WO2024175662A1 - Flavagline derivatives as ras inhibitors - Google Patents
Flavagline derivatives as ras inhibitors Download PDFInfo
- Publication number
- WO2024175662A1 WO2024175662A1 PCT/EP2024/054426 EP2024054426W WO2024175662A1 WO 2024175662 A1 WO2024175662 A1 WO 2024175662A1 EP 2024054426 W EP2024054426 W EP 2024054426W WO 2024175662 A1 WO2024175662 A1 WO 2024175662A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compounds
- formulae
- compound
- pharmaceutically acceptable
- formula
- Prior art date
Links
- 229940078123 Ras inhibitor Drugs 0.000 title claims description 8
- 150000001875 compounds Chemical class 0.000 claims abstract description 416
- 239000000203 mixture Substances 0.000 claims abstract description 261
- 239000003814 drug Substances 0.000 claims abstract description 73
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 67
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 64
- 102000016914 ras Proteins Human genes 0.000 claims abstract description 62
- 238000000034 method Methods 0.000 claims abstract description 50
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 31
- 230000002401 inhibitory effect Effects 0.000 claims abstract description 30
- 230000002062 proliferating effect Effects 0.000 claims abstract description 25
- 238000009472 formulation Methods 0.000 claims abstract description 23
- 201000011510 cancer Diseases 0.000 claims abstract description 21
- 230000004913 activation Effects 0.000 claims abstract description 18
- 102100030708 GTPase KRas Human genes 0.000 claims abstract description 17
- 101000584612 Homo sapiens GTPase KRas Proteins 0.000 claims abstract description 17
- 102100039788 GTPase NRas Human genes 0.000 claims abstract description 11
- 101000744505 Homo sapiens GTPase NRas Proteins 0.000 claims abstract description 11
- 102100029974 GTPase HRas Human genes 0.000 claims abstract description 10
- 101000584633 Homo sapiens GTPase HRas Proteins 0.000 claims abstract description 10
- 230000035755 proliferation Effects 0.000 claims abstract description 10
- 206010027476 Metastases Diseases 0.000 claims abstract description 4
- 230000009401 metastasis Effects 0.000 claims abstract description 4
- 150000003839 salts Chemical class 0.000 claims description 101
- 229940002612 prodrug Drugs 0.000 claims description 97
- 239000000651 prodrug Substances 0.000 claims description 97
- 238000011282 treatment Methods 0.000 claims description 58
- 208000035475 disorder Diseases 0.000 claims description 41
- 108090000623 proteins and genes Proteins 0.000 claims description 27
- 201000010099 disease Diseases 0.000 claims description 22
- 230000005764 inhibitory process Effects 0.000 claims description 16
- 239000003112 inhibitor Substances 0.000 claims description 14
- 238000011321 prophylaxis Methods 0.000 claims description 13
- 108010014186 ras Proteins Proteins 0.000 claims description 13
- 230000035772 mutation Effects 0.000 claims description 11
- 102000006311 Cyclin D1 Human genes 0.000 claims description 10
- 108010058546 Cyclin D1 Proteins 0.000 claims description 10
- 102100038895 Myc proto-oncogene protein Human genes 0.000 claims description 10
- 101710135898 Myc proto-oncogene protein Proteins 0.000 claims description 10
- 101710150448 Transcriptional regulator Myc Proteins 0.000 claims description 10
- 239000003446 ligand Substances 0.000 claims description 10
- 108010028138 prohibitin Proteins 0.000 claims description 10
- 102000016670 prohibitin Human genes 0.000 claims description 10
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 9
- 230000011664 signaling Effects 0.000 claims description 9
- 208000026350 Inborn Genetic disease Diseases 0.000 claims description 8
- 208000016361 genetic disease Diseases 0.000 claims description 8
- 102200006538 rs121913530 Human genes 0.000 claims description 7
- 208000003019 Neurofibromatosis 1 Diseases 0.000 claims description 6
- 208000024834 Neurofibromatosis type 1 Diseases 0.000 claims description 6
- 229940126062 Compound A Drugs 0.000 claims description 5
- NLDMNSXOCDLTTB-UHFFFAOYSA-N Heterophylliin A Natural products O1C2COC(=O)C3=CC(O)=C(O)C(O)=C3C3=C(O)C(O)=C(O)C=C3C(=O)OC2C(OC(=O)C=2C=C(O)C(O)=C(O)C=2)C(O)C1OC(=O)C1=CC(O)=C(O)C(O)=C1 NLDMNSXOCDLTTB-UHFFFAOYSA-N 0.000 claims description 5
- 230000003213 activating effect Effects 0.000 claims description 5
- 208000027866 inflammatory disease Diseases 0.000 claims description 5
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims description 5
- 239000001257 hydrogen Substances 0.000 claims description 4
- 229910052739 hydrogen Inorganic materials 0.000 claims description 4
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 4
- 102200006655 rs104894230 Human genes 0.000 claims description 4
- 230000003827 upregulation Effects 0.000 claims description 4
- 230000001815 facial effect Effects 0.000 claims description 3
- 102200006532 rs112445441 Human genes 0.000 claims description 3
- 102220011004 rs121913237 Human genes 0.000 claims description 3
- 102220053950 rs121913238 Human genes 0.000 claims description 3
- 102200006525 rs121913240 Human genes 0.000 claims description 3
- 102200006531 rs121913529 Human genes 0.000 claims description 3
- 102200006539 rs121913529 Human genes 0.000 claims description 3
- 102220014328 rs121913535 Human genes 0.000 claims description 3
- 102200007373 rs17851045 Human genes 0.000 claims description 3
- 230000012010 growth Effects 0.000 claims description 2
- 101100232687 Drosophila melanogaster eIF4A gene Proteins 0.000 claims 3
- 230000007783 downstream signaling Effects 0.000 claims 1
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 claims 1
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 claims 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 claims 1
- 210000004027 cell Anatomy 0.000 abstract description 66
- 238000000338 in vitro Methods 0.000 abstract description 9
- 210000000170 cell membrane Anatomy 0.000 abstract description 7
- 101001129610 Homo sapiens Prohibitin 1 Proteins 0.000 abstract description 4
- 102100031169 Prohibitin 1 Human genes 0.000 abstract description 4
- 230000030833 cell death Effects 0.000 abstract description 2
- 230000001939 inductive effect Effects 0.000 abstract description 2
- 235000002639 sodium chloride Nutrition 0.000 description 86
- -1 glucaronate Chemical compound 0.000 description 80
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 49
- 230000000694 effects Effects 0.000 description 41
- 108010056472 Eukaryotic Initiation Factor-4A Proteins 0.000 description 37
- 239000003795 chemical substances by application Substances 0.000 description 37
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 37
- 102000005289 Eukaryotic Initiation Factor-4A Human genes 0.000 description 36
- 229940124597 therapeutic agent Drugs 0.000 description 36
- 229910001868 water Inorganic materials 0.000 description 36
- 239000003937 drug carrier Substances 0.000 description 31
- 239000000725 suspension Substances 0.000 description 30
- HEDRZPFGACZZDS-MICDWDOJSA-N Trichloro(2H)methane Chemical compound [2H]C(Cl)(Cl)Cl HEDRZPFGACZZDS-MICDWDOJSA-N 0.000 description 28
- 239000000243 solution Substances 0.000 description 26
- 239000004480 active ingredient Substances 0.000 description 24
- 229940079593 drug Drugs 0.000 description 24
- RTZKZFJDLAIYFH-UHFFFAOYSA-N ether Substances CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 21
- 241000282414 Homo sapiens Species 0.000 description 20
- 239000007787 solid Substances 0.000 description 20
- 235000019439 ethyl acetate Nutrition 0.000 description 19
- 229940127089 cytotoxic agent Drugs 0.000 description 18
- 239000002552 dosage form Substances 0.000 description 18
- 239000003921 oil Substances 0.000 description 18
- 235000019198 oils Nutrition 0.000 description 18
- 238000002360 preparation method Methods 0.000 description 18
- 239000003826 tablet Substances 0.000 description 18
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 17
- 239000002671 adjuvant Substances 0.000 description 17
- 238000000502 dialysis Methods 0.000 description 17
- 239000003995 emulsifying agent Substances 0.000 description 17
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 17
- 102000004169 proteins and genes Human genes 0.000 description 17
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 16
- 239000002775 capsule Substances 0.000 description 16
- 229940045207 immuno-oncology agent Drugs 0.000 description 16
- 239000002584 immunological anticancer agent Substances 0.000 description 16
- 201000001441 melanoma Diseases 0.000 description 16
- 235000018102 proteins Nutrition 0.000 description 16
- 238000005160 1H NMR spectroscopy Methods 0.000 description 15
- 239000012528 membrane Substances 0.000 description 15
- 239000003755 preservative agent Substances 0.000 description 15
- 239000005557 antagonist Substances 0.000 description 14
- 125000004429 atom Chemical group 0.000 description 14
- 239000000872 buffer Substances 0.000 description 14
- 235000014113 dietary fatty acids Nutrition 0.000 description 14
- 239000000194 fatty acid Substances 0.000 description 14
- 229930195729 fatty acid Natural products 0.000 description 14
- 150000004665 fatty acids Chemical class 0.000 description 14
- 229940127084 other anti-cancer agent Drugs 0.000 description 14
- 239000002904 solvent Substances 0.000 description 14
- 239000002246 antineoplastic agent Substances 0.000 description 13
- 150000002148 esters Chemical class 0.000 description 13
- 239000000796 flavoring agent Substances 0.000 description 13
- 239000000546 pharmaceutical excipient Substances 0.000 description 13
- 230000002829 reductive effect Effects 0.000 description 13
- 239000003765 sweetening agent Substances 0.000 description 13
- 230000014616 translation Effects 0.000 description 13
- 239000000080 wetting agent Substances 0.000 description 13
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 12
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 12
- 239000000839 emulsion Substances 0.000 description 12
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 12
- 239000000463 material Substances 0.000 description 12
- 239000000843 powder Substances 0.000 description 12
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 11
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 11
- 239000007832 Na2SO4 Substances 0.000 description 11
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 11
- 239000003085 diluting agent Substances 0.000 description 11
- 235000013355 food flavoring agent Nutrition 0.000 description 11
- 235000003599 food sweetener Nutrition 0.000 description 11
- 238000004519 manufacturing process Methods 0.000 description 11
- 239000011159 matrix material Substances 0.000 description 11
- 210000004379 membrane Anatomy 0.000 description 11
- 230000036961 partial effect Effects 0.000 description 11
- 108700042226 ras Genes Proteins 0.000 description 11
- 229910052938 sodium sulfate Inorganic materials 0.000 description 11
- 239000003981 vehicle Substances 0.000 description 11
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 10
- 229920000858 Cyclodextrin Polymers 0.000 description 10
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 10
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 10
- 239000007900 aqueous suspension Substances 0.000 description 10
- 239000007859 condensation product Substances 0.000 description 10
- 230000007423 decrease Effects 0.000 description 10
- 235000019197 fats Nutrition 0.000 description 10
- 230000014509 gene expression Effects 0.000 description 10
- 239000008187 granular material Substances 0.000 description 10
- 239000007788 liquid Substances 0.000 description 10
- 239000003208 petroleum Substances 0.000 description 10
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 9
- 241000282412 Homo Species 0.000 description 9
- 239000012267 brine Substances 0.000 description 9
- 230000003993 interaction Effects 0.000 description 9
- 239000002609 medium Substances 0.000 description 9
- 231100000252 nontoxic Toxicity 0.000 description 9
- 230000003000 nontoxic effect Effects 0.000 description 9
- 239000012044 organic layer Substances 0.000 description 9
- 239000011541 reaction mixture Substances 0.000 description 9
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical compound O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 9
- 239000000375 suspending agent Substances 0.000 description 9
- 230000001225 therapeutic effect Effects 0.000 description 9
- PUPZLCDOIYMWBV-UHFFFAOYSA-N (+/-)-1,3-Butanediol Chemical compound CC(O)CCO PUPZLCDOIYMWBV-UHFFFAOYSA-N 0.000 description 8
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 8
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 8
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 8
- 102000004127 Cytokines Human genes 0.000 description 8
- 108090000695 Cytokines Proteins 0.000 description 8
- IAYPIBMASNFSPL-UHFFFAOYSA-N Ethylene oxide Chemical compound C1CO1 IAYPIBMASNFSPL-UHFFFAOYSA-N 0.000 description 8
- 241000124008 Mammalia Species 0.000 description 8
- 241001465754 Metazoa Species 0.000 description 8
- 239000002202 Polyethylene glycol Substances 0.000 description 8
- 239000013543 active substance Substances 0.000 description 8
- 239000003963 antioxidant agent Substances 0.000 description 8
- 235000006708 antioxidants Nutrition 0.000 description 8
- 210000004369 blood Anatomy 0.000 description 8
- 239000008280 blood Substances 0.000 description 8
- 230000037396 body weight Effects 0.000 description 8
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 8
- 229960004316 cisplatin Drugs 0.000 description 8
- 238000002648 combination therapy Methods 0.000 description 8
- 239000002270 dispersing agent Substances 0.000 description 8
- 239000012091 fetal bovine serum Substances 0.000 description 8
- 239000004615 ingredient Substances 0.000 description 8
- 108020004999 messenger RNA Proteins 0.000 description 8
- 229920001223 polyethylene glycol Polymers 0.000 description 8
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 8
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 8
- 230000002335 preservative effect Effects 0.000 description 8
- 102000005962 receptors Human genes 0.000 description 8
- 108020003175 receptors Proteins 0.000 description 8
- 239000000126 substance Substances 0.000 description 8
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 7
- 239000000654 additive Substances 0.000 description 7
- 238000001990 intravenous administration Methods 0.000 description 7
- 239000011780 sodium chloride Substances 0.000 description 7
- 238000013519 translation Methods 0.000 description 7
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 6
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 6
- 102100025221 CD70 antigen Human genes 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 description 6
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 6
- 102100034980 ICOS ligand Human genes 0.000 description 6
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 6
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 description 6
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 description 6
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 6
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 6
- 102100022203 Tumor necrosis factor receptor superfamily member 25 Human genes 0.000 description 6
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 description 6
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 6
- 238000010521 absorption reaction Methods 0.000 description 6
- 239000000556 agonist Substances 0.000 description 6
- 239000007864 aqueous solution Substances 0.000 description 6
- 230000009286 beneficial effect Effects 0.000 description 6
- 230000027455 binding Effects 0.000 description 6
- 239000000969 carrier Substances 0.000 description 6
- 229920002678 cellulose Polymers 0.000 description 6
- 238000006243 chemical reaction Methods 0.000 description 6
- 230000000973 chemotherapeutic effect Effects 0.000 description 6
- 239000003086 colorant Substances 0.000 description 6
- 238000007796 conventional method Methods 0.000 description 6
- 239000007903 gelatin capsule Substances 0.000 description 6
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 6
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 6
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 6
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 description 6
- 238000001727 in vivo Methods 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 238000007918 intramuscular administration Methods 0.000 description 6
- 239000008101 lactose Substances 0.000 description 6
- 235000010445 lecithin Nutrition 0.000 description 6
- 239000000787 lecithin Substances 0.000 description 6
- 229940067606 lecithin Drugs 0.000 description 6
- 229940057995 liquid paraffin Drugs 0.000 description 6
- 235000019359 magnesium stearate Nutrition 0.000 description 6
- 239000004530 micro-emulsion Substances 0.000 description 6
- 238000002156 mixing Methods 0.000 description 6
- 239000000346 nonvolatile oil Substances 0.000 description 6
- 239000004006 olive oil Substances 0.000 description 6
- 235000008390 olive oil Nutrition 0.000 description 6
- DAPAQENNNINUPW-IDAMAFBJSA-N rocaglamide Chemical compound C1=CC(OC)=CC=C1[C@]1([C@@H]([C@H]([C@H]2O)C(=O)N(C)C)C=3C=CC=CC=3)[C@]2(O)C2=C(OC)C=C(OC)C=C2O1 DAPAQENNNINUPW-IDAMAFBJSA-N 0.000 description 6
- 239000011734 sodium Substances 0.000 description 6
- 235000011152 sodium sulphate Nutrition 0.000 description 6
- 230000007928 solubilization Effects 0.000 description 6
- 238000005063 solubilization Methods 0.000 description 6
- 239000003381 stabilizer Substances 0.000 description 6
- 230000004936 stimulating effect Effects 0.000 description 6
- 238000007920 subcutaneous administration Methods 0.000 description 6
- 239000006188 syrup Substances 0.000 description 6
- 235000020357 syrup Nutrition 0.000 description 6
- 238000002560 therapeutic procedure Methods 0.000 description 6
- 238000004809 thin layer chromatography Methods 0.000 description 6
- 229960005486 vaccine Drugs 0.000 description 6
- 239000001993 wax Substances 0.000 description 6
- QFLWZFQWSBQYPS-AWRAUJHKSA-N (3S)-3-[[(2S)-2-[[(2S)-2-[5-[(3aS,6aR)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoylamino]-3-methylbutanoyl]amino]-3-(4-hydroxyphenyl)propanoyl]amino]-4-[1-bis(4-chlorophenoxy)phosphorylbutylamino]-4-oxobutanoic acid Chemical compound CCCC(NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](Cc1ccc(O)cc1)NC(=O)[C@@H](NC(=O)CCCCC1SC[C@@H]2NC(=O)N[C@H]12)C(C)C)P(=O)(Oc1ccc(Cl)cc1)Oc1ccc(Cl)cc1 QFLWZFQWSBQYPS-AWRAUJHKSA-N 0.000 description 5
- 239000002253 acid Substances 0.000 description 5
- 229910052681 coesite Inorganic materials 0.000 description 5
- 229910052906 cristobalite Inorganic materials 0.000 description 5
- 210000000172 cytosol Anatomy 0.000 description 5
- 230000009977 dual effect Effects 0.000 description 5
- 239000012636 effector Substances 0.000 description 5
- 235000019441 ethanol Nutrition 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 238000011534 incubation Methods 0.000 description 5
- 238000003670 luciferase enzyme activity assay Methods 0.000 description 5
- 239000002953 phosphate buffered saline Substances 0.000 description 5
- 230000008569 process Effects 0.000 description 5
- 239000000377 silicon dioxide Substances 0.000 description 5
- 229910052682 stishovite Inorganic materials 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 230000014621 translational initiation Effects 0.000 description 5
- 229910052905 tridymite Inorganic materials 0.000 description 5
- SZUVGFMDDVSKSI-WIFOCOSTSA-N (1s,2s,3s,5r)-1-(carboxymethyl)-3,5-bis[(4-phenoxyphenyl)methyl-propylcarbamoyl]cyclopentane-1,2-dicarboxylic acid Chemical compound O=C([C@@H]1[C@@H]([C@](CC(O)=O)([C@H](C(=O)N(CCC)CC=2C=CC(OC=3C=CC=CC=3)=CC=2)C1)C(O)=O)C(O)=O)N(CCC)CC(C=C1)=CC=C1OC1=CC=CC=C1 SZUVGFMDDVSKSI-WIFOCOSTSA-N 0.000 description 4
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 4
- ZORQXIQZAOLNGE-UHFFFAOYSA-N 1,1-difluorocyclohexane Chemical compound FC1(F)CCCCC1 ZORQXIQZAOLNGE-UHFFFAOYSA-N 0.000 description 4
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 description 4
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 4
- IZHVBANLECCAGF-UHFFFAOYSA-N 2-hydroxy-3-(octadecanoyloxy)propyl octadecanoate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)COC(=O)CCCCCCCCCCCCCCCCC IZHVBANLECCAGF-UHFFFAOYSA-N 0.000 description 4
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 4
- 102000002627 4-1BB Ligand Human genes 0.000 description 4
- 108010082808 4-1BB Ligand Proteins 0.000 description 4
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 4
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 4
- 235000003911 Arachis Nutrition 0.000 description 4
- 244000105624 Arachis hypogaea Species 0.000 description 4
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 4
- 108010074708 B7-H1 Antigen Proteins 0.000 description 4
- 101150013553 CD40 gene Proteins 0.000 description 4
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 4
- 229940045513 CTLA4 antagonist Drugs 0.000 description 4
- 201000009030 Carcinoma Diseases 0.000 description 4
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 4
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 4
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 4
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 4
- 102100037354 Ectodysplasin-A Human genes 0.000 description 4
- 102000004190 Enzymes Human genes 0.000 description 4
- 108090000790 Enzymes Proteins 0.000 description 4
- 108010010803 Gelatin Proteins 0.000 description 4
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 4
- 229920000084 Gum arabic Polymers 0.000 description 4
- 101000934356 Homo sapiens CD70 antigen Proteins 0.000 description 4
- 101000880080 Homo sapiens Ectodysplasin-A Proteins 0.000 description 4
- 101001019455 Homo sapiens ICOS ligand Proteins 0.000 description 4
- 101000610605 Homo sapiens Tumor necrosis factor receptor superfamily member 10A Proteins 0.000 description 4
- 101000610604 Homo sapiens Tumor necrosis factor receptor superfamily member 10B Proteins 0.000 description 4
- 101000679903 Homo sapiens Tumor necrosis factor receptor superfamily member 25 Proteins 0.000 description 4
- 101000679907 Homo sapiens Tumor necrosis factor receptor superfamily member 27 Proteins 0.000 description 4
- 101000920026 Homo sapiens Tumor necrosis factor receptor superfamily member EDAR Proteins 0.000 description 4
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 4
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 4
- 102000000588 Interleukin-2 Human genes 0.000 description 4
- 108010002350 Interleukin-2 Proteins 0.000 description 4
- 102000004083 Lymphotoxin-alpha Human genes 0.000 description 4
- 108090000542 Lymphotoxin-alpha Proteins 0.000 description 4
- 102100028198 Macrophage colony-stimulating factor 1 receptor Human genes 0.000 description 4
- 101710150918 Macrophage colony-stimulating factor 1 receptor Proteins 0.000 description 4
- BZLVMXJERCGZMT-UHFFFAOYSA-N Methyl tert-butyl ether Chemical compound COC(C)(C)C BZLVMXJERCGZMT-UHFFFAOYSA-N 0.000 description 4
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 4
- 239000005642 Oleic acid Substances 0.000 description 4
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 4
- 108700020796 Oncogene Proteins 0.000 description 4
- 229910019142 PO4 Inorganic materials 0.000 description 4
- 235000019483 Peanut oil Nutrition 0.000 description 4
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 4
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 4
- 108010029485 Protein Isoforms Proteins 0.000 description 4
- 102000001708 Protein Isoforms Human genes 0.000 description 4
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 4
- 229920002472 Starch Polymers 0.000 description 4
- 235000021355 Stearic acid Nutrition 0.000 description 4
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 4
- 229930006000 Sucrose Natural products 0.000 description 4
- 230000006044 T cell activation Effects 0.000 description 4
- 230000005867 T cell response Effects 0.000 description 4
- 210000001744 T-lymphocyte Anatomy 0.000 description 4
- 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 4
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 4
- DKGAVHZHDRPRBM-UHFFFAOYSA-N Tert-Butanol Chemical compound CC(C)(C)O DKGAVHZHDRPRBM-UHFFFAOYSA-N 0.000 description 4
- 229920001615 Tragacanth Polymers 0.000 description 4
- 102100040113 Tumor necrosis factor receptor superfamily member 10A Human genes 0.000 description 4
- 102100040112 Tumor necrosis factor receptor superfamily member 10B Human genes 0.000 description 4
- 102100022202 Tumor necrosis factor receptor superfamily member 27 Human genes 0.000 description 4
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 4
- 102100030810 Tumor necrosis factor receptor superfamily member EDAR Human genes 0.000 description 4
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 235000010489 acacia gum Nutrition 0.000 description 4
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 4
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 4
- 235000010443 alginic acid Nutrition 0.000 description 4
- 239000000783 alginic acid Substances 0.000 description 4
- 229920000615 alginic acid Polymers 0.000 description 4
- 229960001126 alginic acid Drugs 0.000 description 4
- 150000004781 alginic acids Chemical class 0.000 description 4
- 235000001014 amino acid Nutrition 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 230000000259 anti-tumor effect Effects 0.000 description 4
- 239000000427 antigen Substances 0.000 description 4
- 108091007433 antigens Proteins 0.000 description 4
- 102000036639 antigens Human genes 0.000 description 4
- 230000003078 antioxidant effect Effects 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- WHGYBXFWUBPSRW-FOUAGVGXSA-N beta-cyclodextrin Chemical compound OC[C@H]([C@H]([C@@H]([C@H]1O)O)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)CO)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1CO WHGYBXFWUBPSRW-FOUAGVGXSA-N 0.000 description 4
- 229960004853 betadex Drugs 0.000 description 4
- 239000011230 binding agent Substances 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 235000019437 butane-1,3-diol Nutrition 0.000 description 4
- 229910000019 calcium carbonate Inorganic materials 0.000 description 4
- 239000001506 calcium phosphate Substances 0.000 description 4
- 229910000389 calcium phosphate Inorganic materials 0.000 description 4
- 235000011010 calcium phosphates Nutrition 0.000 description 4
- 229940022399 cancer vaccine Drugs 0.000 description 4
- 239000001768 carboxy methyl cellulose Substances 0.000 description 4
- 229960005243 carmustine Drugs 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 235000010980 cellulose Nutrition 0.000 description 4
- 239000001913 cellulose Substances 0.000 description 4
- 238000002512 chemotherapy Methods 0.000 description 4
- 238000004440 column chromatography Methods 0.000 description 4
- 229940126543 compound 14 Drugs 0.000 description 4
- 229940125782 compound 2 Drugs 0.000 description 4
- 229940125898 compound 5 Drugs 0.000 description 4
- 239000006071 cream Substances 0.000 description 4
- 229940097362 cyclodextrins Drugs 0.000 description 4
- 239000002254 cytotoxic agent Substances 0.000 description 4
- 231100000599 cytotoxic agent Toxicity 0.000 description 4
- 229960003901 dacarbazine Drugs 0.000 description 4
- 230000008482 dysregulation Effects 0.000 description 4
- 229940088598 enzyme Drugs 0.000 description 4
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 4
- 229920000159 gelatin Polymers 0.000 description 4
- 239000008273 gelatin Substances 0.000 description 4
- 235000019322 gelatine Nutrition 0.000 description 4
- 235000011852 gelatine desserts Nutrition 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- BXWNKGSJHAJOGX-UHFFFAOYSA-N hexadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCO BXWNKGSJHAJOGX-UHFFFAOYSA-N 0.000 description 4
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 4
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 4
- 229940071676 hydroxypropylcellulose Drugs 0.000 description 4
- 210000000987 immune system Anatomy 0.000 description 4
- 238000001802 infusion Methods 0.000 description 4
- 238000010253 intravenous injection Methods 0.000 description 4
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 4
- 238000011068 loading method Methods 0.000 description 4
- 239000000314 lubricant Substances 0.000 description 4
- 238000004020 luminiscence type Methods 0.000 description 4
- 229960001924 melphalan Drugs 0.000 description 4
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 4
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 4
- 239000002480 mineral oil Substances 0.000 description 4
- 235000010446 mineral oil Nutrition 0.000 description 4
- 239000001788 mono and diglycerides of fatty acids Substances 0.000 description 4
- 210000000822 natural killer cell Anatomy 0.000 description 4
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 4
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 4
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 4
- 230000037361 pathway Effects 0.000 description 4
- 239000000312 peanut oil Substances 0.000 description 4
- FPVKHBSQESCIEP-JQCXWYLXSA-N pentostatin Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC[C@H]2O)=C2N=C1 FPVKHBSQESCIEP-JQCXWYLXSA-N 0.000 description 4
- 235000021317 phosphate Nutrition 0.000 description 4
- 239000006187 pill Substances 0.000 description 4
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 4
- 239000008389 polyethoxylated castor oil Substances 0.000 description 4
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 4
- 229920000053 polysorbate 80 Polymers 0.000 description 4
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 4
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 4
- 230000002265 prevention Effects 0.000 description 4
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical compound CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 4
- 238000000746 purification Methods 0.000 description 4
- 230000005855 radiation Effects 0.000 description 4
- 239000008159 sesame oil Substances 0.000 description 4
- 235000011803 sesame oil Nutrition 0.000 description 4
- 150000003384 small molecules Chemical class 0.000 description 4
- 229910052708 sodium Inorganic materials 0.000 description 4
- 235000010413 sodium alginate Nutrition 0.000 description 4
- 239000000661 sodium alginate Substances 0.000 description 4
- 229940005550 sodium alginate Drugs 0.000 description 4
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 4
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 4
- 235000011069 sorbitan monooleate Nutrition 0.000 description 4
- 239000001593 sorbitan monooleate Substances 0.000 description 4
- 229940035049 sorbitan monooleate Drugs 0.000 description 4
- 239000008107 starch Substances 0.000 description 4
- 235000019698 starch Nutrition 0.000 description 4
- 239000007858 starting material Substances 0.000 description 4
- 239000008117 stearic acid Substances 0.000 description 4
- 239000005720 sucrose Substances 0.000 description 4
- 239000004094 surface-active agent Substances 0.000 description 4
- 239000000454 talc Substances 0.000 description 4
- 229910052623 talc Inorganic materials 0.000 description 4
- 235000012222 talc Nutrition 0.000 description 4
- 229960004964 temozolomide Drugs 0.000 description 4
- NRUKOCRGYNPUPR-QBPJDGROSA-N teniposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@@H](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 4
- FPGGTKZVZWFYPV-UHFFFAOYSA-M tetrabutylammonium fluoride Chemical compound [F-].CCCC[N+](CCCC)(CCCC)CCCC FPGGTKZVZWFYPV-UHFFFAOYSA-M 0.000 description 4
- HLZKNKRTKFSKGZ-UHFFFAOYSA-N tetradecan-1-ol Chemical compound CCCCCCCCCCCCCCO HLZKNKRTKFSKGZ-UHFFFAOYSA-N 0.000 description 4
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 4
- 210000001519 tissue Anatomy 0.000 description 4
- AOBORMOPSGHCAX-DGHZZKTQSA-N tocofersolan Chemical compound OCCOC(=O)CCC(=O)OC1=C(C)C(C)=C2O[C@](CCC[C@H](C)CCC[C@H](C)CCCC(C)C)(C)CCC2=C1C AOBORMOPSGHCAX-DGHZZKTQSA-N 0.000 description 4
- 229960000984 tocofersolan Drugs 0.000 description 4
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 4
- 235000015112 vegetable and seed oil Nutrition 0.000 description 4
- 239000008158 vegetable oil Substances 0.000 description 4
- 229960003048 vinblastine Drugs 0.000 description 4
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 4
- 239000002076 α-tocopherol Substances 0.000 description 4
- 235000004835 α-tocopherol Nutrition 0.000 description 4
- UNILWMWFPHPYOR-KXEYIPSPSA-M 1-[6-[2-[3-[3-[3-[2-[2-[3-[[2-[2-[[(2r)-1-[[2-[[(2r)-1-[3-[2-[2-[3-[[2-(2-amino-2-oxoethoxy)acetyl]amino]propoxy]ethoxy]ethoxy]propylamino]-3-hydroxy-1-oxopropan-2-yl]amino]-2-oxoethyl]amino]-3-[(2r)-2,3-di(hexadecanoyloxy)propyl]sulfanyl-1-oxopropan-2-yl Chemical compound O=C1C(SCCC(=O)NCCCOCCOCCOCCCNC(=O)COCC(=O)N[C@@H](CSC[C@@H](COC(=O)CCCCCCCCCCCCCCC)OC(=O)CCCCCCCCCCCCCCC)C(=O)NCC(=O)N[C@H](CO)C(=O)NCCCOCCOCCOCCCNC(=O)COCC(N)=O)CC(=O)N1CCNC(=O)CCCCCN\1C2=CC=C(S([O-])(=O)=O)C=C2CC/1=C/C=C/C=C/C1=[N+](CC)C2=CC=C(S([O-])(=O)=O)C=C2C1 UNILWMWFPHPYOR-KXEYIPSPSA-M 0.000 description 3
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 3
- OJRUSAPKCPIVBY-KQYNXXCUSA-N C1=NC2=C(N=C(N=C2N1[C@H]3[C@@H]([C@@H]([C@H](O3)COP(=O)(CP(=O)(O)O)O)O)O)I)N Chemical compound C1=NC2=C(N=C(N=C2N1[C@H]3[C@@H]([C@@H]([C@H](O3)COP(=O)(CP(=O)(O)O)O)O)O)I)N OJRUSAPKCPIVBY-KQYNXXCUSA-N 0.000 description 3
- 102100025626 GTP-binding protein GEM Human genes 0.000 description 3
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 3
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 3
- 108060001084 Luciferase Proteins 0.000 description 3
- 239000005089 Luciferase Substances 0.000 description 3
- 238000000134 MTT assay Methods 0.000 description 3
- 231100000002 MTT assay Toxicity 0.000 description 3
- 229910017906 NH3H2O Inorganic materials 0.000 description 3
- 102000043276 Oncogene Human genes 0.000 description 3
- 102000005877 Peptide Initiation Factors Human genes 0.000 description 3
- 108010044843 Peptide Initiation Factors Proteins 0.000 description 3
- 239000004698 Polyethylene Substances 0.000 description 3
- 150000001408 amides Chemical class 0.000 description 3
- 239000003242 anti bacterial agent Substances 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 235000010323 ascorbic acid Nutrition 0.000 description 3
- 239000011668 ascorbic acid Substances 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- NDKBVBUGCNGSJJ-UHFFFAOYSA-M benzyltrimethylammonium hydroxide Chemical compound [OH-].C[N+](C)(C)CC1=CC=CC=C1 NDKBVBUGCNGSJJ-UHFFFAOYSA-M 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 238000004364 calculation method Methods 0.000 description 3
- 230000010261 cell growth Effects 0.000 description 3
- 229940125904 compound 1 Drugs 0.000 description 3
- 229940125758 compound 15 Drugs 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- DAPAQENNNINUPW-UHFFFAOYSA-N endo rocaglamide Natural products C1=CC(OC)=CC=C1C1(C(C(C2O)C(=O)N(C)C)C=3C=CC=CC=3)C2(O)C2=C(OC)C=C(OC)C=C2O1 DAPAQENNNINUPW-UHFFFAOYSA-N 0.000 description 3
- 239000001963 growth medium Substances 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 238000010829 isocratic elution Methods 0.000 description 3
- 238000012417 linear regression Methods 0.000 description 3
- 230000004060 metabolic process Effects 0.000 description 3
- 229910052757 nitrogen Inorganic materials 0.000 description 3
- 230000003287 optical effect Effects 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 239000002304 perfume Substances 0.000 description 3
- 230000003285 pharmacodynamic effect Effects 0.000 description 3
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 3
- 239000013612 plasmid Substances 0.000 description 3
- 102000004196 processed proteins & peptides Human genes 0.000 description 3
- 108090000765 processed proteins & peptides Proteins 0.000 description 3
- 238000012545 processing Methods 0.000 description 3
- 238000001243 protein synthesis Methods 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 235000012239 silicon dioxide Nutrition 0.000 description 3
- 239000001488 sodium phosphate Substances 0.000 description 3
- 241000894007 species Species 0.000 description 3
- KDYFGRWQOYBRFD-UHFFFAOYSA-L succinate(2-) Chemical compound [O-]C(=O)CCC([O-])=O KDYFGRWQOYBRFD-UHFFFAOYSA-L 0.000 description 3
- 238000004808 supercritical fluid chromatography Methods 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- LEIMLDGFXIOXMT-UHFFFAOYSA-N trimethylsilyl cyanide Chemical compound C[Si](C)(C)C#N LEIMLDGFXIOXMT-UHFFFAOYSA-N 0.000 description 3
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 3
- GHYOCDFICYLMRF-UTIIJYGPSA-N (2S,3R)-N-[(2S)-3-(cyclopenten-1-yl)-1-[(2R)-2-methyloxiran-2-yl]-1-oxopropan-2-yl]-3-hydroxy-3-(4-methoxyphenyl)-2-[[(2S)-2-[(2-morpholin-4-ylacetyl)amino]propanoyl]amino]propanamide Chemical compound C1(=CCCC1)C[C@@H](C(=O)[C@@]1(OC1)C)NC([C@H]([C@@H](C1=CC=C(C=C1)OC)O)NC([C@H](C)NC(CN1CCOCC1)=O)=O)=O GHYOCDFICYLMRF-UTIIJYGPSA-N 0.000 description 2
- JPSHPWJJSVEEAX-OWPBQMJCSA-N (2s)-2-amino-4-fluoranylpentanedioic acid Chemical compound OC(=O)[C@@H](N)CC([18F])C(O)=O JPSHPWJJSVEEAX-OWPBQMJCSA-N 0.000 description 2
- GVJHHUAWPYXKBD-IEOSBIPESA-N (R)-alpha-Tocopherol Natural products OC1=C(C)C(C)=C2O[C@@](CCC[C@H](C)CCC[C@H](C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-IEOSBIPESA-N 0.000 description 2
- 102100025573 1-alkyl-2-acetylglycerophosphocholine esterase Human genes 0.000 description 2
- JLPULHDHAOZNQI-ZTIMHPMXSA-N 1-hexadecanoyl-2-(9Z,12Z-octadecadienoyl)-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/C\C=C/CCCCC JLPULHDHAOZNQI-ZTIMHPMXSA-N 0.000 description 2
- CHHHXKFHOYLYRE-UHFFFAOYSA-M 2,4-Hexadienoic acid, potassium salt (1:1), (2E,4E)- Chemical compound [K+].CC=CC=CC([O-])=O CHHHXKFHOYLYRE-UHFFFAOYSA-M 0.000 description 2
- OZAIFHULBGXAKX-UHFFFAOYSA-N 2-(2-cyanopropan-2-yldiazenyl)-2-methylpropanenitrile Chemical compound N#CC(C)(C)N=NC(C)(C)C#N OZAIFHULBGXAKX-UHFFFAOYSA-N 0.000 description 2
- BGFTWECWAICPDG-UHFFFAOYSA-N 2-[bis(4-chlorophenyl)methyl]-4-n-[3-[bis(4-chlorophenyl)methyl]-4-(dimethylamino)phenyl]-1-n,1-n-dimethylbenzene-1,4-diamine Chemical compound C1=C(C(C=2C=CC(Cl)=CC=2)C=2C=CC(Cl)=CC=2)C(N(C)C)=CC=C1NC(C=1)=CC=C(N(C)C)C=1C(C=1C=CC(Cl)=CC=1)C1=CC=C(Cl)C=C1 BGFTWECWAICPDG-UHFFFAOYSA-N 0.000 description 2
- NDMPLJNOPCLANR-UHFFFAOYSA-N 3,4-dihydroxy-15-(4-hydroxy-18-methoxycarbonyl-5,18-seco-ibogamin-18-yl)-16-methoxy-1-methyl-6,7-didehydro-aspidospermidine-3-carboxylic acid methyl ester Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 NDMPLJNOPCLANR-UHFFFAOYSA-N 0.000 description 2
- XQDNFAMOIPNVES-UHFFFAOYSA-N 3,5-Dimethoxyphenol Chemical compound COC1=CC(O)=CC(OC)=C1 XQDNFAMOIPNVES-UHFFFAOYSA-N 0.000 description 2
- AOJJSUZBOXZQNB-VTZDEGQISA-N 4'-epidoxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-VTZDEGQISA-N 0.000 description 2
- 108020003589 5' Untranslated Regions Proteins 0.000 description 2
- IDPUKCWIGUEADI-UHFFFAOYSA-N 5-[bis(2-chloroethyl)amino]uracil Chemical compound ClCCN(CCCl)C1=CNC(=O)NC1=O IDPUKCWIGUEADI-UHFFFAOYSA-N 0.000 description 2
- VVIAGPKUTFNRDU-UHFFFAOYSA-N 6S-folinic acid Natural products C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 VVIAGPKUTFNRDU-UHFFFAOYSA-N 0.000 description 2
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 2
- OZAIFHULBGXAKX-VAWYXSNFSA-N AIBN Substances N#CC(C)(C)\N=N\C(C)(C)C#N OZAIFHULBGXAKX-VAWYXSNFSA-N 0.000 description 2
- 244000215068 Acacia senegal Species 0.000 description 2
- 235000006491 Acacia senegal Nutrition 0.000 description 2
- 102100022089 Acyl-[acyl-carrier-protein] hydrolase Human genes 0.000 description 2
- 102100031934 Adhesion G-protein coupled receptor G1 Human genes 0.000 description 2
- 229920001450 Alpha-Cyclodextrin Polymers 0.000 description 2
- 239000005995 Aluminium silicate Substances 0.000 description 2
- 108010024976 Asparaginase Proteins 0.000 description 2
- 108010011485 Aspartame Proteins 0.000 description 2
- 241000416162 Astragalus gummifer Species 0.000 description 2
- NOWKCMXCCJGMRR-UHFFFAOYSA-N Aziridine Chemical class C1CN1 NOWKCMXCCJGMRR-UHFFFAOYSA-N 0.000 description 2
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 2
- 102000006942 B-Cell Maturation Antigen Human genes 0.000 description 2
- 108010008014 B-Cell Maturation Antigen Proteins 0.000 description 2
- 108010006654 Bleomycin Proteins 0.000 description 2
- 102000004506 Blood Proteins Human genes 0.000 description 2
- 108010017384 Blood Proteins Proteins 0.000 description 2
- 101000964894 Bos taurus 14-3-3 protein zeta/delta Proteins 0.000 description 2
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 2
- 102100031151 C-C chemokine receptor type 2 Human genes 0.000 description 2
- 101710149815 C-C chemokine receptor type 2 Proteins 0.000 description 2
- 101710149863 C-C chemokine receptor type 4 Proteins 0.000 description 2
- 101150025841 CCND1 gene Proteins 0.000 description 2
- 102100032976 CCR4-NOT transcription complex subunit 6 Human genes 0.000 description 2
- 108010046080 CD27 Ligand Proteins 0.000 description 2
- 102100027207 CD27 antigen Human genes 0.000 description 2
- 102100038078 CD276 antigen Human genes 0.000 description 2
- 108010029697 CD40 Ligand Proteins 0.000 description 2
- 102100032937 CD40 ligand Human genes 0.000 description 2
- 102100036008 CD48 antigen Human genes 0.000 description 2
- GAGWJHPBXLXJQN-UORFTKCHSA-N Capecitabine Chemical compound C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1[C@H]1[C@H](O)[C@H](O)[C@@H](C)O1 GAGWJHPBXLXJQN-UORFTKCHSA-N 0.000 description 2
- GAGWJHPBXLXJQN-UHFFFAOYSA-N Capecitabine Natural products C1=C(F)C(NC(=O)OCCCCC)=NC(=O)N1C1C(O)C(O)C(C)O1 GAGWJHPBXLXJQN-UHFFFAOYSA-N 0.000 description 2
- 108050000299 Chemokine receptor Proteins 0.000 description 2
- 102000009410 Chemokine receptor Human genes 0.000 description 2
- 208000006332 Choriocarcinoma Diseases 0.000 description 2
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 2
- 229920002261 Corn starch Polymers 0.000 description 2
- 101710093674 Cyclic nucleotide-gated cation channel beta-1 Proteins 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 108010041986 DNA Vaccines Proteins 0.000 description 2
- 229940021995 DNA vaccine Drugs 0.000 description 2
- 108010092160 Dactinomycin Proteins 0.000 description 2
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 2
- ROSDSFDQCJNGOL-UHFFFAOYSA-N Dimethylamine Chemical compound CNC ROSDSFDQCJNGOL-UHFFFAOYSA-N 0.000 description 2
- 206010013911 Dysgeusia Diseases 0.000 description 2
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 2
- HTIJFSOGRVMCQR-UHFFFAOYSA-N Epirubicin Natural products COc1cccc2C(=O)c3c(O)c4CC(O)(CC(OC5CC(N)C(=O)C(C)O5)c4c(O)c3C(=O)c12)C(=O)CO HTIJFSOGRVMCQR-UHFFFAOYSA-N 0.000 description 2
- 239000001856 Ethyl cellulose Substances 0.000 description 2
- ZZSNKZQZMQGXPY-UHFFFAOYSA-N Ethyl cellulose Chemical compound CCOCC1OC(OC)C(OCC)C(OCC)C1OC1C(O)C(O)C(OC)C(CO)O1 ZZSNKZQZMQGXPY-UHFFFAOYSA-N 0.000 description 2
- 101150089023 FASLG gene Proteins 0.000 description 2
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 2
- 108010001498 Galectin 1 Proteins 0.000 description 2
- 102100021736 Galectin-1 Human genes 0.000 description 2
- 102100031351 Galectin-9 Human genes 0.000 description 2
- 101710121810 Galectin-9 Proteins 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
- 239000004471 Glycine Substances 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 2
- 206010066476 Haematological malignancy Diseases 0.000 description 2
- 108010007712 Hepatitis A Virus Cellular Receptor 1 Proteins 0.000 description 2
- 102100034459 Hepatitis A virus cellular receptor 1 Human genes 0.000 description 2
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 2
- 101710083479 Hepatitis A virus cellular receptor 2 homolog Proteins 0.000 description 2
- 101000824278 Homo sapiens Acyl-[acyl-carrier-protein] hydrolase Proteins 0.000 description 2
- 101000775042 Homo sapiens Adhesion G-protein coupled receptor G1 Proteins 0.000 description 2
- 101000864344 Homo sapiens B- and T-lymphocyte attenuator Proteins 0.000 description 2
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 2
- 101000716130 Homo sapiens CD48 antigen Proteins 0.000 description 2
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 2
- 101001138062 Homo sapiens Leukocyte-associated immunoglobulin-like receptor 1 Proteins 0.000 description 2
- 101001023712 Homo sapiens Nectin-3 Proteins 0.000 description 2
- 101000831007 Homo sapiens T-cell immunoreceptor with Ig and ITIM domains Proteins 0.000 description 2
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 2
- 101100207070 Homo sapiens TNFSF8 gene Proteins 0.000 description 2
- 101000764622 Homo sapiens Transmembrane and immunoglobulin domain-containing protein 2 Proteins 0.000 description 2
- 101000764263 Homo sapiens Tumor necrosis factor ligand superfamily member 4 Proteins 0.000 description 2
- 101000610602 Homo sapiens Tumor necrosis factor receptor superfamily member 10C Proteins 0.000 description 2
- 101000610609 Homo sapiens Tumor necrosis factor receptor superfamily member 10D Proteins 0.000 description 2
- 101000798130 Homo sapiens Tumor necrosis factor receptor superfamily member 11B Proteins 0.000 description 2
- 101000795167 Homo sapiens Tumor necrosis factor receptor superfamily member 13B Proteins 0.000 description 2
- 101000795169 Homo sapiens Tumor necrosis factor receptor superfamily member 13C Proteins 0.000 description 2
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 description 2
- 101000801227 Homo sapiens Tumor necrosis factor receptor superfamily member 19 Proteins 0.000 description 2
- 101000679921 Homo sapiens Tumor necrosis factor receptor superfamily member 21 Proteins 0.000 description 2
- 101000611023 Homo sapiens Tumor necrosis factor receptor superfamily member 6 Proteins 0.000 description 2
- 101000597785 Homo sapiens Tumor necrosis factor receptor superfamily member 6B Proteins 0.000 description 2
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 2
- 108091006905 Human Serum Albumin Proteins 0.000 description 2
- 102000008100 Human Serum Albumin Human genes 0.000 description 2
- 101710093458 ICOS ligand Proteins 0.000 description 2
- XDXDZDZNSLXDNA-TZNDIEGXSA-N Idarubicin Chemical compound C1[C@H](N)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XDXDZDZNSLXDNA-TZNDIEGXSA-N 0.000 description 2
- XDXDZDZNSLXDNA-UHFFFAOYSA-N Idarubicin Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XDXDZDZNSLXDNA-UHFFFAOYSA-N 0.000 description 2
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 description 2
- 102000037982 Immune checkpoint proteins Human genes 0.000 description 2
- 108091008036 Immune checkpoint proteins Proteins 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 description 2
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 description 2
- 102000006992 Interferon-alpha Human genes 0.000 description 2
- 108010047761 Interferon-alpha Proteins 0.000 description 2
- 102000014150 Interferons Human genes 0.000 description 2
- 108010050904 Interferons Proteins 0.000 description 2
- 208000007766 Kaposi sarcoma Diseases 0.000 description 2
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 2
- 102000017578 LAG3 Human genes 0.000 description 2
- 101150030213 Lag3 gene Proteins 0.000 description 2
- 240000007472 Leucaena leucocephala Species 0.000 description 2
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 2
- 102100020943 Leukocyte-associated immunoglobulin-like receptor 1 Human genes 0.000 description 2
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 2
- 102000008072 Lymphokines Human genes 0.000 description 2
- 108010074338 Lymphokines Proteins 0.000 description 2
- 206010025323 Lymphomas Diseases 0.000 description 2
- 101150039798 MYC gene Proteins 0.000 description 2
- 201000005505 Measles Diseases 0.000 description 2
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 2
- 206010027406 Mesothelioma Diseases 0.000 description 2
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 2
- 208000005647 Mumps Diseases 0.000 description 2
- 101100207071 Mus musculus Tnfsf8 gene Proteins 0.000 description 2
- 101000597780 Mus musculus Tumor necrosis factor ligand superfamily member 18 Proteins 0.000 description 2
- 102100035487 Nectin-3 Human genes 0.000 description 2
- 108010032605 Nerve Growth Factor Receptors Proteins 0.000 description 2
- 102000004473 OX40 Ligand Human genes 0.000 description 2
- 108010042215 OX40 Ligand Proteins 0.000 description 2
- 208000000474 Poliomyelitis Diseases 0.000 description 2
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 2
- 229920001214 Polysorbate 60 Polymers 0.000 description 2
- 239000004372 Polyvinyl alcohol Substances 0.000 description 2
- 102100033237 Pro-epidermal growth factor Human genes 0.000 description 2
- 102000007327 Protamines Human genes 0.000 description 2
- 108010007568 Protamines Proteins 0.000 description 2
- LCTONWCANYUPML-UHFFFAOYSA-M Pyruvate Chemical compound CC(=O)C([O-])=O LCTONWCANYUPML-UHFFFAOYSA-M 0.000 description 2
- 102000014128 RANK Ligand Human genes 0.000 description 2
- 108010025832 RANK Ligand Proteins 0.000 description 2
- 102000018795 RELT Human genes 0.000 description 2
- 108010052562 RELT Proteins 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 0.000 description 2
- 208000006265 Renal cell carcinoma Diseases 0.000 description 2
- 206010039491 Sarcoma Diseases 0.000 description 2
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 2
- NWGKJDSIEKMTRX-AAZCQSIUSA-N Sorbitan monooleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O NWGKJDSIEKMTRX-AAZCQSIUSA-N 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 2
- 102100039367 T-cell immunoglobulin and mucin domain-containing protein 4 Human genes 0.000 description 2
- 101710174757 T-cell immunoglobulin and mucin domain-containing protein 4 Proteins 0.000 description 2
- 229940126547 T-cell immunoglobulin mucin-3 Drugs 0.000 description 2
- 102100024834 T-cell immunoreceptor with Ig and ITIM domains Human genes 0.000 description 2
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 2
- 108700012920 TNF Proteins 0.000 description 2
- 102000016946 TWEAK Receptor Human genes 0.000 description 2
- 108010014401 TWEAK Receptor Proteins 0.000 description 2
- 208000033133 Testicular seminomatous germ cell tumor Diseases 0.000 description 2
- 102100025946 Transforming growth factor beta activator LRRC32 Human genes 0.000 description 2
- 101710169732 Transforming growth factor beta activator LRRC32 Proteins 0.000 description 2
- 102100026224 Transmembrane and immunoglobulin domain-containing protein 2 Human genes 0.000 description 2
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 2
- 102100024584 Tumor necrosis factor ligand superfamily member 12 Human genes 0.000 description 2
- 101710097155 Tumor necrosis factor ligand superfamily member 12 Proteins 0.000 description 2
- 102100035283 Tumor necrosis factor ligand superfamily member 18 Human genes 0.000 description 2
- 102100026890 Tumor necrosis factor ligand superfamily member 4 Human genes 0.000 description 2
- 102100031988 Tumor necrosis factor ligand superfamily member 6 Human genes 0.000 description 2
- 102100032100 Tumor necrosis factor ligand superfamily member 8 Human genes 0.000 description 2
- 102100040115 Tumor necrosis factor receptor superfamily member 10C Human genes 0.000 description 2
- 102100040110 Tumor necrosis factor receptor superfamily member 10D Human genes 0.000 description 2
- 102100032236 Tumor necrosis factor receptor superfamily member 11B Human genes 0.000 description 2
- 102100029675 Tumor necrosis factor receptor superfamily member 13B Human genes 0.000 description 2
- 102100029690 Tumor necrosis factor receptor superfamily member 13C Human genes 0.000 description 2
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 2
- 102100033725 Tumor necrosis factor receptor superfamily member 16 Human genes 0.000 description 2
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 description 2
- 102100033760 Tumor necrosis factor receptor superfamily member 19 Human genes 0.000 description 2
- 102100033732 Tumor necrosis factor receptor superfamily member 1A Human genes 0.000 description 2
- 101710187743 Tumor necrosis factor receptor superfamily member 1A Proteins 0.000 description 2
- 102100033733 Tumor necrosis factor receptor superfamily member 1B Human genes 0.000 description 2
- 101710187830 Tumor necrosis factor receptor superfamily member 1B Proteins 0.000 description 2
- 102100022205 Tumor necrosis factor receptor superfamily member 21 Human genes 0.000 description 2
- 102100035284 Tumor necrosis factor receptor superfamily member 6B Human genes 0.000 description 2
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 2
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 2
- 229940122803 Vinca alkaloid Drugs 0.000 description 2
- WERKSKAQRVDLDW-ANOHMWSOSA-N [(2s,3r,4r,5r)-2,3,4,5,6-pentahydroxyhexyl] (z)-octadec-9-enoate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO WERKSKAQRVDLDW-ANOHMWSOSA-N 0.000 description 2
- 239000000205 acacia gum Substances 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 2
- 239000011149 active material Substances 0.000 description 2
- 239000002487 adenosine deaminase inhibitor Substances 0.000 description 2
- 210000004100 adrenal gland Anatomy 0.000 description 2
- 125000005907 alkyl ester group Chemical group 0.000 description 2
- 229940045714 alkyl sulfonate alkylating agent Drugs 0.000 description 2
- 150000008052 alkyl sulfonates Chemical class 0.000 description 2
- 229940100198 alkylating agent Drugs 0.000 description 2
- 239000002168 alkylating agent Substances 0.000 description 2
- 125000002947 alkylene group Chemical group 0.000 description 2
- 229940087168 alpha tocopherol Drugs 0.000 description 2
- HFHDHCJBZVLPGP-RWMJIURBSA-N alpha-cyclodextrin Chemical compound OC[C@H]([C@H]([C@@H]([C@H]1O)O)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)CO)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1CO HFHDHCJBZVLPGP-RWMJIURBSA-N 0.000 description 2
- 229940043377 alpha-cyclodextrin Drugs 0.000 description 2
- PNEYBMLMFCGWSK-UHFFFAOYSA-N aluminium oxide Inorganic materials [O-2].[O-2].[O-2].[Al+3].[Al+3] PNEYBMLMFCGWSK-UHFFFAOYSA-N 0.000 description 2
- 235000012211 aluminium silicate Nutrition 0.000 description 2
- CEGOLXSVJUTHNZ-UHFFFAOYSA-K aluminium tristearate Chemical compound [Al+3].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O CEGOLXSVJUTHNZ-UHFFFAOYSA-K 0.000 description 2
- 229940063655 aluminum stearate Drugs 0.000 description 2
- 229960002932 anastrozole Drugs 0.000 description 2
- YBBLVLTVTVSKRW-UHFFFAOYSA-N anastrozole Chemical compound N#CC(C)(C)C1=CC(C(C)(C#N)C)=CC(CN2N=CN=C2)=C1 YBBLVLTVTVSKRW-UHFFFAOYSA-N 0.000 description 2
- 230000003388 anti-hormonal effect Effects 0.000 description 2
- 230000003127 anti-melanomic effect Effects 0.000 description 2
- 230000000340 anti-metabolite Effects 0.000 description 2
- 230000000118 anti-neoplastic effect Effects 0.000 description 2
- 230000002155 anti-virotic effect Effects 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 229940124691 antibody therapeutics Drugs 0.000 description 2
- 229940100197 antimetabolite Drugs 0.000 description 2
- 239000002256 antimetabolite Substances 0.000 description 2
- 229940045719 antineoplastic alkylating agent nitrosoureas Drugs 0.000 description 2
- 239000003443 antiviral agent Substances 0.000 description 2
- 210000001367 artery Anatomy 0.000 description 2
- 229960005070 ascorbic acid Drugs 0.000 description 2
- 239000000605 aspartame Substances 0.000 description 2
- IAOZJIPTCAWIRG-QWRGUYRKSA-N aspartame Chemical compound OC(=O)C[C@H](N)C(=O)N[C@H](C(=O)OC)CC1=CC=CC=C1 IAOZJIPTCAWIRG-QWRGUYRKSA-N 0.000 description 2
- 235000010357 aspartame Nutrition 0.000 description 2
- 229960003438 aspartame Drugs 0.000 description 2
- 239000000305 astragalus gummifer gum Substances 0.000 description 2
- 238000011914 asymmetric synthesis Methods 0.000 description 2
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 2
- 235000013871 bee wax Nutrition 0.000 description 2
- 239000012166 beeswax Substances 0.000 description 2
- 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 2
- 229960001561 bleomycin Drugs 0.000 description 2
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 210000004556 brain Anatomy 0.000 description 2
- 210000000481 breast Anatomy 0.000 description 2
- 229960002092 busulfan Drugs 0.000 description 2
- KVUAALJSMIVURS-ZEDZUCNESA-L calcium folinate Chemical compound [Ca+2].C1NC=2NC(N)=NC(=O)C=2N(C=O)C1CNC1=CC=C(C(=O)N[C@@H](CCC([O-])=O)C([O-])=O)C=C1 KVUAALJSMIVURS-ZEDZUCNESA-L 0.000 description 2
- 159000000007 calcium salts Chemical class 0.000 description 2
- 208000035269 cancer or benign tumor Diseases 0.000 description 2
- 238000009566 cancer vaccine Methods 0.000 description 2
- 229960004117 capecitabine Drugs 0.000 description 2
- GGNALUCSASGNCK-UHFFFAOYSA-N carbon dioxide;propan-2-ol Chemical compound O=C=O.CC(C)O GGNALUCSASGNCK-UHFFFAOYSA-N 0.000 description 2
- 229960004562 carboplatin Drugs 0.000 description 2
- 230000012292 cell migration Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 229920006217 cellulose acetate butyrate Polymers 0.000 description 2
- 210000003679 cervix uteri Anatomy 0.000 description 2
- 229940082500 cetostearyl alcohol Drugs 0.000 description 2
- 229960000541 cetyl alcohol Drugs 0.000 description 2
- 229940044683 chemotherapy drug Drugs 0.000 description 2
- 229960004630 chlorambucil Drugs 0.000 description 2
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 2
- 239000003240 coconut oil Substances 0.000 description 2
- 235000019864 coconut oil Nutrition 0.000 description 2
- 239000008119 colloidal silica Substances 0.000 description 2
- 210000001072 colon Anatomy 0.000 description 2
- 229940125797 compound 12 Drugs 0.000 description 2
- 210000002808 connective tissue Anatomy 0.000 description 2
- 238000013270 controlled release Methods 0.000 description 2
- 239000002285 corn oil Substances 0.000 description 2
- 235000005687 corn oil Nutrition 0.000 description 2
- 239000008120 corn starch Substances 0.000 description 2
- 239000006184 cosolvent Substances 0.000 description 2
- 230000000139 costimulatory effect Effects 0.000 description 2
- 235000012343 cottonseed oil Nutrition 0.000 description 2
- 239000002385 cottonseed oil Substances 0.000 description 2
- 229960004397 cyclophosphamide Drugs 0.000 description 2
- 229960000684 cytarabine Drugs 0.000 description 2
- GVJHHUAWPYXKBD-UHFFFAOYSA-N d-alpha-tocopherol Natural products OC1=C(C)C(C)=C2OC(CCCC(C)CCCC(C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-UHFFFAOYSA-N 0.000 description 2
- 229960000640 dactinomycin Drugs 0.000 description 2
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 2
- 229960000975 daunorubicin Drugs 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- CFCUWKMKBJTWLW-UHFFFAOYSA-N deoliosyl-3C-alpha-L-digitoxosyl-MTM Natural products CC=1C(O)=C2C(O)=C3C(=O)C(OC4OC(C)C(O)C(OC5OC(C)C(O)C(OC6OC(C)C(O)C(C)(O)C6)C5)C4)C(C(OC)C(=O)C(O)C(C)O)CC3=CC2=CC=1OC(OC(C)C1O)CC1OC1CC(O)C(O)C(C)O1 CFCUWKMKBJTWLW-UHFFFAOYSA-N 0.000 description 2
- 239000008121 dextrose Substances 0.000 description 2
- YMWUJEATGCHHMB-UHFFFAOYSA-N dichloromethane Natural products ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 2
- GXGAKHNRMVGRPK-UHFFFAOYSA-N dimagnesium;dioxido-bis[[oxido(oxo)silyl]oxy]silane Chemical compound [Mg+2].[Mg+2].[O-][Si](=O)O[Si]([O-])([O-])O[Si]([O-])=O GXGAKHNRMVGRPK-UHFFFAOYSA-N 0.000 description 2
- ZPWVASYFFYYZEW-UHFFFAOYSA-L dipotassium hydrogen phosphate Chemical compound [K+].[K+].OP([O-])([O-])=O ZPWVASYFFYYZEW-UHFFFAOYSA-L 0.000 description 2
- 229910000396 dipotassium phosphate Inorganic materials 0.000 description 2
- 235000019797 dipotassium phosphate Nutrition 0.000 description 2
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 2
- 239000006185 dispersion Substances 0.000 description 2
- 239000003534 dna topoisomerase inhibitor Substances 0.000 description 2
- 229960004679 doxorubicin Drugs 0.000 description 2
- 238000012377 drug delivery Methods 0.000 description 2
- 239000003596 drug target Substances 0.000 description 2
- 239000003792 electrolyte Substances 0.000 description 2
- 239000003974 emollient agent Substances 0.000 description 2
- 230000001804 emulsifying effect Effects 0.000 description 2
- 239000008387 emulsifying waxe Substances 0.000 description 2
- 210000004696 endometrium Anatomy 0.000 description 2
- 239000003623 enhancer Substances 0.000 description 2
- 239000002702 enteric coating Substances 0.000 description 2
- 238000009505 enteric coating Methods 0.000 description 2
- 229960001904 epirubicin Drugs 0.000 description 2
- 210000003238 esophagus Anatomy 0.000 description 2
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 2
- 235000019325 ethyl cellulose Nutrition 0.000 description 2
- 229920001249 ethyl cellulose Polymers 0.000 description 2
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 2
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 2
- 229960005420 etoposide Drugs 0.000 description 2
- 230000029142 excretion Effects 0.000 description 2
- ODKNJVUHOIMIIZ-RRKCRQDMSA-N floxuridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ODKNJVUHOIMIIZ-RRKCRQDMSA-N 0.000 description 2
- 229960000961 floxuridine Drugs 0.000 description 2
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 2
- 229960005304 fludarabine phosphate Drugs 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 229960002949 fluorouracil Drugs 0.000 description 2
- 239000004052 folic acid antagonist Substances 0.000 description 2
- 235000008191 folinic acid Nutrition 0.000 description 2
- 239000011672 folinic acid Substances 0.000 description 2
- 238000004108 freeze drying Methods 0.000 description 2
- GDSRMADSINPKSL-HSEONFRVSA-N gamma-cyclodextrin Chemical compound OC[C@H]([C@H]([C@@H]([C@H]1O)O)O[C@H]2O[C@@H]([C@@H](O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O[C@H]3O[C@H](CO)[C@H]([C@@H]([C@H]3O)O)O3)[C@H](O)[C@H]2O)CO)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H]3O[C@@H]1CO GDSRMADSINPKSL-HSEONFRVSA-N 0.000 description 2
- 229940080345 gamma-cyclodextrin Drugs 0.000 description 2
- 206010017758 gastric cancer Diseases 0.000 description 2
- 208000010749 gastric carcinoma Diseases 0.000 description 2
- 210000001035 gastrointestinal tract Anatomy 0.000 description 2
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 2
- 229960005277 gemcitabine Drugs 0.000 description 2
- 208000005017 glioblastoma Diseases 0.000 description 2
- 238000003881 globally optimized alternating phase rectangular pulse Methods 0.000 description 2
- 125000005456 glyceride group Chemical group 0.000 description 2
- 229940074045 glyceryl distearate Drugs 0.000 description 2
- 229940075507 glyceryl monostearate Drugs 0.000 description 2
- 229960002449 glycine Drugs 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 239000003966 growth inhibitor Substances 0.000 description 2
- LHGVFZTZFXWLCP-UHFFFAOYSA-N guaiacol Chemical class COC1=CC=CC=C1O LHGVFZTZFXWLCP-UHFFFAOYSA-N 0.000 description 2
- 230000003394 haemopoietic effect Effects 0.000 description 2
- 239000007902 hard capsule Substances 0.000 description 2
- 210000003128 head Anatomy 0.000 description 2
- 229940022353 herceptin Drugs 0.000 description 2
- UBHWBODXJBSFLH-UHFFFAOYSA-N hexadecan-1-ol;octadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCO.CCCCCCCCCCCCCCCCCCO UBHWBODXJBSFLH-UHFFFAOYSA-N 0.000 description 2
- FBPFZTCFMRRESA-UHFFFAOYSA-N hexane-1,2,3,4,5,6-hexol Chemical compound OCC(O)C(O)C(O)C(O)CO FBPFZTCFMRRESA-UHFFFAOYSA-N 0.000 description 2
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 2
- 230000002977 hyperthermial effect Effects 0.000 description 2
- 229960000908 idarubicin Drugs 0.000 description 2
- 229960001101 ifosfamide Drugs 0.000 description 2
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 239000000367 immunologic factor Substances 0.000 description 2
- 229960003444 immunosuppressant agent Drugs 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 238000009169 immunotherapy Methods 0.000 description 2
- 239000003701 inert diluent Substances 0.000 description 2
- 108091008042 inhibitory receptors Proteins 0.000 description 2
- 229940102223 injectable solution Drugs 0.000 description 2
- 229940102213 injectable suspension Drugs 0.000 description 2
- 229940047124 interferons Drugs 0.000 description 2
- 238000000185 intracerebroventricular administration Methods 0.000 description 2
- 150000002500 ions Chemical class 0.000 description 2
- UWKQSNNFCGGAFS-XIFFEERXSA-N irinotecan Chemical compound C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 UWKQSNNFCGGAFS-XIFFEERXSA-N 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 230000003907 kidney function Effects 0.000 description 2
- 229960003881 letrozole Drugs 0.000 description 2
- HPJKCIUCZWXJDR-UHFFFAOYSA-N letrozole Chemical compound C1=CC(C#N)=CC=C1C(N1N=CN=C1)C1=CC=C(C#N)C=C1 HPJKCIUCZWXJDR-UHFFFAOYSA-N 0.000 description 2
- 229960001691 leucovorin Drugs 0.000 description 2
- 208000032839 leukemia Diseases 0.000 description 2
- 210000000265 leukocyte Anatomy 0.000 description 2
- 229950011263 lirilumab Drugs 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 230000003908 liver function Effects 0.000 description 2
- 229960002247 lomustine Drugs 0.000 description 2
- 239000007937 lozenge Substances 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 210000001165 lymph node Anatomy 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 239000000395 magnesium oxide Substances 0.000 description 2
- CPLXHLVBOLITMK-UHFFFAOYSA-N magnesium oxide Inorganic materials [Mg]=O CPLXHLVBOLITMK-UHFFFAOYSA-N 0.000 description 2
- 239000000391 magnesium silicate Substances 0.000 description 2
- 229910000386 magnesium trisilicate Inorganic materials 0.000 description 2
- 235000019793 magnesium trisilicate Nutrition 0.000 description 2
- 229940099273 magnesium trisilicate Drugs 0.000 description 2
- AXZKOIWUVFPNLO-UHFFFAOYSA-N magnesium;oxygen(2-) Chemical compound [O-2].[Mg+2] AXZKOIWUVFPNLO-UHFFFAOYSA-N 0.000 description 2
- 230000000873 masking effect Effects 0.000 description 2
- 229960004961 mechlorethamine Drugs 0.000 description 2
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 2
- 229960001428 mercaptopurine Drugs 0.000 description 2
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 2
- 229960000485 methotrexate Drugs 0.000 description 2
- 229940016286 microcrystalline cellulose Drugs 0.000 description 2
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 2
- 239000008108 microcrystalline cellulose Substances 0.000 description 2
- CFCUWKMKBJTWLW-BKHRDMLASA-N mithramycin Chemical compound O([C@@H]1C[C@@H](O[C@H](C)[C@H]1O)OC=1C=C2C=C3C[C@H]([C@@H](C(=O)C3=C(O)C2=C(O)C=1C)O[C@@H]1O[C@H](C)[C@@H](O)[C@H](O[C@@H]2O[C@H](C)[C@H](O)[C@H](O[C@@H]3O[C@H](C)[C@@H](O)[C@@](C)(O)C3)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@@H](O)[C@H](O)[C@@H](C)O1 CFCUWKMKBJTWLW-BKHRDMLASA-N 0.000 description 2
- 229960004857 mitomycin Drugs 0.000 description 2
- 229960001156 mitoxantrone Drugs 0.000 description 2
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 210000004400 mucous membrane Anatomy 0.000 description 2
- 208000010805 mumps infectious disease Diseases 0.000 description 2
- 210000003205 muscle Anatomy 0.000 description 2
- 229940043348 myristyl alcohol Drugs 0.000 description 2
- 239000006070 nanosuspension Substances 0.000 description 2
- 229930014626 natural product Natural products 0.000 description 2
- 210000003739 neck Anatomy 0.000 description 2
- 238000013546 non-drug therapy Methods 0.000 description 2
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 2
- 239000012457 nonaqueous media Substances 0.000 description 2
- GYCKQBWUSACYIF-UHFFFAOYSA-N o-hydroxybenzoic acid ethyl ester Natural products CCOC(=O)C1=CC=CC=C1O GYCKQBWUSACYIF-UHFFFAOYSA-N 0.000 description 2
- 239000007764 o/w emulsion Substances 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- 239000003883 ointment base Substances 0.000 description 2
- 230000006508 oncogene activation Effects 0.000 description 2
- 239000007935 oral tablet Substances 0.000 description 2
- 210000001672 ovary Anatomy 0.000 description 2
- 210000000496 pancreas Anatomy 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 229960002340 pentostatin Drugs 0.000 description 2
- 230000010412 perfusion Effects 0.000 description 2
- 239000008177 pharmaceutical agent Substances 0.000 description 2
- 239000008024 pharmaceutical diluent Substances 0.000 description 2
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 2
- XHXFXVLFKHQFAL-UHFFFAOYSA-N phosphoryl trichloride Chemical compound ClP(Cl)(Cl)=O XHXFXVLFKHQFAL-UHFFFAOYSA-N 0.000 description 2
- 239000002504 physiological saline solution Substances 0.000 description 2
- 229960000952 pipobroman Drugs 0.000 description 2
- NJBFOOCLYDNZJN-UHFFFAOYSA-N pipobroman Chemical compound BrCCC(=O)N1CCN(C(=O)CCBr)CC1 NJBFOOCLYDNZJN-UHFFFAOYSA-N 0.000 description 2
- 229910052697 platinum Inorganic materials 0.000 description 2
- 229960003171 plicamycin Drugs 0.000 description 2
- 229920000058 polyacrylate Polymers 0.000 description 2
- 229920000573 polyethylene Polymers 0.000 description 2
- 229940113116 polyethylene glycol 1000 Drugs 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 2
- 229920000136 polysorbate Polymers 0.000 description 2
- 229920002451 polyvinyl alcohol Polymers 0.000 description 2
- 229910000027 potassium carbonate Inorganic materials 0.000 description 2
- 235000010241 potassium sorbate Nutrition 0.000 description 2
- 239000004302 potassium sorbate Substances 0.000 description 2
- 229940069338 potassium sorbate Drugs 0.000 description 2
- 239000002243 precursor Substances 0.000 description 2
- 238000002953 preparative HPLC Methods 0.000 description 2
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 2
- 229960000624 procarbazine Drugs 0.000 description 2
- 235000013772 propylene glycol Nutrition 0.000 description 2
- 210000002307 prostate Anatomy 0.000 description 2
- 229950008679 protamine sulfate Drugs 0.000 description 2
- 150000003212 purines Chemical class 0.000 description 2
- 150000003230 pyrimidines Chemical class 0.000 description 2
- 230000003439 radiotherapeutic effect Effects 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 210000000664 rectum Anatomy 0.000 description 2
- CVHZOJJKTDOEJC-UHFFFAOYSA-N saccharin Chemical compound C1=CC=C2C(=O)NS(=O)(=O)C2=C1 CVHZOJJKTDOEJC-UHFFFAOYSA-N 0.000 description 2
- 235000019204 saccharin Nutrition 0.000 description 2
- 229940081974 saccharin Drugs 0.000 description 2
- 239000000901 saccharin and its Na,K and Ca salt Substances 0.000 description 2
- 229920006395 saturated elastomer Polymers 0.000 description 2
- HFHDHCJBZVLPGP-UHFFFAOYSA-N schardinger α-dextrin Chemical compound O1C(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC(C(O)C2O)C(CO)OC2OC(C(C2O)O)C(CO)OC2OC2C(O)C(O)C1OC2CO HFHDHCJBZVLPGP-UHFFFAOYSA-N 0.000 description 2
- 238000007789 sealing Methods 0.000 description 2
- 239000008299 semisolid dosage form Substances 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 210000002966 serum Anatomy 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 102000030938 small GTPase Human genes 0.000 description 2
- 208000000587 small cell lung carcinoma Diseases 0.000 description 2
- 229940126586 small molecule drug Drugs 0.000 description 2
- 229910001467 sodium calcium phosphate Inorganic materials 0.000 description 2
- 229910000029 sodium carbonate Inorganic materials 0.000 description 2
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 2
- 239000007901 soft capsule Substances 0.000 description 2
- 239000007909 solid dosage form Substances 0.000 description 2
- 235000010199 sorbic acid Nutrition 0.000 description 2
- 239000004334 sorbic acid Substances 0.000 description 2
- 229940075582 sorbic acid Drugs 0.000 description 2
- 229940083466 soybean lecithin Drugs 0.000 description 2
- 239000003549 soybean oil Substances 0.000 description 2
- 235000012424 soybean oil Nutrition 0.000 description 2
- 239000007921 spray Substances 0.000 description 2
- 238000010922 spray-dried dispersion Methods 0.000 description 2
- 230000006641 stabilisation Effects 0.000 description 2
- 238000011105 stabilization Methods 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 230000001954 sterilising effect Effects 0.000 description 2
- 238000004659 sterilization and disinfection Methods 0.000 description 2
- 210000002784 stomach Anatomy 0.000 description 2
- 201000000498 stomach carcinoma Diseases 0.000 description 2
- 239000012089 stop solution Substances 0.000 description 2
- 229960001052 streptozocin Drugs 0.000 description 2
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 2
- 125000001424 substituent group Chemical group 0.000 description 2
- 229960004793 sucrose Drugs 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N sulfuric acid group Chemical class S(O)(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 238000001356 surgical procedure Methods 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 238000013268 sustained release Methods 0.000 description 2
- 239000012730 sustained-release form Substances 0.000 description 2
- 229960001603 tamoxifen Drugs 0.000 description 2
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 2
- 229960001674 tegafur Drugs 0.000 description 2
- WFWLQNSHRPWKFK-ZCFIWIBFSA-N tegafur Chemical compound O=C1NC(=O)C(F)=CN1[C@@H]1OCCC1 WFWLQNSHRPWKFK-ZCFIWIBFSA-N 0.000 description 2
- 229960001278 teniposide Drugs 0.000 description 2
- 208000024662 testicular seminoma Diseases 0.000 description 2
- 210000001550 testis Anatomy 0.000 description 2
- WYURNTSHIVDZCO-UHFFFAOYSA-N tetrahydrofuran Substances C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 2
- OULAJFUGPPVRBK-UHFFFAOYSA-N tetratriacontyl alcohol Natural products CCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCO OULAJFUGPPVRBK-UHFFFAOYSA-N 0.000 description 2
- 230000004797 therapeutic response Effects 0.000 description 2
- 239000002562 thickening agent Substances 0.000 description 2
- 210000001685 thyroid gland Anatomy 0.000 description 2
- 229960003087 tioguanine Drugs 0.000 description 2
- 229940044693 topoisomerase inhibitor Drugs 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 229960000575 trastuzumab Drugs 0.000 description 2
- 229950001353 tretamine Drugs 0.000 description 2
- IUCJMVBFZDHPDX-UHFFFAOYSA-N tretamine Chemical compound C1CN1C1=NC(N2CC2)=NC(N2CC2)=N1 IUCJMVBFZDHPDX-UHFFFAOYSA-N 0.000 description 2
- 150000004654 triazenes Chemical class 0.000 description 2
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 2
- 229940121358 tyrosine kinase inhibitor Drugs 0.000 description 2
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 2
- 229910021642 ultra pure water Inorganic materials 0.000 description 2
- 239000012498 ultrapure water Substances 0.000 description 2
- 229960001055 uracil mustard Drugs 0.000 description 2
- 210000003932 urinary bladder Anatomy 0.000 description 2
- 235000013311 vegetables Nutrition 0.000 description 2
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 2
- 229960004528 vincristine Drugs 0.000 description 2
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 2
- UGGWPQSBPIFKDZ-KOTLKJBCSA-N vindesine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(N)=O)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1N=C1[C]2C=CC=C1 UGGWPQSBPIFKDZ-KOTLKJBCSA-N 0.000 description 2
- 229960004355 vindesine Drugs 0.000 description 2
- 229960004854 viral vaccine Drugs 0.000 description 2
- 210000001835 viscera Anatomy 0.000 description 2
- 238000009736 wetting Methods 0.000 description 2
- 210000002268 wool Anatomy 0.000 description 2
- 239000012224 working solution Substances 0.000 description 2
- 150000003751 zinc Chemical class 0.000 description 2
- UAYWVJHJZHQCIE-UHFFFAOYSA-L zinc iodide Chemical compound I[Zn]I UAYWVJHJZHQCIE-UHFFFAOYSA-L 0.000 description 2
- ZGGHKIMDNBDHJB-NRFPMOEYSA-M (3R,5S)-fluvastatin sodium Chemical compound [Na+].C12=CC=CC=C2N(C(C)C)C(\C=C\[C@@H](O)C[C@@H](O)CC([O-])=O)=C1C1=CC=C(F)C=C1 ZGGHKIMDNBDHJB-NRFPMOEYSA-M 0.000 description 1
- GHOKWGTUZJEAQD-ZETCQYMHSA-N (D)-(+)-Pantothenic acid Chemical compound OCC(C)(C)[C@@H](O)C(=O)NCCC(O)=O GHOKWGTUZJEAQD-ZETCQYMHSA-N 0.000 description 1
- SCYULBFZEHDVBN-UHFFFAOYSA-N 1,1-Dichloroethane Chemical compound CC(Cl)Cl SCYULBFZEHDVBN-UHFFFAOYSA-N 0.000 description 1
- MICMHFIQSAMEJG-UHFFFAOYSA-N 1-bromopyrrolidine-2,5-dione Chemical compound BrN1C(=O)CCC1=O.BrN1C(=O)CCC1=O MICMHFIQSAMEJG-UHFFFAOYSA-N 0.000 description 1
- HYGDHSYCSJKRFX-UHFFFAOYSA-N 1h-cyclopenta[b][1]benzofuran Chemical class O1C2=CC=CC=C2C2=C1C=CC2 HYGDHSYCSJKRFX-UHFFFAOYSA-N 0.000 description 1
- WXTMDXOMEHJXQO-UHFFFAOYSA-N 2,5-dihydroxybenzoic acid Chemical compound OC(=O)C1=CC(O)=CC=C1O WXTMDXOMEHJXQO-UHFFFAOYSA-N 0.000 description 1
- SGUAFYQXFOLMHL-UHFFFAOYSA-N 2-hydroxy-5-{1-hydroxy-2-[(4-phenylbutan-2-yl)amino]ethyl}benzamide Chemical compound C=1C=C(O)C(C(N)=O)=CC=1C(O)CNC(C)CCC1=CC=CC=C1 SGUAFYQXFOLMHL-UHFFFAOYSA-N 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-M 3-carboxy-2,3-dihydroxypropanoate Chemical compound OC(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-M 0.000 description 1
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 1
- DKVRNHPCAOHRSI-KQYNXXCUSA-N 7-methyl-GTP Chemical compound C1=2N=C(N)NC(=O)C=2[N+](C)=CN1[C@@H]1O[C@H](COP(O)(=O)OP(O)(=O)OP(O)([O-])=O)[C@@H](O)[C@H]1O DKVRNHPCAOHRSI-KQYNXXCUSA-N 0.000 description 1
- 108091006112 ATPases Proteins 0.000 description 1
- 101001082110 Acanthamoeba polyphaga mimivirus Eukaryotic translation initiation factor 4E homolog Proteins 0.000 description 1
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 1
- 208000030090 Acute Disease Diseases 0.000 description 1
- 208000005641 Adenomyosis Diseases 0.000 description 1
- 102000057290 Adenosine Triphosphatases Human genes 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 206010048610 Cardiotoxicity Diseases 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- 208000017667 Chronic Disease Diseases 0.000 description 1
- DSLZVSRJTYRBFB-LLEIAEIESA-N D-glucaric acid Chemical compound OC(=O)[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C(O)=O DSLZVSRJTYRBFB-LLEIAEIESA-N 0.000 description 1
- RGHNJXZEOKUKBD-SQOUGZDYSA-M D-gluconate Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O RGHNJXZEOKUKBD-SQOUGZDYSA-M 0.000 description 1
- 108010033333 DEAD-box RNA Helicases Proteins 0.000 description 1
- 102000007120 DEAD-box RNA Helicases Human genes 0.000 description 1
- 101001082109 Danio rerio Eukaryotic translation initiation factor 4E-1B Proteins 0.000 description 1
- 102100022874 Dexamethasone-induced Ras-related protein 1 Human genes 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 239000006145 Eagle's minimal essential medium Substances 0.000 description 1
- 201000009273 Endometriosis Diseases 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 102000003782 Eukaryotic Initiation Factor-4F Human genes 0.000 description 1
- 108010057194 Eukaryotic Initiation Factor-4F Proteins 0.000 description 1
- 102000002241 Eukaryotic Initiation Factors Human genes 0.000 description 1
- 108010014863 Eukaryotic Initiation Factors Proteins 0.000 description 1
- 102100039950 Eukaryotic initiation factor 4A-I Human genes 0.000 description 1
- 102100022462 Eukaryotic initiation factor 4A-II Human genes 0.000 description 1
- 102100022461 Eukaryotic initiation factor 4A-III Human genes 0.000 description 1
- 101710091919 Eukaryotic translation initiation factor 4G Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- BDAGIHXWWSANSR-UHFFFAOYSA-M Formate Chemical compound [O-]C=O BDAGIHXWWSANSR-UHFFFAOYSA-M 0.000 description 1
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 1
- 102100037941 GTP-binding protein Di-Ras1 Human genes 0.000 description 1
- 102100037949 GTP-binding protein Di-Ras2 Human genes 0.000 description 1
- 102100037948 GTP-binding protein Di-Ras3 Human genes 0.000 description 1
- 102100033962 GTP-binding protein RAD Human genes 0.000 description 1
- 102100037880 GTP-binding protein REM 1 Human genes 0.000 description 1
- 102100027362 GTP-binding protein REM 2 Human genes 0.000 description 1
- 102100027988 GTP-binding protein Rhes Human genes 0.000 description 1
- 102100022871 GTPase ERas Human genes 0.000 description 1
- 101100382952 Homo sapiens CCND1 gene Proteins 0.000 description 1
- 101000620808 Homo sapiens Dexamethasone-induced Ras-related protein 1 Proteins 0.000 description 1
- 101000959666 Homo sapiens Eukaryotic initiation factor 4A-I Proteins 0.000 description 1
- 101001044475 Homo sapiens Eukaryotic initiation factor 4A-II Proteins 0.000 description 1
- 101001044466 Homo sapiens Eukaryotic initiation factor 4A-III Proteins 0.000 description 1
- 101000951240 Homo sapiens GTP-binding protein Di-Ras1 Proteins 0.000 description 1
- 101000951231 Homo sapiens GTP-binding protein Di-Ras2 Proteins 0.000 description 1
- 101000951235 Homo sapiens GTP-binding protein Di-Ras3 Proteins 0.000 description 1
- 101000856606 Homo sapiens GTP-binding protein GEM Proteins 0.000 description 1
- 101001132495 Homo sapiens GTP-binding protein RAD Proteins 0.000 description 1
- 101001095995 Homo sapiens GTP-binding protein REM 1 Proteins 0.000 description 1
- 101000581787 Homo sapiens GTP-binding protein REM 2 Proteins 0.000 description 1
- 101000578396 Homo sapiens GTP-binding protein Rhes Proteins 0.000 description 1
- 101000620820 Homo sapiens GTPase ERas Proteins 0.000 description 1
- 101001037191 Homo sapiens Hyaluronan synthase 1 Proteins 0.000 description 1
- 101000997257 Homo sapiens NF-kappa-B inhibitor-interacting Ras-like protein 1 Proteins 0.000 description 1
- 101000997252 Homo sapiens NF-kappa-B inhibitor-interacting Ras-like protein 2 Proteins 0.000 description 1
- 101000677111 Homo sapiens Ras-like protein family member 10A Proteins 0.000 description 1
- 101000677113 Homo sapiens Ras-like protein family member 10B Proteins 0.000 description 1
- 101000677110 Homo sapiens Ras-like protein family member 11A Proteins 0.000 description 1
- 101000700393 Homo sapiens Ras-like protein family member 11B Proteins 0.000 description 1
- 101001061889 Homo sapiens Ras-like protein family member 12 Proteins 0.000 description 1
- 101001061661 Homo sapiens Ras-related and estrogen-regulated growth inhibitor-like protein Proteins 0.000 description 1
- 101000744515 Homo sapiens Ras-related protein M-Ras Proteins 0.000 description 1
- 101000686246 Homo sapiens Ras-related protein R-Ras Proteins 0.000 description 1
- 101000686227 Homo sapiens Ras-related protein R-Ras2 Proteins 0.000 description 1
- 101001130465 Homo sapiens Ras-related protein Ral-A Proteins 0.000 description 1
- 101001130458 Homo sapiens Ras-related protein Ral-B Proteins 0.000 description 1
- 101000584600 Homo sapiens Ras-related protein Rap-1b Proteins 0.000 description 1
- 101001130441 Homo sapiens Ras-related protein Rap-2a Proteins 0.000 description 1
- 101001130437 Homo sapiens Ras-related protein Rap-2b Proteins 0.000 description 1
- 101001130433 Homo sapiens Ras-related protein Rap-2c Proteins 0.000 description 1
- 102100040203 Hyaluronan synthase 1 Human genes 0.000 description 1
- 102000004157 Hydrolases Human genes 0.000 description 1
- 108090000604 Hydrolases Proteins 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 108091027974 Mature messenger RNA Proteins 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 1
- 108091057508 Myc family Proteins 0.000 description 1
- 108010052185 Myotonin-Protein Kinase Proteins 0.000 description 1
- 102100022437 Myotonin-protein kinase Human genes 0.000 description 1
- PCLIMKBDDGJMGD-UHFFFAOYSA-N N-bromosuccinimide Substances BrN1C(=O)CCC1=O PCLIMKBDDGJMGD-UHFFFAOYSA-N 0.000 description 1
- 102100034306 NF-kappa-B inhibitor-interacting Ras-like protein 1 Human genes 0.000 description 1
- 102100034325 NF-kappa-B inhibitor-interacting Ras-like protein 2 Human genes 0.000 description 1
- 229910002651 NO3 Inorganic materials 0.000 description 1
- NHNBFGGVMKEFGY-UHFFFAOYSA-N Nitrate Chemical compound [O-][N+]([O-])=O NHNBFGGVMKEFGY-UHFFFAOYSA-N 0.000 description 1
- 229910019213 POCl3 Inorganic materials 0.000 description 1
- NFHFRUOZVGFOOS-UHFFFAOYSA-N Pd(PPh3)4 Substances [Pd].C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1.C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 NFHFRUOZVGFOOS-UHFFFAOYSA-N 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-L Phosphate ion(2-) Chemical compound OP([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-L 0.000 description 1
- 101710163354 Potassium voltage-gated channel subfamily H member 2 Proteins 0.000 description 1
- 102000052575 Proto-Oncogene Human genes 0.000 description 1
- 108700020978 Proto-Oncogene Proteins 0.000 description 1
- 102100033479 RAF proto-oncogene serine/threonine-protein kinase Human genes 0.000 description 1
- 102100021577 Ras-like protein family member 10A Human genes 0.000 description 1
- 102100021578 Ras-like protein family member 10B Human genes 0.000 description 1
- 102100021586 Ras-like protein family member 11A Human genes 0.000 description 1
- 102100029518 Ras-like protein family member 11B Human genes 0.000 description 1
- 102100029559 Ras-like protein family member 12 Human genes 0.000 description 1
- 102100028429 Ras-related and estrogen-regulated growth inhibitor Human genes 0.000 description 1
- 102100028428 Ras-related and estrogen-regulated growth inhibitor-like protein Human genes 0.000 description 1
- 102100039789 Ras-related protein M-Ras Human genes 0.000 description 1
- 102100024683 Ras-related protein R-Ras Human genes 0.000 description 1
- 102100025003 Ras-related protein R-Ras2 Human genes 0.000 description 1
- 102100031424 Ras-related protein Ral-A Human genes 0.000 description 1
- 102100031425 Ras-related protein Ral-B Human genes 0.000 description 1
- 102100030706 Ras-related protein Rap-1A Human genes 0.000 description 1
- 102100030705 Ras-related protein Rap-1b Human genes 0.000 description 1
- 102100031420 Ras-related protein Rap-2a Human genes 0.000 description 1
- 102100031421 Ras-related protein Rap-2b Human genes 0.000 description 1
- 102100031422 Ras-related protein Rap-2c Human genes 0.000 description 1
- 108700005075 Regulator Genes Proteins 0.000 description 1
- 108700008625 Reporter Genes Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- JLRGJRBPOGGCBT-UHFFFAOYSA-N Tolbutamide Chemical compound CCCCNC(=O)NS(=O)(=O)C1=CC=C(C)C=C1 JLRGJRBPOGGCBT-UHFFFAOYSA-N 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 108700005077 Viral Genes Proteins 0.000 description 1
- 101710101493 Viral myc transforming protein Proteins 0.000 description 1
- 150000008043 acidic salts Chemical class 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 150000001447 alkali salts Chemical class 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 150000001450 anions Chemical class 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 229940034982 antineoplastic agent Drugs 0.000 description 1
- 229940041181 antineoplastic drug Drugs 0.000 description 1
- 229940072107 ascorbate Drugs 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 229940077388 benzenesulfonate Drugs 0.000 description 1
- SRSXLGNVWSONIS-UHFFFAOYSA-M benzenesulfonate Chemical compound [O-]S(=O)(=O)C1=CC=CC=C1 SRSXLGNVWSONIS-UHFFFAOYSA-M 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- 150000004657 carbamic acid derivatives Chemical class 0.000 description 1
- 235000013877 carbamide Nutrition 0.000 description 1
- 125000004432 carbon atom Chemical group C* 0.000 description 1
- 150000004649 carbonic acid derivatives Chemical class 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 150000001733 carboxylic acid esters Chemical class 0.000 description 1
- 239000012659 cardioprotective agent Substances 0.000 description 1
- 230000003293 cardioprotective effect Effects 0.000 description 1
- 231100000259 cardiotoxicity Toxicity 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 230000004715 cellular signal transduction Effects 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 239000012069 chiral reagent Substances 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 229940001468 citrate Drugs 0.000 description 1
- 230000009137 competitive binding Effects 0.000 description 1
- 239000012043 crude product Substances 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- DEZRYPDIMOWBDS-UHFFFAOYSA-N dcm dichloromethane Chemical compound ClCCl.ClCCl DEZRYPDIMOWBDS-UHFFFAOYSA-N 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-M dihydrogenphosphate Chemical compound OP(O)([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-M 0.000 description 1
- JMRYOSQOYJBDOI-UHFFFAOYSA-N dilithium;di(propan-2-yl)azanide Chemical compound [Li+].CC(C)[N-]C(C)C.CC(C)N([Li])C(C)C JMRYOSQOYJBDOI-UHFFFAOYSA-N 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 229960001760 dimethyl sulfoxide Drugs 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 238000010864 dual luciferase reporter gene assay Methods 0.000 description 1
- 108010093366 eIF-4B Proteins 0.000 description 1
- 230000002500 effect on skin Effects 0.000 description 1
- 238000003821 enantio-separation Methods 0.000 description 1
- 201000009274 endometriosis of uterus Diseases 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- 150000002170 ethers Chemical class 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 210000004265 eukaryotic small ribosome subunit Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 238000001640 fractional crystallisation Methods 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 229940050410 gluconate Drugs 0.000 description 1
- 229930195712 glutamate Natural products 0.000 description 1
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical group O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 1
- 229940093915 gynecological organic acid Drugs 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 235000003642 hunger Nutrition 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-M hydrogensulfate Chemical compound OS([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-M 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 239000003999 initiator Substances 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 230000000266 injurious effect Effects 0.000 description 1
- 239000013038 irreversible inhibitor Substances 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- TWBYWOBDOCUKOW-UHFFFAOYSA-M isonicotinate Chemical compound [O-]C(=O)C1=CC=NC=C1 TWBYWOBDOCUKOW-UHFFFAOYSA-M 0.000 description 1
- 108091008792 l-Myc Proteins 0.000 description 1
- 229960001632 labetalol Drugs 0.000 description 1
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 1
- ZCSHNCUQKCANBX-UHFFFAOYSA-N lithium diisopropylamide Substances [Li+].CC(C)[N-]C(C)C ZCSHNCUQKCANBX-UHFFFAOYSA-N 0.000 description 1
- PNVKCUSCADAAMP-UHFFFAOYSA-M magnesium;methanolate;methyl carbonate Chemical compound [Mg+2].[O-]C.COC([O-])=O PNVKCUSCADAAMP-UHFFFAOYSA-M 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000003228 microsomal effect Effects 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 108091006026 monomeric small GTPases Proteins 0.000 description 1
- 230000000869 mutational effect Effects 0.000 description 1
- LLYKPZOWCPVRPD-UHFFFAOYSA-N n,n-dimethylpyridin-2-amine;n,n-dimethylpyridin-4-amine Chemical compound CN(C)C1=CC=NC=C1.CN(C)C1=CC=CC=N1 LLYKPZOWCPVRPD-UHFFFAOYSA-N 0.000 description 1
- 108091008800 n-Myc Proteins 0.000 description 1
- WOOWBQQQJXZGIE-UHFFFAOYSA-N n-ethyl-n-propan-2-ylpropan-2-amine Chemical compound CCN(C(C)C)C(C)C.CCN(C(C)C)C(C)C WOOWBQQQJXZGIE-UHFFFAOYSA-N 0.000 description 1
- 108700043045 nanoluc Proteins 0.000 description 1
- 230000035407 negative regulation of cell proliferation Effects 0.000 description 1
- 230000000324 neuroprotective effect Effects 0.000 description 1
- 238000006386 neutralization reaction Methods 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 231100000590 oncogenic Toxicity 0.000 description 1
- 230000002246 oncogenic effect Effects 0.000 description 1
- 230000005959 oncogenic signaling Effects 0.000 description 1
- 102000027450 oncoproteins Human genes 0.000 description 1
- 108091008819 oncoproteins Proteins 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- WLJNZVDCPSBLRP-UHFFFAOYSA-L pamoate(2-) Chemical class C1=CC=C2C(CC3=C4C=CC=CC4=CC(=C3O)C([O-])=O)=C(O)C(C([O-])=O)=CC2=C1 WLJNZVDCPSBLRP-UHFFFAOYSA-L 0.000 description 1
- 229940014662 pantothenate Drugs 0.000 description 1
- 235000019161 pantothenic acid Nutrition 0.000 description 1
- 239000011713 pantothenic acid Substances 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 235000015320 potassium carbonate Nutrition 0.000 description 1
- 244000144977 poultry Species 0.000 description 1
- 238000003672 processing method Methods 0.000 description 1
- 230000006920 protein precipitation Effects 0.000 description 1
- 108010077182 raf Kinases Proteins 0.000 description 1
- 102000009929 raf Kinases Human genes 0.000 description 1
- 108010036805 rap1 GTP-Binding Proteins Proteins 0.000 description 1
- 238000009790 rate-determining step (RDS) Methods 0.000 description 1
- 230000009712 regulation of translation Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 239000013037 reversible inhibitor Substances 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- RRVZOJQBRVGMMK-HCBGRYSISA-N rocaglaol Chemical class C1=CC(OC)=CC=C1[C@]1([C@@H](C[C@H]2O)C=3C=CC=CC=3)[C@]2(O)C2=C(OC)C=C(OC)C=C2O1 RRVZOJQBRVGMMK-HCBGRYSISA-N 0.000 description 1
- YGSDEFSMJLZEOE-UHFFFAOYSA-M salicylate Chemical compound OC1=CC=CC=C1C([O-])=O YGSDEFSMJLZEOE-UHFFFAOYSA-M 0.000 description 1
- 229960001860 salicylate Drugs 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 108060007624 small GTPase Proteins 0.000 description 1
- 239000012279 sodium borohydride Substances 0.000 description 1
- 229910000033 sodium borohydride Inorganic materials 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 239000011537 solubilization buffer Substances 0.000 description 1
- 238000003797 solvolysis reaction Methods 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 230000037351 starvation Effects 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000008399 tap water Substances 0.000 description 1
- 235000020679 tap water Nutrition 0.000 description 1
- 229940095064 tartrate Drugs 0.000 description 1
- WHRNULOCNSKMGB-UHFFFAOYSA-N tetrahydrofuran thf Chemical compound C1CCOC1.C1CCOC1 WHRNULOCNSKMGB-UHFFFAOYSA-N 0.000 description 1
- WROMPOXWARCANT-UHFFFAOYSA-N tfa trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F.OC(=O)C(F)(F)F WROMPOXWARCANT-UHFFFAOYSA-N 0.000 description 1
- 229960005371 tolbutamide Drugs 0.000 description 1
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- DTQVDTLACAAQTR-UHFFFAOYSA-N trifluoroacetic acid Substances OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 150000003672 ureas Chemical class 0.000 description 1
- 230000035899 viability Effects 0.000 description 1
- 239000003643 water by type Substances 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 239000011592 zinc chloride Substances 0.000 description 1
- JIAARYAFYJHUJI-UHFFFAOYSA-L zinc dichloride Chemical compound [Cl-].[Cl-].[Zn+2] JIAARYAFYJHUJI-UHFFFAOYSA-L 0.000 description 1
- 229940121641 zotatifin Drugs 0.000 description 1
- QYCXWOACFWMQFO-WZWZCULESA-N zotatifin Chemical compound CN(C)C[C@@H]1[C@H]([C@]2([C@](C=3C(=NC(=CC=3O2)OC)OC)([C@@H]1O)O)C1=CC=C(C#N)C=C1)C1=CC=CC=C1 QYCXWOACFWMQFO-WZWZCULESA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D307/00—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
- C07D307/77—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
- C07D307/93—Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems condensed with a ring other than six-membered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Definitions
- Novel RAS inhibitors The present invention relates to novel compounds and their use as a medicament, in particular for use in treating proliferative disorders.
- the present invention relates further to a pharmaceutical composition comprising the novel compounds.
- the present invention relates to a method of inhibiting proliferation or metastasis of cancer cells or inducing their cell death in a subject in need thereof.
- the present invention relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting RAS, in particular KRAS, HRAS and NRAS, activation in vitro.
- the present invention relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting eIF4A complex or engaging PHB1/2 complex in the plasma membrane or cytosol in vitro.
- the present invention relates to a kit containing a formulation comprising a pharmaceutical composition comprising a compound according to the invention.
- flavaglines like rocaglamide, a class of natural anti-tumour drugs and chemical ligands of prohibitins, inhibit RAS activation in cells by uncoupling the interaction between RAS and its effectors in the plane of the plasma membrane.
- rocaglamide inhibits RAS-activation in KRAS-mutated cell lines, there is still a demand for compounds with a better activity towards RAS oncogenes.
- various flavagline derivatives exhibit cytotoxic properties.
- WO 2005/113529 A2 describes cyclopenta[b]benzofuran derivatives and their utilization for the production of medicaments, especially for the prophylaxis and/or therapy of acute or chronic diseases.
- WO 2010/060891 A1 describes rocaglaol derivatives and the use of these derivatives to prevent or to limit the cardiotoxicity of an anti-neoplastic agent.
- WO 2012/0666002 A1 describes flavagline derivatives and their use as neuroprotective and/or cardioprotective and/or anti-tumor agents.
- WO 2017/058768 A1 describes compounds having activity as inhibitors of G12C mutant KRAS proteins.
- WO 2020/078975 A1 relates to inhibitors of KRAS oncogene activation, which are flavagline derivatives with the ability to target prohibitin to inhibit KRAS activation.
- N. Ribeiro et al., J. Med. Chem., 2012, 55, 100064 and Thuaud et al. Med. Chem., 2011, 54, 411 relates to flavagline derivates, in particular FL3 and FL23 which are effective in inhibition of cell proliferation and enhancement of viability at lower doses compared to rocaglalol.
- H. Yurugi et. al, Journal of Cell Science,133, 1, 2020, relates to a study of a subset of flavaglines, which may inhibits KRAS nanoclustering and activation.
- Eukaryotic initiation factor 4A is a DEAD-box protein containing ATPase and ATP- dependent RNA helicase required to melt local secondary structure and facilitate access of the ribosome to the mRNA template. The factor regulates the cap-dependent protein synthesis.
- eIF4A In mammals, there are three isoforms of eIF4A (eIF4AI, II and III), wherein eIF4AII and eIF4AIII share ⁇ 90% and ⁇ 65% identity, respectively, with the most abundant cellular factor eIF4AI. All isoforms are DEAD-box RNA helicase family members but only the paralogs eIF4AI and eIF4AII are found in the eIF4F complex and participate in translation initiation.
- WO 2017/091585 describes compounds having activity as inhibitors of eIF4A. However, the disclosed compounds have a different structure compared to the compounds according to the present invention.
- Prohibitins are evolutionarily conserved proteins and recent studies revealed a critical role for prohibitins in the activation of RAS by enabling RAS-effector interaction in the plane of the plasma membrane. Polier et al, Chemistry and Biology, 19, 1093–1104, 2012 showed that rocaglamides target this interaction (PHB1-CRAF) to inhibit RAS-CRAF interaction. These are several follow up studies confirming these effects. Ernst et al, J. Med. Chem.2020, 63, 5879 describes that the flavagline compounds rocaglamide A and Zotatifin show inhibition properties towards protein synthesis by stabilizing a translation-incompetent complex for selecting messenger RNAs (mRNAs) with eIF4A.
- mRNAs messenger RNAs
- the activated RAS can also influence of the functioning of the eIF4A complexes through one of the effector pathways (MAPK cascades).
- MPK cascades effector pathways
- the invention relates to a compound of formula (I) prodrug, or isotope enriched forms thereof, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from Br and CN, R 2 is selected from hydrogen and methyl, R 3 is selected from methyl and methoxy.
- the invention further relates to compounds of formula (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrug, or isotope enriched forms thereof, or the pharmaceutically acceptable salts thereof , or prodrugs, or isotope enriched forms thereof, or a pharmaceutically acceptable salt thereof, wherein R 1 is selected from Br and CN, R 2 is selected from hydrogen and methyl, R 3 is selected from methyl and methoxy.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use as a medicament.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment and/or prophylaxis of proliferative disorders or genetic disorders or inflammatory disorders.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or in form of a prodrug or isotope enriched forms thereof, for use in the treatment and/or prophylaxis of inflammatory disorders.
- the inflammatory disorders are selected from endometriosis and adenomyosis, in particular where RAS is mutated.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof as defined above or below, for use in the treatment and/or prophylaxisof proliferative disorders or genetic disorders or inflammatory disorders which involves oncogenic RAS or eIF4A complex that controls the expression of genes selected from c-myc and cyclin D1.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment and/or prophylaxis genetic disorders, wherein RAS signaling is pathologically involved, in particular CFC (Cranio Facial Cutaneous) and NF1 (Neurofibromatosis Type 1).
- CFC Cirranio Facial Cutaneous
- NF1 Neurofibromatosis Type 1
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment and/or prophylaxis genetic disorders, wherein Myc signaling is pathologically involved.
- the genetic disorders are selected from RASOpathies, in particular CFC (Cranio Facial Cutaneous) and NF1 (Neurofibromatosis Type 1).
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment of cancer.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use as inhibitor of RAS protein activation.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment or prevention of any diseases or conditions that are associated with the activity of RAS protein (RAS oncogene).
- RAS oncogene RAS protein
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein RAS-signaling is involved, preferably wherein KRAS G12V, NRAS G12V, HRAS G12V, KRAS G12C, KRAS G12D, KRAS G12C/Y96D, KRAS G13C, KRAS G13D, KRASG13S, KRAS Q61H, KRAS Q61R or KRAS Q61K or wherein any activating mutation in KRAS, HRAS and NRAS is involved or wherein any mutation that acquires resistance to RAS inhibitors.
- the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein RAS-signaling is involved.
- the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, KRAS G12V, NRAS G12V, HRAS G12V, KRAS G12C, KRAS G12D, KRAS G12C/Y96D, KRAS G13C, KRAS G13D, KRASG13S, KRAS Q61H, KRAS Q61R or KRAS Q61K is involved.
- the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein any activating mutation in KRAS, HRAS and NRAS is involved.
- the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein eIF4A-Myc-signaling is involved.
- the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein any mutation that acquires resistance to RAS inhibitors or eIF4Ainhibotors.
- the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, as defined above and below, for use as inhibitor of eIF4A complex or a ligand eIF4A complex or a ligand of Prohibitin.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug or isotope enriched forms thereof, for use as inhibitor of eukaryotic initiation factor 4A (eIF4A).
- eIF4A eukaryotic initiation factor 4A
- the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use as inhibitor of eukaryotic initiation factor 4A (eIF4A), which controls expression of cancer driving genes, especially which controls expression c-myc and cyclin D1.
- eIF4A eukaryotic initiation factor 4A
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt, for use as a ligand of Prohibitins (PHB1/2 complex) in the plasma membrane or cytosol.
- a pharmaceutically acceptable salt for use as a ligand of Prohibitins (PHB1/2 complex) in the plasma membrane or cytosol.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein dysregulation of eIF4A is involved, preferably wherein EIF4A1, EIF4A2 or EIF4A3 or EIF4F complex is involved.
- the invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein overexpression of Prohibitins (PHB/2) is involved.
- PHB/2 Prohibitins
- the invention further relates to a pharmaceutical composition, comprising a pharmaceutical effective amount of at least one compound selected from compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined above and below, and one or more pharmaceutical acceptable carrier.
- a pharmaceutical composition comprising a pharmaceutical effective amount of at least one compound selected from compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined above and below, and one or more pharmaceutical acceptable carrier.
- the invention further relates to a pharmaceutical composition, comprising a pharmaceutical effective amount of at least one compound selected from compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, for use in the prophylaxis and/or treatment of proliferative disorders or genetic disorders.
- a pharmaceutical composition comprising a pharmaceutical effective amount of at least one compound selected from compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, for use in the prophylaxis and/or treatment of proliferative disorders or genetic disorders.
- the invention further relates to a method of inhibiting growth, proliferation, or metastasis of cancer cells in a subject in need thereof, said method comprising administering to the subject a therapeutically effective amount of at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, or a pharmaceutical composition as defined above or below.
- the invention further relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting RAS activation, in particular inhibiting KRAS, HRAS or NRAS activation in vitro or ex vivo, the method comprising contacting the cell population with at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof or a pharmaceutical composition as defined above and below.
- the invention further relates to method of inhibiting proliferation of a cell population sensitive towards inhibiting eIF4A or its downstream targets, in particular c-myc and/or cyclin D1 in vitro or ex vivo, the method comprising contacting the cell population with at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof a pharmaceutical composition as defined above and below.
- the invention further relates to a kit comprising formulation comprising: a1) at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof or a2) a pharmaceutical composition comprising at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable, or a prodrug, or isotope enriched forms thereof or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of RAS activation is effective in treating the disorder.
- the invention further relates to a kit comprising formulation comprising: a1) at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof or a2) a pharmaceutical composition comprising at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable, or a prodrug, or isotope enriched forms thereof or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of dysregulation of protein translation, wherein eIF4A is involved,
- the invention further relates to a kit comprising formulation comprising: a1) at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof or a2) a pharmaceutical composition comprising at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable, or a prodrug, or isotope enriched forms thereof or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of dysregulation of protein translation, wherein eIF4A is involved,
- the invention has the following advantages: -
- the compounds according to the invention exhibit advantageous RAS inhibition properties.
- the compounds according to the invention qualify as inhibitors of RAS oncogene activation by inhibiting the prohibitin pathway, in particular inhibiting EGF-induced RAS-GTP loading in cells which is measured by the ability of RAS to bind to its effector proteins like RAF kinases.
- the compounds according to the invention prevent the activation of RAS as the interaction between RAS and its effectors is uncoupled due to defects in nanoclustering of RAS in the plane of the plasma membrane.
- salts may form salts which are also within the scope of this invention.
- salt(s) denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases.
- Pharmaceutically acceptable (i.e. non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful, e.g., in isolation or purification steps which may be employed during preparation.
- Salts of the compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), or an enantiomeric mixture thereof may be formed, for example, by reacting a compound of formulae (I), (I.b), (A), (B), (C), or an enantiomeric mixture thereof with at least one acid or base.
- the acid or base is added in an amount suitable for partial or complete neutralization, e.g. an equivalent amount.
- salts containing pharmacologically acceptable anions or cations such as chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, hydrogen phosphate, dihydrogen phosphate, isonicotinate, acetate, lactate, salicylate, citrate, acid citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, sulfate, benzenesulfonate, p-toluenesulfonate and palmoate [i.e.4,4'- methylene-bis-(3-hydroxy-2-naphthoate)] salts
- a suitable prodrug has chemically or metabolically cleavable group(s) and becomes, by solvolysis or under physiological conditions, a compound that is pharmaceutically active in vivo.
- a prodrug can be formed in a conventional manner by reaction of a functional group of the compound (such as an amino, hydroxy or carboxy group). Prodrug often offer advantages of better metabolism, potency, solubility, tissue compatibility, or delayed release in mammals.
- the term “prodrug” refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable.
- a prodrug may be inactive when administered to a subject in need thereof but is converted in vivo to an active compound of the invention.
- Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood.
- prodrug are compounds which are metabolized in vivo to give the compounds of the invention of formula I.
- Typical examples for prodrug are for example described in C.G. Wermeth (editor): The Practice of Medicinal Chemistry, Academic Press, San Diego, 1996, pages 671-715, as well as J. Rautio et al., NATURE REVIEWS, Drug Discovery, Vol.17, 2018, p.559.
- suitable prodrugs can be compounds of formula I wherein the hydroxy group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy group.
- Myc is a family of regulator genes and proto-oncogenes that code for transcription factors. The Myc family consists of three related human genes: c-myc (MYC), l-myc (MYCL), and n-myc (MYCN).
- Cyclin D1 is a protein that in humans is encoded by the CCND1 gene.
- the CCND1 gene encodes the cyclin D1 protein.
- the human CCND1 gene is located on the long arm of chromosome 11 (band 11q13). It is 13,388 base pairs long, and translates into 295 amino acids.
- Cyclin D1 is expressed in all adult human tissues with the exception of cells derived from bone marrow stem cell lines (both lymphoid and myeloid).
- a chemical structure that does not explicitly show a specific stereochemical orientation usually means all possible stereoisomers and mixtures thereof, unless indicated otherwise, for example, in which * designates the asymmetry centers.
- “Chiral compounds” in the sense of the invention are compounds that contain no improper axis of rotation (S n ). In the context of the present invention, they are in particular compounds with at least five chirality centers and without Sn-symmetry.
- “Stereoisomers” in the context of the invention are compounds of identical constitution but different atomic arrangement in the three-dimensional space.
- Enantiomers are stereoisomers which behave like image to mirror image to one another, e.g. compounds of formulae (I.a) and (I.b) are enantiomers.
- R and S are the descriptors of the CIP system for the two enantiomers and describe the absolute configuration on the asymmetric atom.
- “Diastereomers” are stereoisomers which are not enantiomeric to one another.
- the compound of the invention can exist in various isomeric forms, as well as in one or more tautomeric forms, including both single tautomers and mixtures of tautomers.
- the term “isomer” is intended to encompass all isomeric forms of a compound of this invention, including tautomeric forms of the compound. Some compounds described here can have asymmetric centers and therefore exist in different enantiomeric and diastereomeric forms.
- a compound of the invention can be in the form of an optical isomer or a diastereomer. Accordingly, the invention encompasses compounds of the invention and their uses as described herein in the form of their optical isomers, diastereoisomers and mixtures thereof, including a racemic mixture.
- Optical isomers of the compounds of the invention can be obtained by known techniques such as asymmetric synthesis, chiral chromatography, or via chemical separation of stereoisomers through the employment of optically active resolving agents.
- “stereoisomer” means one stereoisomer of a compound that is substantially free of other stereoisomers of that compound.
- a stereomerically pure compound having one chiral center will be substantially free of the opposite enantiomer of the compound.
- a stereomerically pure compound having two chiral centers will be substantially free of other diastereomers of the compound.
- a typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, for example greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, or greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, or greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound.
- A“stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
- the present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposable mirror images of one another.
- Compounds of the invention or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids.
- the present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms.
- Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization.
- Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography.
- Relative configuration in stereochemistry is the arrangement of atoms or groups of atoms that is described relative to other atoms or groups of atoms in the molecule.
- this term describes the position of atoms or groups of atoms in space in relation to other atoms or groups of atoms that are located elsewhere in the molecule.
- Absolute configuration in stereochemistry is the arrangement of atoms or group of atoms that is described independently of any other atom or group of atoms in the molecule. This type of configuration is defined for chiral molecular entities and their stereochemical descriptions (e.g. R or S). Syn means that with regard to the orientation of the substituents on the 5-membered ring they are bound to (5 asymmetric carbon atoms) all substituents point in the same direction relative to the plane of the 5-membered ring.
- the compounds of formula (I) in which * designates the asymmetry centers represent the isomers of formulae (I.1), (I.2), (I.3), (I.4), (I.5), (I.6), (I.7), (I.8), (I.9), (I.10), (I.11), (I.12), (I.13), (I.14), (I.15), (I.16), (I.17), (I.18(, (I.19), (I.20), (I.21), (I.22), (I.23), (I.24), (I.25), (I.26), (I.27), (I.28), (I.29), (I.30), (I.31) and (I.32): 10 (I.
- the compounds of formulae (I.1) to (I.32) are specified by their absolute stereochemistry.
- the compound of formula (I) is a mixture of at least two enantiomers (I.1) to (I.32) or a mixture of the prodrugs, or isotope enriched forms thereof, or a mixture of the pharmaceutically acceptable salt thereof, wherein one enantiomer is enriched.
- the compound of formula (I) is a mixture of (I.a) and (I.b) or a mixture of the prodrug, or isotope enriched forms thereof or a mixture of the pharmaceutically acceptable salt thereof, wherein the enantiomer excess (ee) of the enantiomer of formula (I.a) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%.
- compound of formula (I) according to the invention, or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, wherein R 2 is methyl and R 3 is methoxy.
- Another special embodiment are the compounds of formula (I), wherein R 1 , R 2 and R 3 are selected from the definition given in one line of table 1:
- Another special embodiment are the compounds selected from A, B, C, D, E, F and the mixture of the each of compounds A to F with its respective enantiomer:
- Preferred is a compound of formula (A) or an enantiomeric mixture comprising the compounds of formula (A) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (A) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%.
- a compound of formula (F) or an enantiomeric mixture comprising the compounds of formula (F) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (F) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%.
- compound A is especially preferred.
- compound F is especially preferred.
- the compounds of the present invention can be synthesized using the methods known in the prior art and together with methods known from synthetic organic chemistry, or variations thereof as appreciated by those skilled in the art.
- compositions are employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response or other problem or complication, commensurate with a reasonable benefit/risk ratio.
- therapeutically effective is intended to qualify the amount of each agent, which will achieve the goal of improvement in disorder severity and the frequency of incidence, while avoiding adverse side-effects typically associated with alternative therapies. For example, effective anticancer agents prolong the survivability of the patient or his/her life quality, inhibit the rapidly proliferating cell growth associated with the neoplasm, or effect a regression of the neoplasm.
- treat refers to any type of intervention or process performed on, or administering an active agent to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting or slowing down or preventing the progression, development, severity or recurrence of a symptom, complication, condition or biochemical indicia associated with a disease.
- prophylaxis or “prevention” refers to administration to a subject who does not have a disease to prevent the disease from occurring.
- the term "cell” is meant to refer to a cell that is in vitro, ex vivo or in vivo.
- an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal.
- an in vitro cell can be a cell in a cell culture.
- an in vivo cell is a cell living in an organism such as a mammal.
- patient includes humans and animals that receive either therapeutic or prophylactic treatment.
- subject includes any human or animal.
- the methods and compositions herein disclosed can be used to treat a subject having cancer.
- a (non-human) animal includes all vertebrates, e.g.
- the subject is a human subject.
- pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid diluent, solvent, excipient, manufacturing aid (e.g. lubricant) or encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body.
- Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
- Suitable other ingredients are the afore-mentioned carrier and further additives, including adjuvants, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, bittering agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents, dispensing agents, etc..
- Suitable additives are selected depending on the nature of the mode of administration and dosage forms; and not injurious to the patient.
- pharmaceutical composition means a composition comprising a compound of the invention in combination with at least one further compound selected from a) at least one further pharmaceutically active substance and b) at least one additional pharmaceutically acceptable carrier and or additive.
- RAS inhibitor refers to an agent capable of decreasing RAS protein levels, decreasing RAS activity levels and/or inhibiting RAS expression levels in the cells.
- the RAS inhibitor may be a reversible or irreversible inhibitor.
- RAS protein refers to a protein that is a member of a family of related proteins that are expressed in all human and animal cell lineages and organs. All RAS protein family members belong to a class of proteins called small GTPase (also known as small G proteins, a family of hydrolase enzymes that can bind and hydrolyse GTP), and are involved in transmitting signals within cells (cellular signal transduction).
- RAS is the prototypical member of the RAS superfamily of proteins, which are all related in three-dimensional structure and regulate diverse cell behaviours. When RAS is 'switched on' by incoming signals, it subsequently switches on other proteins, which ultimately turn on genes involved in cell growth, differentiation, and survival. Mutations in RAS genes can lead to the production of permanently activated RAS proteins, which can cause unintended and overactive signaling inside the cell, even in the absence of incoming signals. Because these signals result in cell growth and division, overactive RAS signaling can ultimately lead to cancer.
- the three RAS genes in humans (HRAS, KRAS, and NRAS) are the most common oncogenes in human cancer.
- the clinically most notable members of the RAS subfamily are HRAS, KRAS and NRAS.
- members of this subfamily which are e.g. selected from DIRAS1, DIRAS2, DIRAS3, ERAS, GEM, MRAS, NKIRAS1, NKIRAS2, NRAS, RALA, RALB, RAP1A, RAP1B, RAP2A, RAP2B, RAP2C, RASD1, RASD2, RASL10A, RASL10B, RASL11A, RASL11B, RASL12, REM1, REM2, RERG, RERGL, RRAD, RRAS, RRAS2.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and a pharmaceutical composition comprising in at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be administered to humans and animals, preferably humans.
- any method of administration may be used to deliver the compound or pharmaceutical composition according to the invention to a subject. Suitable methods of administration are orally, enterally, parenterally, intravenously, topically, intramuscular, subcutaneous/dermal routes.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above can selectively decrease RAS protein levels, decrease RAS activity levels and/or inhibit RAS expression levels in the cells.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above can be used to selectively decrease RAS activity levels and/or inhibit RAS expression levels in cells or in an individual in need of a decrease in RAS protein levels, decrease in RAS activity levels and/or inhibition of RAS expression levels by administering an inhibiting amount of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above or a salt thereof.
- the present invention provides a combined preparation of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or a pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and an additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases, preferably of proliferative disorders (e.g. cancer), in particular disorders associated with the activity of RAS protein.
- Additional therapeutic agent(s) are selected from chemotherapeutic agents, radiotherapeutic agents, immuno-oncology agents, and combinations thereof.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above are sequentially administered prior to administration of the immuno-oncology agent.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above are administered concurrently with the immuno-oncology agent.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above are sequentially administered after administration of the immuno-oncology agent.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be co-formulated with an immuno- oncology agent.
- Immuno-oncology agents include, for example, a small molecule drug, antibody or other biologic or small molecule.
- biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines.
- the antibody is a monoclonal antibody.
- the monoclonal antibody is humanized or human.
- the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators).
- Suitable of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF).
- B7 family includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
- B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6 includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
- TNF family of molecules that bind to cognate TNF receptor family members which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTpR, LIGHT, DcR3, HVEM, VEGETL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin a/TNFp, TNFR2, TNFa, LTpR, Lymphotoxin a 1b2, FAS
- T cell responses can be stimulated by a combination of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4- 1BB (CD 137), 4-1BBL, ICOS, ICOS-L, 0X40, OX40L
- antagonists of KIR such as Lirilumab.
- agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155.
- the combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner.
- Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents.
- Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues.
- the therapeutic agents can be administered by the same route or by different routes.
- a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally.
- all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection.
- Combination therapy can also embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g surgery or radiation treatment.)
- the combination therapy further comprises a non-drug treatment
- the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved.
- the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks.
- Types of cancers that may be treated with the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above include, but are not limited to, prostate, colon, rectum, pancreas, cervix, stomach, endometrium, brain, liver, bladder, ovary, testis, head, neck, skin (including melanoma and basal carcinoma), mesothelial lining, white blood cell (including lymphoma and leukemia), esophagus, breast, muscle, connective tissue, lung (including small cell lung carcinoma and non- small-cell carcinoma), adrenal gland, thyroid, kidney, or bone; or glioblastoma, mesothelioma, renal cell carcinoma, gastric carcinoma, sarcoma (including Kaposi's sarcoma), choriocarcinoma, cutaneous basocellular carcinoma, haematological malignancies (including
- One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g ., IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above for treatment of RAS protein associated diseases, disorders or conditions.
- cytokine therapy e.g ., IL2 and GM-CSF
- tyrosine kinase inhibitors can be optionally used in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (
- chemotherapeutic or other anti-cancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine, cyclophosphamide (CYTOXAN®), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.
- alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes
- alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and
- suitable agents for use in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the "Dartmouth regimen", which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOYTM.
- DTIC dacarbazine
- BCNU carmustine
- cisplatin cisplatin
- tamoxifen a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOYTM.
- Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may also be combined with immunotherapy drugs, including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in the treatment of melanoma.
- immunotherapy drugs including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in the treatment of melanoma.
- Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may also be used in combination with vaccine therapy in the treatment of melanoma.
- Antimelanoma vaccines are, in some ways, similar to the anti-virus vaccines which are used to prevent diseases caused by viruses such as polio, measles, and mumps.
- Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body's immune system to destroy melanoma cells.
- Melanomas that are confined to the arms or legs may also be treated with a combination of agents including one or more compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined, using a hyperthermic isolated limb perfusion technique.
- This treatment protocol temporarily separates the circulation of the involved limb from the rest of the body and injects high doses of chemotherapy into the artery feeding the limb, thus providing high doses to the area of the tumor without exposing internal organs to these doses that might otherwise cause severe side effects.
- the fluid is warmed to 38.9 °C to 40 °C.
- Melphalan is the drug most often used in this chemotherapy procedure. This can be given with another agent called tumor necrosis factor (TNF).
- TNF tumor necrosis factor
- Suitable chemotherapeutic or other anti-cancer agents include, for example, antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine.
- antimetabolites including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
- methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
- methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
- Suitable chemotherapeutic or other anti-cancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxyco-formycin, mitomycin-C, L- asparaginase, interferons (especially IFN-a), etoposide, and teniposide.
- certain natural products and their derivatives for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins
- vinblastine vincristine, vindesine
- bleomycin dactinomycin, daunorubicin, dox
- cytotoxic agents include navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafme, and droloxafme.
- cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors.
- anti-cancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-IO or TGF-b).
- Other anti-cancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4.
- Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer.
- Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.
- At least one compound of (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and at least one chemotherapeutic agent are administered to the patient concurrently or sequentially.
- At least one compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be administered first, at least one chemotherapeutic agent may be administered first, or at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be administered at the same time.
- the compounds may be administered in any order.
- the invention also provides pharmaceutically compositions which comprise a therapeutically effective amount of one or more of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally, one or more additional therapeutic agents described above.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
- the compound(s) and compositions of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) an enantiomeric mixture as defined above, can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques ( e.g.
- the pharmaceutical composition may be in the form of, for example, a tablet, capsule, liquid capsule, suspension, or liquid.
- the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
- the pharmaceutical composition may be provided as a tablet or capsule comprising an amount of active ingredient in the range of from about 0.1 to 1000 mg, preferably from about 0.25 to 250 mg, and more preferably from about 0.5 to 100 mg.
- a suitable daily dose for a human or animal may vary widely depending on the condition of the patient and other factors, but, can be determined using routine methods.
- Any pharmaceutical composition contemplated herein can, for example, be delivered orally via any acceptable and suitable oral preparations.
- Exemplary oral preparations include, but are not limited to, for example, tablets, troches, lozenges, aqueous and oily suspensions, dispersible powders or granules, emulsions, hard and soft capsules, liquid capsules, syrups, and elixirs.
- compositions intended for oral administration can be prepared according to any methods known in the art for manufacturing pharmaceutical compositions intended for oral administration.
- a pharmaceutical composition in accordance with the invention can contain at least one agent selected from sweetening agents, flavoring agents, bittering agents, coloring agents, demulcents, antioxidants, and preserving agents.
- a tablet can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one non-toxic pharmaceutically acceptable excipient suitable for the manufacture of tablets.
- excipients include, but are not limited to, for example, inert diluents, such as, for example, calcium carbonate, sodium carbonate, lactose, calcium phosphate, and sodium phosphate; granulating and disintegrating agents, such as, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, and alginic acid; binding agents, such as, for example, starch, gelatin, polyvinyl-pyrrolidone, and acacia; and lubricating agents, such as, for example, magnesium stearate, stearic acid, and talc.
- inert diluents such as, for example, calcium carbonate, sodium carbonate, lactose, calcium phosphate, and sodium phosphate
- granulating and disintegrating agents such as, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, and alginic acid
- binding agents such as, for example, starch, gelatin, polyvinyl-pyrrol
- a tablet can either be uncoated, or coated by known techniques to either mask the bad taste of an unpleasantly tasting drug, or delay disintegration and absorption of the active ingredient in the gastrointestinal tract thereby sustaining the effects of the active ingredient for a longer period.
- exemplary water soluble taste masking materials include, but are not limited to, hydroxypropyl-methylcellulose and hydroxypropyl- cellulose.
- Exemplary time delay materials include, but are not limited to, ethyl cellulose and cellulose acetate butyrate.
- Hard gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or at least one salt thereof with at least one inert solid diluent, such as, for example, calcium carbonate; calcium phosphate; and kaolin.
- at least one inert solid diluent such as, for example, calcium carbonate; calcium phosphate; and kaolin.
- Soft gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one water soluble carrier, such as, for example, polyethylene glycol; and at least one oil medium, such as, for example, peanut oil, liquid paraffin, and olive oil.
- at least one water soluble carrier such as, for example, polyethylene glycol
- at least one oil medium such as, for example, peanut oil, liquid paraffin, and olive oil.
- An aqueous suspension can be prepared, for example, by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one excipient suitable for the manufacture of an aqueous suspension.
- excipients suitable for the manufacture of an aqueous suspension include, but are not limited to, for example, suspending agents, such as, for example, sodium carboxymethylcellulose, hydroxypropyl- methylcellulose and hydroxypropyl- cellulose, sodium alginate, alginic acid, polyvinyl- pyrrolidone, gum tragacanth, and gum acacia; dispersing or wetting agents, such as, for example, a naturally-occurring phosphatide, e.g., lecithin; condensation products of alkylene oxide with fatty acids, such as, for example, polyoxyethylene stearate; condensation products of ethylene oxide with long chain aliphatic alcohols, such as, for example heptadecaethylene-oxycetanol; condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol, such as, for example, polyoxyethylene sorbitol monooleate; and condensation products of ethylene oxide with partial esters derived from
- An aqueous suspension can also contain at least one preservative, such as, for example, ethyl and n-propyl p-hydroxybenzoate; at least one coloring agent; at least one flavoring agent; and/or at least one sweetening agent, including but not limited to, for example, sucrose, saccharin, and aspartame.
- Oily suspensions can, for example, be prepared by suspending at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof in either a vegetable oil, such as, for example, arachis oil, olive oil, sesame oil and coconut oil or in mineral oil, such as, for example, liquid paraffin.
- An oily suspension can also contain at least one thickening agent, such as, for example, beeswax, hard paraffin and cetyl alcohol.
- At least one of the sweetening agents already described hereinabove, and/or at least one flavoring agent can be added to the oily suspension.
- An oily suspension can further contain at least one preservative, including, but not limited to, for example, an anti- oxidant, such as, for example, butylated hydroxyanisol, and alpha-tocopherol.
- Dispersible powders and granules can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one dispersing and/or wetting agent; at least one suspending agent; and/or at least one preservative.
- Suitable dispersing agents, wetting agents, and suspending agents are as already described above.
- Exemplary preservatives include, but are not limited to, for example, anti-oxidants, e.g., ascorbic acid.
- dispersible powders and granules can also contain at least one excipient, including, but not limited to, for example, sweetening agents; flavoring agents; and coloring agents.
- An emulsion of at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, be prepared as an oil-in-water emulsion.
- the oily phase of the emulsions comprising compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be constituted from known ingredients in a known manner.
- the oil phase can be provided by, but is not limited to, for example, a vegetable oil, such as, for example, olive oil and arachis oil; a mineral oil, such as, for example, liquid paraffin; and mixtures thereof. While the phase may comprise merely an emulsifier, it may comprise a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil.
- Suitable emulsifying agents include, but are not limited to, for example, naturally-occurring phosphatides, e.g., soy bean lecithin; esters or partial esters derived from fatty acids and hexitol anhydrides, such as, for example, sorbitan monooleate; and condensation products of partial esters with ethylene oxide, such as, for example, polyoxyethylene sorbitan monooleate.
- a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat.
- emulsifier(s) with or without stabilize make- up the so-called emulsifying wax
- the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
- An emulsion can also contain a sweetening agent, a flavoring agent, a preservative, and/or an antioxidant.
- Emulsifiers and emulsion stabilizers suitable for use in the formulation of the present invention include Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate, sodium lauryl sulfate, glyceryl distearate alone or with a wax, or other materials well known in the art.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, also be delivered intravenously, subcutaneously, and/or intramuscularly via any pharmaceutically acceptable and suitable injectable form.
- Exemplary injectable forms include, but are not limited to, for example, sterile aqueous solutions comprising acceptable vehicles and solvents, such as, for example, water, Ringer’s solution, and isotonic sodium chloride solution; sterile oil-in-water microemulsions and aqueous or oleaginous suspensions.
- Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in the formulations for oral administration or by using other suitable dispersing or wetting agents and suspending agents.
- the compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum, and/or various buffers.
- Other adjuvants and modes of administration are well and widely known in the pharmaceutical art.
- the active ingredient may also be administered by injection as a composition with suitable carriers, including saline, dextrose, water or with cyclodextrin solubilization (i.e. Captisol), cosolvent solubilization (i.e. propylene glycol) or micellar solubilization (i.e. Tween 80).
- the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3- butanediol.
- a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3- butanediol.
- acceptable vehicles and solvents that may be employed are water, Ringer’s solution, and isotonic sodium chloride solution.
- sterile, fixed oils are conventionally employed as a solvent or suspending medium.
- any bland fixed oil may be employed, including synthetic mono- or diglycerides.
- fatty acids such as oleic acid find use in the preparation of injectables.
- a sterile injectable oil-in-water microemulsion can, for example, be prepared by 1) dissolving at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above in an oily phase, such as, for example, a mixture of soybean oil and lecithin; 2) combining the compound(s) of formulae (I), (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above containing oil phase with a water and glycerol mixture; and 3) processing the combination to form a microemulsion.
- an oily phase such as, for example, a mixture of soybean oil and lecithin
- a sterile aqueous or oleaginous suspension can be prepared in accordance with methods already known in the art.
- a sterile aqueous solution or suspension can be prepared with a non-toxic parenterally-acceptable diluent or solvent, such as, for example, 1,3-butane diol; and a sterile oleaginous suspension can be prepared with a sterile non- toxic acceptable solvent or suspending medium, such as, for example, sterile fixed oils, e.g., synthetic mono- or diglycerides; and fatty acids, such as, for example, oleic acid.
- Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art.
- Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Allen, L. V. Jr.
- compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-alpha-tocopherol poly ethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, polyethoxylated castor oil such as CREMOPHOR surfactant (BASF), or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts,
- Cyclodextrins such as alpha-, beta-, and gamma-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-cyclodextrins, or other solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein.
- the pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals.
- the pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc.
- Tablets and pills can additionally be prepared with enteric coatings.
- Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents.
- adjuvants such as wetting, sweetening, flavoring, and perfuming agents.
- the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration.
- the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration.
- Such capsules or tablets may contain a controlled-release formulation as may be provided in a dispersion of active compound in hydroxypropylmethyl cellulose.
- the amounts of compounds that are administered and the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention depend on a variety of factors, including the age, weight, sex, the medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods.
- the daily dose can be administered in one to four doses per day. Other dosing schedules include one dose per week and one dose per two day cycle.
- compositions of this invention comprise at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and optionally an additional agent selected from any pharmaceutically acceptable carrier, adjuvant, and vehicle.
- compositions of this invention comprise a compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, or a prodrug, or isotope enriched forms thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
- the present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of RAS protein-associated diseases.
- the present invention also relates to a kit containing a formulation comprising: a) a pharmaceutical composition comprising a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, or a therapeutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of RAS activation is effective in treating the disorder.
- kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art.
- kit components such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art.
- Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
- the dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
- the daily oral dosage of each active ingredient when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day.
- the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.
- Compound(s) of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
- the compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g.
- Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 200 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.1-95 % by weight based on the total weight of the composition.
- a typical capsule for oral administration contains at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture (250 mg), lactose (75 mg), and magnesium stearate (15 mg).
- a typical injectable preparation is produced by aseptically placing at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture (250 mg) into a vial, aseptically freeze-drying and sealing.
- the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation.
- compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, alone or in combination with a pharmaceutical carrier.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above can be used alone, in combination with other compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, or in combination with one or more other therapeutic agent(s), e.g. an anticancer agent or other pharmaceutically active material.
- therapeutic agent(s) e.g. an anticancer agent or other pharmaceutically active material.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
- Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
- the selected dosage level will depend upon a variety of factors including the activity of the particular compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
- a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
- the physician or veterinarian could start doses of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
- a suitable daily dose of compound(s) of formulae (I), (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect.
- Such an effective dose will generally depend upon the factors described above.
- oral, intravenous, intracerebroventricular and subcutaneous doses of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above for a patient will range from about 0.01 to about 50 mg per kilogram of body weight per day.
- the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
- dosing is one administration per day. While it is possible for compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition).
- Human translational control is of increasing research interest as it has connotations in a range of diseases. Orthologs of many of the factors involved in human translation are shared by a range of eukaryotic organisms. Synthesis of protein from mature messenger RNA in eukaryotes is divided into translation initiation, elongation, and termination of these stages; the initiation of translation is the rate limiting step. Within the process of translation initiation; the bottleneck occurs shortly before the ribosome binds to the 5’ m7GTP facilitated by a number of proteins; it is at this stage that constrictions born of stress, amino acid starvation etc. take effect.
- Eukaryotic initiation factor complex 2 (eIF2) forms a ternary complex with GTP and the initiator Met-tRNA – this process is regulated by guanine nucleotide exchange and phosphorylation and serves as the main regulatory element of the bottleneck of gene expression.
- a number of initiation factors must facilitate the synergy of the ribosome and the mRNA and ensure that the 5’ UTR of the mRNA is sufficiently devoid of secondary structure. Binding in this way is facilitated by group 4 eukaryotic initiation factors; eIF4F has implications in the normal regulation of translation as well as the transformation and progression of cancerous cells.
- eIF4F is responsible for the binding of capped mRNA to the 40S ribosomal subunit via eIF3.
- the mRNA cap is bound by eIF4E (25 kDa)
- eIF4G (185 kDa) acts as a scaffold for the complex whilst the ATP-dependent RNA helicase eIF4A (46 kDa) processes the secondary structure of the mRNA 5’ UTR to render it more conducive to ribosomal binding and subsequent translation.
- eIF4F For maximal activity; eIF4A also requires eIF4B (80 kDa), which itself is enhanced by eIF4H (25 kDa).
- this 48S complex searches for the (usually) AUG start codon and translation begins.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and a pharmaceutical composition comprising in at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be administered to humans and animals, preferably humans. In principle any method of administration may be used to deliver the compound or pharmaceutical composition according to the invention to a subject.
- Suitable methods of administration are orally, enterally, parenterally, intravenously, topically, intramuscular, subcutaneous routes.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above can selectively decrease activity of eIF4A, decrease eIF4A activity levels and/or inhibit activity of eIF4 in the cytosol.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above can be used to selectively decrease eIF4A activity levels and/or inhibit eIF4A in cytosol or in an individual in need of a decrease in activity of eIF4A, decrease in eIF4A activity and/or inhibition of eIF4A activity levels by administering an inhibiting amount of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above or a salt or a prodrug, or isotope enriched forms thereof.
- the present invention provides a combined preparation of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, and/or a pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and an additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases, preferably of proliferative disorders (e.g. cancer), in particular disorders associated with the activity of eIF4A.
- Additional therapeutic agent(s) are selected from chemotherapeutic agents, radiotherapeutic agents, immuno-oncology agents, and combinations thereof.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above are sequentially administered prior to administration of the immuno-oncology agent.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above are administered concurrently with the immuno-oncology agent.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above are sequentially administered after administration of the immuno-oncology agent.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be co-formulated with an immuno- oncology agent.
- Immuno-oncology agents include, for example, a small molecule drug, antibody or other biologic or small molecule.
- biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines.
- the antibody is a monoclonal antibody.
- the monoclonal antibody is humanized or human.
- the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators).
- Suitable of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF).
- B7 family includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
- B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6 includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6.
- TNF family of molecules that bind to cognate TNF receptor family members which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTpR, LIGHT, DcR3, HVEM, VEGETL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin a/TNFp, TNFR2, TNFa, LTpR, Lymphotoxin a 1b2, FAS
- T cell responses can be stimulated by a combination of compound(s) of formulae (I), (I-A),(I.a’), (I-A.a’), (I.a), (I-A.a), (A), (B), (C), (D) , (E), (F), (G), (H), (I), (J), (K), (L), (M), (N), (O), (P), (Q), (R), (S), (T), (U), (V) and an enantiomeric mixture as defined above and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and
- antagonists of KIR such as Lirilumab.
- agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155.
- the combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner.
- Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents.
- Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues.
- the therapeutic agents can be administered by the same route or by different routes.
- a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally.
- all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection.
- Combination therapy can also embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g surgery or radiation treatment.)
- the combination therapy further comprises a non-drug treatment
- the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved.
- the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks.
- Types of cancers that may be treated with the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above include, but are not limited to, prostate, colon, rectum, pancreas, cervix, stomach, endometrium, brain, liver, bladder, ovary, testis, head, neck, skin (including melanoma and basal carcinoma), mesothelial lining, white blood cell (including lymphoma and leukemia), esophagus, breast, muscle, connective tissue, lung (including small cell lung carcinoma and non- small-cell carcinoma), adrenal gland, thyroid, kidney, or bone; or glioblastoma, mesothelioma, renal cell carcinoma, gastric carcinoma, sarcoma (including Kaposi's sarcoma), choriocarcinoma, cutaneous basocellular carcinoma, haematological malignancies (including blood, bone
- One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g ., IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above for treatment of eIF4A associated diseases, disorders or conditions.
- cytokine therapy e.g IL2 and GM-CSF
- tyrosine kinase inhibitors can be optionally used in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an
- chemotherapeutic or other anti-cancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine, cyclophosphamide (CYTOXAN®), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.
- alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes
- alkylating agents including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and
- suitable agents for use in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the "Dartmouth regimen", which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOYTM.
- Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may also be combined with immunotherapy drugs, including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in the treatment of melanoma.
- immunotherapy drugs including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in the treatment of melanoma.
- Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may also be used in combination with vaccine therapy in the treatment of melanoma.
- Antimelanoma vaccines are, in some ways, similar to the anti-virus vaccines which are used to prevent diseases caused by viruses such as polio, measles, and mumps.
- Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body's immune system to destroy melanoma cells.
- Melanomas that are confined to the arms or legs may also be treated with a combination of agents including one or more compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined, using a hyperthermic isolated limb perfusion technique.
- This treatment protocol temporarily separates the circulation of the involved limb from the rest of the body and injects high doses of chemotherapy into the artery feeding the limb, thus providing high doses to the area of the tumor without exposing internal organs to these doses that might otherwise cause severe side effects.
- the fluid is warmed to 38.9 °C to 40 °C.
- Melphalan is the drug most often used in this chemotherapy procedure. This can be given with another agent called tumor necrosis factor (TNF).
- TNF tumor necrosis factor
- Suitable chemotherapeutic or other anti-cancer agents include, for example, antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine.
- antimetabolites including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
- methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
- methotrexate including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
- Suitable chemotherapeutic or other anti-cancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxyco-formycin, mitomycin-C, L- asparaginase, interferons (especially IFN-a), etoposide, and teniposide.
- certain natural products and their derivatives for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins
- vinblastine vincristine, vindesine
- bleomycin dactinomycin, daunorubicin, dox
- cytotoxic agents include navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafme, and droloxafme.
- cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors.
- anti-cancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-IO or TGF-b).
- Other anti-cancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4.
- Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer.
- Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.
- At least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and at least one chemotherapeutic agent are administered to the patient concurrently or sequentially.
- At least one compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be administered first, at least one chemotherapeutic agent may be administered first, or at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be administered at the same time.
- the compounds may be administered in any order.
- the invention also provides pharmaceutically compositions which comprise a therapeutically effective amount of one or more of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally, one or more additional therapeutic agents described above.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
- the compound(s) and compositions of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques (e.g.
- the pharmaceutical composition may be in the form of, for example, a tablet, capsule, liquid capsule, suspension, or liquid.
- the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
- the pharmaceutical composition may be provided as a tablet or capsule comprising an amount of active ingredient in the range of from about 0.1 to 1000 mg, preferably from about 0.25 to 250 mg, and more preferably from about 0.5 to 100 mg.
- a suitable daily dose for a human or animal may vary widely depending on the condition of the patient and other factors, but, can be determined using routine methods.
- Any pharmaceutical composition contemplated herein can, for example, be delivered orally via any acceptable and suitable oral preparations.
- Exemplary oral preparations include, but are not limited to, for example, tablets, troches, lozenges, aqueous and oily suspensions, dispersible powders or granules, emulsions, hard and soft capsules, liquid capsules, syrups, and elixirs.
- compositions intended for oral administration can be prepared according to any methods known in the art for manufacturing pharmaceutical compositions intended for oral administration.
- a pharmaceutical composition in accordance with the invention can contain at least one agent selected from sweetening agents, flavoring agents, bittering agents, coloring agents, demulcents, antioxidants, and preserving agents.
- a tablet can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one non-toxic pharmaceutically acceptable excipient suitable for the manufacture of tablets.
- excipients include, but are not limited to, for example, inert diluents, such as, for example, calcium carbonate, sodium carbonate, lactose, calcium phosphate, and sodium phosphate; granulating and disintegrating agents, such as, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, and alginic acid; binding agents, such as, for example, starch, gelatin, polyvinyl-pyrrolidone, and acacia; and lubricating agents, such as, for example, magnesium stearate, stearic acid, and talc.
- inert diluents such as, for example, calcium carbonate, sodium carbonate, lactose, calcium phosphate, and sodium phosphate
- granulating and disintegrating agents such as, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, and alginic acid
- binding agents such as, for example, starch, gelatin, polyvinyl-pyrrol
- a tablet can either be uncoated, or coated by known techniques to either mask the bad taste of an unpleasantly tasting drug, or delay disintegration and absorption of the active ingredient in the gastrointestinal tract thereby sustaining the effects of the active ingredient for a longer period.
- exemplary water soluble taste masking materials include, but are not limited to, hydroxypropyl-methylcellulose and hydroxypropyl- cellulose.
- Exemplary time delay materials include, but are not limited to, ethyl cellulose and cellulose acetate butyrate.
- Hard gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, and/or at least one salt thereof with at least one inert solid diluent, such as, for example, calcium carbonate; calcium phosphate; and kaolin.
- Soft gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one water soluble carrier, such as, for example, polyethylene glycol; and at least one oil medium, such as, for example, peanut oil, liquid paraffin, and olive oil.
- water soluble carrier such as, for example, polyethylene glycol
- oil medium such as, for example, peanut oil, liquid paraffin, and olive oil.
- An aqueous suspension can be prepared, for example, by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one excipient suitable for the manufacture of an aqueous suspension.
- excipients suitable for the manufacture of an aqueous suspension include, but are not limited to, for example, suspending agents, such as, for example, sodium carboxymethylcellulose, hydroxypropyl- methylcellulose and hydroxypropyl- cellulose, sodium alginate, alginic acid, polyvinyl- pyrrolidone, gum tragacanth, and gum acacia; dispersing or wetting agents, such as, for example, a naturally-occurring phosphatide, e.g., lecithin; condensation products of alkylene oxide with fatty acids, such as, for example, polyoxyethylene stearate; condensation products of ethylene oxide with long chain aliphatic alcohols, such as, for example heptadecaethylene-oxycetanol; condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol, such as, for example, polyoxyethylene sorbitol monooleate; and condensation products of ethylene oxide with partial esters derived from
- An aqueous suspension can also contain at least one preservative, such as, for example, ethyl and n-propyl p-hydroxybenzoate; at least one coloring agent; at least one flavoring agent; and/or at least one sweetening agent, including but not limited to, for example, sucrose, saccharin, and aspartame.
- Oily suspensions can, for example, be prepared by suspending at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof in either a vegetable oil, such as, for example, arachis oil, olive oil, sesame oil and coconut oil or in mineral oil, such as, for example, liquid paraffin.
- An oily suspension can also contain at least one thickening agent, such as, for example, beeswax, hard paraffin and cetyl alcohol.
- At least one of the sweetening agents already described hereinabove, and/or at least one flavoring agent can be added to the oily suspension.
- An oily suspension can further contain at least one preservative, including, but not limited to, for example, an anti- oxidant, such as, for example, butylated hydroxyanisol, and alpha-tocopherol.
- Dispersible powders and granules can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one dispersing and/or wetting agent; at least one suspending agent; and/or at least one preservative.
- Suitable dispersing agents, wetting agents, and suspending agents are as already described above.
- Exemplary preservatives include, but are not limited to, for example, anti-oxidants, e.g., ascorbic acid.
- dispersible powders and granules can also contain at least one excipient, including, but not limited to, for example, sweetening agents; flavoring agents; and coloring agents.
- An emulsion of at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, be prepared as an oil-in-water emulsion.
- the oily phase of the emulsions comprising compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be constituted from known ingredients in a known manner.
- the oil phase can be provided by, but is not limited to, for example, a vegetable oil, such as, for example, olive oil and arachis oil; a mineral oil, such as, for example, liquid paraffin; and mixtures thereof. While the phase may comprise merely an emulsifier, it may comprise a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil.
- Suitable emulsifying agents include, but are not limited to, for example, naturally-occurring phosphatides, e.g., soy bean lecithin; esters or partial esters derived from fatty acids and hexitol anhydrides, such as, for example, sorbitan monooleate; and condensation products of partial esters with ethylene oxide, such as, for example, polyoxyethylene sorbitan monooleate.
- a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat.
- emulsifier(s) with or without stabilize make- up the so-called emulsifying wax
- the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations.
- An emulsion can also contain a sweetening agent, a flavoring agent, a preservative, and/or an antioxidant.
- Emulsifiers and emulsion stabilizers suitable for use in the formulation of the present invention include Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate, sodium lauryl sulfate, glyceryl distearate alone or with a wax, or other materials well known in the art.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, also be delivered intravenously, subcutaneously, and/or intramuscularly via any pharmaceutically acceptable and suitable injectable form.
- Exemplary injectable forms include, but are not limited to, for example, sterile aqueous solutions comprising acceptable vehicles and solvents, such as, for example, water, Ringer’s solution, and isotonic sodium chloride solution; sterile oil-in-water microemulsions and aqueous or oleaginous suspensions.
- Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in the formulations for oral administration or by using other suitable dispersing or wetting agents and suspending agents.
- the compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum, and/or various buffers.
- Other adjuvants and modes of administration are well and widely known in the pharmaceutical art.
- the active ingredient may also be administered by injection as a composition with suitable carriers, including saline, dextrose, water or with cyclodextrin solubilization (i.e. Captisol), cosolvent solubilization (i.e. propylene glycol) or micellar solubilization (i.e. Tween 80).
- the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3- butanediol.
- a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3- butanediol.
- acceptable vehicles and solvents that may be employed are water, Ringer’s solution, and isotonic sodium chloride solution.
- sterile, fixed oils are conventionally employed as a solvent or suspending medium.
- any bland fixed oil may be employed, including synthetic mono- or diglycerides.
- fatty acids such as oleic acid find use in the preparation of injectables.
- a sterile injectable oil-in-water microemulsion can, for example, be prepared by 1) dissolving at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above in an oily phase, such as, for example, a mixture of soybean oil and lecithin; 2) combining the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above containing oil phase with a water and glycerol mixture; and 3) processing the combination to form a microemulsion.
- an oily phase such as, for example, a mixture of soybean oil and lecithin
- a sterile aqueous or oleaginous suspension can be prepared in accordance with methods already known in the art.
- a sterile aqueous solution or suspension can be prepared with a non-toxic parenterally-acceptable diluent or solvent, such as, for example, 1,3-butane diol; and a sterile oleaginous suspension can be prepared with a sterile non- toxic acceptable solvent or suspending medium, such as, for example, sterile fixed oils, e.g., synthetic mono- or diglycerides; and fatty acids, such as, for example, oleic acid.
- Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art.
- Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Allen, L. V. Jr.
- compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-alpha-tocopherol poly ethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, polyethoxylated castor oil such as CREMOPHOR surfactant (BASF), or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts,
- Cyclodextrins such as alpha-, beta-, and gamma-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-cyclodextrins, or other solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein.
- the pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals.
- the pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc.
- Tablets and pills can additionally be prepared with enteric coatings.
- Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents.
- adjuvants such as wetting, sweetening, flavoring, and perfuming agents.
- the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration.
- the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration.
- Such capsules or tablets may contain a controlled-release formulation as may be provided in a dispersion of active compound in hydroxypropylmethyl cellulose.
- the amounts of compounds that are administered and the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention depend on a variety of factors, including the age, weight, sex, the medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods.
- the daily dose can be administered in one to four doses per day. Other dosing schedules include one dose per week and one dose per two day cycle.
- compositions of this invention comprise at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and optionally an additional agent selected from any pharmaceutically acceptable carrier, adjuvant, and vehicle.
- compositions of this invention comprise a compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, or a prodrug, or isotope enriched forms thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
- the present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of eIF4a-associated diseases.
- the present invention also relates to a kit containing a formulation comprising: a) a pharmaceutical composition comprising a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, or a therapeutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of the activity eIF4A, is effective in treating the disorder.
- a pharmaceutical composition comprising a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, or a therapeutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for
- kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art.
- kit components such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art.
- Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
- the dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired.
- the daily oral dosage of each active ingredient when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day.
- the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion.
- Compound(s) of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
- the compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g. oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices.
- Dosage forms may contain from about 1 milligram to about 200 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.1-95 % by weight based on the total weight of the composition.
- a typical capsule for oral administration contains at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture (250 mg), lactose (75 mg), and magnesium stearate (15 mg). The mixture is passed through a 60 mesh sieve and packed into a no.1 gelatin capsule.
- a typical injectable preparation is produced by aseptically placing at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture (250 mg) into a vial, aseptically freeze-drying and sealing.
- the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation.
- compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, alone or in combination with a pharmaceutical carrier.
- compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above can be used alone, in combination with other compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, or in combination with one or more other therapeutic agent(s), e.g. an anticancer agent or other pharmaceutically active material.
- therapeutic agent(s) e.g. an anticancer agent or other pharmaceutically active material.
- the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
- Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
- the selected dosage level will depend upon a variety of factors including the activity of the particular compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
- a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required.
- the physician or veterinarian could start doses of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
- a suitable daily dose of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect.
- Such an effective dose will generally depend upon the factors described above.
- oral, intravenous, intracerebroventricular and subcutaneous doses of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above for a patient will range from about 0.01 to about 50 mg per kilogram of body weight per day.
- the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
- dosing is one administration per day, every other day, twice per week or per week. While it is possible for compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition).
- FIG. 1 MTT assay for cell viability in 96 well cell culture plate
- Cells with RAS mutations were treated with compounds 1, 2 or C-1 for 48h and cell viability was evaluated by MTT assay. The results shown are mean from 3 independent experiments.
- Figure 2 The effect of the compounds in RAS activation. NanoBiT assay for the RAS GTP-loading was performed in HeLa cells transfected with LgBit-K, N and HRAS G12V and SmBit-CRAF-RBD. Cells were treated with compounds 1, 2 or C-1 for 2 h in serum-free DMEM (Dulbecco's modified Eagle's Medium).
- DMEM Dens modified Eagle's Medium
- sNF96.2 were purchased from ATCC and cultured in DMEM (10 % heat inactivated FBS+ 1 mM Na Pyruvate).
- ASPC-1 cells were purchased from DSMZ and cultured in RPMI-1640 (10 % heat inactivated FBS).
- NCI-H358 were purchased from ATCC and cultured in RPMI-1640 (10 % heat inactivated FBS).
- T24 and HCT-116 cells were cultured in Mccoy’s 5A medium (10% heat inactivated FBS).
- HT-1080 cells were cultured in EMEM (10 % heat inactivated FBS + 1 mM Na Pyruvate).
- HeLa cells were harvested with 0.05% Trypsin/0.02% EDTA in PBS and seeded in 6 well or 12 well cell culture plates at the concentration of 5x10 4 cells in complete DMEM (2 ml for 6 well plate and 1 ml for 12 well plate). After 1 day from seeding, the DNA were transfected with Flag tagged Ras or NanoBiT plasmid using PEI/PBS solution. NanoBit assay. The plasmids were transfected to HeLa cells and the cells were harvested after 1 day of transfection. The cells were seeded to 96 well white plate. Next day, the medium was changed to serum free DMEM for 2h with compounds. After incubation, Nano Glo assay was performed.
- Dual-Glo Luciferase Assay was performed according to the manufacturer’s instructions (Promega, N2920). The luminescence was measured using Tecan infinite (Tecan). IC50 calculations by non-linear regression were conducted with GraphPad Prism 9.
- MTT assay Cancer cells were seeded in 96 well plates (5x10 3 cells/well) in 80 ⁇ l in growth medium and cultured for 1 day in the incubator. Next day, 20 ⁇ l of compounds containing growth medium was added to each well and the cells were cultured for 48h. After incubation with compounds, 10 ⁇ l of MTT solution was added to the wells and incubated for 2-4 hours. After incubation with MTT, solubilization buffer was added and incubated over night.
- Compound A Compound 15 (0.1 g) was purified by SFC (column: DAICEL CHIRALCEL OJ(250 mm * 30 mm,10 um);mobile phase: [CO 2 -IPA(0.1%NH 3 H 2 O)]; B%:30%, isocratic elution mode) and SFC (column: REGIS (s,s) WHELK-O1 (250 mm * 30 mm,5 um);mobile phase: [CO2- MeOH(0.1%NH 3 H 2 O)];B%:50%, isocratic elution mode) to give two peaks. Peak 1 ent-A (7.9 mg, 15.24 ⁇ mol, 8% yield) as a white solid.
- the protocol for determining the unbound fraction and pharmacokinetics is routine practice and known to the skilled person.
- the compounds according to the invention have a much higher free fraction in human plasma. There is much more drug available for binding to a target compared to the compounds of the prior art.
- the human microsomal clearance (hMic Clint) is lower for compound according to the invention compared to C-1. Since the exposure (AUC, area under the concentration-time curve) of a drug depends, inter alia, on its clearance (the lower the clearance of a drug, the higher the exposure).
- the beneficial effect of a drug depends on its potency and exposure, so a lower clearance corresponds to a lower dose to provide the same beneficial effect.
- Table 1 It shows that 2 has higher AUC and Cave than C-1 when dosed at 2 mg/kg iv (intravenous).
- Table 2 It shows that 2 has higher AUC and Cave than C-1 when dosed at 10 mg/kg po (oral).
- Table 3 It shows that 2 has lower unbound clearance than C-1 which is why it has a higher unbound AUC at this dose.
- HT-Dialysis plates (Model HTD 96 b, Cat# 1006) and the dialysis membrane (molecular weight cut off 12-14 KDa, Cat# 1101) were purchased from HT Dialysis LLC (Gales Ferry, CT).
- Dialysis Buffer 100 mM sodium phosphate and 150 mM NaCl, pH 7.4 ⁇ 0.1
- Stop Solution (Acetonitrile containing tolbutamide at 200 ng/mL, labetalol at 200 ng/mL) Processing Procedure of Dialysis Membrane and Matrix: On the day of experiment, the plasma was thawed under running cold tap water and centrifuged at 3220 ⁇ g for 5 min to remove any clots.
- the pH value was checked and recorded. Only plasma within a range of pH 7.0 to pH 8.0 was used.
- the dialysis membrane was pretreated according to the manufacturer's instructions: the dialysis membrane strips were soaked in ultra-pure water at room temperature for approximately 1 hr. Afterwards, each membrane strip that contains 2 membranes was separated and soaked in ethanol:water (20:80 v:v) for approximately 20 min, after which it was ready for use or was stored in the solution at 2-8°C for up to 1 month. Prior to the experiment, the membrane was rinsed and soaked for 20 min in ultra-pure water for use. Dilution Procedure of Test Compound and Control Compound: Working solutions (400 ⁇ M) of test compound and control compound were prepared.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention relates to novel compounds and their use as a medicament, in particular for use in treating proliferative disorders. The present invention relates further to a pharmaceutical composition comprising the novel compounds. Moreover, the present invention relates to a method of inhibiting proliferation or metastasis of cancer cells or inducing their cell death in a subject in need thereof. In addition, the present invention relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting RAS, in particular KRAS, HRAS and NRAS, activation in vitro. Furthermore, the present invention relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting elF4A complex or engaging PHB1/2 complex in the plasma membrane in vitro. Furthermore, the present invention relates to a kit containing a formulation comprising a pharmaceutical composition comprising a compound according to the invention.
Description
Novel RAS inhibitors The present invention relates to novel compounds and their use as a medicament, in particular for use in treating proliferative disorders. The present invention relates further to a pharmaceutical composition comprising the novel compounds. Moreover, the present invention relates to a method of inhibiting proliferation or metastasis of cancer cells or inducing their cell death in a subject in need thereof. In addition, the present invention relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting RAS, in particular KRAS, HRAS and NRAS, activation in vitro. Furthermore, the present invention relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting eIF4A complex or engaging PHB1/2 complex in the plasma membrane or cytosol in vitro. Furthermore, the present invention relates to a kit containing a formulation comprising a pharmaceutical composition comprising a compound according to the invention. BACKGROUND OF THE INVENTION Several approaches are known for the treatment of tumor diseases. A first possibility is the inhibition of RAS proteins, one of the major oncogenes which are mutationally activated in a large section of human cancers. The RAS oncogenes are frequently mutated in human cancers and among the three isoforms (KRAS, HRAS and NRAS), KRAS is the most frequently mutated oncogene. It is known that some of flavaglines like rocaglamide, a class of natural anti-tumour drugs and chemical ligands of prohibitins, inhibit RAS activation in cells by uncoupling the interaction between RAS and its effectors in the plane of the plasma membrane. Although a treatment with rocaglamide inhibits RAS-activation in KRAS-mutated cell lines, there is still a demand for compounds with a better activity towards RAS oncogenes. It is also known that various flavagline derivatives exhibit cytotoxic properties. WO 2005/113529 A2 describes cyclopenta[b]benzofuran derivatives and their utilization for the production of medicaments, especially for the prophylaxis and/or therapy of acute or chronic diseases. WO 2010/060891 A1 describes rocaglaol derivatives and the use of these derivatives to prevent or to limit the cardiotoxicity of an anti-neoplastic agent. However, to reach the
compounds of formula (I) according to the invention several selections from lists have to be made. WO 2012/0666002 A1 describes flavagline derivatives and their use as neuroprotective and/or cardioprotective and/or anti-tumor agents. WO 2017/058768 A1 describes compounds having activity as inhibitors of G12C mutant KRAS proteins. WO 2020/078975 A1 relates to inhibitors of KRAS oncogene activation, which are flavagline derivatives with the ability to target prohibitin to inhibit KRAS activation. However, to reach the compounds of formula (I) according to the invention several selections from lists have to be made. N. Ribeiro et al., J. Med. Chem., 2012, 55, 100064 and Thuaud et al. Med. Chem., 2011, 54, 411 relates to flavagline derivates, in particular FL3 and FL23 which are effective in inhibition of cell proliferation and enhancement of viability at lower doses compared to rocaglalol. H. Yurugi et. al, Journal of Cell Science,133, 1, 2020, relates to a study of a subset of flavaglines, which may inhibits KRAS nanoclustering and activation. However, to reach the compounds of formula (I) according to the invention several selections from lists have to be made. Furthermore, it is known that patients frequently develop resistance to RAS oncogene inhibitors, like KRAS G12 C inhibitors (Tanaka et al., Cancer Discov, 2021, PMID 33824136). In addition, the patients treated with KRAS G12 C inhibitors often develop secondary mutations in other RAS isoforms (Awad MM et al. New England J. of Med., 2021, PMID34161704). As mentioned before, there are already some approaches to inhibit RAS oncogenes. However, effective targeting of this gene with small molecules is still a challenge. Another option for the treatment of tumor diseases is the inhibition of dysregulation of protein translation. In other words, there are some developments targeting eukaryotic initiation factor (eIF4A complex) that integrates multiple oncogenic signalling inputs to the
translation apparatus which leads to the expression of cancer driving genes like c-myc, cyclin D1 etc. Eukaryotic initiation factor 4A (eIF4A) is a DEAD-box protein containing ATPase and ATP- dependent RNA helicase required to melt local secondary structure and facilitate access of the ribosome to the mRNA template. The factor regulates the cap-dependent protein synthesis. In mammals, there are three isoforms of eIF4A (eIF4AI, II and III), wherein eIF4AII and eIF4AIII share ~90% and ~65% identity, respectively, with the most abundant cellular factor eIF4AI. All isoforms are DEAD-box RNA helicase family members but only the paralogs eIF4AI and eIF4AII are found in the eIF4F complex and participate in translation initiation. WO 2017/091585 describes compounds having activity as inhibitors of eIF4A. However, the disclosed compounds have a different structure compared to the compounds according to the present invention. Prohibitins are evolutionarily conserved proteins and recent studies revealed a critical role for prohibitins in the activation of RAS by enabling RAS-effector interaction in the plane of the plasma membrane. Polier et al, Chemistry and Biology, 19, 1093–1104, 2012 showed that rocaglamides target this interaction (PHB1-CRAF) to inhibit RAS-CRAF interaction. These are several follow up studies confirming these effects. Ernst et al, J. Med. Chem.2020, 63, 5879 describes that the flavagline compounds rocaglamide A and Zotatifin show inhibition properties towards protein synthesis by stabilizing a translation-incompetent complex for selecting messenger RNAs (mRNAs) with eIF4A. Accordingly, while advances have been made in this field there remains a significant need for compounds that specifically inhibit RAS and eFI4A activity, particularly with regard to eIF4A’s role in regulation of cancer pathways, as well as for associated compositions and methods. In contrast to the inhibition of RAS oncogenes, which takes place at the plasma membrane possibly by engaging prohibitins driven nanoclusters among others (H. Yurugi et. al, Journal of Cell Science,133, 1, 2020), the inhibition of the eukaryotic factor 4A
occurs in the cytosol. Therefore, two different oncogenic protein complexes are possibly targeted for profound inhibition of the tumour cell proliferation and survival. Further the activated RAS can also influence of the functioning of the eIF4A complexes through one of the effector pathways (MAPK cascades). As mentioned before, there are already some approaches to inhibit RAS oncogenes or to inhibit eFI4A activity. However, effective targeting of this genes with small molecules is still a challenge. It is therefore the object of the present invention to provide pharmaceutically active compounds that have the capability to inhibit the activation of RAS oncogenes, in particular in cells at nanomolar concentrations with high specificity. Furthermore, it is the object of the present invention to provide pharmaceutically active compounds that have the capability to inhibit the RAS and eFI4A activity, in particular in cells at nanomolar concentrations with high specificity, especially, with regard to both RAS’s and eIF4A’s role in regulation of cancer pathways. This object is achieved by the compounds of formula (I). SUMMARY OF THE INVENTION The invention relates to a compound of formula (I)
prodrug, or isotope enriched forms thereof, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from Br and CN, R2 is selected from hydrogen and methyl, R3 is selected from methyl and methoxy.
The invention further relates to compounds of formula (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrug, or isotope enriched forms thereof, or the pharmaceutically acceptable salts thereof
, or prodrugs, or isotope enriched forms thereof, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from Br and CN, R2 is selected from hydrogen and methyl, R3 is selected from methyl and methoxy. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use as a medicament. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment and/or prophylaxis of proliferative disorders or genetic disorders or inflammatory disorders. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or in form of a prodrug or isotope enriched forms thereof, for use in the treatment and/or prophylaxis of inflammatory disorders. In particular the inflammatory disorders are selected from endometriosis and adenomyosis, in particular where RAS is mutated. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a
prodrug, or isotope enriched forms thereof as defined above or below, for use in the treatment and/or prophylaxisof proliferative disorders or genetic disorders or inflammatory disorders which involves oncogenic RAS or eIF4A complex that controls the expression of genes selected from c-myc and cyclin D1. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment and/or prophylaxis genetic disorders, wherein RAS signaling is pathologically involved, in particular CFC (Cranio Facial Cutaneous) and NF1 (Neurofibromatosis Type 1). The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment and/or prophylaxis genetic disorders, wherein Myc signaling is pathologically involved. In particular the genetic disorders are selected from RASOpathies, in particular CFC (Cranio Facial Cutaneous) and NF1 (Neurofibromatosis Type 1). The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment of cancer. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use as inhibitor of RAS protein activation. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in the treatment or prevention of any diseases or conditions that are associated with the activity of RAS protein (RAS oncogene).
The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein RAS-signaling is involved, preferably wherein KRAS G12V, NRAS G12V, HRAS G12V, KRAS G12C, KRAS G12D, KRAS G12C/Y96D, KRAS G13C, KRAS G13D, KRASG13S, KRAS Q61H, KRAS Q61R or KRAS Q61K or wherein any activating mutation in KRAS, HRAS and NRAS is involved or wherein any mutation that acquires resistance to RAS inhibitors. In particular the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein RAS-signaling is involved. In particular the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, KRAS G12V, NRAS G12V, HRAS G12V, KRAS G12C, KRAS G12D, KRAS G12C/Y96D, KRAS G13C, KRAS G13D, KRASG13S, KRAS Q61H, KRAS Q61R or KRAS Q61K is involved. In particular the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein any activating mutation in KRAS, HRAS and NRAS is involved. In particular the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein eIF4A-Myc-signaling is involved.
In particular the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein any mutation that acquires resistance to RAS inhibitors or eIF4Ainhibotors. In particular the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, as defined above and below, for use as inhibitor of eIF4A complex or a ligand eIF4A complex or a ligand of Prohibitin. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug or isotope enriched forms thereof, for use as inhibitor of eukaryotic initiation factor 4A (eIF4A). In particular the invention relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use as inhibitor of eukaryotic initiation factor 4A (eIF4A), which controls expression of cancer driving genes, especially which controls expression c-myc and cyclin D1. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt, for use as a ligand of Prohibitins (PHB1/2 complex) in the plasma membrane or cytosol. The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein dysregulation of eIF4A is involved, preferably wherein EIF4A1, EIF4A2 or EIF4A3 or EIF4F complex is involved.
The invention further relates to a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, for use in treating proliferative disorders, wherein overexpression of Prohibitins (PHB/2) is involved. The invention further relates to a pharmaceutical composition, comprising a pharmaceutical effective amount of at least one compound selected from compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined above and below, and one or more pharmaceutical acceptable carrier. The invention further relates to a pharmaceutical composition, comprising a pharmaceutical effective amount of at least one compound selected from compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, for use in the prophylaxis and/or treatment of proliferative disorders or genetic disorders. The invention further relates to a method of inhibiting growth, proliferation, or metastasis of cancer cells in a subject in need thereof, said method comprising administering to the subject a therapeutically effective amount of at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof, or a pharmaceutical composition as defined above or below. The invention further relates to a method of inhibiting proliferation of a cell population sensitive towards inhibiting RAS activation, in particular inhibiting KRAS, HRAS or NRAS activation in vitro or ex vivo, the method comprising contacting the cell population with at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof or a pharmaceutical composition as defined above and below.
The invention further relates to method of inhibiting proliferation of a cell population sensitive towards inhibiting eIF4A or its downstream targets, in particular c-myc and/or cyclin D1 in vitro or ex vivo, the method comprising contacting the cell population with at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof a pharmaceutical composition as defined above and below. The invention further relates to a kit comprising formulation comprising: a1) at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof or a2) a pharmaceutical composition comprising at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable, or a prodrug, or isotope enriched forms thereof or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of RAS activation is effective in treating the disorder. The invention further relates to a kit comprising formulation comprising: a1) at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof or a2) a pharmaceutical composition comprising at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable, or a prodrug, or isotope enriched forms thereof or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of dysregulation of protein translation, wherein eIF4A is involved, is effective in treating the disorder. The invention further relates to a kit comprising formulation comprising: a1) at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable salt or form of a prodrug, or isotope enriched forms thereof
or a2) a pharmaceutical composition comprising at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) as defined above and below, an enantiomeric mixture thereof, wherein each of the compounds can be in the form of a pharmaceutically acceptable, or a prodrug, or isotope enriched forms thereof or a therapeutically acceptable salt thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of dysregulation of protein translation, wherein eIF4A is involved, is effective in treating the disorder, in particular including C-Myc upregulation and/ cyclin D1 upregulation is effective in treating the disorder. The invention has the following advantages: - The compounds according to the invention exhibit advantageous RAS inhibition properties. In other words, the compounds according to the invention qualify as inhibitors of RAS oncogene activation by inhibiting the prohibitin pathway, in particular inhibiting EGF-induced RAS-GTP loading in cells which is measured by the ability of RAS to bind to its effector proteins like RAF kinases. - The compounds according to the invention prevent the activation of RAS as the interaction between RAS and its effectors is uncoupled due to defects in nanoclustering of RAS in the plane of the plasma membrane. - It is possible to identify potential candidate substances, by conducting a screening of compounds according to the invention that are able to inhibit the activation of RAS by directly disrupting the interaction between activated RAS (both by EGF and mutational activation) and the RAS binding domain (RBD) of the CRAF kinase in cells or other RAS interaction domains like the RA (RAS associating) domain. - The compounds are obtainable on reasonable scale for further development. - The compounds are soluble with good pharmacokinetic/pharmacodynamic (PK/PD) properties. These compounds inhibit RAS irrespective of the mutations at nanomolar range, e.gand exhibit inhibitory effects on KRAS, NRAS and HRAS. - In addition, the compounds, in particular at increasing concentrations and at longer time points post treatment, inhibit eIF4A complex, which is responsible for the
translation of several oncogenes. This complex has been targeted by the so called STIs (Selective translational inhibitors). - A dual luciferase assay has been established to measure eIF4A activity in vitro and the compounds are further screened for the inhibition of eIF4A. DESCRIPTION OF THE INVENTION Compounds of formula (I) Unless specifically stated otherwise herein, references made in the singular may also include the plural. For example, “a” and “an” may refer to either one, or one or more. The compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), an enantiomeric mixture thereof, may form salts which are also within the scope of this invention. The term "salt(s)" as employed herein, denotes acidic and/or basic salts formed with inorganic and/or organic acids and bases. Pharmaceutically acceptable (i.e. non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful, e.g., in isolation or purification steps which may be employed during preparation. Salts of the compounds of formulae (I.a), (I.b), (A), (B), (C), (D), (E), or an enantiomeric mixture thereof may be formed, for example, by reacting a compound of formulae (I), (I.b), (A), (B), (C), or an enantiomeric mixture thereof with at least one acid or base. The acid or base is added in an amount suitable for partial or complete neutralization, e.g. an equivalent amount. The phrase "pharmaceutically acceptable salt(s)" as used herein, unless otherwise indicated, includes salts containing pharmacologically acceptable anions or cations, such as chloride, bromide, iodide, nitrate, sulfate, bisulfate, phosphate, hydrogen phosphate, dihydrogen phosphate, isonicotinate, acetate, lactate, salicylate, citrate, acid citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, sulfate, benzenesulfonate, p-toluenesulfonate and palmoate [i.e.4,4'- methylene-bis-(3-hydroxy-2-naphthoate)] salts. Any compound according to the invention can be prepared and used in prodrug form. A suitable prodrug has chemically or metabolically cleavable group(s) and becomes, by solvolysis or under physiological conditions, a compound that is pharmaceutically active in vivo. A prodrug can be formed in a conventional manner by reaction of a functional group of the compound (such as an amino, hydroxy or carboxy group). Prodrug often offer
advantages of better metabolism, potency, solubility, tissue compatibility, or delayed release in mammals. The term “prodrug” refers to a metabolic precursor of a compound of the invention that is pharmaceutically acceptable. A prodrug may be inactive when administered to a subject in need thereof but is converted in vivo to an active compound of the invention. Prodrugs are typically rapidly transformed in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. In other words, in the terms of the present invention, "prodrug" are compounds which are metabolized in vivo to give the compounds of the invention of formula I. Typical examples for prodrug are for example described in C.G. Wermeth (editor): The Practice of Medicinal Chemistry, Academic Press, San Diego, 1996, pages 671-715, as well as J. Rautio et al., NATURE REVIEWS, Drug Discovery, Vol.17, 2018, p.559. Examples are phosphates, carbonates, carbamates, aminoacids, esters (carboxylic acid esters), ethers, amides, peptides, ureas and the like. In the present case, suitable prodrugs can be compounds of formula I wherein the hydroxy group is bonded to any group that, when the prodrug of the compound of the invention is administered to a mammalian subject, cleaves to form a free hydroxy group. Myc is a family of regulator genes and proto-oncogenes that code for transcription factors. The Myc family consists of three related human genes: c-myc (MYC), l-myc (MYCL), and n-myc (MYCN). c-myc (also sometimes referred to as MYC) was the first gene to be discovered in this family, due to homology with the viral gene v-myc. In cancer, c-myc is often constitutively (persistently) expressed. This leads to the increased expression of many genes, some of which are involved in cell proliferation, contributing to the formation of cancer. Cyclin D1 is a protein that in humans is encoded by the CCND1 gene. The CCND1 gene encodes the cyclin D1 protein. The human CCND1 gene is located on the long arm of chromosome 11 (band 11q13). It is 13,388 base pairs long, and translates into 295 amino acids. Cyclin D1 is expressed in all adult human tissues with the exception of cells derived from bone marrow stem cell lines (both lymphoid and myeloid). In the context of the present invention, a chemical structure that does not explicitly show a specific stereochemical orientation usually means all possible stereoisomers and mixtures thereof, unless indicated otherwise, for example,
in which * designates the asymmetry centers. “Chiral compounds” in the sense of the invention are compounds that contain no improper axis of rotation (Sn). In the context of the present invention, they are in particular compounds with at least five chirality centers and without Sn-symmetry. “Stereoisomers” in the context of the invention are compounds of identical constitution but different atomic arrangement in the three-dimensional space. “Enantiomers” are stereoisomers which behave like image to mirror image to one another, e.g. compounds of formulae (I.a) and (I.b) are enantiomers. The “enantiomeric excess” (ee) achieved during asymmetric synthesis is given here by the following formula: ee [%]=(R−S)/(R+S)×100. R and S are the descriptors of the CIP system for the two enantiomers and describe the absolute configuration on the asymmetric atom. The enantiomerically pure compound (ee=100%) is also referred to as “homochiral compound”. “Diastereomers” are stereoisomers which are not enantiomeric to one another. The compound of the invention can exist in various isomeric forms, as well as in one or more tautomeric forms, including both single tautomers and mixtures of tautomers. The term “isomer” is intended to encompass all isomeric forms of a compound of this invention, including tautomeric forms of the compound. Some compounds described here can have asymmetric centers and therefore exist in different enantiomeric and diastereomeric forms. A compound of the invention can be in the form of an optical isomer or a diastereomer. Accordingly, the invention encompasses compounds of the invention and their uses as described herein in the form of their optical isomers, diastereoisomers and mixtures thereof, including a racemic mixture. Optical isomers of the compounds of the invention can be obtained by known techniques such as
asymmetric synthesis, chiral chromatography, or via chemical separation of stereoisomers through the employment of optically active resolving agents. Unless otherwise indicated“stereoisomer” means one stereoisomer of a compound that is substantially free of other stereoisomers of that compound. Thus, a stereomerically pure compound having one chiral center will be substantially free of the opposite enantiomer of the compound. A stereomerically pure compound having two chiral centers will be substantially free of other diastereomers of the compound. A typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, for example greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, or greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, or greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound. A“stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable. The present invention contemplates various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposable mirror images of one another. Compounds of the invention or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography. Relative configuration in stereochemistry (relative stereochemistry) is the arrangement of atoms or groups of atoms that is described relative to other atoms or groups of atoms in
the molecule. In other words, this term describes the position of atoms or groups of atoms in space in relation to other atoms or groups of atoms that are located elsewhere in the molecule. Absolute configuration in stereochemistry (absolute stereochemistry) is the arrangement of atoms or group of atoms that is described independently of any other atom or group of atoms in the molecule. This type of configuration is defined for chiral molecular entities and their stereochemical descriptions (e.g. R or S). Syn means that with regard to the orientation of the substituents on the 5-membered ring they are bound to (5 asymmetric carbon atoms) all substituents point in the same direction relative to the plane of the 5-membered ring. Racemic mixture or racemate is defined as a mixture of compounds consisting of two molecules structured like image and mirror image (= enantiomers) and which are present in an equimolar mixture, i.e. in the ratio 1:1 (50:50). Further, the compounds of formula (I)
in which * designates the asymmetry centers, represent the isomers of formulae (I.1), (I.2), (I.3), (I.4), (I.5), (I.6), (I.7), (I.8), (I.9), (I.10), (I.11), (I.12), (I.13), (I.14), (I.15), (I.16), (I.17), (I.18(, (I.19), (I.20), (I.21), (I.22), (I.23), (I.24), (I.25), (I.26), (I.27), (I.28), (I.29), (I.30), (I.31) and (I.32):
10
(I.7) (I.8)
5 10 (I.15) (I.16)
5 10 (I.23) (I.24)
wherein, R1, R2 and R3 have one of the meanings as defined above or below.
The compounds of formulae (I.1) to (I.32) are specified by their absolute stereochemistry. In a preferred embodiment the compound of formula (I) is a mixture of at least two enantiomers (I.1) to (I.32) or a mixture of the prodrugs, or isotope enriched forms thereof, or a mixture of the pharmaceutically acceptable salt thereof, wherein one enantiomer is enriched. Preferably the compound of formula (I) is a mixture of (I.a) and (I.b) or a mixture of the prodrug, or isotope enriched forms thereof or a mixture of the pharmaceutically acceptable salt thereof, wherein the enantiomer excess (ee) of the enantiomer of formula (I.a) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%. Preferred are compound of formula (I) according to the invention, or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, wherein R1 is CN. Preferred are compound of formula (I) according to the invention, or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, wherein R1 is Br. Further, preferred are compound of formula (I) according to the invention, or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, wherein R2 and R3 are both methyl. Further, preferred are compound of formula (I) according to the invention, or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, wherein R2 is hydrogen and R3 is methoxy. Further, preferred are compound of formula (I) according to the invention, or a prodrug, or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, wherein R2 is methyl and R3 is methoxy. Another special embodiment are the compounds of formula (I), wherein R1, R2 and R3 are selected from the definition given in one line of table 1:
Another special embodiment are the compounds selected from A, B, C, D, E, F and the mixture of the each of compounds A to F with its respective enantiomer:
Preferred is a compound of formula (A) or an enantiomeric mixture comprising the compounds of formula (A) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (A) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%.
Preferred is a compound of formula (B) or an enantiomeric mixture comprising the compounds of formula (B) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (B) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%. Preferred is a compound of formula (C) or an enantiomeric mixture comprising the compounds of formula (C) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (C) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%. Preferred is a compound of formula (D) or an enantiomeric mixture comprising the compounds of formula (D) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (D) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%. Preferred is a compound of formula (E) or an enantiomeric mixture comprising the compounds of formula (E) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (E) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%. Preferred is a compound of formula (F) or an enantiomeric mixture comprising the compounds of formula (F) and its enantiomer, in particular wherein the enantiomer excess (ee) of the enantiomer of formula (F) is at least 20%, preferably at least 50%, in particular at least 80%, especially at least 99%. Especially preferred is compound A. Further, especially preferred is compound F. The compounds of the present invention can be synthesized using the methods known in the prior art and together with methods known from synthetic organic chemistry, or variations thereof as appreciated by those skilled in the art. Pharmaceutical composition The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of
sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response or other problem or complication, commensurate with a reasonable benefit/risk ratio. The phrase "therapeutically effective" is intended to qualify the amount of each agent, which will achieve the goal of improvement in disorder severity and the frequency of incidence, while avoiding adverse side-effects typically associated with alternative therapies. For example, effective anticancer agents prolong the survivability of the patient or his/her life quality, inhibit the rapidly proliferating cell growth associated with the neoplasm, or effect a regression of the neoplasm. The terms “treat,” “treating,” and “treatment,” as used herein, refer to any type of intervention or process performed on, or administering an active agent to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting or slowing down or preventing the progression, development, severity or recurrence of a symptom, complication, condition or biochemical indicia associated with a disease. By contrast, “prophylaxis” or “prevention” refers to administration to a subject who does not have a disease to prevent the disease from occurring. As used herein, the term "cell" is meant to refer to a cell that is in vitro, ex vivo or in vivo. In the sense of the invention, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In the sense of the invention, an in vitro cell can be a cell in a cell culture. In the sense of the invention, an in vivo cell is a cell living in an organism such as a mammal. The term “patient” includes humans and animals that receive either therapeutic or prophylactic treatment. The term “subject” includes any human or animal. For example, the methods and compositions herein disclosed can be used to treat a subject having cancer. A (non-human) animal includes all vertebrates, e.g. mammals and non-mammals, including cows, sheep, pigs, goats, horses, poultry, dogs, cats, non-human primates, rodents etc. In one embodiment, the subject is a human subject. The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid
diluent, solvent, excipient, manufacturing aid (e.g. lubricant) or encapsulating material, involved in carrying or transporting the subject compound from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation. Suitable other ingredients are the afore-mentioned carrier and further additives, including adjuvants, preserving agents, fillers, flow regulating agents, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, bittering agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents, dispensing agents, etc.. Suitable additives are selected depending on the nature of the mode of administration and dosage forms; and not injurious to the patient. The term "pharmaceutical composition" means a composition comprising a compound of the invention in combination with at least one further compound selected from a) at least one further pharmaceutically active substance and b) at least one additional pharmaceutically acceptable carrier and or additive. RAS The term "RAS inhibitor" refers to an agent capable of decreasing RAS protein levels, decreasing RAS activity levels and/or inhibiting RAS expression levels in the cells. The RAS inhibitor may be a reversible or irreversible inhibitor. As used herein, “RAS” protein refers to a protein that is a member of a family of related proteins that are expressed in all human and animal cell lineages and organs. All RAS protein family members belong to a class of proteins called small GTPase (also known as small G proteins, a family of hydrolase enzymes that can bind and hydrolyse GTP), and are involved in transmitting signals within cells (cellular signal transduction). RAS is the prototypical member of the RAS superfamily of proteins, which are all related in three-dimensional structure and regulate diverse cell behaviours. When RAS is 'switched on' by incoming signals, it subsequently switches on other proteins, which ultimately turn on genes involved in cell growth, differentiation, and survival. Mutations in RAS genes can lead to the production of permanently activated RAS proteins, which can cause unintended and overactive signaling inside the cell, even in the absence of incoming signals. Because these signals result in cell growth and division, overactive RAS signaling can ultimately lead to cancer. The three RAS genes in humans (HRAS, KRAS, and NRAS) are the most common oncogenes in human cancer. As mentioned, the clinically most notable members of the RAS subfamily are HRAS, KRAS and NRAS. However, there are other members of this
subfamily, which are e.g. selected from DIRAS1, DIRAS2, DIRAS3, ERAS, GEM, MRAS, NKIRAS1, NKIRAS2, NRAS, RALA, RALB, RAP1A, RAP1B, RAP2A, RAP2B, RAP2C, RASD1, RASD2, RASL10A, RASL10B, RASL11A, RASL11B, RASL12, REM1, REM2, RERG, RERGL, RRAD, RRAS, RRAS2. The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and a pharmaceutical composition comprising in at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be administered to humans and animals, preferably humans. In principle any method of administration may be used to deliver the compound or pharmaceutical composition according to the invention to a subject. Suitable methods of administration are orally, enterally, parenterally, intravenously, topically, intramuscular, subcutaneous/dermal routes. The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above can selectively decrease RAS protein levels, decrease RAS activity levels and/or inhibit RAS expression levels in the cells. For example, the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above can be used to selectively decrease RAS activity levels and/or inhibit RAS expression levels in cells or in an individual in need of a decrease in RAS protein levels, decrease in RAS activity levels and/or inhibition of RAS expression levels by administering an inhibiting amount of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above or a salt thereof. In one embodiment, the present invention provides a combined preparation of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or a pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and an additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases, preferably of proliferative disorders (e.g. cancer), in particular disorders associated with the activity of RAS protein. Additional therapeutic agent(s) are selected from chemotherapeutic agents, radiotherapeutic agents, immuno-oncology agents, and combinations thereof.
In one aspect, the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above are sequentially administered prior to administration of the immuno-oncology agent. In another aspect, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above are administered concurrently with the immuno-oncology agent. In yet another aspect, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above are sequentially administered after administration of the immuno-oncology agent. In another aspect, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be co-formulated with an immuno- oncology agent. Immuno-oncology agents include, for example, a small molecule drug, antibody or other biologic or small molecule. Examples of biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In one aspect, the antibody is a monoclonal antibody. In another aspect, the monoclonal antibody is humanized or human. In one aspect, the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators). Suitable of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to co- stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTpR, LIGHT, DcR3, HVEM, VEGETL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin a/TNFp, TNFR2, TNFa, LTpR, Lymphotoxin a 1b2, FAS, FASL, RELT, DR6, TROY, NGFR.
In one aspect, T cell responses can be stimulated by a combination of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4- 1BB (CD 137), 4-1BBL, ICOS, ICOS-L, 0X40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H. Other agents that can be combined with compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells. For example, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above can be combined with antagonists of KIR, such as Lirilumab. Yet other agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155. The combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally. Alternatively, for example, all therapeutic agents may be administered
orally or all therapeutic agents may be administered by intravenous injection. Combination therapy can also embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g surgery or radiation treatment.) Where the combination therapy further comprises a non-drug treatment, the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks. Types of cancers that may be treated with the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above include, but are not limited to, prostate, colon, rectum, pancreas, cervix, stomach, endometrium, brain, liver, bladder, ovary, testis, head, neck, skin (including melanoma and basal carcinoma), mesothelial lining, white blood cell (including lymphoma and leukemia), esophagus, breast, muscle, connective tissue, lung (including small cell lung carcinoma and non- small-cell carcinoma), adrenal gland, thyroid, kidney, or bone; or glioblastoma, mesothelioma, renal cell carcinoma, gastric carcinoma, sarcoma (including Kaposi's sarcoma), choriocarcinoma, cutaneous basocellular carcinoma, haematological malignancies (including blood, bone marrow and lymph nodes) or testicular seminoma. One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g ., IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above for treatment of RAS protein associated diseases, disorders or conditions. The agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms. Suitable chemotherapeutic or other anti-cancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine, cyclophosphamide (CYTOXAN®), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.
In the treatment of melanoma, suitable agents for use in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the "Dartmouth regimen", which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOY™. Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may also be combined with immunotherapy drugs, including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in the treatment of melanoma. Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may also be used in combination with vaccine therapy in the treatment of melanoma. Antimelanoma vaccines are, in some ways, similar to the anti-virus vaccines which are used to prevent diseases caused by viruses such as polio, measles, and mumps. Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body's immune system to destroy melanoma cells. Melanomas that are confined to the arms or legs may also be treated with a combination of agents including one or more compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined, using a hyperthermic isolated limb perfusion technique. This treatment protocol temporarily separates the circulation of the involved limb from the rest of the body and injects high doses of chemotherapy into the artery feeding the limb, thus providing high doses to the area of the tumor without exposing internal organs to these doses that might otherwise cause severe side effects. Usually the fluid is warmed to 38.9 °C to 40 °C. Melphalan is the drug most often used in this chemotherapy procedure. This can be given with another agent called tumor necrosis factor (TNF). Suitable chemotherapeutic or other anti-cancer agents include, for example, antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine. Suitable chemotherapeutic or other anti-cancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine,
vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxyco-formycin, mitomycin-C, L- asparaginase, interferons (especially IFN-a), etoposide, and teniposide. Other cytotoxic agents include navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafme, and droloxafme. Also suitable are cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors. Other anti-cancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-IO or TGF-b). Other anti-cancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4. Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer. Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses. In a specific embodiment of the present invention, at least one compound of (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and at least one chemotherapeutic agent are administered to the patient concurrently or sequentially. In other words, at least one compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be administered first, at least one chemotherapeutic agent may be administered first, or at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be administered at the same time. Additionally, when more than one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or chemotherapeutic agent is used, the compounds may be administered in any order.
The invention also provides pharmaceutically compositions which comprise a therapeutically effective amount of one or more of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally, one or more additional therapeutic agents described above. The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. The compound(s) and compositions of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) an enantiomeric mixture as defined above, can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques ( e.g. as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice. For oral administration, the pharmaceutical composition may be in the form of, for example, a tablet, capsule, liquid capsule, suspension, or liquid. The pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient. For example, the pharmaceutical composition may be provided as a tablet or capsule comprising an amount of active ingredient in the range of from about 0.1 to 1000 mg, preferably from about 0.25 to 250 mg, and more preferably from about 0.5 to 100 mg. A suitable daily dose for a human or animal may vary widely depending on the condition of the patient and other factors, but, can be determined using routine methods. Any pharmaceutical composition contemplated herein can, for example, be delivered orally via any acceptable and suitable oral preparations. Exemplary oral preparations, include, but are not limited to, for example, tablets, troches, lozenges, aqueous and oily
suspensions, dispersible powders or granules, emulsions, hard and soft capsules, liquid capsules, syrups, and elixirs. Pharmaceutical compositions intended for oral administration can be prepared according to any methods known in the art for manufacturing pharmaceutical compositions intended for oral administration. In order to provide pharmaceutically palatable preparations, a pharmaceutical composition in accordance with the invention can contain at least one agent selected from sweetening agents, flavoring agents, bittering agents, coloring agents, demulcents, antioxidants, and preserving agents. A tablet can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one non-toxic pharmaceutically acceptable excipient suitable for the manufacture of tablets. Exemplary excipients include, but are not limited to, for example, inert diluents, such as, for example, calcium carbonate, sodium carbonate, lactose, calcium phosphate, and sodium phosphate; granulating and disintegrating agents, such as, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, and alginic acid; binding agents, such as, for example, starch, gelatin, polyvinyl-pyrrolidone, and acacia; and lubricating agents, such as, for example, magnesium stearate, stearic acid, and talc. Additionally, a tablet can either be uncoated, or coated by known techniques to either mask the bad taste of an unpleasantly tasting drug, or delay disintegration and absorption of the active ingredient in the gastrointestinal tract thereby sustaining the effects of the active ingredient for a longer period. Exemplary water soluble taste masking materials, include, but are not limited to, hydroxypropyl-methylcellulose and hydroxypropyl- cellulose. Exemplary time delay materials, include, but are not limited to, ethyl cellulose and cellulose acetate butyrate. Hard gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or at least one salt thereof with at least one inert solid diluent, such as, for example, calcium carbonate; calcium phosphate; and kaolin. Soft gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one water soluble carrier, such as,
for example, polyethylene glycol; and at least one oil medium, such as, for example, peanut oil, liquid paraffin, and olive oil. An aqueous suspension can be prepared, for example, by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one excipient suitable for the manufacture of an aqueous suspension. Exemplary excipients suitable for the manufacture of an aqueous suspension, include, but are not limited to, for example, suspending agents, such as, for example, sodium carboxymethylcellulose, hydroxypropyl- methylcellulose and hydroxypropyl- cellulose, sodium alginate, alginic acid, polyvinyl- pyrrolidone, gum tragacanth, and gum acacia; dispersing or wetting agents, such as, for example, a naturally-occurring phosphatide, e.g., lecithin; condensation products of alkylene oxide with fatty acids, such as, for example, polyoxyethylene stearate; condensation products of ethylene oxide with long chain aliphatic alcohols, such as, for example heptadecaethylene-oxycetanol; condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol, such as, for example, polyoxyethylene sorbitol monooleate; and condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, such as, for example, polyethylene sorbitan monooleate. An aqueous suspension can also contain at least one preservative, such as, for example, ethyl and n-propyl p-hydroxybenzoate; at least one coloring agent; at least one flavoring agent; and/or at least one sweetening agent, including but not limited to, for example, sucrose, saccharin, and aspartame. Oily suspensions can, for example, be prepared by suspending at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof in either a vegetable oil, such as, for example, arachis oil, olive oil, sesame oil and coconut oil or in mineral oil, such as, for example, liquid paraffin. An oily suspension can also contain at least one thickening agent, such as, for example, beeswax, hard paraffin and cetyl alcohol. In order to provide a palatable oily suspension, at least one of the sweetening agents already described hereinabove, and/or at least one flavoring agent can be added to the oily suspension. An oily suspension can further contain at least one preservative, including, but not limited to, for example, an anti- oxidant, such as, for example, butylated hydroxyanisol, and alpha-tocopherol.
Dispersible powders and granules can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one dispersing and/or wetting agent; at least one suspending agent; and/or at least one preservative. Suitable dispersing agents, wetting agents, and suspending agents are as already described above. Exemplary preservatives include, but are not limited to, for example, anti-oxidants, e.g., ascorbic acid. In addition, dispersible powders and granules can also contain at least one excipient, including, but not limited to, for example, sweetening agents; flavoring agents; and coloring agents. An emulsion of at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, be prepared as an oil-in-water emulsion. The oily phase of the emulsions comprising compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above may be constituted from known ingredients in a known manner. The oil phase can be provided by, but is not limited to, for example, a vegetable oil, such as, for example, olive oil and arachis oil; a mineral oil, such as, for example, liquid paraffin; and mixtures thereof. While the phase may comprise merely an emulsifier, it may comprise a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Suitable emulsifying agents include, but are not limited to, for example, naturally-occurring phosphatides, e.g., soy bean lecithin; esters or partial esters derived from fatty acids and hexitol anhydrides, such as, for example, sorbitan monooleate; and condensation products of partial esters with ethylene oxide, such as, for example, polyoxyethylene sorbitan monooleate. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilize) make- up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations. An emulsion can also contain a sweetening agent, a flavoring agent, a preservative, and/or an antioxidant. Emulsifiers and emulsion stabilizers suitable for use in the formulation of the present invention include Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate, sodium lauryl sulfate, glyceryl distearate alone or with a wax, or other materials well known in the art.
The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, also be delivered intravenously, subcutaneously, and/or intramuscularly via any pharmaceutically acceptable and suitable injectable form. Exemplary injectable forms include, but are not limited to, for example, sterile aqueous solutions comprising acceptable vehicles and solvents, such as, for example, water, Ringer’s solution, and isotonic sodium chloride solution; sterile oil-in-water microemulsions and aqueous or oleaginous suspensions. Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in the formulations for oral administration or by using other suitable dispersing or wetting agents and suspending agents. The compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum, and/or various buffers. Other adjuvants and modes of administration are well and widely known in the pharmaceutical art. The active ingredient may also be administered by injection as a composition with suitable carriers, including saline, dextrose, water or with cyclodextrin solubilization (i.e. Captisol), cosolvent solubilization (i.e. propylene glycol) or micellar solubilization (i.e. Tween 80). The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer’s solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. A sterile injectable oil-in-water microemulsion can, for example, be prepared by 1) dissolving at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above in an oily phase, such as, for example, a mixture of soybean oil and lecithin; 2) combining the compound(s) of formulae (I), (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above containing oil phase with a water and glycerol mixture; and 3) processing the combination to form a microemulsion.
A sterile aqueous or oleaginous suspension can be prepared in accordance with methods already known in the art. For example, a sterile aqueous solution or suspension can be prepared with a non-toxic parenterally-acceptable diluent or solvent, such as, for example, 1,3-butane diol; and a sterile oleaginous suspension can be prepared with a sterile non- toxic acceptable solvent or suspending medium, such as, for example, sterile fixed oils, e.g., synthetic mono- or diglycerides; and fatty acids, such as, for example, oleic acid. Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include, without limitation: the type and nature of the active agent being formulated; the subject to which the agent- containing composition is to be administered; the intended route of administration of the composition; and the therapeutic indication being targeted. Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Allen, L. V. Jr. et al. Remington: The Science and Practice of Pharmacy (2 Volumes), 22nd Edition (2012), Pharmaceutical Press. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-alpha-tocopherol poly ethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, polyethoxylated castor oil such as CREMOPHOR surfactant (BASF), or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins such as alpha-, beta-, and gamma-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-cyclodextrins, or other
solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein. The pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals. The pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. Tablets and pills can additionally be prepared with enteric coatings. Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents. For therapeutic purposes, the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration. If administered orally, the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration. Such capsules or tablets may contain a controlled-release formulation as may be provided in a dispersion of active compound in hydroxypropylmethyl cellulose. The amounts of compounds that are administered and the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention depend on a variety of factors, including the age, weight, sex, the medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods. A daily dose of about 0.001 to 100 mg/kg body weight, preferably between about 0.0025 and about 50 mg/kg body weight and most preferably between about 0.005 to 10 mg/kg body weight, may be appropriate. The daily dose can be administered in one to four doses per day. Other dosing schedules include one dose per week and one dose per two day cycle. Pharmaceutical compositions of this invention comprise at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and optionally an additional agent selected
from any pharmaceutically acceptable carrier, adjuvant, and vehicle. Alternate compositions of this invention comprise a compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, or a prodrug, or isotope enriched forms thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of RAS protein-associated diseases. Thus, the present invention also relates to a kit containing a formulation comprising: a) a pharmaceutical composition comprising a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, or a therapeutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of RAS activation is effective in treating the disorder. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit. The dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired. By way of general guidance, the daily oral dosage of each active ingredient, when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day. Intravenously, the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion. Compound(s) of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g. oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices. Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 200 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.1-95 % by weight based on the total weight of the composition. A typical capsule for oral administration contains at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture (250 mg), lactose (75 mg), and magnesium stearate (15 mg). The mixture is passed through a 60 mesh sieve and packed into a no.1 gelatin capsule. A typical injectable preparation is produced by aseptically placing at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture (250 mg) into a vial, aseptically freeze-drying and sealing. For use, the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation. The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, alone or in combination with a pharmaceutical carrier. Optionally, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above can be used alone, in combination with other compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, or in combination with one or more other therapeutic agent(s), e.g. an anticancer agent or other pharmaceutically active material. Regardless of the route of administration selected, the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F), (F) and an enantiomeric mixture as defined above, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of factors including the activity of the particular compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, a suitable daily dose of compound(s) of formulae (I), (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, oral, intravenous, intracerebroventricular and subcutaneous doses of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above for a patient will range from about 0.01 to about 50 mg per kilogram of body weight per day. If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In certain aspects of the invention, dosing is one administration per day.
While it is possible for compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition). The above other therapeutic agents, when employed in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, may be used, for example, in those amounts indicated in the Physicians’ Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art. In the methods of the present invention, such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the inventive compounds. Eukaryotic initiation factor 4A The mechanisms governing the basic subsistence of eukaryotic cells are immensely complex; it is therefore unsurprising that regulation occurs at a number of stages of protein synthesis. Human translational control is of increasing research interest as it has connotations in a range of diseases. Orthologs of many of the factors involved in human translation are shared by a range of eukaryotic organisms. Synthesis of protein from mature messenger RNA in eukaryotes is divided into translation initiation, elongation, and termination of these stages; the initiation of translation is the rate limiting step. Within the process of translation initiation; the bottleneck occurs shortly before the ribosome binds to the 5’ m7GTP facilitated by a number of proteins; it is at this stage that constrictions born of stress, amino acid starvation etc. take effect. Eukaryotic initiation factor complex 2 (eIF2) forms a ternary complex with GTP and the initiator Met-tRNA – this process is regulated by guanine nucleotide exchange and phosphorylation and serves as the main regulatory element of the bottleneck of gene expression. Before translation can progress to the elongation stage, a number of initiation factors must facilitate the synergy of the ribosome and the mRNA and ensure that the 5’ UTR of the mRNA is sufficiently devoid of secondary structure. Binding in this way is facilitated by group 4 eukaryotic initiation factors; eIF4F has implications in the normal regulation of translation as well as the transformation and progression of cancerous cells. eIF4F is responsible for the binding of capped mRNA to the 40S ribosomal subunit via eIF3. The mRNA cap is bound by eIF4E (25 kDa), eIF4G (185 kDa) acts as a scaffold for the complex whilst the ATP-dependent RNA helicase eIF4A (46 kDa) processes the
secondary structure of the mRNA 5’ UTR to render it more conducive to ribosomal binding and subsequent translation. Together these three proteins are referred to as eIF4F. For maximal activity; eIF4A also requires eIF4B (80 kDa), which itself is enhanced by eIF4H (25 kDa). Once bound to the 5’ cap of mRNA, this 48S complex then searches for the (usually) AUG start codon and translation begins. The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and a pharmaceutical composition comprising in at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be administered to humans and animals, preferably humans. In principle any method of administration may be used to deliver the compound or pharmaceutical composition according to the invention to a subject. Suitable methods of administration are orally, enterally, parenterally, intravenously, topically, intramuscular, subcutaneous routes. The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above can selectively decrease activity of eIF4A, decrease eIF4A activity levels and/or inhibit activity of eIF4 in the cytosol. For example, the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above can be used to selectively decrease eIF4A activity levels and/or inhibit eIF4A in cytosol or in an individual in need of a decrease in activity of eIF4A, decrease in eIF4A activity and/or inhibition of eIF4A activity levels by administering an inhibiting amount of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above or a salt or a prodrug, or isotope enriched forms thereof. In one embodiment, the present invention provides a combined preparation of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, and/or a pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and an additional therapeutic agent(s) for simultaneous, separate or sequential use in the treatment and/or prophylaxis of multiple diseases, preferably of proliferative disorders (e.g. cancer), in particular disorders associated with the activity of eIF4A.
Additional therapeutic agent(s) are selected from chemotherapeutic agents, radiotherapeutic agents, immuno-oncology agents, and combinations thereof. In one aspect, the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above are sequentially administered prior to administration of the immuno-oncology agent. In another aspect, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above are administered concurrently with the immuno-oncology agent. In yet another aspect, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above are sequentially administered after administration of the immuno-oncology agent. In another aspect, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be co-formulated with an immuno- oncology agent. Immuno-oncology agents include, for example, a small molecule drug, antibody or other biologic or small molecule. Examples of biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In one aspect, the antibody is a monoclonal antibody. In another aspect, the monoclonal antibody is humanized or human. In one aspect, the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co- inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators). Suitable of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to co- stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTpR, LIGHT, DcR3, HVEM, VEGETL1A, TRAMP/DR3,
EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin a/TNFp, TNFR2, TNFa, LTpR, Lymphotoxin a 1b2, FAS, FASL, RELT, DR6, TROY, NGFR. In one aspect, T cell responses can be stimulated by a combination of compound(s) of formulae (I), (I-A),(I.a’), (I-A.a’), (I.a), (I-A.a), (A), (B), (C), (D) , (E), (F), (G), (H), (I), (J), (K), (L), (M), (N), (O), (P), (Q), (R), (S), (T), (U), (V) and an enantiomeric mixture as defined above and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4- 1BB (CD 137), 4-1BBL, ICOS, ICOS-L, 0X40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H. Other agents that can be combined with compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells. For example, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above can be combined with antagonists of KIR, such as Lirilumab. Yet other agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155. The combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The
therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally. Alternatively, for example, all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection. Combination therapy can also embrace the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g surgery or radiation treatment.) Where the combination therapy further comprises a non-drug treatment, the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks. Types of cancers that may be treated with the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above include, but are not limited to, prostate, colon, rectum, pancreas, cervix, stomach, endometrium, brain, liver, bladder, ovary, testis, head, neck, skin (including melanoma and basal carcinoma), mesothelial lining, white blood cell (including lymphoma and leukemia), esophagus, breast, muscle, connective tissue, lung (including small cell lung carcinoma and non- small-cell carcinoma), adrenal gland, thyroid, kidney, or bone; or glioblastoma, mesothelioma, renal cell carcinoma, gastric carcinoma, sarcoma (including Kaposi's sarcoma), choriocarcinoma, cutaneous basocellular carcinoma, haematological malignancies (including blood, bone marrow and lymph nodes) or testicular seminoma. One or more additional pharmaceutical agents or treatment methods such as, for example, anti-viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g ., IL2 and GM-CSF), and/or tyrosine kinase inhibitors can be optionally used in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above for treatment of eIF4A associated diseases, disorders or conditions. The agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms. Suitable chemotherapeutic or other anti-cancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl
sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine, cyclophosphamide (CYTOXAN®), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide. In the treatment of melanoma, suitable agents for use in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the "Dartmouth regimen", which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC, temozolomide or YERVOY™. Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may also be combined with immunotherapy drugs, including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in the treatment of melanoma. Compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may also be used in combination with vaccine therapy in the treatment of melanoma. Antimelanoma vaccines are, in some ways, similar to the anti-virus vaccines which are used to prevent diseases caused by viruses such as polio, measles, and mumps. Weakened melanoma cells or parts of melanoma cells called antigens may be injected into a patient to stimulate the body's immune system to destroy melanoma cells. Melanomas that are confined to the arms or legs may also be treated with a combination of agents including one or more compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined, using a hyperthermic isolated limb perfusion technique. This treatment protocol temporarily separates the circulation of the involved limb from the rest of the body and injects high doses of chemotherapy into the artery feeding the limb, thus providing high doses to the area of the tumor without exposing internal organs to these doses that might otherwise cause severe side effects. Usually, the fluid is warmed to 38.9 °C to 40 °C. Melphalan is the drug most often used in this chemotherapy procedure. This can be given with another agent called tumor necrosis factor (TNF). Suitable chemotherapeutic or other anti-cancer agents include, for example, antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate, pentostatine, and gemcitabine.
Suitable chemotherapeutic or other anti-cancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara-C, paclitaxel (Taxol), mithramycin, deoxyco-formycin, mitomycin-C, L- asparaginase, interferons (especially IFN-a), etoposide, and teniposide. Other cytotoxic agents include navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafme, and droloxafme. Also suitable are cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cisplatin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors. Other anti-cancer agent(s) include antibody therapeutics such as trastuzumab (HERCEPTIN®), antibodies to costimulatory molecules such as CTLA-4, 4-1BB and PD-1, or antibodies to cytokines (IL-IO or TGF-b). Other anti-cancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4. Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer. Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses. In a specific embodiment of the present invention, at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and at least one chemotherapeutic agent are administered to the patient concurrently or sequentially. In other words, at least one compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be administered first, at least one chemotherapeutic agent may be administered first, or at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be administered at the same time. Additionally, when more than one
compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or chemotherapeutic agent is used, the compounds may be administered in any order. The invention also provides pharmaceutically compositions which comprise a therapeutically effective amount of one or more of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally, one or more additional therapeutic agents described above. The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. The compound(s) and compositions of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, can be administered for any of the uses described herein by any suitable means, for example, orally, such as tablets, capsules (each of which includes sustained release or timed release formulations), pills, powders, granules, elixirs, tinctures, suspensions (including nanosuspensions, microsuspensions, spray-dried dispersions), syrups, and emulsions; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection, or infusion techniques (e.g. as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories. They can be administered alone, but generally will be administered with a pharmaceutical carrier selected on the basis of the chosen route of administration and standard pharmaceutical practice. For oral administration, the pharmaceutical composition may be in the form of, for example, a tablet, capsule, liquid capsule, suspension, or liquid. The pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient. For example, the pharmaceutical composition may be provided as a tablet or capsule comprising an amount of active ingredient in the range of from about 0.1 to 1000 mg, preferably from about 0.25 to 250 mg, and more preferably from about 0.5 to 100 mg. A suitable daily dose for a human or animal may vary widely depending on the condition of the patient and other factors, but, can be determined using routine methods.
Any pharmaceutical composition contemplated herein can, for example, be delivered orally via any acceptable and suitable oral preparations. Exemplary oral preparations, include, but are not limited to, for example, tablets, troches, lozenges, aqueous and oily suspensions, dispersible powders or granules, emulsions, hard and soft capsules, liquid capsules, syrups, and elixirs. Pharmaceutical compositions intended for oral administration can be prepared according to any methods known in the art for manufacturing pharmaceutical compositions intended for oral administration. In order to provide pharmaceutically palatable preparations, a pharmaceutical composition in accordance with the invention can contain at least one agent selected from sweetening agents, flavoring agents, bittering agents, coloring agents, demulcents, antioxidants, and preserving agents. A tablet can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one non-toxic pharmaceutically acceptable excipient suitable for the manufacture of tablets. Exemplary excipients include, but are not limited to, for example, inert diluents, such as, for example, calcium carbonate, sodium carbonate, lactose, calcium phosphate, and sodium phosphate; granulating and disintegrating agents, such as, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, and alginic acid; binding agents, such as, for example, starch, gelatin, polyvinyl-pyrrolidone, and acacia; and lubricating agents, such as, for example, magnesium stearate, stearic acid, and talc. Additionally, a tablet can either be uncoated, or coated by known techniques to either mask the bad taste of an unpleasantly tasting drug, or delay disintegration and absorption of the active ingredient in the gastrointestinal tract thereby sustaining the effects of the active ingredient for a longer period. Exemplary water soluble taste masking materials, include, but are not limited to, hydroxypropyl-methylcellulose and hydroxypropyl- cellulose. Exemplary time delay materials, include, but are not limited to, ethyl cellulose and cellulose acetate butyrate. Hard gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, and/or at least one salt thereof with at least one inert solid diluent, such as, for example, calcium carbonate; calcium phosphate; and kaolin. Soft gelatin capsules can, for example, be prepared by mixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined
above, and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one water soluble carrier, such as, for example, polyethylene glycol; and at least one oil medium, such as, for example, peanut oil, liquid paraffin, and olive oil. An aqueous suspension can be prepared, for example, by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one excipient suitable for the manufacture of an aqueous suspension. Exemplary excipients suitable for the manufacture of an aqueous suspension, include, but are not limited to, for example, suspending agents, such as, for example, sodium carboxymethylcellulose, hydroxypropyl- methylcellulose and hydroxypropyl- cellulose, sodium alginate, alginic acid, polyvinyl- pyrrolidone, gum tragacanth, and gum acacia; dispersing or wetting agents, such as, for example, a naturally-occurring phosphatide, e.g., lecithin; condensation products of alkylene oxide with fatty acids, such as, for example, polyoxyethylene stearate; condensation products of ethylene oxide with long chain aliphatic alcohols, such as, for example heptadecaethylene-oxycetanol; condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol, such as, for example, polyoxyethylene sorbitol monooleate; and condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, such as, for example, polyethylene sorbitan monooleate. An aqueous suspension can also contain at least one preservative, such as, for example, ethyl and n-propyl p-hydroxybenzoate; at least one coloring agent; at least one flavoring agent; and/or at least one sweetening agent, including but not limited to, for example, sucrose, saccharin, and aspartame. Oily suspensions can, for example, be prepared by suspending at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof in either a vegetable oil, such as, for example, arachis oil, olive oil, sesame oil and coconut oil or in mineral oil, such as, for example, liquid paraffin. An oily suspension can also contain at least one thickening agent, such as, for example, beeswax, hard paraffin and cetyl alcohol. In order to provide a palatable oily suspension, at least one of the sweetening agents already described hereinabove, and/or at least one flavoring agent can be added to the oily suspension. An oily suspension can further contain at least one preservative, including, but not limited to, for example, an anti- oxidant, such as, for example, butylated hydroxyanisol, and alpha-tocopherol.
Dispersible powders and granules can, for example, be prepared by admixing at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof with at least one dispersing and/or wetting agent; at least one suspending agent; and/or at least one preservative. Suitable dispersing agents, wetting agents, and suspending agents are as already described above. Exemplary preservatives include, but are not limited to, for example, anti-oxidants, e.g., ascorbic acid. In addition, dispersible powders and granules can also contain at least one excipient, including, but not limited to, for example, sweetening agents; flavoring agents; and coloring agents. An emulsion of at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, be prepared as an oil-in-water emulsion. The oily phase of the emulsions comprising compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above may be constituted from known ingredients in a known manner. The oil phase can be provided by, but is not limited to, for example, a vegetable oil, such as, for example, olive oil and arachis oil; a mineral oil, such as, for example, liquid paraffin; and mixtures thereof. While the phase may comprise merely an emulsifier, it may comprise a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Suitable emulsifying agents include, but are not limited to, for example, naturally-occurring phosphatides, e.g., soy bean lecithin; esters or partial esters derived from fatty acids and hexitol anhydrides, such as, for example, sorbitan monooleate; and condensation products of partial esters with ethylene oxide, such as, for example, polyoxyethylene sorbitan monooleate. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilize) make- up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations. An emulsion can also contain a sweetening agent, a flavoring agent, a preservative, and/or an antioxidant. Emulsifiers and emulsion stabilizers suitable for use in the formulation of the present invention include Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate, sodium lauryl sulfate, glyceryl distearate alone or with a wax, or other materials well known in the art.
The compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof can, for example, also be delivered intravenously, subcutaneously, and/or intramuscularly via any pharmaceutically acceptable and suitable injectable form. Exemplary injectable forms include, but are not limited to, for example, sterile aqueous solutions comprising acceptable vehicles and solvents, such as, for example, water, Ringer’s solution, and isotonic sodium chloride solution; sterile oil-in-water microemulsions and aqueous or oleaginous suspensions. Formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in the formulations for oral administration or by using other suitable dispersing or wetting agents and suspending agents. The compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum, and/or various buffers. Other adjuvants and modes of administration are well and widely known in the pharmaceutical art. The active ingredient may also be administered by injection as a composition with suitable carriers, including saline, dextrose, water or with cyclodextrin solubilization (i.e. Captisol), cosolvent solubilization (i.e. propylene glycol) or micellar solubilization (i.e. Tween 80). The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer’s solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. A sterile injectable oil-in-water microemulsion can, for example, be prepared by 1) dissolving at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above in an oily phase, such as, for example, a mixture of soybean oil and lecithin; 2) combining the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above containing oil phase with a water and glycerol mixture; and 3) processing the combination to form a microemulsion.
A sterile aqueous or oleaginous suspension can be prepared in accordance with methods already known in the art. For example, a sterile aqueous solution or suspension can be prepared with a non-toxic parenterally-acceptable diluent or solvent, such as, for example, 1,3-butane diol; and a sterile oleaginous suspension can be prepared with a sterile non- toxic acceptable solvent or suspending medium, such as, for example, sterile fixed oils, e.g., synthetic mono- or diglycerides; and fatty acids, such as, for example, oleic acid. Pharmaceutically acceptable carriers are formulated according to a number of factors well within the purview of those of ordinary skill in the art. These include, without limitation: the type and nature of the active agent being formulated; the subject to which the agent- containing composition is to be administered; the intended route of administration of the composition; and the therapeutic indication being targeted. Pharmaceutically acceptable carriers include both aqueous and non-aqueous liquid media, as well as a variety of solid and semi-solid dosage forms. Such carriers can include a number of different ingredients and additives in addition to the active agent, such additional ingredients being included in the formulation for a variety of reasons, e.g., stabilization of the active agent, binders, etc., well known to those of ordinary skill in the art. Descriptions of suitable pharmaceutically acceptable carriers, and factors involved in their selection, are found in a variety of readily available sources such as, for example, Allen, L. V. Jr. et al. Remington: The Science and Practice of Pharmacy (2 Volumes), 22nd Edition (2012), Pharmaceutical Press. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, self-emulsifying drug delivery systems (SEDDS) such as d-alpha-tocopherol poly ethyleneglycol 1000 succinate, surfactants used in pharmaceutical dosage forms such as Tweens, polyethoxylated castor oil such as CREMOPHOR surfactant (BASF), or other similar polymeric delivery matrices, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat. Cyclodextrins such as alpha-, beta-, and gamma-cyclodextrin, or chemically modified derivatives such as hydroxyalkylcyclodextrins, including 2- and 3-hydroxypropyl-cyclodextrins, or other
solubilized derivatives may also be advantageously used to enhance delivery of compounds of the formulae described herein. The pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals. The pharmaceutical compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. Tablets and pills can additionally be prepared with enteric coatings. Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents. For therapeutic purposes, the active compounds of this invention are ordinarily combined with one or more adjuvants appropriate to the indicated route of administration. If administered orally, the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and then tableted or encapsulated for convenient administration. Such capsules or tablets may contain a controlled-release formulation as may be provided in a dispersion of active compound in hydroxypropylmethyl cellulose. The amounts of compounds that are administered and the dosage regimen for treating a disease condition with the compounds and/or compositions of this invention depend on a variety of factors, including the age, weight, sex, the medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods. A daily dose of about 0.001 to 100 mg/kg body weight, preferably between about 0.0025 and about 50 mg/kg body weight and most preferably between about 0.005 to 10 mg/kg body weight, may be appropriate. The daily dose can be administered in one to four doses per day. Other dosing schedules include one dose per week and one dose per two day cycle. Pharmaceutical compositions of this invention comprise at least one compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above and/or at least one pharmaceutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof, and optionally an additional agent selected from any
pharmaceutically acceptable carrier, adjuvant, and vehicle. Alternate compositions of this invention comprise a compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, or a prodrug, or isotope enriched forms thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The present invention also includes pharmaceutical kits useful, for example, in the treatment or prevention of eIF4a-associated diseases. Thus, the present invention also relates to a kit containing a formulation comprising: a) a pharmaceutical composition comprising a compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, or a therapeutically acceptable salt thereof and/or a prodrug, or isotope enriched forms thereof and a pharmaceutically acceptable carrier; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of the activity eIF4A, is effective in treating the disorder. Such kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, as will be readily apparent to those skilled in the art. Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit. The dosage regimen for the compounds of the present invention will, of course, vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent and its mode and route of administration; the species, age, sex, health, medical condition, and weight of the recipient; the nature and extent of the symptoms; the kind of concurrent treatment; the frequency of treatment; the route of administration, the renal and hepatic function of the patient, and the effect desired. By way of general guidance, the daily oral dosage of each active ingredient, when used for the indicated effects, will range between about 0.001 to about 5000 mg per day, preferably between about 0.01 to about 1000 mg per day, and most preferably between about 0.1 to about 250 mg per day. Intravenously, the most preferred doses will range from about 0.01 to about 10 mg/kg/minute during a constant rate infusion. Compound(s) of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three, or four times daily.
The compounds are typically administered in admixture with suitable pharmaceutical diluents, excipients, or carriers (collectively referred to herein as pharmaceutical carriers) suitably selected with respect to the intended form of administration, e.g. oral tablets, capsules, elixirs, and syrups, and consistent with conventional pharmaceutical practices. Dosage forms (pharmaceutical compositions) suitable for administration may contain from about 1 milligram to about 200 milligrams of active ingredient per dosage unit. In these pharmaceutical compositions the active ingredient will ordinarily be present in an amount of about 0.1-95 % by weight based on the total weight of the composition. A typical capsule for oral administration contains at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture (250 mg), lactose (75 mg), and magnesium stearate (15 mg). The mixture is passed through a 60 mesh sieve and packed into a no.1 gelatin capsule. A typical injectable preparation is produced by aseptically placing at least one of the compound of the formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture (250 mg) into a vial, aseptically freeze-drying and sealing. For use, the contents of the vial are mixed with 2 mL of physiological saline, to produce an injectable preparation. The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compound of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, alone or in combination with a pharmaceutical carrier. Optionally, compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above can be used alone, in combination with other compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, or in combination with one or more other therapeutic agent(s), e.g. an anticancer agent or other pharmaceutically active material. Regardless of the route of administration selected, the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is
effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of factors including the activity of the particular compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, a suitable daily dose of compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, oral, intravenous, intracerebroventricular and subcutaneous doses of the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above for a patient will range from about 0.01 to about 50 mg per kilogram of body weight per day. If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In certain aspects of the invention, dosing is one administration per day, every other day, twice per week or per week.
While it is possible for compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above to be administered alone, it is preferable to administer the compound as a pharmaceutical formulation (composition). The above other therapeutic agents, when employed in combination with the compound(s) of formulae (I), (I.a), (I.b), (A), (B), (C), (D), (E), (F) and an enantiomeric mixture as defined above, may be used, for example, in those amounts indicated in the Physicians’ Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art. In the methods of the present invention, such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the inventive compounds. The invention will be illustrated further with reference to the examples that follow, without restricting the scope to the specific embodiments described. The invention includes all combinations of described and especially of preferred features that do not exclude each other. DESCRIPTION OF THE DRAWINGS Figure 1: MTT assay for cell viability in 96 well cell culture plate Cells with RAS mutations were treated with compounds 1, 2 or C-1 for 48h and cell viability was evaluated by MTT assay. The results shown are mean from 3 independent experiments. Figure 2: The effect of the compounds in RAS activation. NanoBiT assay for the RAS GTP-loading was performed in HeLa cells transfected with LgBit-K, N and HRAS G12V and SmBit-CRAF-RBD. Cells were treated with compounds 1, 2 or C-1 for 2 h in serum-free DMEM (Dulbecco's modified Eagle's Medium). After incubation, the substrate for NanoLuc was added, and the luminescence was measured using a multiplate reader. Data were normalized to cells transfected with the indicated mutant and exposed to DMSO for 2 h. DMSO-treated cells were set as 1. The results shown are mean from 3 independent experiments. Figure 3: Dual luciferase assay for Cap-dependent translation initiation
Dual luciferase assay for Cap-dependent translation initiation was performed in HeLa cells transfected with dual luciferase reporter gene based on pFR_HCV_xb. Cells were treated with compounds 1, 2 or C-1 for 24 h in serum-free DMEM (Dulbecco's modified Eagle's Medium). After incubation, Dual luciferase reporter assay was performed according to the manufacturer’s instruction and the luminescence was measured using a multiplate reader. Data were normalized to transfected cells treated with DMSO for 24 h. DMSO-treated cells were set as 1. The results shown are mean from 3 independent experiments. Figure 4: Structure of compounds C-1, 1 and 2. EXAMPLES Abbreviations ACN acetonitrile AIBN azo-bis-(isobutylonitrile) DCE Dichloroethane DMEM Dulbecco's modified Eagle's Medium DCM dichloromethane DIPEA Diisopropylethylamine DMAP Dimethylaminopyridine DMSO dimethylsulfoxid FA Formic acid FBS fetal bovine serum IPA Isopropyl alcohol LDA lithium diisopropylamide NBS N-bromosuccinimide PBS phosphate-buffered saline PE Pet-ether RLU relative light unit (luciferase activity)/relative luciferase units SD standard deviation SEM trimethylsilylethoxymethyl TBAF tetra-n-butylammonium fluoride TFA Trifluoroacetic acid THF tetrahydrofuran TLC thin-layer chromatography TMSCN trimethylsilyl cyanide
Triton B benzyltrimethylammonium hydroxide Materials and Methods Cell: HCT-116 were authenticated by Eurofin genomics and cultured in DMEM (10 % heat inactivated FBS). sNF96.2 were purchased from ATCC and cultured in DMEM (10 % heat inactivated FBS+ 1 mM Na Pyruvate). ASPC-1 cells were purchased from DSMZ and cultured in RPMI-1640 (10 % heat inactivated FBS). NCI-H358 were purchased from ATCC and cultured in RPMI-1640 (10 % heat inactivated FBS). T24 and HCT-116 cells were cultured in Mccoy’s 5A medium (10% heat inactivated FBS). HT-1080 cells were cultured in EMEM (10 % heat inactivated FBS + 1 mM Na Pyruvate). DNA Transfection: HeLa cells were harvested with 0.05% Trypsin/0.02% EDTA in PBS and seeded in 6 well or 12 well cell culture plates at the concentration of 5x104 cells in complete DMEM (2 ml for 6 well plate and 1 ml for 12 well plate). After 1 day from seeding, the DNA were transfected with Flag tagged Ras or NanoBiT plasmid using PEI/PBS solution. NanoBit assay. The plasmids were transfected to HeLa cells and the cells were harvested after 1 day of transfection. The cells were seeded to 96 well white plate. Next day, the medium was changed to serum free DMEM for 2h with compounds. After incubation, Nano Glo assay was performed. The luminescence was measured using Tecan SPARKS (Tecan). IC50 calculations by non-linear regression were conducted with GraphPad Prism 9. Dual luciferase assay: Hela cells were seeded at 1x106 cells/ml, 2 ml in 6 well cell culture plate in growth medium.2 µg of plasmids were transfected into cells with 0.5mM of PEI reagent in 200 µl PBS. One day after transfection, cells were harvested and seeded into 96 well white plates, half area (Greiner). After an additional day the medium was changed to serum free DMEM and the cells were incubated for 24 h with Compounds. Dual-Glo Luciferase Assay was performed according to the manufacturer’s instructions (Promega, N2920). The luminescence was measured using Tecan infinite (Tecan). IC50 calculations by non-linear regression were conducted with GraphPad Prism 9.
MTT assay: Cancer cells were seeded in 96 well plates (5x103 cells/well) in 80 µl in growth medium and cultured for 1 day in the incubator. Next day, 20 µl of compounds containing growth medium was added to each well and the cells were cultured for 48h. After incubation with compounds, 10 µl of MTT solution was added to the wells and incubated for 2-4 hours. After incubation with MTT, solubilization buffer was added and incubated over night. MTT was measured at O.D.570 nm with a plate reader (Tecan). IC50 calculations by non- linear regression were conducted with GraphPad Prism 9. Synthesis: Compound F: (1R,2R,3S,3aR,8bS)-3a-(4-bromophenyl)-3-(3-fluorophenyl)-1,8b- dihydroxy-6,8-dimethoxy-N,N-dimethyl-2,3,3a,8b-tetrahydro-1H-cyclopenta[b]benzofuran- 2-carboxamide
Compound 2 To a solution of compound 1 (50 g, 219.3 mmol, 1 eq) in DCE (500 mL) was added AIBN (3.23 g, 19.64 mmol, 0.09 eq) and NBS (46.62 g, 261.93 mmol, 1.2 eq) at 25°C. Then the mixture was stirred at 65°C under N2 for 16 hrs. TLC (Petroleum ether/Ethyl acetate = 10/1, Rf = 0.8) showed compound 1 was consumed completely and one main peak with desired mass was detected. The reaction mixure was quenched by water (500 mL) at 25°C, extracted with DCM (1 L*2). The combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give to compound 2 (74 g, crude) as white solid which was used for the next step without further purification. Compound 4 To a solution of crude compound 2 (60 g, ~176.5 mmol, 90% purity, 1 eq) in acetone (420 mL) was added K2CO3 (41.20 g, 298.08 mmol, 1.7 eq) and 3, 5-dimethoxyphenol (30.00 g, 194.63 mmol, 1.11 eq) at 25°C. Then the mixture was stirred at 70°C under N2 for 16 hrs. TLC (Petroleum ether/Ethyl acetate = 10/1, Rf = 0.60) indicated compound 2 was consumed completely and one new spot formed. The reaction mixture was filtered and concentrated, diluted with H2O, extracted with EtOAc three times. The combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give crude compound 4. The residue was purified by column (SiO2, petroleum ether/ethyl acetate = 100/1 to 5/1) to give compound 4 (57.6 g, 151.6 mmol, 86% yield over 2 steps) as a white solid.1H NMR (400 MHz, CDCl3) δ = 7.59 - 7.50 (m, 2H), 7.49 - 7.41 (m, 2H), 6.11 (s, 3H), 5.57 (s, 1H), 3.82 - 3.70 (m, 9H). Compound 5 To a solution of compound 4 (60 g, 157.9 mmol, 1 eq) in MeOH (420 mL) and H2O (42 mL) was added K2CO3 (26.10 g, 188.87 mmol, 1.2 eq) at 25°C. The mixture was stirred at 50°C under N2 for 16 hrs. TLC (Petroleum ether/Ethyl acetate = 1:1, Rf = 0.60) indicated compound 4 was consumed completely and one new spot formed. Four parallel reactions were combined and worked up together. The reaction mixture was quenched by water (1 L) at 25°C, then the reaction was adjusted pH with HCl (6 mol/L), extracted with EtOAc (1
L*3). The combined organic layers were washed with brine (1 L), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. Then the residue was triturated by Petroleum ether/Ethyl acetate = 20:1, filtered and concentrated under reduced pressure to give compound 5 (325 g, 888.0 mmol, 71%) as a white solid.1H NMR (400 MHz, CDCl3) δ = 7.60 - 7.50 (m, 2H), 7.49 - 7.40 (m, 2H), 6.21 - 6.01 (m, 3H), 5.58 (s, 1H), 3.75 (s, 6H). Compound 6 To a mixture of compound 5 (25 g, 68.3 mmol, 1 eq) and ZnCl2 (12.99 g, 95.32 mmol, 4.46 mL, 1.4 eq) was added POCl3 (100 mL) at 25 °C, then the mixture was stirred at 25 °C for 16 hrs. TLC (Petroleum ether/Ethyl acetate = 2:1, Rf = 0.50) indicated compound 5 was consumed completely and one new spot formed. Four parallel reactions were combined and worked up together. The reaction mixture was quenched by slowly dropping into water (1000 mL) at 25-40°C, filtered to give a residue. Then the residue was washed with water (3 times), filtered and concentrated under reduced pressure to give a crude product. Then the residue was triturated with methyl alcohol, filtered and concentrated under reduced pressure to give compound 6 (83 g, 238.50 mmol, 87% yield) as a white solid.1H NMR (400 MHz, CDCl3) δ = 7.53 - 7.48 (m, 2H), 7.31 (d, J = 8.3 Hz, 2H), 6.27 (d, J = 1.9 Hz, 1H), 6.06 (d, J = 1.8 Hz, 1H), 5.43 (s, 1H), 3.91 (d, J = 2.0 Hz, 6H). Compound 8 To a solution of compound 6 (10 g, 28.7 mmol, 1 eq) in t-BuOH (360 mL) was added triton B (1.20 g, 2.86 mmol, 1.30 mL, 40% purity, 0.1 eq) and the solution of compound 7 (5.16 g, 34.37 mmol, 1.2 eq) in t-BuOH (40 mL) at 25℃. The mixture was stirred at 60°C under N2 for 2 hrs. LCMS showed compound 6 was consumed completely and two peaks with desired mass was detected. The solvent was removed in vacuo, then the mixture was quenched by water (50 mL) and acidified with 6 M HCl to pH =1. The remained liquid was extracted with ethyl acetate (100 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 10/1 to 5/1) to give compound 8 (3.01 g, 6.0 mmol, 21% yield) as a white solid and its diastereoisomer 8A (1.32 g, 2.65 mmol, 9% yield) as white solid. Compound 8: 1H NMR (400 MHz, CDCl3) δ = 9.44 (d, J = 1.1 Hz, 1H), 7.67 - 7.58 (m, 2H), 7.54 - 7.44 (m, 2H), 7.17 - 7.09 (m, 2H), 7.06 - 6.99 (m, 1H), 6.84 - 6.76 (m, 1H), 6.24 (d, J = 1.9 Hz, 1H), 5.85 (d, J = 1.8 Hz, 1H), 4.20 (dd, J = 3.8, 10.8 Hz, 1H), 3.87 (s, 3H), 3.73 (s, 3H), 3.06 (ddd, J = 2.1, 10.9, 17.7 Hz, 1H), 2.75 - 2.51 (m, 1H). Compound 8A: 1H NMR (400 MHz, CDCl3) δ = 9.51 (d, J = 2.1 Hz, 1H), 7.39
- 7.34 (m, 2H), 7.32 - 7.28 (m, 2H), 7.09 (dt, J = 6.1, 7.9 Hz, 1H), 7.02 - 6.96 (m, 1H), 6.89 (d, J = 7.9 Hz, 1H), 6.82 (dt, J = 2.6, 8.3 Hz, 1H), 6.37 (d, J = 1.8 Hz, 1H), 6.05 (d, J = 1.8 Hz, 1H), 4.16 (dd, J = 3.4, 11.1 Hz, 1H), 3.94 (s, 3H), 3.89 (s, 3H), 2.97 (ddd, J = 2.5, 11.0, 17.2 Hz, 1H), 2.65 (dd, J = 3.4, 17.3 Hz, 1H). Compound 9 To a solution of compound 8 (22.5 g, 45.2 mmol, 1 eq) in CH3CN (225 mL) was added TMSCN (14.31 g, 144.19 mmol, 18.04 mL, 3.2 eq) and ZnI2 (1.44 g, 4.51 mmol, 0.1 eq) at 25°C. The mixture was stirred at 25°C under N2 for 16 hrs. LCMS showed compound 8 was consumed completely and one main peak with desired mass was detected. The reaction mixture was quenched by water (200 mL), extracted with EtOAc (150 mL x 2). The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give compound 9 (27.15 g, crude) as yellow solid. The residue was used to the next step without further purification. 1H NMR (400 MHz, CDCl3) δ = 7.63 - 7.55 (m, 2H), 7.54 - 7.49 (m, 2H), 7.23 - 7.10 (m, 2H), 7.10 - 6.95 (m, 2H), 6.91 - 6.82 (m, 1H), 6.26 (t, J = 2.2 Hz, 1H), 5.85 (dd, J = 1.9, 3.0 Hz, 1H), 3.88 (s, 4H), 3.72 (d, J = 1.8 Hz, 3H), 2.01 (s, 3H), 0.05 (d, J = 3.8 Hz, 9H). Compound 11 To a solution of compound 9 (21.6 g, 36.2 mmol, 1 eq) in THF (432 mL) was added dropwise LDA (2 M, 54.13 mL, 3 eq) at -78°C. The mixture was stirred at -78°C under N2 for 1 hr. LCMS showed compound 9 was consumed completely and one main peak with desired mass was detected. The reaction mixture was quenched by H2O/ice (500 mL), extracted with EtOAc (500 mL x 2). The combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 10/1 to 5/1) to give compound 11 (14.2 g, 23.7 mmol, 66% yield) as yellow solid and INT.10 (2.5 g, 5.0 mmol, 14% yield) as yellow solid. Compound 111H NMR (400 MHz, CDCl3) δ = 7.29 (br d, J = 8.4 Hz, 1H), 7.20 (d, J = 8.3 Hz, 2H), 7.11 (q, J = 7.3 Hz, 1H), 7.01 - 6.94 (m, 2H), 6.85 (d, J = 7.8 Hz, 1H), 6.82 - 6.73 (m, 2H), 6.21 (s, 1H), 6.05 (s, 1H), 4.32 (dd, J = 5.6, 14.4 Hz, 1H), 3.85 (s, 3H), 3.82 (s, 3H), 3.17 - 2.99 (m, 2H), 2.68 (dd, J = 5.7, 12.7 Hz, 1H), 0.11 (s, 9H). INT.10 To a solution of compound 11 (14.2 g, 23.8 mmol, 1 eq) in THF (142 mL) was added TBAF (1 M, 29.66 mL, 1.25 eq) at 0℃. The mixture was stirred at 25°C under N2 for 2 hrs. LCMS showed compound 11 was consumed completely and one main peak with desired mass
was detected. The reaction mixture was quenched by water (150 mL), extracted with EtOAc (150 mL x 2). The combined organic layers were washed with brine (150 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 10/1 to 5/1) to give INT.10 (10.2 g, 20.5 mmol, 86% yield) as yellow solid. LCMS: M+23 (520.9), 92.7%. 1H NMR (400 MHz, CDCl3) δ = 7.35 - 7.28 (m, 2H), 7.15 - 7.03 (m, 2H), 6.95 (d, J = 8.6 Hz, 2H), 6.83 (br t, J = 8.1 Hz, 1H), 6.74 (br d, J = 8.1 Hz, 2H), 6.36 (d, J = 1.8 Hz, 1H), 6.13 (d, J = 1.7 Hz, 1H), 5.31 (s, 1H), 3.97 - 3.91 (m, 1H), 3.87 (s, 3H), 3.85 (s, 1H), 3.83 (s, 3H), 3.29 (br s, 1H), 3.13 - 3.02 (m, 1H), 3.02 - 2.90 (m, 1H). Compound 12 The mixture of INT.10 (2 g, 4.01 mmol, 1 eq) in methoxy(methoxycarbonyloxy)magnesium (2 M, 20.00 mL, 9.99 eq) was stirred at 100 °C for 16 hr. LCMS showed the starting material was consumed completely and two main peaks with desired mass was detected. The reaction solution was cooled to 0°C, added ice-cold MTBE (100 mL), quenched with ice-cold HCl (6M) to pH=3, extracted with ice-cold MTBE (50 mL*2), dried over Na2SO4 and filtered. The MTBE solution of compound 1 was used for the next step without concentration and further purification. LCMS: M-17 (525.0), 91.2%. Compound 13 To a solution of compound 12 (~2 g, 3.68 mmol, 1 eq) in MTBE was added DCM (50 mL), PyBOP (3.83 g, 7.36 mmol, 2 eq) and N-methylmethanamine (2 M, 18.40 mL, 10 eq) at - 30°C, then the mixture was stirred at -30°C for 2 hr. LCMS showed the starting material was consumed completely and one peak with desired mass was detected. The reaction mixture was quenched by water (200 mL) at 0°C, extracted with EtOAc (100 mL x 2).The combined organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, petroleum ether/ethyl acetate = 100/1 to 2/1) to give compound 13 (0.82 g, 1.44 mmol, 39% yield) as yellow solid. LCMS: M+1 (570.0), 29.4%. 1H NMR (400 MHz, CDCl3) δ = 7.34 (d, J = 8.6 Hz, 2H), 7.11 - 7.03 (m, 1H), 6.98 (d, J = 8.6 Hz, 2H), 6.82 (br t, J = 7.8 Hz, 1H), 6.63 (br d, J = 8.1 Hz, 2H), 6.35 (d, J = 1.7 Hz, 1H), 6.10 (d, J = 1.6 Hz, 1H), 4.57 (d, J = 13.1 Hz, 1H), 4.30 (d, J = 13.1 Hz, 1H), 3.86 (s, 3H), 3.81 (s, 3H), 3.32 (s, 3H), 3.06 (s, 1H), 2.94 (s, 3H) Compound 14 To a solution of compound 13 (0.82 g, 1.44 mmol, 1 eq) in MeOH (20 mL) was added NaBH4 (1.09 g, 28.75 mmol, 20 eq) at 0°C, then the mixture was stirred at 25°C for
16 hr. LCMS showed the starting material was consumed completely and one main peak with desired mass was detected. The reaction mixture was quenched by water (100 mL) at 25°C, extracted with EtOAc (50 mL x 2).The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue which was purified by prep-HPLC (column: Phenomenex Luna C18 75*30mm*3um;mobile phase: [H2O(0.1%TFA)-ACN];gradient:30%-55% B over 8.0 min) to give compound 14 (250 mg, 30.4%) as a white solid. LCMS: M+1 (572.1), 45.5%.1H NMR (400 MHz, CDCl3) δ = 7.30 (d, J = 8.5 Hz, 2H), 7.08 (d, J = 8.6 Hz, 2H), 7.06 - 6.99 (m, 1H), 6.78 - 6.71 (m, 1H), 6.67 - 6.57 (m, 2H), 6.30 (d, J = 1.8 Hz, 1H), 6.13 (d, J = 1.8 Hz, 1H), 4.95 (dd, J = 1.2, 6.6 Hz, 1H), 4.57 (d, J = 13.5 Hz, 1H), 4.03 - 3.95 (m, 2H), 3.86 (d, J = 7.6 Hz, 6H), 3.33 (s, 3H), 2.96 (s, 3H), 1.85 (s, 1H). Compound F Separation of 80 mg of compound 14 by SFC (column: DAICEL CHIRALPAK AD(250 mm * 30 mm,10 um); mobile phase: [CO2-IPA(0.1%NH3H2O)];B%:50%, isocratic elution mode) gave two peaks. Peak 1 was assigned as compound ent-F (7.9 mg, 13.8 μmol, 9.9%) as a white solid. LCMS: M+1 (572.1), 99.0%.1H NMR (400 MHz, CDCl3) δ = 7.32 - 7.28 (m, 2H), 7.11 - 7.06 (m, 2H), 7.02 (dt, J = 6.2, 7.9 Hz, 1H), 6.74 (dt, J = 2.3, 8.4 Hz, 1H), 6.67 - 6.57 (m, 2H), 6.30 (d, J = 1.9 Hz, 1H), 6.13 (d, J = 2.0 Hz, 1H), 4.95 (dd, J = 1.9, 6.5 Hz, 1H), 4.57 (d, J = 13.5 Hz, 1H), 4.03 - 3.98 (m, 1H), 3.97 (d, J = 2.0 Hz, 1H), 3.86 (d, J = 7.6 Hz, 6H), 3.33 (s, 3H), 2.96 (s, 3H), 1.85 (s, 1H). Peak 2 was assigned as compound F (8.8 mg, 15.4 μmol, 11% yield) as a white solid. LCMS: M+1 (572.1), 98.8%.1H NMR (400 MHz, CDCl3) δ = 7.30 (d, J = 8.6 Hz, 2H), 7.08 (d, J = 8.6 Hz, 2H), 7.06 - 6.99 (m, 1H), 6.74 (dt, J = 2.2, 8.3 Hz, 1H), 6.67 - 6.56 (m, 2H), 6.30 (d, J = 1.8 Hz, 1H), 6.13 (d, J = 1.8 Hz, 1H), 4.95 (dd, J = 1.6, 6.5 Hz, 1H), 4.57 (d, J = 13.5 Hz, 1H), 4.06 - 3.94 (m, 2H), 3.86 (d, J = 7.6 Hz, 6H), 3.33 (s, 3H), 2.96 (s, 3H), 1.85 (s, 1H). Compound A
Compound 15 To a solution of compound 14 (0.160 g, 0.28 mmol, 1 eq) in DMF (4 mL) was added Zn(CN)2 (65.64 mg, 559.03 μmol, 35.48 μL, 2 eq) and Pd(PPh3)4 (32.30 mg, 27.95 μmol, 0.1 eq), then the mixture was stirred at 120°C for 16 hr under N2. LCMS showed the starting material was consumed completely and one main peak with desired mass was detected. The reaction mixture was quenched by water (50 mL) at 0°C, extracted with EtOAc (50 mL x 2). The combined organic layers were washed with brine (30 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge Prep OBD C18150 * 40 mm * 10 um; mobile phase: [H2O (10mM NH4HCO3)-ACN];gradient: 30%-60% B over 8.0 min) to give compound 15 (0.1 g, 0.19 mmol, 68% yield) as white solid. LCMS: M+1 (519.1), 38%.1H NMR (400 MHz, DMSO-d6) δ = 7.52 (d, J = 8.5 Hz, 2H), 7.36 (d, J = 8.4 Hz, 2H), 7.13 - 7.00 (m, 1H), 6.83 - 6.70 (m, 2H), 6.66 (br d, J = 10.4 Hz, 1H), 6.33 (d, J = 1.8 Hz, 1H), 6.13 (d, J = 1.8 Hz, 1H), 5.34 (br s, 1H), 4.84 - 4.70 (m, 2H), 4.38 (d, J = 13.3 Hz, 1H), 4.18 (dd, J = 5.5, 13.4 Hz, 1H), 3.82 - 3.70 (m, 6H), 3.27 (s, 3H), 2.77 (s, 3H). Compound A Compound 15 (0.1 g) was purified by SFC (column: DAICEL CHIRALCEL OJ(250 mm * 30 mm,10 um);mobile phase: [CO2-IPA(0.1%NH3H2O)]; B%:30%, isocratic elution mode) and SFC (column: REGIS (s,s) WHELK-O1 (250 mm * 30 mm,5 um);mobile phase: [CO2- MeOH(0.1%NH3H2O)];B%:50%, isocratic elution mode) to give two peaks. Peak 1 ent-A (7.9 mg, 15.24 μmol, 8% yield) as a white solid. LCMS: M+1 (519.2), 98.0%. 1H NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 8.4 Hz, 2H), 7.33 (d, J = 8.5 Hz, 2H), 7.05 - 6.97 (m, 1H), 6.78 - 6.70 (m, 1H), 6.65 - 6.57 (m, 2H), 6.31 (d, J = 1.8 Hz, 1H), 6.14 (d, J = 1.8 Hz, 1H), 4.94 (dd, J = 1.7, 6.3 Hz, 1H), 4.64 (d, J = 13.5 Hz, 1H), 4.03 (dd, J = 6.4, 13.5 Hz, 1H), 3.93 (d, J = 1.8 Hz, 1H), 3.87 (d, J = 5.9 Hz, 6H), 3.35 (s, 3H), 2.97 (s, 3H), 2.00 (s, 1H). Peak 2 Compound A (21.7 mg) as a white solid. LCMS M+1 (519.2), 98.8%.1H NMR (400 MHz, CDCl3) δ = 7.45 (d, J = 8.3 Hz, 2H), 7.33 (d, J = 8.5 Hz, 2H), 7.08 - 6.95 (m, 1H), 6.74 (dt, J = 2.3, 8.4 Hz, 1H), 6.65 - 6.54 (m, 2H), 6.31 (d, J = 1.9 Hz, 1H), 6.14 (d, J = 1.8 Hz, 1H), 4.94 (dd, J = 1.5, 6.3 Hz, 1H), 4.64 (d, J = 13.5 Hz, 1H), 4.02 (dd, J = 6.3, 13.6 Hz, 1H), 3.94 (d, J = 1.8 Hz, 1H), 3.86 (d, J = 5.9 Hz, 6H), 3.35 (s, 3H), 2.97 (s, 3H), 2.00 (s, 1H).
Results: Figure 1a):
Figure 1b):
Figure 1c):
Figure 1d):
Figure 1e):
Figure 1f):
Figure 2a):
Figure 2b):
Figure 2c):
Figure 3:
Unbound fraction in human plasma and pharmacokinetics (DMPK) (table 1 table 3) After administration, a certain amount of drug is in the blood or plasma. Of the total amount in the blood or plasma, some is bound to proteins (or other species in the blood or plasma). Only unbound drug is available e.g., for drug-target interactions. Therefore, the higher the unbound fraction that is available in the blood or plasma, the more is available for binding the drug to a target. The less unbound drug is available in the blood or plasma e.g., because it is involved in competitive binding to, for example, albumin, the less is available for desired drug-target interaction. The protocol for determining the unbound fraction and pharmacokinetics is routine practice and known to the skilled person. Surprisingly the compounds according to the invention have a much higher free fraction in human plasma. There is much more drug available for binding to a target compared to the compounds of the prior art. The human microsomal clearance (hMic Clint) is lower for compound according to the invention compared to C-1. Since the exposure (AUC, area under the concentration-time curve) of a drug depends, inter alia, on its clearance (the lower the clearance of a drug, the higher the exposure). Typically, the beneficial effect of a drug depends on its potency and exposure, so a lower clearance corresponds to a lower dose to provide the same beneficial effect. AUC=(F.D)/CL where F is bioavailability (=100% for iv dosing), D is dose and CL is clearance. Table 1: It shows that 2 has higher AUC and Cave than C-1 when dosed at 2 mg/kg iv (intravenous).
Table 2: It shows that 2 has higher AUC and Cave than C-1 when dosed at 10 mg/kg po (oral).
Table 3: It shows that 2 has lower unbound clearance than C-1 which is why it has a higher unbound AUC at this dose.
Protocol for unbound fraction Dialysis Setup: HT-Dialysis plates (Model HTD 96 b, Cat# 1006) and the dialysis membrane (molecular weight cut off 12-14 KDa, Cat# 1101) were purchased from HT Dialysis LLC (Gales Ferry, CT). Dialysis Buffer (100 mM sodium phosphate and 150 mM NaCl, pH 7.4 ± 0.1) Stop Solution (Acetonitrile containing tolbutamide at 200 ng/mL, labetalol at 200 ng/mL) Processing Procedure of Dialysis Membrane and Matrix:
On the day of experiment, the plasma was thawed under running cold tap water and centrifuged at 3220 ×g for 5 min to remove any clots. The pH value was checked and recorded. Only plasma within a range of pH 7.0 to pH 8.0 was used. The dialysis membrane was pretreated according to the manufacturer's instructions: the dialysis membrane strips were soaked in ultra-pure water at room temperature for approximately 1 hr. Afterwards, each membrane strip that contains 2 membranes was separated and soaked in ethanol:water (20:80 v:v) for approximately 20 min, after which it was ready for use or was stored in the solution at 2-8°C for up to 1 month. Prior to the experiment, the membrane was rinsed and soaked for 20 min in ultra-pure water for use. Dilution Procedure of Test Compound and Control Compound: Working solutions (400 μM) of test compound and control compound were prepared. Aliquots of working solutions (3 μL) were spiked into blank matrix (597 μL) to achieve final concentrations and mixed thoroughly. Procedure of Assay Aliquots of 50 μL loading matrix containing test compound or control compound in triplicate was transferred to Sample Collection Plate, respectively. The samples were matched with opposite blank buffer to obtain a final volume of 100 μL with a volume ratio of matrix :Dialysis Buffer (1:1, v:v) in each well immediately. The stop solution was added to these T0 samples of test compound and control compound. The plate was sealed and shaken at 800 rpm for 10 min. Then these T0 samples were stored at 2-8°C pending further process along with other post-dialysis samples. The dialysis instrument was assembled following manufacturer's instructions. An aliquot of 100 μL of the loading matrix containing test compound or control compound was transferred to the donor side of each dialysis well in triplicate and 100 μL of the Dialysis Buffer was loaded to the receiver side of the well. Then the plate was rotated at approximately 100 rpm in a humidified incubator with 5% CO2 at 37±1°C for 4 hours. At the end of the dialysis, aliquots of 50 μL samples from the buffer side and matrix side of the dialysis device were taken into new 96-well plates (Sample Collection Plates). An equal volume of opposite blank matrix (buffer or matrix) in each sample was added to reach a final volume of 100 μL with volume ratio of matrix : dialysis buffer at 1:1 (v:v) in each well. All samples were further processed by protein precipitation for LC-MS/MS analysis. The detailed processing method of samples were described in the study record.
Data Analysis: The %Unbound, %Bound and %Recovery were calculated using the following equations: %Unbound = 100 × F / T %Bound = 100 - %Unbound %Recovery = 100 × (F + T) / T0 F = Free compound concentration as determined by the calculated concentration on the buffer side of the membrane T = Total compound concentration as determined by the calculated concentration on the matrix side of the membrane T0 = Total compound concentration as determined by the calculated concentration in matrix before dialysis
Claims
Claims 1. A compound of formula (I)
prodrugs, or isotope enriched forms thereof, or a pharmaceutically acceptable salt thereof, wherein R1 is selected from Br and CN, R2 is selected from hydrogen and methyl, R3 is selected from methyl and methoxy. 2. The compounds of formula (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or the pharmaceutically acceptable salts thereof
, wherein R1, R2 and R3 have the same meanings as defined in claim 1. 3. The compounds of formula (I) according to any of the preceding claims, being a mixture of (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salts thereof, wherein the enantiomer excess (ee) of the enantiomer of formula (I.a) is at least 20%.
4. The compounds of formula (I) according to any of the preceding claims, being a mixture of (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salts thereof, wherein the enantiomer excess (ee) of the enantiomer of formula (I.a) at least 50%. 5. The compounds of formula (I) according to any of the preceding claims, being a mixture of (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salts thereof, wherein the enantiomer excess (ee) of the enantiomer of formula (I.a) is at least 80%. 6. The compounds of formula (I) according to any of the preceding claims, being a mixture of (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salts thereof, wherein the enantiomer excess (ee) of the enantiomer of formula (I.a) is at least 99%. 7. The compounds of formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, according to any of the preceding claims, wherein R2 and R3 are methyl. 8. The compounds according to any of the preceding claims, selected from compounds of the formulae A, B, C, D, E and F, the mixture of each of compound A to F with its respective enantiomer
. 9. Compounds of formulae (I), (I.a), (I.b), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined in anyone of claims 1 to 8 for use as a medicament. 10. The compounds of formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined in anyone of claims 1 to 8, for use in the treatment and/or prophylaxis of diseases. 11. The compounds of formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined in anyone of claims 1 to 8, for use according to claim 10 for use in the treatment and/or prophylaxis of proliferative disorders or genetic disorders or inflammatory disorders. 12. The compounds of formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, wherein the disease is selected from neoproliferative diseases, cancer and RASOpathies in particular CFC (Cranio Facial Cutaneous) and NF1 (Neurofibromatosis Type 1). 13. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, for use as inhibitor of RAS protein activation or inhibitor of eIF4A complex or as ligand of eIF4A complex or as ligand of Prohibitin.
14. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, for use in treating proliferative disorders, wherein RAS-signaling is involved. 15. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined in anyone of claims 1 to 8, use in treating proliferative disorders, wherein eIF4A-Myc-signaling is involved. 16. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, for use according to claim 13 in treating proliferative disorders, wherein KRAS G12V, NRAS G12V, HRAS G12V, KRAS G12C, KRAS G12D, KRAS G12C/Y96D, KRAS G13C, KRAS G13D, KRASG13S, KRAS Q61H, KRAS Q61R or KRAS Q61K is involved. 17. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined in anyone of claims 1 to 9, use according to claim 11 in treating proliferative disorders, wherein c-myc is overexpressed and/or wherein and cyclin D1 is overexpressed. 18. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, for use according to claim 13 in treating proliferative disorders, wherein any activating mutation in KRAS, HRAS and NRAS is involved. 19. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, for use according to claim 13 in treating proliferative disorders, wherein any mutation that acquires resistance to RAS inhibitors is involved.
20. The compounds formulae (I) or (I.a) or (I.b) or an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, for use according to claim 13 in treating proliferative disorders, wherein any mutation in EGFR or its related genes that contributes to acquired resistance to RAS inhibitors or eIF4A inhibotors. 21. A method of inhibiting growth, proliferation, or metastasis of cancer cells in a subject in need thereof, said method comprising administering to the subject a therapeutically effective amount of at least one compound selected from compounds of formulae (I), (I.a), (I.b), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8. 22. A pharmaceutical composition, comprising a pharmaceutical effective amount of at least one compound selected from compounds of formulae (I), (I.a), (I.b), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof as defined in anyone of claims 1 to 8, and one or more pharmaceutical acceptable carrier. 23. The pharmaceutical composition according to claim 22, for use in the prophylaxis and/or treatment of proliferative disorders or genetic disorders. 24. A kit containing a formulation comprising: a1) at least one compound selected from compounds of formulae (I), (I.a), (I.b), an enantiomeric mixture comprising the compounds of formula (I.a) and (I.b) or prodrugs or isotope enriched forms thereof or a pharmaceutically acceptable salt thereof, as defined in anyone of claims 1 to 8, or a2) a pharmaceutical composition according to claim 22; and b) instructions for dosing of the pharmaceutical composition for the treatment of a disorder in which inhibition of RAS activation or the downstream signaling pathways , in particular C- Myc upregulation and/ cyclin D1 upregulation is effective in treating the disorder.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP23158213 | 2023-02-23 | ||
EP23158213.1 | 2023-02-23 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024175662A1 true WO2024175662A1 (en) | 2024-08-29 |
Family
ID=85381080
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2024/054426 WO2024175662A1 (en) | 2023-02-23 | 2024-02-21 | Flavagline derivatives as ras inhibitors |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024175662A1 (en) |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2005113529A2 (en) | 2004-05-18 | 2005-12-01 | Bayer Healthcare Ag | Novel cylopenta[b]benzofuran derivatives and the utilization thereof |
WO2010060891A1 (en) | 2008-11-25 | 2010-06-03 | Universite De Strasbourg | Rocaglaol derivatives as cardioprotectant agents and as antineoplastic agents |
WO2017058768A1 (en) | 2015-09-28 | 2017-04-06 | Araxes Pharma Llc | Inhibitors of kras g12c mutant proteins |
WO2017091585A1 (en) | 2015-11-25 | 2017-06-01 | Effector Therapeutics, Inc. | Eif4-a-inhibiting compounds and methods related thereto |
WO2020078975A1 (en) | 2018-10-16 | 2020-04-23 | Universitätsmedizin Der Johannes Gutenberg-Universität Mainz | Flavagline derivatives for inhibition of kras oncogene activation |
-
2024
- 2024-02-21 WO PCT/EP2024/054426 patent/WO2024175662A1/en unknown
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2005113529A2 (en) | 2004-05-18 | 2005-12-01 | Bayer Healthcare Ag | Novel cylopenta[b]benzofuran derivatives and the utilization thereof |
WO2010060891A1 (en) | 2008-11-25 | 2010-06-03 | Universite De Strasbourg | Rocaglaol derivatives as cardioprotectant agents and as antineoplastic agents |
WO2017058768A1 (en) | 2015-09-28 | 2017-04-06 | Araxes Pharma Llc | Inhibitors of kras g12c mutant proteins |
WO2017091585A1 (en) | 2015-11-25 | 2017-06-01 | Effector Therapeutics, Inc. | Eif4-a-inhibiting compounds and methods related thereto |
WO2020078975A1 (en) | 2018-10-16 | 2020-04-23 | Universitätsmedizin Der Johannes Gutenberg-Universität Mainz | Flavagline derivatives for inhibition of kras oncogene activation |
Non-Patent Citations (12)
Title |
---|
"The Practice of Medicinal Chemistry", 1996, ACADEMIC PRESS, pages: 671 - 715 |
ALLEN, L. V. JR. ET AL.: "Remington: The Science and Practice of Pharmacy", vol. 2, 2012, PHARMACEUTICAL PRESS. |
AWAD MM ET AL., NEW ENGLAND J. OF MED., 2021 |
ERNST ET AL., J. MED. CHEM., vol. 63, 2020, pages 5879 |
H. YURUGI, JOURNAL OF CELL SCIENCE, vol. 133, no. 1, 2020, pages 1 |
J. RAUTIO ET AL., NATURE REVIEWS, vol. 17, 2018, pages 559 |
N. RIBEIRO ET AL., J. MED. CHEM., vol. 55, 2012, pages 100064 |
NIGEL RIBEIRO ET AL: "Flavaglines as Potent Anticancer and Cytoprotective Agents", JOURNAL OF MEDICINAL CHEMISTRY, vol. 55, no. 22, 26 November 2012 (2012-11-26), pages 10064 - 10073, XP055122607, ISSN: 0022-2623, DOI: 10.1021/jm301201z * |
POLIER ET AL., CHEMISTRY AND BIOLOGY, vol. 19, 2012, pages 1093 - 1104 |
TANAKA ET AL., CANCER DISCOV, 2021 |
THUAUD ET AL., MED. CHEM., vol. 54, 2011, pages 411 |
YURUGI HAJIME ET AL: "A subset of flavaglines inhibits KRAS nanoclustering and activation", 24 June 2020 (2020-06-24), Cambridge, XP055890835, ISSN: 0021-9533, Retrieved from the Internet <URL:http://journals.biologists.com/jcs/article-pdf/doi/10.1242/jcs.244111/2068496/jcs_244111v1.pdf> DOI: 10.1242/jcs.244111 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10590079B2 (en) | Cyano-substituted indoles as LSD1 inhibitors | |
CN104395319B (en) | The Thienopyridinone derivatives of activator as AMPK | |
KR20190020753A (en) | The crystalline form of the triazolopyrimidine compound | |
KR20170095882A (en) | Triazolopyrimidine compounds and uses thereof | |
CN111978319A (en) | Aryl receptor modulators and methods of making and using the same | |
JP2016528197A (en) | IDO inhibitor | |
US11578058B2 (en) | Heterocyclic compounds for inhibiting TYK2 activities | |
CA2854836C (en) | Tricyclic amino containing compounds for treatment or prevention of symptoms associated with endocrine dysfunction | |
WO2018019222A1 (en) | Heterocyclic compound as jak inhibitor, and salts and therapeutic use thereof | |
KR20210102887A (en) | Crystalline spirocyclic compound inhibitors of tryptophan hydroxylase 1 (TPH1) for treating diseases or disorders associated with peripheral serotonin | |
US9884874B2 (en) | Bromodomain-inhibiting compounds and pharmaceutical composition comprising same for preventing or treating a cancer | |
KR20200041954A (en) | Compounds, pharmaceutical compositions thereof, and uses and applications thereof | |
WO2022233286A1 (en) | Nitrogen-containing heterocyclic pyridine compound | |
WO2019011170A1 (en) | Oxadiazole derivative, preparation method therefor and medical application thereof | |
KR20190041509A (en) | Inhibition of OLIG2 activity | |
KR20130118731A (en) | Piperazinotriazines as pi3k inhibitors for use in the treatment antiproliferative disorders | |
EP1911451A1 (en) | Protein-kinase CK2 inhibitors and their therapeutic applications | |
WO2019057112A1 (en) | 2-substituted pyrazole amino-4-substituted amino-5-pyrimidine formamide compound, composition, and application thereof | |
EP4395897A1 (en) | Novel ras inhibitors | |
CN103313707A (en) | Diphenyl-amine derivatives: uses, process of synthesis and pharmaceutical compositions | |
WO2023242100A1 (en) | Novel ras inhibitors | |
WO2024175662A1 (en) | Flavagline derivatives as ras inhibitors | |
TW201922690A (en) | Inhibitors of cyclic-AMP response element-binding protein | |
WO2024175659A1 (en) | 3ah-cyclopenta[b]benzofuran-3a-yl as ras inhibitors | |
WO2015014283A1 (en) | Protein tyrosine kinase inhibitor and application thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24706125 Country of ref document: EP Kind code of ref document: A1 |