz, CDCl; 6 9.15 (d, J= 1.1 Hz, 1H), 8.08 (d, J=4.9 Hz, 1H), 7.76 (dd, J=4.8, 1.2 Hz, 1H), 5.09 (it, J = 8.7, 6.2 Hz, 1H), 4.70 (dd, J = 8.7, 6.1 Hz, 1H), 4.49 (dd, J= 10.1, 6.3 Hz, 1H), 4.42 (t, J = 8.7 Hz, 1H), 4.32 (t, J = 9.4 Hz, 1H}, 1.92 (s, 3H). 13C NMR (101 MHz, CDCls) & 170.82, 166.56, 166.26, 156.15, 145.15, 138.97, 125.40, 117.09, 54.53, 52.63, 38.79, 19.06. LC-MS (ESI) m/z calcd for C12H11N3O3 [M + H]* 245.1, found 246.1. HPLC (method B): 7.78 min, purity 96%. (S)-4-methyl-2-(4-nitro-1,3-dioxoisocindolin-2-yl)pentancic acid: 2 #
NO, 2 Q
The compound. was synthesized according to general procedure D using the following reagents: ethyl 4- nitro-1,3-dioxoiscindoline-2-carboxylate (0.4 g, 1.5 mmol), triethylamine (0.42 mL, 2 equiv.), and L-leucine (0.20 g, 1 equiv.). After stirring at room temperature overnight the reaction mixture was subsequently heated to 130 °C for 15 minutes. Yield: 52% (0.24 g, 0.78 mmol) as colourless oil. 'H NMR (400 MHz,
CDClz) 6 8.17 — 8.11 {m, 2H), 7.94 (t, J = 7.8 Hz, 1H), 5.00 (ddd, J = 11.5, 4.4, 1.4 Hz, 1H), 1.97 (dddd, J =14.5,10.3, 4.4, 1.2 Hz, 1H), 1.55 — 1.43 (m, 1H), 1.34 — 1.20 (m, 1H), 1.02 — 0.87 (m, 6H}. (S)-2-{4-amino-1,3-dioxoisoindolin-2-yl}-4-methylpentanoic acid (17): 20 NH, QQ
P352801NL -33-
The compound was synthesized according to general procedure C using the following conditions: (S)-4- methyl-2-(4-nitro-1,3-dioxoisoindolin-2-yl)pentancic acid (0.23 g, 0.75 mmol) and tin(ll} chloride dihydrate (0.68 g, 4 equiv.) were stirred in ethyl acetate (10 mL) at 40 °C for 16 hours. Product purified by flash column chromatography using a C18 cartridge, eluting with a gradient of 10-90% CH3CN in H20. Yield: 63% (0.13 g, 0.47 mmol) as a bright yellow solid. "H NMR (400 MHz, CDCls) 6 7.40 (t, J = 7.7 Hz, 1H), 715, J=7.1Hz, 1H), 6.87 (d, J=8.3 Hz, 1H), 4.92 (dd, J= 11.5, 4.3 Hz, 1H), 2.31 (ddd, J = 15.1, 11.6, 4.1 Hz, 1H), 1.90 (ddd, J = 14.4, 10.2, 4.4 Hz, 1H), 1.57 — 1.40 (m, 1H), 0.92 (t, J = 6.8 Hz, 6H). °C
NMR (101 MHz, CDCl) ò 175.88, 169.50, 168.03, 145.10, 135.55, 132.46, 121.68, 113.50, 111.34, 50.15, 37.20, 25.14, 23.26, 21.10. LC-MS (ESI) m/z calcd for C14H1sN204 [M + H]* 276.1, found 277.0.
HPLC (method A): 8.43 min, purity 97%. (S)-4,4-dimethyl-2-{4-nitro-1,3-dioxoisoindolin-2-yl)pentanoic acid:
Rpg, Se
SUH
NO, d
The compound was synthesized according to general procedure D using the following reagents: ethyl 4- nitro-1,3-dioxoisoindoline-2-carboxylate (0.91 g, 3.44 mmol), triethylamine (0.96 mL, 2 equiv}, and (S)-2- amino-4,4-dimethylpentanoic acid (0.50 g, 1 equiv.). After stirring at room temperature under a Nz atmosphere for 4 hours the reaction mixture was subsequently irradiated in a MW synthesizer at 130 °C for 10 minutes. Yield: 92% (1.02 g, 3.18 mmol) as light yellow oil. "TH NMR (400 MHz, CDCl) & 11.24 — 10.69 (m, 1H), 8.19 = 8.11 (m, 2H), 7.95 (t, J = 7.9 Hz, 1H), 5.05 — 4.97 (m, 1H), 2.32 — 2.17 {m, 2H), 0.93 (d,J=1.7 Hz, 9H). (S)-2-{4-amino-1,3-dioxoisoindolin-2-yl)-4,4-dimethylpentanoic acid (30):
The compound was synthesized according to general procedure C using the following conditions: (S)-4,4- dimethyl-2-(4-nitro-1,3-dioxcisoindolin-2-yl)pentanocic acid (0.25 g, 0.78 mmol) and tin(ll) chloride dihydrate (0.70 g, 4 equiv.) were stirred in ethyl acetate (20 mL) at reflux for 6 hours. Crude product was purified by column chromatography eluting with a solution of 1% MeOH and 0.5% AcOH in dichloromethane, affording the title compound as a yellow solid. Yield: 88% (0.20 g, 0.70 mmol). 'H NMR (400 MHz, CDCl:)} 6 7.39 (dd, J= 8.4, 7.1 Hz, 1H), 7.14 (d, J = 7.1 Hz, 1H), 6.84 (d, J = 8.2 Hz, 1H), 4.93 (dd, J=9.6, 2.9 Hz, 1H), 2.40 — 2.11 (m, 2H), 0.92 (s, 9H). 13C NMR (101 MHz, CDClz) § 176.43, 169.57, 168.07, 145.63, 135.51, 132.52, 121.43, 113.18, 111.10, 49.08, 41.30, 30.51, 29.26. LC-MS (ESI) m/z calcd for CisH1sNzO4 [M + H]* 290.1, found 291.0. HPLC (method A): 9.80 min, purity 88%.
P352801NL -34- (S)-3-cyclopropyl-2-(4-nitro-1,3-dioxoisoindolin-2-yl}propanoic acid:
Q
Sg SO
NG, DD
The compound was synthesized according to general procedure D using the following reagents: ethyl 4- nitro-1,3-dioxoiscindoline-2-carboxylate (1.02 g, 3.87 mmol), triethylamine (1.08 mL, 2 equiv.), and (S)-2- amino-3-cyclopropylpropanoic acid (0.50 g, 1 equiv.). After stirring at room temperature under a Nz atmosphere for 2 hours the reaction mixture was subsequently irradiated in a MW synthesizer at 130 °C for 5 minutes. Yield: 25% (0.30 g, 1.0 mmol) as a colourless oil. "TH NMR (400 MHz, CDCls) & 11.11 — 10.84 (m, 1H), 8.18 (dd, J = 3.2, 0.9 Hz, 1H), 8.17 — 8.13 (m, 1H), 8.02 — 7.93 (m, 1H), 5.05 (dd, J = 10.3, 5.4 Hz, 1H), 2.24 2.12 (m, 2H), 0.73 — 0.57 (m, 1H), 0.49 — 0.32 (m, 2H), 0.12 (td, J=9.1, 8.7, 4.9 Hz, 1H), 0.08 — 0.00 (m, 1H). (S)-2-{4-amino-1,3-dioxoisoindolin-2-yl}-3-cyclopropylpropanoic acid (31):
A Nn ee eH
NH, a 0
The compound was synthesized according to general procedure C using the following conditions: (S)-3- cyclopropyl-2-(4-nitro-1,3-dioxcisoindolin-2-yl}preopanoic acid (0.30 g, 1.0 mmol} and tin(ll) chloride dihydrate (0.89 g, 4 equiv.) were stirred in ethyl acetate (40 mL) at reflux for 6 hours. Crude product was purified by column chromatography eluting with a solution of 2% MeOH and 0.5% AcOH in dichloromethane, affording the title compound as a yellow solid. Yield: 89% (0.24 g, 0.88 mmol). 'H (400
MHz, CDCls) 6 7.40 (dd, J=8.3, 7.1 Hz, 1H), 7.15 (dd, J = 7.2, 0.7 Hz, 1H), 6.86 (dd, J = 8.3, 0.7 Hz, 1H), 4.96 (dd, J= 10.8, 4.8 Hz, 1H), 2.17 (ddd, J = 14.4, 10.8, 7.2 Hz, 1H), 2.04 (ddd, J= 14.4, 7.0, 4.8
Hz, 1H), 0.73 — 0.58 (m, 1H), 0.47 — 0.32 (m, 2H), 0.14 — 0.01 (m, 2H). 13C NMR (101 MHz, CDCl:) § 175.43, 169.56, 168.07, 145.63, 135.51, 132.45, 121.44, 113.18, 110.97, 52.03, 33.45, 8.25, 4.76, 3.64.
LC-MS (ESI) m/z calcd for C1aH14N204 [M + H]* 275.1, found 275.0. HPLC (method A): 8.81 min, purity 99%. (S)-2-{4-nitro-1,3-dioxoisoindolin-2-yl}-3-(thiophen-2-yl}propanoic acid: {3 ew ee Nes
Sg OH
NG, DD
The compound was synthesized according tc general procedure D using the following reagents: ethyl 4- nitro-1,3-dioxoisoindoline-2-carboxylate (0.77 g, 2.92 mmol), triethylamine (0.81 mL, 2 equiv.), and (S)-2-
P352801NL -35- amino-3-(thiophen-2-yl)propanoic acid (0.50 g, 1 equiv.). After stirring at room temperature under a Nz atmosphere for 2 hours the reaction mixture was subsequently irradiated in a MW synthesizer at 130 °C for 5 minutes. Yield: 78% {0.79 g, 2.28 mmol) as a light brown solid. *H NMR (400 MHz, CDCls) ò 10.51 (s, 1H), 8.14 (dd, J=8.1,1.0 Hz, 1H), 8.10 (dd, J = 7.5, 0.9 Hz, 1H), 7.92 (dd, J = 8.1, 7.5 Hz, 1H), 7.09 (dd, J=4.7, 1.7 Hz, 1H), 6.87 — 6.80 (m, 2H), 5.22 (dd, J = 11.5, 4.8 Hz, 1H), 3.97 — 3.71 {m, 2H). (S)-2-{(4-amino-1,3-dioxoisoindolin-2-yl}-3-(thiophen-2-yl)propanoic acid (32): 0 Im
L ge A Í
NH, OQ
The compound was synthesized according to general procedure C using the following conditions: (S)-2- (4-nitro-1,3-dioxoiscindolin-2-yl}-3-(thiophen-2-ylypropanoic acid (0.35 g, 1.0 mmol} and tin(ll} chloride dihydrate (0.92 g, 4 equiv.) were stirred in ethyl acetate (40 mL) at reflux for 6 hours. Crude product was purified by column chromatography eluting with a gradient of 2-10% MeOH in dichloromethane, followed by washing of the resulted solids with hot dichloromethane, affording the title compound as a yellow solid.
Yield: 46% (0.15 g, 0.47 mmol). *H NMR (400 MHz, DMSO-Ds) ò 13.36 (brs, 1H), 7.43 (dd, /=8.5,7.0
Hz, 1H), 7.26 (dd, J= 5.0, 1.4 Hz, 1H), 6.96 (dd, J = 14.9, 7.7 Hz, 2H), 6.90 — 6.82 (m, 2H), 6.51 (br s, 2H), 4.92 (dd, J= 9.7, 6.4 Hz, 1H), 3.70 — 3.55 (m, 2H). *C NMR (101 MHz, DMSO-Ds) 6 170.00, 168.65, 167.46, 146.73, 139.26, 135.51, 131.81, 126.94, 126.19, 124.93, 121.79, 110.98, 108.25, 52.57, 28.26.
LC-MS (ESI) m/z calcd for C1sH1zN204S [M + H]* 316.1, found 317.0. HPLC (method A): 8.11 min, purity 99%. tert-butyl 3-(4-nitro-1,3-dioxoisoindolin-2-yljazetidine-1-carboxylate: 0
Sg SN wd
No, UO \
The compound was synthesized according to general procedure D using the following reagents: ethyl 4- nitro-1,3-dioxoisoindoline-2-carboxylate (0.1 g, 0.38 mmol), triethylamine (0.11 mL, 2 equiv.), and tert- butyl 3-aminocazetidine-1-carboxylate hydrochloride (0.07 g. 1 equiv.}. Product purified by column purification using a gradient of 0-1% MeOH in dichloromethane. Yield: 82% (0.11 g, 0.31 mmol) as white solid. *H NMR (400 MHz, CDCl3) ò 8.18 — 8.15 (m, 1H), 8.15 - 8.13 (m, 1H), 7.96 (dd, J = 8.2, 7.4 Hz, 1H), 5.05 (tt, J = 8.5, 6.2 Hz, 1H), 4.51 (dd, J = 9.0, 6.3 Hz, 2H), 4.26 (t, J = 8.9 Hz, 2H), 1.48 (s, 9H).
P352801NL -36- tert-butyl 3-(4-amino-1,3-dioxoisoindolin-2-yl)azetidine-1-carboxylate (21):
D eee vay
Se yg es 0 —_— }
Ng, © >
The compound was synthesized according to general procedure C using the following conditions: tert butyl 3-(4-nitro-1,3-dioxoisoindolin-2-yl)azetidine-1-carboxylate (0.09 9, 0.27 mmol) and tin(ll) chloride dihydrate (0.37 g, 6 equiv.) were stirred in ethyl acetate (15 mL) at 40 °C for 3 hours. Product purified by flash column chromatography using a C18 cartridge, eluting with a gradient of 10-90% CHsCN in H20.
Yield: 40% (0.03 g, 0.11 mmol) as a bright yellow solid. 'H NMR (400 MHz, CDCl3) § 7.41 (dd, J = 8.2, 7.3 Hz, 1H), 7.12 (d, J=7.1 Hz, 1H), 6.86 (d, J = 8.4 Hz, 1H), 4.96 (tt, J = 8.6, 6.2 Hz, 1H), 4.50 (dd, J = 8.9,6.2 Hz, 2H), 4.20 (t, J = 8.7 Hz, 2H), 1.47 (5, 9H). 13C NMR (101 MHz, CDCls) ò 169.76, 168.10, 156.58, 145.65, 135.58, 132.40, 121.45, 112.97, 110.86, 79.92, 54.11, 38.30, 28.51. LC-MS (ESI) m/z calcd for CisHisN3O4 [M + H]* 317.1, found 218.0 [M — Boc + H]t. HPLC (method A): 9.92 min, purity 94%. 4-amino-2-{azetidin-3-yl}isoindoline-1,3-dione hydrochloride salt (34):
P
Ll N— NH -xXHCI
So ie
NH, © tert-butyl 3-(4-amino-1,3-dioxoisoindolin-2-yl)azetidine-1-carboxylate (0.10 g, 0.32 mmol) was stirred in a 4N HCI solution in MeOH (20 mL) for 2 hours. The resulting suspension was concentrated in vacuo, and co-evaporated with EtOAc (2x), yielding the title compound as an HCI salt. Yield: 88% (0.08 g, 0.28 mmol) as a bright yellow solid. 'H NMR (400 MHz, DMSO-Ds) ò 9.63 (brs, 1H}, 9.18 (brs, 1H), 7.45 (dd,
J=8.5,7.0Hz 1H), 7.02 (dd, J = 8.5, 0.7 Hz, 1H), 6.98 (dd, J = 7.0, 0.7 Hz, 1H), 6.36 (br s, 3H), 5.01 (p,
J =8.3 Hz, 1H), 4.59 — 4.46 (m, 2H), 4.25 — 4.08 (m, 2H). 13C NMR (101 MHz, DMSO-Ds) & 168.90, 167.72, 146.78, 135.42, 132.09, 121.68, 110.92, 108.65, 49.84, 39.99. LC-MS (ESI) m/z calcd for
C11H11N302 [M + H]* 217.1, found 218.0 [M + H]*. HPLC (method B): 1.19 min, purity 95%. tert-butyl (S)-3-{4-nitro-1,3-dioxoisoindolin-2-yl)piperidine-1-carboxylate: 3 ’ 0 x
The compound was synthesized according tc general procedure D using the following reagents: ethyl 4- nitro-1,3-dioxoisoindoline-2-carboxylate (0.66 g, 2.50 mmol), triethylamine (0.70 mL, 2 equiv.}, and fert-
P352801NL - 37 - butyl {S)-3-amincpiperidine-1-carboxylate (0.50 g, 1 equiv.). Yield: 89% (0.93 g, 2.48 mmol) as a colourless oil. '"H NMR (400 MHz, CDCls) ò 8.17 — 8.08 (m, 2H), 7.94 (dd, J= 8.2, 7.4 Hz, 1H), 4.88 — 4.68 (m, 1H), 4.22 (tt, J= 11.9, 4.3 Hz, 1H), 3.865 — 3.30 (m, 1H), 2.88 — 2.55 (m, 1H), 2.32 (tt, J = 12.5, 6.3 Hz, 1H), 1.93 - 1.78 (m, 2H), 1.68 — 1.56 (m, 1H), 1.46 (d, J = 1.4 Hz, SH). (S)-3-(4-nitro-1,3-dioxoisoindolin-2-yl)piperidin-1-ium chloride:
Se I NNO
No, ©
To a stirring solution of fert-Butyl (S)-3-(4-nitro-1,3-dicoxoisoindelin-2-yhpiperidine-1-carboxylate (0.90 g, 2.40 mmol) in methanol (10 mL) was carefully added 4N HCI in dioxane (10 mL) and subsequently stirred for 1 hour. The reaction mixture was then concentrated in vacuo, affording the title compound as a white solid. Yield: 75% (0.56 g, 1.81 mmol). *H NMR (400 MHz, DMSO-Ds) ò 9.57 (brs, 1H), 9.42 (brs, 1H), 8.29 (dd, J = 8.0, 0.9 Hz, 1H), 8.17 (dd, J = 7.5, 0.9 Hz, 1H), 8.09 — 8.04 (m, 1H), 4.47 (ddt, J= 16.2, 10.1, 3.7 Hz, 1H), 3.34 (br s, 2H), 3.25 (d, J=12.5 Hz, 1H), 2.83 (t, J = 12.9 Hz, 1H), 2.17 (gd, J= 13.1, 12.3, 4.3 Hz, 1H), 2.03 — 1.68 (m, 3H). (S)-2-{1-acetylpiperidin-3-yl})-4-nitroisoindoline-1,3-dione: 0
Q
To a stirring suspension of (S)-3-(4-nitro-1,3-dioxoiscindolin-2-yl)piperidin-1-ium chloride (0.16 g, 0.50 mmol) in DCM (12.5 mL) were added triethylamine (0.17 mL, 2.4 equiv.) and acetyl chloride (0.08 mL, 2.3 equiv.), and the solution was stirred for 1 hour. The reaction mixture was then concentrated onto silica and purified by column chromatography, eluting with 1% MeQH in dichloromethane as eluent affording the title compound as a colourless oil. Yield: 81% (0.15 g, 0.46 mmol). 'H NMR (400 MHz, CDCls) § 8.12 (dd, J= 8.5, 7.7 Hz, 2H), 8.01 — 7.87 (m, 1H), 4.73 - 4.61 (m, 1H), 4.28 — 4.15 (m, 1H), 3.90 — 3.72 (m, 1H), 3.33 (t, J=12.0 Hz, 0.5H), 3.11 (td, J=13.3, 2.6 Hz, 0.5H), 2.81 — 2.23 (m, 2H), 212d, J=11.2
Hz, 3H), 1.99 - 1.83 (m, 2H), 1.70 — 1.50 (m, 1H).
P352801NL -38- (S)-2-(1-acetylpiperidin-3-yl}-4-aminoisoindoline-1,3-dione (26):
Ng Nl 3
The compound was synthesized according to general procedure C using the following conditions: (S)-2- (1-acetylpiperidin-3-yl}-4-nitroisoindoline-1,3-dione (0.15 g, 0.46 mmol) and tin(ll) chloride dihydrate (0.41 g, 4 equiv.) were stirred in ethyl acetate (30 mL) at a reflux for 16 hours. The crude product was purified by column chromatography, eluting with EtOAc as eluent, affording the title compound as a yellow solid.
Yield: 93% (0.12 g, 0.43 mmol) as a bright yellow solid. 'H NMR (400 MHz, DMSO-Ds) ò 7.42 (ddd, J = 8.4,7.0,2.4 Hz, 1H), 7.00 - 6.91 (m, 2H), 6.47 (brs, 2H), 4.44 — 4.33 (m, 1H), 3.98 — 3.75 (m, 2H), 3.58 (dd, J=12.9,11.4 Hz, 0.5H), 3.08 (t, J= 11.9 Hz, 0.5H), 2.97 (td, J= 13.3, 2.6 Hz, 0.5H), 243 (id, J = 13.1,2.8 Hz, 0.5H), 2.33 - 2.16 (m, 1H), 2.00 (d, J = 22.8 Hz, 3H), 1.89 — 1.68 (m, 2H), 1.60 — 1.44 (m, 0.5H), 1.43 — 1.29 (m, 0.5H). 13C NMR (101 MHz, DMSO-Ds) & 169.29, 168.31, 168.22, 167.87, 146.51, 135.18, 132.16, 132.09, 121.40, 121.37, 110.68, 110.64, 108.69, 47.98, 47.56, 46.68, 45.86, 43.11, 40.91, 27.59, 27.49, 25.27, 24.44, 21.35, 21.32. "H NMR (500 MHz, DMSO-Ds, 100 °C) 6 7.43 (dd, J = 8.4,7.0Hz, 1H), 7.01 (d, J= 8.4 Hz, 1H), 6.97 (d, J = 7.0 Hz, 1H), 6.24 (s, 2H), 4.00 — 3.87 (m, 1H), 2.31 (qd, J=12.6, 4.2 Hz, 1H), 2.01 (s, 3H), 1.89 — 1.78 (m, 2H), 1.49 (br s, 1H). LC-MS (ESI) m/z calcd for
C15H17N3O3 [M + H]* 287.1, found 288.1. HPLC (method A): 7.99 min, purity 99%. tert-butyl (R}-3-(4-nitro-1,3-dioxoisoindolin-2-yl}piperidine-1-carboxylate: 3
Spey Set
Np, © 0
G J
The compound was synthesized according to general procedure D using the following reagents: ethyl 4- nitro-1,3-dioxoisoindoline-2-carboxylate (0.66 g, 2.50 mmol), triethylamine (0.70 mL, 2 equiv.), and fert- butyl (R)-3-aminopiperidine-1-carboxylate (0.50 g, 1 equiv.). Yield: 82% (0.77 g, 2.05 mmol) as a colourless oil. *H NMR (400 MHz, CDCls) 6 8.17 — 8.08 (m, 2H), 7.94 (dd, J= 8.2, 7.3 Hz, 1H), 4.78 (br s, 1H), 4.30 — 4.19 (m, 1H), 4.01 (brs, 1H), 3.49 (d, J= 14.9 Hz, 1H), 2.72 (brs, 1H), 2.33 (qd, /=12.8, 4.0
Hz, 1H), 1.85-1.78 (m, 2H), 1.61 (tt, J= 13.3, 3.9 Hz, 1H), 1.46 (d, J = 1.3 Hz, SH).
P352801NL -39- (R)-3-(4-nitro-1,3-dioxoisoindolin-2-yl)piperidin-1-ium chloride: &
Sot NH
NO, ©
To a stirring solution of fert-Butyl (R)-3-(4-nitro-1,3-dioxoisoindolin-2-yhpiperidine-1-carboxylate (0.77 g, 2.05 mmol) in EtOAc (10 mL) was carefully added 4N HCI in dioxane (10 mL) and subsequently stirred for 1 hour. The reaction mixture was then filtered and the solids washed with EtOAc, affording the title compound as a white solid. Yield: 75% (0.48 g, 1.54 mmol). "H NMR (400 MHz, DMSO-Ds) § 9.57 (br s, 1H), 9.40 (br s, 1H), 8.29 (dd, J = 8.0, 0.9 Hz, 1H), 8.17 (dd, J = 7.5, 0.9 Hz, 1H), 8.11 — 8.02 (m, 1H), 4.47 (tdd, J = 12.3, 8.2, 4.0 Hz, 1H), 3.42 — 3.33 (m, 3H), 3.29 - 3.21 (m, 1H), 2.83 (q, J = 11.9 Hz, 1H), 217(qd, J=13.1, 12.3, 4.3 Hz, 1H), 1.96 — 1.73 (m, 3H). (R)-2-(1-acetylpiperidin-3-yl}-4-nitroisoindoline-1,3-dione:
NN en 4 a
To a stirring suspension of (8}-3-(4-nitro-1,3-dioxoisoindolin-2-yhpiperidin-1-ium chloride (0.16 g, 0.50 mmol) in DCM (12.5 mL) were added triethylamine (0.17 mL, 2.4 equiv.) and acetyl chloride (0.08 mL, 2.3 equiv.), and the solution was stirred for 1 hour. The reaction mixture was then diluted with dichloromethane and subsequently washed with 0.5N HCI (aq.) and brine. The organics were dried over
MgSO. and solvents were removed in vacuo, affording the title compound as a colourless film. Yield: 99% (0.16 g, 0.50 mmol). 'H NMR (400 MHz, CDCl:) 6 8.12 (dd, J= 8.5, 7.7 Hz, 2H), 8.03 — 7.89 (m, 1H), 4.73 —4.81(m, 1H), 4.30 -4.13 (m, 1H), 3.89 — 3.74 (m, 1.5H), 3.33 (t, J= 12.0 Hz, 0.5H), 3.10 (id, J = 13.3, 2.5 Hz, 0.5H), 2.55 (td, J=13.2, 3.0 Hz, 0.5H), 2.50 - 2.31 (m, 1H), 2.12 (d, J= 10.9 Hz, 3H), 2.01 - 1.83 (m, 2H), 1.71 — 1.50 (m, 1H). {R)-2-(1-acetylpiperidin-3-yl}-4-aminoisoindoline-1,3-dione (28): ae € NN
G
The compound was synthesized according to general procedure C using the following conditions: (R)-2- (1-acetylpiperidin-3-yl)-4-nitroisoindoline-1,3-dione (0.16 g, 0.50 mmol) and tin{ll) chloride dihydrate (0.45 g, 4 equiv.) were stirred in ethyl acetate (30 mL) at a reflux for 16 hours. The crude product was purified
P352801NL -40- by column chromatography, eluting with EtOAc as eluent, affording the title compound as a yellow solid.
Yield: 95% (0.14 g, 0.47 mmol) as a bright yellow solid. 'H NMR (400 MHz, DMSO-Ds) 6 7.53 — 7.29 (m, 1H), 7.05 — 6.83 (m, 2H), 6.47 (brs, 2H), 4.44 — 4.33 (m, 1H), 3.99 — 3.77 (m, 2H}, 3.58 (dd, J= 12.9, 11.4 Hz, 0.5H), 3.08 (t, J = 11.8 Hz, 0.5H), 2.97 (td, J = 13.5, 2.6 Hz, 0.5H), 2.43 (id, J = 13.0, 2.8 Hz, 0.5H),2.26 (qd, J=12.7, 4.2 Hz, 1H), 2.00 (d, J = 22.8 Hz, 3H), 1.89 — 1.69 (m, 2H), 1.63 — 1.43 (m, 0.5H), 1.43 — 1.29 (m, 0.5H). *C NMR (101 MHz, DMSO-Ds) ò 169.29, 168.31, 168.22, 167.87, 146.51, 135.18, 132.16, 132.09, 121.40, 121.37, 110.68, 110.64, 108.75, 108.69, 47.98, 47.56, 46.68, 45.86, 43.11,40.91, 27.59, 27.49, 25.27, 24.44, 21.34, 21.31. 'H NMR (500 MHz, DMSO-Ds, 100 °C) ò 7.43 (dd,
J=84,7.0Hz, 1H), 7.01 (d, J= 8.4 Hz, 1H), 6.97 (d, J = 7.0 Hz, 1H), 6.24 (brs, 2H), 4.01 — 3.88 (m, 1H), 2.31 (gd, J = 12.3, 3.9 Hz, 1H), 2.01 (s, 3H), 1.80 — 1.78 (m, 2H), 1.50 (br s, 1H). LC-MS (ESI) m/z calcd for C1sH17N3O3 [M + H]* 287.1, found 288.1. HPLC (method A): 8.07 min, purity 99%. 3-{4-nitro-1,3-dioxoisoindolin-2-yl}azetidin-1-ium chloride:
Q
© TC NHC no, © tert-Butyl 3-(4-nitro-1,3-dioxoisoindolin-2-yhazetidine-1-carboxylate (0.29 g, 0.83 mmol) was stirred in. a freshly prepared solution of HCI in MeOH (2M, 25 mL) at room temperature for 2 hours. After completion (TLC) the solvents were removed in vacuo and the solids were washed with hot MeOH. Yield: 67% (0.16 g, 0.55 mmol) as light yellow crystals. 'H NMR (400 MHz, DMSO-Ds) 6 9.24 (br s, 2H), 8.33 (d, J=8.0
Hz, 1H), 8.22 (d, J= 7.4 Hz, 1H), 8.10 (t, J = 7.8 Hz, 1H), 5.16 — 5.04 (m, 1H), 4.51 (dd, J = 11.6, 7.3 Hz, 2H), 4.22 (dd, J= 11.5, 9.1 Hz, 2H). 2-(1-acetylazetidin-3-yl)-4-nitroisoindoline-1,3-dione: {x
No, ©
The compound was synthesized according to general procedure E using the following conditions: 3-(4- nitro-1,3-dioxoisoindolin-2-yl)azetidin-1-ium chloride (0.054 g, 0.198 mmol), triethylamine (0.05 mL, 2 equiv.), and acetic anhydride (0.02 mL, 1.3 equiv.) were stirred in dichloromethane (5 mL) at room temperature until completion. Yield: 92% (0.05 g, 0.18 mmol) as a colourless solid. *H NMR (400 MHz,
CDCl3) ò 8.20 — 8.13 (m, 2H), 7.99 (dd, J= 8.3, 7.3 Hz, 1H), 5.14 (it, J=8.7,6 2 Hz, 1H), 4.76 (dd, J = 8.6,6.1 Hz, 1H), 4.58 — 4.42 (m, 2H), 4.36 (t, J = 9.2 Hz, 1H), 1.95 (s, 3H).
P352801NL -41- 2-(1-acetylazetidin-3-yl}-4-aminoisoindoline-1,3-dione (5): a
Nn
Sg ~~ v 3
Ni, ©
The compound was synthesized according to general procedure C using the following conditions: 2-(1- acetylazetidin-3-yl)-4-nitroiscindoline-1,3-dione (0.05 g, 0.17 mmol) and tin{ll) chloride dihydrate (0.15 g, 4 equiv.) were stirred in ethyl acetate (20 mL) at 40 °C for 16 hours. Product purified by flash column chromatography using a C18 cartridge, eluting with a gradient of 0-50% CHsCN in H2O. Yield: 77% (0.03 g, 0.13 mmol) as a bright yellow solid. "H NMR (400 MHz, Acetone-Ds) ö 7.46 (dd, J= 8.4, 7.0 Hz, 1H), 7.07-6.99 (m, 2H), 5.04 (tt, J=8.7, 6.1 Hz, 1H), 4.71 (dd, J/= 8.5, 8.0 Hz, 1H), 4.52 — 4.41 (m, 2H), 4.19 (t, J=9.2 Hz, 1H), 1.84 (s, 3H). LC-MS (ESI) m/z calcd for C13H13N3O3 [M + H]* 259.1, found 260.0.
HPLC (method A): 6.54 min, purity 99%. 4-nitro-2-(1-propionylazetidin-3-yl)isoindoline-1,3-dione: £3
No, O
The compound was synthesized according to general procedure E using the following conditions: 3-(4- nitro-1,3-dioxoiscindolin-2-yl)azetidin-1-ium chloride (0.05 g, 0.18 mmol), triethylamine (0.05 mL, 2.2 equiv.), and proprionyl chloride (0.02 mL, 1.2 equiv.) were stirred in dichloromethane (2 mL) at room temperature until completion. Yield: 86% (0.05 g, 0.15 mmol) as a colourless solid. '"H NMR (400 MHz,
CDCl3) ò 8.20 — 8.12 (m, 2H), 7.99 (dd, J= 8.3, 7.3 Hz, 1H), 5.14 (it, J=8.6,6.1 Hz, 1H), 4.78 — 4.69 (m, 1H), 4.56 — 4.41 (m, 2H), 4.35 (t, J = 8.9 Hz, 1H), 2.28 — 2.08 (m, 2H), 1.16 (t, J = 7.5 Hz, 3H). 4-amino-2-(1-propionylazetidin-3-yl}isoindoline-1,3-dione (18): a
Seg i Se
Ni, ©
The compound was synthesized according to general procedure C using the following conditions: 4-nitro- 2-(1-propionylazetidin-3-yl}isoindoline-1,3-dione (0.05 g, 0.15 mmol) and tin(ll} chloride dihydrate (0.14 g, 4 equiv.) were stirred in ethyl acetate (5 mL) at 40 °C for 72 hours. Product purified by column chromatography, eluting with ethyl acetate. Yield: 88% (0.04 g, 0.15 mmol) as a bright yellow solid. 'H
NMR (400 MHz, CDCla) 6 7.42 (dd, J= 8.3, 7.1 Hz, 1H), 7.13 (dd, J=7.1, 0.7 Hz, 1H), 6.89 (dd, J = 8.4, 0.7 Hz, 1H), 5.04 (tt, J = 8.7, 6.2 Hz, 1H), 4.67-4.60 (m, 4H), 4.36 (t, J = 9.0 Hz, 2H), 2.19 (q, J = 7.6 Hz, 2H), 1.17 (t, J= 7.5 Hz, 3H). 13C NMR (101 MHz, CDCl:) & 174.27, 169.67, 188.04, 145.87, 135.65,
P352801NL -42- 132.22, 121.62, 112.91, 110.50, 38.30, 24.97, 9.01. LC-MS (ESI) m/z calcd for C14H1sN3O: [M + H]J* 273.1, found 274.0. HPLC (method A): 7.50 min, purity 89%. 2-(1-butyrylazetidin-3-yl)-4-nitroisoindoline-1,3-dione: 9 : No, 0 :
The compound was synthesized according to general procedure E using the following conditions: 3-(4- nitro-1,3-dioxoiscindolin-2-yl}azetidin-1-ium chloride (0.05 g, 0.18 mmol), triethylamine (0.05 mL, 2.2 equiv.), and butyryl chloride (0.02 mL, 1.2 equiv.) were stirred in dichloromethane (2 mL) at room temperature until completion. Yield: 84% (0.05 g, 0.15 mmol) as a colourless solid. 'H NMR (400 MHz,
CDCl:) 5 8.22 8.12 (m, 2H), 7.99 (dd, J= 8.3, 7.3 Hz, 1H), 5.14 (tt, J=8.7,6.2 Hz, 1H), 4.74 (dd, J = 8.6,6.2 Hz, 1H), 4.55 -4.41 (m, 2H), 4.35 (t, J= 9.4 Hz, 1H), 2.22 — 2.05 (m, 2H), 1.70 (p, J = 7.1 Hz, 2H), 0.98 (t, J = 7.4 Hz, 3H). 4-amin0-2-{1-butyrylazetidin-3-yl}isoindoline-1,3-dione (23): £3 ba Be oN UN “SL
Y 3 x 16 ni, © N
The compound was synthesized according to general procedure C using the following conditions: 2-(1- butyrylazetidin-3-yl)-4-nitroisoindoline-1,3-dione (0.05 g, 0.15 mmol) and tin(ll) chloride dihydrate (0.14 g, 4 equiv.) were stirred in ethyl acetate (5 mL) at 40 °C for 72 hours. Product purified by column chromatography, eluting with ethyl acetate. Yield: 97% (0.04 g, 0.15 mmol} as a bright yellow solid. 'H
NMR (400 MHz, CDCl) 6 7.41 (dd, y= 8.3, 7.1 Hz, 1H), 7.12 (dd, J= 7.1, 0.7 Hz, 1H), 6.89 (dd, J = 8.4, 0.7 Hz, 1H), 5.03 (tt, J= 8.7, 6.1 Hz, 1H), 4.89 (br ss, 2H), 4.62 (br s, 2H), 4.36 (t, J = 9.0 Hz, 2H), 2.14 (dd, J=84, 6.6 Hz, 2H), 1.69 (h, J = 7.4 Hz, 2H}, 0.97 (t, J = 7.4 Hz, 3H). 13C NMR (101 MHz, methanol-
Da) & 175.67, 170.72, 169.83, 148.14, 136.37, 133.55, 122.55, 112.48, 110.73, 56.16, 53.93, 39.31, 34.27, 19.40, 14.08. LC-MS (ESI) m/z calcd for CisH17N3Os [M + H]* 287.1, found 288.0. HPLC (method
A): 8.22 min, purity 98%.
P352801NL -43- 2-(1-(3,3-dimethylbutanoyljazetidin-3-yl}-4-nitroisoindoline-1,3-dione:
D
Ol,
No, © >
The compound was synthesized according to general procedure E using the following conditions: 3-(4- nitro-1,3-dioxoiscindolin-2-yl)azetidin-1-ium chloride (0.10 g, 0.35 mmol), triethylamine (0.11 mL, 2.2 equiv.), and 3,3-dimethylbutyryl chloride (0.08 mL, 1.1 equiv.) were stirred in acetonitrile (2 mL) at room temperature until completion. Yield: 93% (0.11 g, 0.33 mmol) as a colourless solid. "H NMR (400 MHz,
CDCl:) © 8.20 — 8.12 (m, 2H), 7.99 (dd, J=8.2, 7.3 Hz, 1H), 5.11 (tt, J= 8.7, 86.2 Hz, 1H), 4.79 - 4.70 (m, 1H), 4.58 — 4.41 (m, 2H), 4.41 — 4.31 (m, 1H}, 2.05 (d, J = 5.0 Hz, 2H), 1.08 (s, SH). 4-amino-2-(1-(3,3-dimethylbutanoyljazetidin-3-yl)isoindoline-1,3-dione (24):
U ee AB
Nit, ò >
The compound was synthesized according to general procedure C using the following conditions: 2-(1- (3.3-dimethylbutanoyl)azetidin-3-yl)-4-nitroisoindoline-1,3-dione (0.05 g, 0.15 mmol) and tin(ll) chloride dihydrate (0.13 g, 4 equiv.) were stirred in ethyl acetate (10 mL) at 40 °C for 72 hours. Product purified by column chromatography, eluting with a gradient of 0-5% MeOH in dichloromethane. Yield: 98% (0.05 g, 0.14 mmol) as a bright yellow solid. 'H NMR (400 MHz, CDCls) 6 7.44 — 7.35 (m, 1H), 7.10 (dd, J = 7.1, 0.7 Hz, 1H), 6.88 (dd, J = 8.4, 0.7 Hz, 1H), 5.00 (tt, J = 8.8, 6.2 Hz, 1H), 4.87 — 4.54 (m, 4H), 4.46 — 4.24 (m, 2H), 2.04 (s, 2H), 1.06 (s, 9H). 3C NMR (101 MHz, CDCl3) 6 172.36, 169.60, 188.07, 145.84, 135.62, 132.24, 121.60, 112.89, 110.53, 44.49, 37.83, 31.62, 29.97. LC-MS (ESI) m/z calcd for C17H21N3O3 [M +
H]* 315.2, found 316.1. HPLC (method A): 9.25 min, purity 96%. 2-(1-(cyclopropanecarbonyljazetidin-3-yl}-4-nitroiscindoline-1,3-dione: 3 erk EN Ô
NO, © u
The compound was synthesized according tc general procedure E using the following conditions: 3-(4- nitro-1,3-dioxoisoindolin-2-yl)azetidin-1-ium chloride (0.07 g, 0.25 mmol), triethylamine (0.08 mL, 2.2 equiv.), and cyclopropanecarbonyl chloride (0.03 g, 1.2 equiv.) were stirred in acetonitrile (3 mL) at room temperature until completion. Yield: 78% (0.06 g, 0.19 mmol) as a colourless solid. '"H NMR (400 MHz,
CDCl3) 6 8.20 — 8.13 (m, 2H), 7.98 (dd, J=8.2, 7.4 Hz, 1H), 5.17 (it, J=8.7,6.2 Hz, 1H), 4.88 (dd, J =
P352801NL -44- 8.5,6.2 Hz, 1H), 4.60 (t, J = 8.6 Hz, 1H), 4.53 (dd, J= 10.0, 6.3 Hz, 1H), 4.37 (t, J = 9.4 Hz, 1H), 1.44 (it,
J= 7.98, 4.8 Hz, 1H), 1.06 — 0.96 (m, 2H), 0.86 — 0.74 (m, 2H). 4-amino-2-(1-(cyclopropanecarbonyl)azetidin-3-yl}isoindoline-1,3-dione (19): © < Ni, © =
The compound was synthesized according to general procedure C using the following conditions: 2-(1- (cyclopropanecarbonyl)}azetidin-3-yl)-4-nitroisoindoline-1,3-dicne (0.06 g, 0.198 mmol) and tin(ll) chloride dihydrate (0.18 g, 4 equiv.) were stirred in ethyl acetate (10 mL) at 40 °C for 48 hours. Product purified by column chromatography, eluting with 1.5% MeOH in dichloromethane. Yield: 73% (0.04 g, 0.14 mmol) as a bright yellow solid. "H NMR (400 MHz, CDCl3) § 7.42 (dd, J=8.3, 7.1 Hz, 1H), 7.13 (dd, J= 7.1, 0.7 Hz, 1H}, 6.88 (dd, J = 8.3, 0.7 Hz, 1H), 5.08 (tt, J= 8.7, 6.2 Hz, 1H), 4.76-4.70 (m, 4H), 4.44 (t, J = 9.0 Hz, 2H), 1.44 (tt, J=7.9, 4.6 Hz, 1H), 1.01 (dt, J = 4.6, 3.3 Hz, 2H), 0.79 (dt, J = 8.0, 3.4 Hz, 2H). 13C NMR (101 MHz, CDCl3) 6 174.11, 169.75, 168.01, 145.80, 135.69, 132.37, 121.58, 113.02, 110.63, 53.92, 38.52, 10.45, 7.64. LC-MS (ESI) m/z calcd for C1sH1sN3Oa [M + HJ" 285.1, found 286.0. HPLC (method
A): 7.70 min, purity 98%. 2-(1-acryloylazetidin-3-yl}-4-nitroisoindoline-1,3-dione: 9
No, ©
The compound was synthesized according to general procedure E using the following conditions: 3-(4- nitro-1,3-dioxoisoindolin-2-yl}azetidin-1-ium chloride (0.07 g, 0.25 mmol), triethylamine (0.08 mL, 2.2 equiv.), and acryloyl chloride (0.02 mL, 1.2 equiv.) were stirred in acetonitrile (3 mL) at room temperature until completion. Yield: 46% (0.03 g, 0.11 mmol) as a colourless solid. 'H NMR (400 MHz, CDCls) § 8.20 -8.12 (m, 2H), 7.99 (dd, J = 8.3, 7.3 Hz, 1H), 6.37 (dd, J= 17.0, 1.9 Hz, 1H), 6.22 (dd, J= 17.0, 10.3 Hz, 1H), 5.72 (dd, J = 10.3, 1.9 Hz, 1H), 5.18 (tt, J= 8.7, 6.2 Hz, 1H), 4.87 — 4.79 (m, 1H), 4.65 — 4.52 (m, 2H), 4.44 (t, J=9.1 Hz, 1H).
P352801NL -45- 2-(1-acryloylazetidin-3-yl}-4-aminoisoindoline-1,3-dione (20): 0 ni, 0
The compound was synthesized according to general procedure C using the following conditions: 2-(1- acryloylazetidin-3-yl)-4-nitroiscindoline-1,3-dione (0.03 g, 0.11 mmol) and tin(ll} chloride dihydrate (0.10 g, 4 equiv.) were stirred in ethyl acetate (3 mL) at 40 °C for 48 hours. Product purified by column chromatography, eluting with 1.5% MeOH in dichloromethane. Yield: 82% (0.03 g, 0.09 mmol) as a bright yellow solid. "TH NMR (400 MHz, CDCl:) 6 7.41 (dd, J=8.4, 7.1 Hz, 1H), 7.12 (d, J=7.0 Hz, 1H), 6.92 — 6.85 (m, 1H), 6.37 (dd, J = 17.0, 1.9 Hz, 1H), 6.22 (dd, J = 17.0, 10.3 Hz, 1H), 5.70 (dd, J = 10.3, 1.9 Hz, 1H), 5.08 (it, J = 8.7, 6.2 Hz, 1H), 4.71 (brs, 2H), 4.53-4.4.45 (m, 4H). '3C NMR (101 MHz, CDCl) & 169.57, 168.16, 165.75, 145.82, 135.72, 132.24, 127.95, 125.95, 121.63, 113.04, 38.53. LC-MS (ESI) m/z calcd for C14H13NaO3 [M + H]* 271.1, found 272.0. HPLC (method A}: 7.31 min, purity 99%. 2-(1-benzoylazetidin-3-yl)-4-nitroiscindoline-1,3-dione:
Q
Eek ©
Y 1 STR
NO © SD
The compound was synthesized according to general procedure E using the following conditions: 3-(4- nitro-1,3-dioxoisoindolin-2-yhazetidin-1-ium chloride (0.05 g, 0.18 mmol), triethylamine (0.05 mL, 2.2 equiv.), and benzoyl chloride (0.03 mL, 1.2 equiv.) were stirred in dichloromethane (2 mL) at room temperature until completion. Yield: 99% (0.06 g, 0.17 mmol) as a colourless solid. TH NMR (400 MHz,
CDCl3) ò 8.17 — 8.10 (m, 2H), 7.96 (dd, J = 8.3, 7.3 Hz, 1H), 7.72 — 7.63 (m, 2H), 7.52 — 7.36 (m, 3H), 5.18 (it, J=8.7,6.1 Hz, 1H), 4.91 — 4.83 (m, 1H), 4.74 — 4.65 (m, 1H), 4.62 — 4.53 (m, 2H). 4-amino-2-(1-benzoylazetidin-3-yl}isoindoline-1,3-dione (22): bo 5 mel , 3 3 ST ni, © Le
The compound was synthesized according to general procedure C using the following conditions: 2-(1- benzoylazetidin-3-yl)-4-nitroiscindoline-1,3-dione (6.06 g, 0.18 mmol) and tin(ll) chloride dihydrate (0.16 g, 4 equiv.) were stirred in ethyl acetate (5 mL) at 40 °C for 48 hours. Product purified by column chromatography, eluting with a gradient of 50-75% ethyl acetate in pentane. Yield: 84% (0.05 g, 0.15 mmol) as a bright yellow solid. "TH NMR (400 MHz, DMSO-Ds) ò 7.69 — 7.62 (m, 2H), 7.58 — 7.39 (m, 4H), 6.97 (dd, J=7.7,6.2 Hz, 2H), 6.51 (brs, 2H), 5.00 (tt, J= 8.6, 5.8 Hz, 1H), 4.72 — 4.57 (m, 2H), 4.50 (dd,
P352801NL -46-
J= 10.2, 5.9 Hz, 1H}, 4.34 (t, J = 9.5 Hz, 1H). LC-MS (ESD m/z calcd for C1sH15N3O3 [M + HJ* 321.1, found 322.0. HPLC (method A): 8.91 min, purity 99%. tert-butyl (1-acetylazetidin-3-yljcarbamate:
HNN <£
XD
SA
3-N-Boc-amino-azetidine hydrochloride (2.0 g, 9.58 mmol) was suspended in dichloromethane (40 mL) and the suspension was cooled to 0 °C using an icebath. To this was added triethylamine (4.0 mL, 3 equiv.) followed by a dropwise addition of acetyl chloride (1.02 mL, 1.5 equiv. in 10 mL dichloromethane).
Upon completion of the addition (TLC) the reaction mixture was allowed to warm to room temperature at which it was then maintained for 1.5 hours. HzO was added, layers separated and the organics were washed with brine and dried over MgSOa. The crude product was purified by column chromatography eluting first with 50/50 EtOAc in pentane and then 100% EtOAc. Yield: 83% (1.71 g, 8.0 mmol) as a colourless oil. "TH NMR (400 MHz, CDClas) 6 6.07 (brs, 1H), 4.43 — 4.18 (m, 2H), 4.16 — 4.03 (m, 1H), 3.97 — 3.82 (m, 1H), 3.78 — 3.85 (m, 1H), 1.71 (s, 3H), 1.30 (s, 9H). 1-acetylazetidin-3-aminium chloride:
CIN nl tert-Butyl (1-acetylazetidin-3-yl)carbamate (1.71 g, 8.0 mmol} was taken up in 40 mL DCM to which was carefully added HCI (4.0 M in dioxane, 27 mL). This solution was stirred at room temperature for 1 hour after which diethylether (40 mL) was added, resulting in the precipitation of the product. The flask was left overnight to complete the precipitation after which the solids were filtered and washed with ether. Yield: 99% (1.21 g, 8.0 mmol), as a off-white solid. "H NMR (400 MHz, DMSO-Ds) & 8.81 (s, 3H), 4.37 - 4.28 (m, 1H), 4.12 (dd, J = 9.6, 4.5 Hz, 1H}, 4.06 — 3.88 (m, 2H}, 3.84 (dd, J= 9.8, 4.4 Hz, 1H), 1.75 (s, 3H). 2-(1-acetylazetidin-3-yl}-5-nitroisoindoline-1,3-dione: 3
NX on &
JE ON
{3
Step one: To a stirring suspension of 5-nitroisobenzofuran-1,3-dione (0.13 g, 0.66 mmol), and 1- acetylazetidin-3-aminium chloride (0.15 g, 1.5 equiv.) in THF (5 mL) was added triethylamine (0.11 mL, 1.2 equiv.). The addition of triethylamine induced instant precipitation. The suspension was stirred at room temperature for 4 hours. The reaction mixture was filtered, solids washed with THF and subsequently dried in vacuo. Step two: The solids were suspended in acetic anhydride (5 mL) to which was then added sodium acetate trihydrate (0.08 g, 1 equiv.). The reaction mixture was heated to 80 °C
P352801NL - 47 - and maintained for 40 minutes, after which TLC indicated full conversion. The reaction mixture was poured onto ice, extracted from EtOAc (3x 50 mL). The combined organics were dried over MgSO. and solvents were subsequently removed in vacuo. Yield: 41% (0.07 g, 0.25 mmol} over two steps, as a light brown solid. 'H NMR (400 MHz, CDCls) § 8.69 — 8.59 (m, 2H), 8.07 (dd, J= 8.1, 0.7 Hz, 1H), 5.11 (it, J = 87,81Hz, 1H), 4.72 (t, J=7.6 Hz, 1H), 4.80 — 4.39 (m, 2H), 4.33 (t, J= 9.8 Hz, 1H), 1.92 (s, 3H). 2-(1-acetylazetidin-3-yl)-56-aminoiscindoline-1,3-dione (6):
G
U
The compound was synthesized according to general procedure C using the following conditions: 2-(1- acetylazetidin-3-yl}-5-nitroisoindoline-1,3-dione (0.07 g, 0.25 mmol) and tin(ll} chloride dihydrate (0.22 g, 4 equiv.) were stirred in ethyl acetate (12 mL) at 40 °C for 24 hours. Product purified by column chromatography, eluting with 2% MeOH in chloroform. Yield: 62% (0.04 g, 0.16 mmol} as a bright yellow solid. "H NMR (400 MHz, DMSQ-Ds) 6 7.50 (d, J = 8.2 Hz, 1H), 6.92 (d, J = 2.0 Hz, 1H), 6.80 (dd, J= 8.2, 2.1 Hz, 1H), 6.52 (br s, 2H), 4.90 (tt, J = 8.7, 6.0 Hz, 1H), 4.52 (dd, J = 8.6, 6.0 Hz, 1H), 4.37 (td, J= 8.7, 0.9Hz, 1H), 4.26 (dd, J =9.7, 6.1 Hz, 1H), 4.09 (t, J = 9.2 Hz, 1H), 1.79 (s, 3H). 13C NMR (101 MHz,
DMSO-Ds) ò 169.47, 168.03, 167.55, 155.13, 134.34, 125.04, 116.81, 116.37, 106.92, 54.71, 52.39, 37.66, 18.87. LC-MS (ESI) mz calcd for C13H13N3O3 [M + H]* 259.1, found 260.1, HPLC (method B): 9.13 min, purity 88%. 5-(1-acetylazetidin-3-yl)-4H-thieno[2,3-c]pyrrole-4,6(5H)-dione (9): 0
Ng | EL < iN A, {3
Step one: To a stirring suspension of thieno[2,3-c]furan-4,6-dione (0.09 g, 0.9 equiv.), and 1- acetylazetidin-3-aminium chloride (0.1 g, 0.66 mmol) in THF (5 mL) was added triethylamine (0.18 mL, 2 equiv.). The addition of triethylamine induced instant precipitation. The suspension was stirred overnight at room temperature after which mixture was concentrated in vacuo. Step two: The crude was suspended in acetic anhydride (5 mL) to which was then added sodium acetate trihydrate (0.10 g, 1 equiv.). The reaction mixture was heated to 80 °C and maintained for 1 hour, after which LC-MS indicated full conversion. The reaction mixture was concentrated in vacuo and residual acetic anhydride was removed by co-evaporation with toluene (3x). The crude product was purified by flash column chromatography, eluting with a gradient of 0-5% MeOH in chloroform. Yield: 42% (0.07 g, 0.28 mmol) over two steps, as a white solid. "TH NMR (400 MHz, CDCls) 6 7.86 (d, J=4.7 Hz, 1H), 7.35 (d, J= 4.7 Hz, 1H), 5.03 (tt, J = 8.7,6.3 Hz, 1H), 4.73 (dd, J = 8.6, 6.1 Hz, 1H), 4.51 (dd, J= 10.0, 6.4 Hz, 1H), 4.43 (t, J = 8.6, 1H), 4.33 (t, J=9.4 Hz, 1H), 1.96 (s, 3H). 13C NMR (101 MHz, CDCls) ô 170.60, 163.16, 162.03, 144.38, 140.61,
P352801NL -48 - 138.50, 121.39, 55.02, 53.04, 38.74, 19.04. LC-MS (ESI) nvz calcd for C11H1oN203S [M + H]* 250.0, found 251.0. HPLC (method B}: 9.31 min, purity 99%.
N-(2-(1-acetylazetidin-3-yl}-1,3-dioxoisoindolin-4-yljacetamide (7}:
JEN Cy, BS ol o : ò
To a stirring solution of 2-(1-acetylazetidin-3-y[}-4-nitroisoindoline-1,3-dione (0.1 g, 0.33 mmol) in EtOAc (10 mL) was then added tin(ll} chloride dihydrate (0.3 g, 4 equiv.). The solution was then maintained at reflux for 1.5 hours, after which LC/MS showed full conversion to the aniline. The reaction mixture was cooled to room temperature, and acetic anhydride {0.3 mL, 10 equiv.) was carefully added. The mixture was then heated and maintained at reflux for another 1.5 hours, after which the reaction mixture was cooled to room temperature, diluted with EtOAc and quenched by the addition of a saturated aqueuous solution of NaHCO:3. Layers were separated and the aqueous was extracted once from EtOAc. The combined organics were washed with brine and dried over MgSOa. The crude product was purified by column chromatography, eluting with a gradient of 0-3% MeOH in dichloromethane affording the title compound as a light yellow solid. Yield: 84% (0.07 g, 0.22 mmol). !H NMR (400 MHz, CDCl) ò 9.43 (s, 1H), 8.75 (d, J=8.3 Hz, 1H), 7.66 (dd, J=8.4, 7.5 Hz, 1H), 7.48 (dd, J = 7.3, 0.7 Hz, 1H), 5.03 (it, J = 8.7,6.2Hz, 1H), 4.72 — 4.64 (m, 1H), 4.48 (dd, J=9.7, 6.4 Hz, 1H), 4.41 (t, J=8.6 Hz, 1H), 4.31 (t, J= 9.4 Hz, 1H), 2.25 (s, 3H), 1.92 (s, 3H). 13C NMR (101 MHz, CDCl3) § 170.61, 169.69, 189.33, 167.02, 137.75, 136.43, 130.97, 125.17, 118.30, 115.08, 54.77, 52.78, 38.36, 25.03, 19.06. LC-MS (ESI) m/z calcd for CisH1sN3Oa4 [M + H]* 301.1, found 302.1. HPLC (method B): 9.97 min, purity 95.1%. 5-chloro-2-methyl-3-nitrobenzoic acid: 9 on
Pg
NU, 5-chloro-2-methylbenzoic acid (5.0 g, 29.3 mmol) was dissolved portionwise in sulfuric acid (34 mL) at -10 °C. After dissolving the material a 2:1 mixture (8.5 mL) of sulfuric acid and nitric acid (65%) was added dropwise, while maintaining a temperature between -10 °C and 0 °C. After addition the reaction mixture was stirred for a further couple of minutes before it was poured onto ice and subsequently extracted from
EtOAc. The organics were dried over MgSO:4, solvents were evaporated in vacuo and the crude product was recrystallized from hot methanol affording the title compound as a light yellow solid. Yield: 37% (2.30 9, 10.7 mmol). !H NMR (400 MHz, CDCI3} 6 11.44 (s, 1H), 8.19 (d, J=2.3 Hz, 1H), 7.90 (d, J = 2.4 Hz, 1H), 2.67 (s, 3H).
P352801NL -49- 5-chloro-3-nitrophthalic acid: 0 rr en
Sy OH
NO, © 5-chloro-2-methyl-3-nitrobenzoic acid (2.16 g, 10 mmol) was suspended in H20 (40 mL) to which was then added sodium hydroxide (0.8 g, 2 equiv.}. The solution was then stirred at 85 °C and to it was added portionwise over 3 hours potassium permanganate (3.16 g, 2 equiv.). After completion of the addition the reaction mixture was maintained at 85 °C one hour. The reaction mixture was cooled tc room temperature, filtered and the solids washed with hot H20 (2x 30 mL). The filtrate was acidified to pH = 1-2 using 2N HCI (aq.), EtOAc added, and the crude product was extracted (3x 100 mL). The combined organics were dried over MgSQ4 and concentrated in vacuo, after which the product was purified by column chromatography eluting first with 99:1:1 DCM/MeOH/AcOH and then 48:48:4 DCM/MeOH/AcOH, affording the title phthalic acid as a yellow solid. Yield: 46% (1.13 g, 4.60 mmol). 2-(1-acetylazetidin-3-yl}-6-chloro-4-nitroisoindoline-1,3-dione: 9 id eed Ea 3 fm Ng, ©
Step one: 5-chloro-3-nitrophthalic acid (0.15 g, 0.61 mmol) and sodium acetate trihydrate (0.08 g, 1 equiv.) were taken up in acetic anhydride (10 mL) and stirred at 80 °C for 1 hour. The solvents were subsequently removed in vacuo and any residual acetic anhydride was removed by co-evaporation with toluene (3x). The crude phthalic anhydride was suspended in THF (4 mL) to which were then added 1- acetylazetidin-3-aminium chloride (0.1 g, 1.1 equiv.), and triethylamine (0.17 mL, 2 equiv.). The resulting suspension was stirred at room temperature for 4 hours. The reaction mixture was concentrated in vacuo, and carried further without any purification. Step two: The crude 2-((1-acetylazetidin-3-yl)carbamoyl)-4- chloro-6-nitrobenzoic acid was taken up in acetic anhydride (5 mL) and to it was added sodium acetate trihydrate (0.08 g, 1 equiv.). This solution was heated to 80 °C at which it was maintained for 1 hour.
LC/MS showed full conversion of the benzoic acid to the phthalimide, so the reaction mixture was concentrated in vacuo, co-evaporated with toluene (2x) and finally purified by flash column chromatography, eluting using a gradient of 0-10% MeOH in DCM affording the title compound as a light yellow oil. Yield: 27% over 2 steps (0.05 g, 0.15 mmol}. *H NMR (400 MHz, CDCls) 6 8.14 (d, J = 1.7 Hz, 1H), 8.12 (d, J=1.7 Hz, 1H), 5.13 (it, J = 8.7, 6.1 Hz, 1H), 4.74 (dd, J = 8.8, 6.2 Hz, 1H}, 4.55 - 4.41 (m, 2H), 4.36 (dd, J= 10.2, 8.7 Hz, 1H), 1.95 (s, 3H).
P352801NL -50- 2-(1-acetylazetidin-3-yl}-4-amino-6-chloreisoindoline-1,3-dione (10): 2
Ch on PE
Nh, 0
The compound was synthesized according to general procedure C using the following conditions: 2-(1- acetylazetidin-3-yl)-6-chloro-4-nitroisoindoeline-1,3-dione (54 mg, 0.17 mmol) was dissolved in ethyl acetate (10 mL}, to which was then added tin(ll} chloride dihydrate (0.35 g, 9 equiv.). The resulting solution was then stirred at a reflux for 8 hours. The title compound was purified by flash chomatography eluting with a gradient of 0-5% MeOH in chloroform, yielded a bright yellow solid. Yield: 92% (0.05 g, 0.15 mmol). 'H NMR (400 MHz, CDCI:) & 7.07 (s, 1H), 6.90 (s, 1H), 5.50 (br s, 2H), 5.07 — 4.95 (m, 1H), 4.69 t, J=7.4 Hz 1H), 4.57 —4.45 (m, 1H), 4.40 (t, J= 8.6 Hz, 1H), 4.30 (t, J = 9.4 Hz, 1H), 1.93 (s, 3H). °C
NMR (101 MHz, CDCls} ò 170.80, 168.94, 166.94, 146.39, 141.98, 133.71, 120.54, 113.50, 109.20, 54.94, 52.91, 38.24, 19.14. LC-MS (ESI) m/z calcd for C1aH12CIN3Oa [M + H]* 293.1, found 294.0. HPLC (method A): 7.88 min, purity 95%. 3-chloro-6-nitrophthalic acid: eo 0
Nog OH
Nix OQ
To a stirring solution of nitric acid in conc. sulfuric acid (1:2, 9 mL) at 0 °C was added portionwise 3- chlorophthalic anydride (2.5 g, 13.7 mmol). After addition the suspension was allowed to warm to room temperature at which it was then maintained for 3 hours. The reaction mixture was then cooled back down to 0 °C after which ice was added to the mixture. The slurry was filtered and the filtrate was extracted from EtOAc (3x). The combined organics were dried over MgSO4 and the solvents were removed in vacuo, and the crude product was further purified by column chromatography, eluting with 5%
MeOH in dichloromethane affording the title phthalic acid as an off-white solid. Yield: 56% (1.87 g, 7.61 mmol). 'H NMR (400 MHz, DMSO-Ds) 6 14.39 (brs, 2H), 8.16 (d, J = 8.7 Hz, 1H), 7.93 (d, J = 8.7 Hz, 1H). 2-(1-acetylazetidin-3-yl}-4-chloro-7-nitroisoindoline-1,3-dione:
Hog
NO, ©
Step one: 3-chloro-6-nitrophthalic acid (0.2 g, 0.81 mmol) and sodium acetate trihydrate (0.11 g, 1 equiv.) were taken up in acetic anhydride (5 mL} and stirred at 80 °C for 1 hour. The resulting suspension was
P352801NL -51- concentrated in vacuo and any residual acetic anhydride was removed by co-evaporation with toluene (3x). The crude phthalic anhydride was then taken up in THF/H20 (1:1, 25 mL), and to it were added 1- acetylazetidin-3-aminium chloride (0.13 g, 1.06 equiv.) and triethylamine (0.23 mL, 2 equiv.). The solution was stirred at room temperature for 2 hours after which the solvents were removed in vacuo, and residual water was removed by co-evaporation with toluene. Step two: The crude 2-((1-acetylazetidin-3- yhycarbamoyl)-3-chlore-6-nitrobenzoic acid was suspended in acetic anhydride (5 mL) and to it was added sodium acetate trihydrate (0.11 g, 1 equiv.). The resulting solution was stirred at 80 °C for approx. 45 minutes. LC/MS showed full conversion to the desired phthalimide, so the reaction mixture was cooled to room temperature, after which it was diluted with water and the residual acetic anhydride was carefully quenched by tritrating with sat. aq. NaHCOs. Ethyl acetate was then added and the product was extracted (3%). The combined organics were dried over MgSOa and concentrated in vacuo. The title compound was finally purified by column chromatography, eluting with 2% MeOH in DCM affording a white solid. Yield: 19% (0.05 g, 0.15 mmol). 'H NMR (400 MHz, CDCl) ò 8.05 (d, J = 8.6 Hz, 1H), 7.87 (d, J = 8.7 Hz, 1H), 512 (qt, J=8.9, 6.2 Hz, 1H), 4.79 — 4.71 (m, 1H), 4.57 — 4.30 (m, 3H), 1.95 (d, J = 1.6 Hz, 3H). 2-(1-acetylazetidin-3-yl)-4-amino-7-chloroisoindoline-1,3-dione (11): “oo gg AS N ni ©
The compound was synthesized according to general procedure C using the following conditions: 2-(1- acetylazetidin-3-yl)-4-chloro-7-nitroisoindoline-1,3-dione (0.05 g, 0.15 mmol) and tin(il) chloride dihydrate (0.20 g, 6 equiv.) taken up in ethyl acetate (10 mL) and stirred at refluxing temperature for 6 hours, after which LC/MS and TLC showed full conversion to the desired aniline. The crude product was purified by column chromatography eluting with 1.5% MeOH in chloroform, affording the title compound as a bright yellow solid. Yield: 75% (0.03 g, 6.11 mmol). "H NMR (400 MHz, CDCls) 6 7.34 (d, J = 8.8 Hz, 1H), 6.86 (d, J=8.8 Hz, 1H), 5.44 (s, 2H), 5.06 (tt, J = 8.7, 6.2 Hz, 1H), 4.74 (dd, J= 8.4, 6.2 Hz, 1H), 4.53 (dd, J = 10.0,6.3 Hz, 1H), 4.41 (id, J = 8.6, 1.0 Hz, 1H), 4.32 (t, J = 9.4 Hz, 1H), 1.94 (s, 3H). 13C NMR (101 MHz,
CDCls) 6 214.52, 170.71, 165.64, 144.71, 137.44, 127.10, 123.08, 119.36, 54.81, 52.89, 38.22, 19.15.
LC-MS (ESI) m/z calcd for C1aH12CIN3O3 [M + H]* 283.1, found 294.0. HPLC (method A): 7.53 min, purity 98.6%.
P352801NL -52- 4-methyl-3-nitrophthalic acid:
NE
RO; 0
To a stirring solution of nitric acid in conc. sulfuric acid (1:2, 8 mL) was added portion wise at 0 °C 4- methylphthalic acid (2.51 g, 13.9 mmol). The suspension was stirred at 0 °C for 15 minutes, and then allowed to reach room temperature over a period of 2 hours. The reaction mixture was then poured onto ice, solides filtered off and washed with cold water. The combined filtrates were extracted with ethyl acetate (2x), and the combined organics were dried over MgSO.. Solvents were removed in vacuo, and the obtained solids were combined with the precipitated solids, affording a 60/40 mixture of 4-methyl-5- nitrophthalic acid and 4-methyl-3-nitrophthalic acid, respectively, as white solids. Yield: 70% (2.20 g, 10.0 mmol). 2-(1-acetylazetidin-3-yl}-5-methyl-4-nitroisoindoline-1,3-dione: 0 io, ©
Step one: The mixture of 4-methyl-5-nitrophthalic acid and 4-methyl-3-nitrophthalic acid (2.2 g, 10.0 mmol) was taken up in acetic anhydride (16 mL) and stirred at 90 °C for 4 hours. The obtained clear solution was then cooled to room temperature and concentrated in vacuo. Residual amounts of acetic anhydride were removed by co-evaporation with toluene (3x). The intermediate anhydride (0.3 g, 1.45 mmol) was then taken up in THF (5 mL), and to it were added 1-acetylazetidin-3-aminium chloride (0.22 g. 1 equiv.), triethylamine (1 mL, 5 equiv.), and H20 (5 mL). The resulted solution was stirred at room temperature for 1 hour, after which the THF was removed in vacuo, and the reaction mixture was then acidified to pH = 1 by titration with HCI (aq, 2.0 M). The product was extracted with EtOAc (2x), and the combined organics dried over MgSO4 and solvents evaporated to dryness in vacuo. Step two: the intermediate was taken up in acetic anydride (10 mL), and stirred at 80 °C for 1 hour, after which LC/MS showed full conversion to the desired phthalimide. The reaction mixture was concentrated onto silica and purified by column chromatography, eluting with a gradient of 1-2% MeOH in DCM. The title compound was isolated as a 1:1 mixture between 2-(1-acetylazetidin-3-yl)-5-methyl-4-nitroisoindoline-1,3-dione and 2-(1-acetylazetidin-3-yl)-5-methyl-8-nitroisoindoline-1,3-dione, as a colourless oil. Yield: 25% (0.11 g, 0.36 mmol). 'H NMR (400 MHz, CDCls) 6 8.33 (s, 1H), 7.92 (d, J=7.7 Hz, 1H), 7.89 (s, 1H), 7.76 (dd, J = 7.7, 0.5 Hz, 1H), 5.31 (s, 1H), 5.11 (dit, J = 15.0, 8.7, 6.2 Hz, 2H), 4.75 (dt, J = 8.7, 6.0 Hz, 2H), 4.63 — 4.30 (m, 6H), 3.95-3.87 (m, 1H), 2.74 (s, 3H), 2.49 (s, 3H), 1.97 (d, J = 8.5 Hz, 6H).
P352801NL -53- 2-(1-acetylazetidin-3-yl}-4-amino-5-methylisoindoline-1,3-dione (12): © oo, 0
The compound was synthesized according to general procedure C using the following conditions: The mixture of the nitro-substituted methylphthalimides (0.11 g, 0.36 mmol, ~1:1 ratio of isomers) and tin(ll) chloride dihydrate (0.33 g, 4 equiv.) taken up in ethyl acetate (15 mL) and stirred at refluxing temperature for 4 hours. The crude product was purified by column chromatography eluting with a gradient of 1-2%
MeOH in chloroform, affording the title compound as a bright yellow solid. Yield: 45% (0.02 g, 0.08 mmol). 'H NMR (400 MHz, CDCl3) § 7.31 (d, J=7.4 Hz, 1H), 7.08 (d, J = 7.3 Hz, 1H), 5.46-4.82 (m, 2H), 5.03 (it,
J=8.7,6.2 Hz, 1H), 4.81 — 4.66 (m, 1H), 4.62 — 4.47 (m, 1H), 4.46 — 4.28 (m, 2H), 2.24 (s, 3H), 1.94 (s, 3H). BC NMR (101 MHz, CDCl) 6 170.74, 170.03, 168.03, 144.45, 135.89, 130.05, 113.01, 110.49, 55.02, 52.96, 38.07, 19.12, 17.18. LC-MS (ESI) m/z calcd for C14H1sN3O3 [M + H]+ 273.1, found 274.0.
HPLC (method A): 7.31 min, purity 96.9%. 2-(1-acetylazetidin-3-yl}-5-amino-6-methylisoindoline-1,3-dione (13): {3
Hal onl eN 3 0
The compound was synthesized according tc general procedure C using the following conditions: The mixture of the nitro-substituted methylphthalimides (0.11 g, 0.36 mmol, ~1:1 ratio of isomers) and tin(ll} chloride dihydrate (0.33 g, 4 equiv.) taken up in ethyl acetate (15 mL) and stirred at refluxing temperature for 4 hours. The crude product was purified by column chromatography eluting with a gradient of 1-2%
MeOH in chloroform, affording the title compound as an orange-yellow solid. Yield: 55% (0.03 g, 0.10 mmol}, "H NMR (400 MHz, CDCls) 6 7.49 (s, 1H), 7.05 (s, 1H), 5.02 (tt, J= 8.7, 6.2 Hz, 1H), 4.76 — 4.68 t, J=7.2Hz, 1H), 4.62 — 4.23 (m, 5H), 2.24 (s, 3H), 1.93 (s, 3H). 3C NMR (101 MHz, CDCl) ò 170.67, 168.44, 168.18, 150.97, 132.35, 126.91, 126.05, 120.16, 108.33, 55.03, 53.05, 38.16, 19.16, 18.08.
LC-MS (ESI) m/z calcd for C414H1sN2O: [M + H]+ 273.1, found 274.0. HPLC (method A): 6.87 min, purity 97.7%. ethyl 1,3-dioxoisoindoline-2-carboxylate: £3
JEN | 0 he LS
Re ~ | h 0 S
To a stirring solution of phthalimide (7.36 g, 50 mmol) and triethylamine (8.0 mL, 1.3 eq.) in DMF (25 mL) was added at 0 °C in dropwise fashion ethyl chloroformate (5.76 mL, 1.2 eq.). After completion of the
P352801NL - 54 - addition (20 minutes) the resulting suspension was stirred at room temperature for 1.5 hours. The reaction mixture was then poured onto ice and the resulting white precipitate was filtered off, washed with water and subsequently dried tc afford the title compound as a white solid. Yield: 87% (9.0 g, 43,6 mmol). 'H NMR (400 MHz, CDCls) 6 7.98 (dd, J= 5.5, 3.1 Hz, 2H), 7.84 (dd, J = 5.6, 3.0 Hz, 2H), 4.50 (q, J = 7.1
Hz,‚2H), 1.45 (t, J= 7.1 Hz, 3H). tert-butyl 3-(1,3-dioxoisoindolin-2-yl}azetidine-1-carboxylate: <
TN ts a. & x
The compound was synthesized according to general procedure B using the following conditions: tert- butyl 3-aminoazetidine-1-carboxylate (1.0 g, 581 mmol), ethyl 1,3-dioxoisoindoline-2-carboxylate (1.27 g, 1 equiv.), and triethylamine (1.62 ml, 2 equiv.) were taken up in THF (20 mL) and the resulting solution was stirred for 16 hours. The solvents were then removed in vacuo, the crude taken up in EtOAc and washed subsequently with HCL (0.5M, aq.) and brine. The organics were dried over MgSO: and concentrated in vacuo after which column purification eluting with 0-1% MeOH in DCM afforded the title compound as a white solid. Yield: 95% (1.67 g, 5.52 mmol). *H NMR (400 MHz, CDCl:) 6 7.91 — 7.82 (m, 2H), 7.80 — 7.70 (m, 2H), 5.09 — 4.97 (m, 1H), 4.53 (dd, J = 8.9, 6.3 Hz, 2H), 4.24 (t, J = 8.9 Hz, 2H), 1.49 (s, SH). 3-(1,3-dioxoisoindolin-2-yljazetidin-1-ium chloride: 3 ee. SNOT
U tert-butyl 3-(1,3-dioxoisoindolin-2-yl)azetidine-1-carboxylate (1.76 g, 5.82 mmol) was stirred in a solution of HCI in MeOH (2.0 M, 30 mL). After 2 hours TLC indicated full conversion so solvents were removed in vacuo and residual HCI was removed by co-evaporation with toluene (3x), affording the title compound as an off-white solid. Yield: 99% (1.40 g, 5.82 mmol). 'H NMR (400 MHz, DMSO-Ds) 6 10.06 — 8.81 (m, 2H), 7.95-7.83 (m, 4H), 5.15 — 5.02 (m, 1H), 4.60 — 4.51 (m, 2H), 4.22 — 4.08 (m, 2H). 2-(1-acetylazetidin-3-yl}isoindoline-1,3-dione (3): 9
Sl i
The compound synthesized according to general procedure E using the following conditions: 3-(1,3- dioxoisoindolin-2-yl)azetidin-1-ium chloride (0.35 g, 1.45 mmol), acetyl chloride (0.12 mL, 1.2 equiv.}, and
P352801NL -55- triethylamine (0.42 mL, 2.1 equiv.) were stirred in DCM (10 mL) for 1 hour. Column purification eluting with 0-2% MeOH in DCM and a subsequent recrystallization from a mixture of hexanes in EtOAc afforded the title compound as a white solid. Yield: 22% (0.08 g, 0.32 mmol. *H NMR (400 MHz, CDCl:)} 6 7.90 — 7.81 (m, 2H), 7.80 — 7.70 (m, 2H), 5.09 (tt, J = 8.7, 6.2 Hz, 1H), 4.81 — 4.62 (m, 1H), 4.62 — 4.47 (m, 1H), 447-424 (m, 2H), 1.94 (s, 3H). 13C NMR (101 MHz, CDCls) 6 170.66, 167.80, 153.51, 134.61, 131.66, 123.71, 119.93, 38.45, 19.10. LC-MS (ESI) m/z calcd for C13H12N2O: [M + H]+ 244.3, found 245.1. HPLC (method B): 9.77 min, purity 99%.
Material and methods
Mouse models
Cdh5(PAC)-CREERT2: Eng mice were provided by Prof. HM. Arthur (Biosciences Institute, Faculty of medical Sciences, Newcastle University, UK). All experiments were maintained on a C57BL6/J genetic background.
C57BL6/JRj mice were provided by Janvier lab.
For Eng-iKO®. Male and female mice at 8-12 weeks of age were given tamoxifen (2.5 mg per day, total volume 50ml per injection) by intraperitoneal injection for 5 consecutive days to deplete endoglin in endothelial cells. Tamoxifen was purchased from Sigma-Aldrich, (cat. number T5648) and dissolved in 1:4 ethanol (Merck, cat. number 107017)/coil oil (Sigma-Aldrich, cat. number C8267) vehicle. Intraperitoneal injections of either vehicle or thalidomide or analogues began on post-endoglin depletion day 6 (day 0 corresponding to the first injection of tamoxifen) for 2 consecutive days and animals were harvested on day 9.
For irradiation of the head including the eyes, animals were immobilized in a containment device without anesthesia. The heads of adult mice were exposed to an X-ray generator Philips/YXYLON type
MG325 (200kV voltage, 21mA intensity). The X-ray beam was collimated by a bloc of lead located at 50 cm from the source. Four mice were irradiated by run. A total dose of 20Gy was delivered in a single fraction at a dose rate of 0.9Gy/min. Mice were intraperitoneally injected of either vehicle (DMSO), thalidomide or lenalidomide (concentration: 200 mmol kg, total volume 50 ml per injection) on days, -2, -1, +1, +2, +5, +6, and +7, from day O where they received a single fraction of X-ray ionizing radiation. Mice were harvested at day +8, day +90 or day +180 after receiving a single dose of radiation therapy on day 0.
Patch-clamp recordings of pericytes
Patch-clamp experiments were performed as described*. Briefly, fresh mouse retinas were dissected, superfused with bicarbonate-buffered solution containing 124 mM NaCl, 26 mM NaHCOs, 1 mM NaHz2PO,4, 2.5 mM KCI, 1 mM MgClz, 2.5 mM CaCl: and 10 mM glucose, bubbled with 85% O2/5% CO: and were maintained in pH 7.3 at room temperature. An electrode was filled with the same bicarbonate-buffered solution and used to stimulate pericytes by pressing the electrode against the soma to apply voltage pulses {pulses 0.02ms, 10Hz for 5 seconds) and to deliver current intensities ranging from 1 to 3.5 mA. Resistance
P352801NL -56- of the pipette was measured before each experiment in order to define the input voltage. Pericytes were stimulated by applying voltage pulses 20 seconds after patch-clamping and the vascular constriction was recorded during 210 seconds with a total of 300 images taken.
Vascular permeability experiments
Vascular permeability experiments were performed as described. Briefly, Lysine-fixable cadaverine conjugated to Alexa Fluor-555 (5mg.ml” in saline) (Invitrogen, cat. number A30677) was injected intravenously into the tail vein in adult C57BL6/JRj mice at 12 weeks of age. Circulation time was 2 hours for cadaverine Alexa Fluor-555. Animals were harvested through perfusion of HEPES buffer saline for fluorescence tracer quantification or through perfusion of 4% PFA for whole-brain pictures. For fluorescence quantification, the quantity of cadaverine Alexa Fluor-555 injected was 120 mg per 20 grams body weight, respectively. Brains were removed and homogenized in 1% tritonX-100 in PBS pH 7.2. Brains were centrifuged at 16,000 rpm for 20 min and the relative fluorescence of the supernatant was measured on fluorometer POLARstar Omega (BMG Labtech) (ex/em 544/590 nm).
For whole-brain pictures, the quantity of Alexa Fluor-555 or BSA Alexa Fluor-555 injected was 500 mg or 1850 mg per 20 grams body weight, respectively. Brains were removed, post-fixed in 4% PFA overnight and images were taken with an Axio Zoom.V16 fluorescence Stereo Microscope (Zeiss). Images were taken with the exposure time where the fluorescence signal deriving from the brain of irradiated (RT) animals was not saturated.
Antibodies and immunofluorescence staining
All images were acquired with a Leica SP5 confocal microscope (Leica Microsystems). Images were adjusted for brightness and contrast using ImageJ and are presented as maximum intensity projections.
For each image, the endothelial cell surface was defined as Glut1 (+) pixels and determined by the total grey value.
To quantify the number of neurons, 25 microns maximal projection z-stacks were reconstructed and the number of NeuN positive neurons per mm2 was determined as previously described? using ImageJ software Cell Counter plugin analysis tool. In each animal, 6 randomly selected fields (388.74 x 388.74 mm) from the primary somatosensory barrel cortex were analyzed in 3 non adjacent sections (~ 250 mm apart).
Statistical analyses
Statistical analyses were performed with Prism 9 software (GraphPad) using 1-way ANOVA. Results are expressed as mean + SEM. For post hoc pairwise comparisons, the Dunnett's test was used. A value of *P< 0.05, P< 0.01, **P< 0.001 or ***P< 0.001 denoted statistical significance.
P352801NL -57-
References 1-Lebrin, F.; Srun, S.; Raymond, K.; Martin, S.; van den Brink, S.; Freitas, C.; Bréant, C.; Mathivet, T.;
Larrivee, B.; Thomas, J. L.; Arthur, H. M.; Westermann, C. J.; Disch, F.; Mager, J. J.; Snijder, R. J.;
Eichmann, A.; Mummery, C. L. Thalidomide stimulates vessel maturation and reduces epistaxis in individuals with hereditary hemorrhagic telangiectasia. Nat Med 2010, 16, 420-8. 2-Lebrin, F. S., C.; Thalgott, J. Use of thalidomide or analogs thereof for preventing neurologic disorders induced by brain irradiation. 2015. 3-Cheng J, Jiang J, He B, Lin WJ, Li Y, Duan J, Li H, Huang X, Cai J, Xie J, Zhang Z, Yang Y, Xu Y, Hu Xx,
Wu M, Zhuo X, Liu Q, Shi Z, Yu P, Rong X, Ye X, Saw PE, Wu LJ, Simone CB 2nd, Chua MLK, Mai HQ,
Tang Y. A phase 2 study of thalidomide for the treatment of radiation-induced-blood-brain barrier injury.
Sci Transl Med. 2023 Feb 22;15(684):eabm6543. doi: 10.1126/scitransimed.abm6543. Epub 2023 Feb 22.PMID: 36812346 Clinical Trial. 4-Mishra, A.; O'Farrell, F. M.; Reynell, C.; Hamilton, N. B.; Hall, C. N.; Attwell, D. Imaging pericytes and capillary diameter in brain slices and isolated retinae. Naf Profoc 2014, 9, 323-36. 5-Armulik, A.; Genové, G.; Mae, M.; Nisancioglu, M. H.; Wallgard, E.; Niaudet, C.; He, L.; Norlin, J;
Lindblom, P.; Strittmatter, K.; Johansson, B. R.; Betsholtz, C. Pericytes regulate the blood-brain barrier.
Nature 2010, 468, 557-61. 6-Montagne, A.; Nikolakopoulou, A. M.; Zhao, Z.; Sagare, A. P.; Si, G.; Lazic, D.; Barnes, S. R.; Daianu,
M.; Ramanathan, A.; Go, A.; Lawson, E. J.; Wang, Y.; Mack, W. J.; Thompson, P. M.; Schneider, J. A;
Varkey, J.; Langen, R.; Mullins, E.; Jacobs, R. E.; Zlokovic, B. V. Pericyte degeneration causes white matter dysfunction in the mouse central nervous system. Naf Med 2018, 24, 326-337.
Claims (22)
Priority Applications (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
NL2035306A NL2035306B1 (en) | 2023-07-07 | 2023-07-07 | 1,3-isoindolinedione derivatives and heteroaromatic analogues for stimulating endothelial cell-pericyte interaction |
PCT/NL2024/050360 WO2025014359A1 (en) | 2023-07-07 | 2024-07-03 | 1,3-isoindolinedione derivatives and heteroaromatic analogues for stimulating endothelial cell-pericyte interaction |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
NL2035306A NL2035306B1 (en) | 2023-07-07 | 2023-07-07 | 1,3-isoindolinedione derivatives and heteroaromatic analogues for stimulating endothelial cell-pericyte interaction |
Publications (1)
Publication Number | Publication Date |
---|---|
NL2035306B1 true NL2035306B1 (en) | 2025-01-14 |
Family
ID=87514260
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
NL2035306A NL2035306B1 (en) | 2023-07-07 | 2023-07-07 | 1,3-isoindolinedione derivatives and heteroaromatic analogues for stimulating endothelial cell-pericyte interaction |
Country Status (2)
Country | Link |
---|---|
NL (1) | NL2035306B1 (en) |
WO (1) | WO2025014359A1 (en) |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2008021369A2 (en) * | 2006-08-14 | 2008-02-21 | Chembridge Research Laboratories, Inc. | Tricyclic compounds and its use as tyrosine kinase modulators |
WO2011163518A1 (en) * | 2010-06-24 | 2011-12-29 | Gilead Sciences, Inc. | Pyrazolo [1, 5 -a] pyrimidines as antiviral agents |
US20140228350A1 (en) * | 2006-08-14 | 2014-08-14 | Chembridge Corporation | Tricyclic compound derivatives useful in the treatment of neoplastic diseases, inflammatory disorders and immunomodulatory disorders |
EP3147283A1 (en) * | 2014-05-22 | 2017-03-29 | North China Pharmaceutical Company., Ltd. | HYDRAZINE COMPOUND AS BLOOD COAGULATION FACTOR Xa INHIBITOR |
WO2018068017A1 (en) * | 2016-10-07 | 2018-04-12 | Araxes Pharma Llc | Heterocyclic compounds as inhibitors of ras and methods of use thereof |
WO2020031040A1 (en) * | 2018-08-06 | 2020-02-13 | Richter Gedeon Nyrt. | Process and intermediates for the preparation of boc-linagliptin |
WO2022224166A1 (en) * | 2021-04-20 | 2022-10-27 | Integral Biosciences Private Limited | Compounds as lxr agonists |
-
2023
- 2023-07-07 NL NL2035306A patent/NL2035306B1/en active
-
2024
- 2024-07-03 WO PCT/NL2024/050360 patent/WO2025014359A1/en unknown
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2008021369A2 (en) * | 2006-08-14 | 2008-02-21 | Chembridge Research Laboratories, Inc. | Tricyclic compounds and its use as tyrosine kinase modulators |
US20140228350A1 (en) * | 2006-08-14 | 2014-08-14 | Chembridge Corporation | Tricyclic compound derivatives useful in the treatment of neoplastic diseases, inflammatory disorders and immunomodulatory disorders |
WO2011163518A1 (en) * | 2010-06-24 | 2011-12-29 | Gilead Sciences, Inc. | Pyrazolo [1, 5 -a] pyrimidines as antiviral agents |
EP3147283A1 (en) * | 2014-05-22 | 2017-03-29 | North China Pharmaceutical Company., Ltd. | HYDRAZINE COMPOUND AS BLOOD COAGULATION FACTOR Xa INHIBITOR |
WO2018068017A1 (en) * | 2016-10-07 | 2018-04-12 | Araxes Pharma Llc | Heterocyclic compounds as inhibitors of ras and methods of use thereof |
WO2020031040A1 (en) * | 2018-08-06 | 2020-02-13 | Richter Gedeon Nyrt. | Process and intermediates for the preparation of boc-linagliptin |
WO2022224166A1 (en) * | 2021-04-20 | 2022-10-27 | Integral Biosciences Private Limited | Compounds as lxr agonists |
Non-Patent Citations (7)
Title |
---|
ARMULIK, A.GENOVE, G.MAE, M.NISANCIOGLU, M. H.WALLGARD, E.NIAUDET, C.HE, L.NORLIN, J.LINDBLOM, P.STRITTMATTER, K.: "Pericytes regulate the blood-brain barrier", NATURE, vol. 468, 2010, pages 557 - 61, XP055179629, DOI: 10.1038/nature09522 |
CHENG JJIANG JHE BLIN WJLI YDUAN JLI HHUANG XCAI JXIE J: "A phase 2 study of thalidomide for the treatment of radiation-induced-blood-brain barrier injury", SCI TRANSL MED. 2023 FEB 22, vol. 15, no. 684, 22 February 2023 (2023-02-22), pages eabm6543 |
LEBRIN, F. S., C.THALGOTT, J., USE OF THALIDOMIDE OR ANALOGS THEREOF FOR PREVENTING NEUROLOGIC DISORDERS INDUCED BY BRAIN IRRADIATION, 2015 |
LEBRIN, F.SRUN, S.RAYMOND, KMARTIN, S.VAN DEN BRINK, S.FREITAS, C.BREANT, C.MATHIVET, T.LARRIVEE, B.THOMAS, J. L.: "Thalidomide stimulates vessel maturation and reduces epistaxis in individuals with hereditary hemorrhagic telangiectasia", NAT MED, vol. 16, 2010, pages 420 - 8 |
LIER SVENJA ET AL: "A novel Cereblon E3 ligase modulator with antitumor activity in gastrointestinal cancer", BIOORGANIC CHEMISTRY, vol. 119, 1 February 2022 (2022-02-01), US, pages 105505, XP093012700, ISSN: 0045-2068, DOI: 10.1016/j.bioorg.2021.105505 * |
MISHRA, A.O'FARRELLF. M.REYNELL, C.HAMILTON, N. B.HALL, C. N.ATTWELL, D.: "Imaging pericytes and capillary diameter in brain slices and isolated retinae", NAT PROTOC, vol. 9, 2014, pages 323 - 36 |
MONTAGNE, A.NIKOLAKOPOULOU, A. M.ZHAO, Z.SAGARE, A. P.SI, G.LAZIC, D.BARNES, S. R.DAIANU, M.RAMANATHAN, A.GO, A.: "Pericyte degeneration causes white matter dysfunction in the mouse central nervous system", NAT MED, vol. 24, 2018, pages 326 - 337 |
Also Published As
Publication number | Publication date |
---|---|
WO2025014359A1 (en) | 2025-01-16 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
KR102350357B1 (en) | Compositions and methods for modulating farnesoid x receptors | |
JP6629947B2 (en) | Treatment of symptoms associated with hyperinsulinemia | |
ES2640063T3 (en) | N-phenyl-carboxamide derivatives and their use as medicines for the treatment of hepatitis B | |
JP2022547014A (en) | Heterocyclic RIP1 kinase inhibitor | |
JP7026787B2 (en) | A novel heteroarylamide derivative as a selective inhibitor of histone deacetylase 1 and / or 2 (HDAC1-2) | |
DK2928474T3 (en) | Diazollactamer | |
CN107660202B (en) | Water-soluble prodrugs | |
JP7383598B2 (en) | Inhibitors of Rho-related coiled-coil-containing protein kinases | |
JP7089596B2 (en) | 7-substituted sulfonimide ylprinone compounds and derivatives for the treatment and prevention of liver cancer | |
BR112021014514A2 (en) | STRESS INTEGRATED RESPONSE PATHWAY INHIBITORS | |
KR20210102887A (en) | Crystalline spirocyclic compound inhibitors of tryptophan hydroxylase 1 (TPH1) for treating diseases or disorders associated with peripheral serotonin | |
WO2018214796A1 (en) | A class of isoindolone-imide ring-1,3-dione-2-ene compounds, composition and use thereof | |
TW202341983A (en) | Compounds for mutant kras protein degradation and uses thereof | |
IL315399A (en) | MTORC modulators and their uses | |
AU2004237130A1 (en) | Positive modulators of nicotinic acetylcholine receptors | |
NL2035306B1 (en) | 1,3-isoindolinedione derivatives and heteroaromatic analogues for stimulating endothelial cell-pericyte interaction | |
CN111801322B (en) | Kinase inhibitors for the treatment of central and peripheral nervous system disorders | |
EP4052706A1 (en) | Composition for preventing or inhibiting axonal degeneration | |
JP2023033316A (en) | Inhibitors of rho associated coiled-coil containing protein kinase | |
CN111108083A (en) | Use of aminomethylene cyclohexane 1, 3-dione compounds | |
CA3209219A1 (en) | Small molecule regulators of alveolar type 2 cell proliferation for the treatment of pulmonary diseases | |
CN115867277A (en) | Novel fused heterocyclyl-carbohydrazone acyldinitrile compounds and use thereof | |
CN105085491B (en) | Substituted benzazolyl compounds and its application method and purposes | |
WO2024125551A1 (en) | Cyclin k degrader and use thereof | |
JP2012521395A (en) | Anticancer compounds, their preparation and their therapeutic use |