CN118164966A - MAT2A inhibitor and application thereof - Google Patents
MAT2A inhibitor and application thereof Download PDFInfo
- Publication number
- CN118164966A CN118164966A CN202211569257.XA CN202211569257A CN118164966A CN 118164966 A CN118164966 A CN 118164966A CN 202211569257 A CN202211569257 A CN 202211569257A CN 118164966 A CN118164966 A CN 118164966A
- Authority
- CN
- China
- Prior art keywords
- compound
- cancer
- pharmaceutically acceptable
- tautomer
- stereoisomer
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- ZTNQNZDNHUAVEI-UHFFFAOYSA-N CC=1SC2=C(N=1)C=CC(=C2)C1=C(NC=2N(C1=O)N=C(C=2C1=CC=CC=C1)C1=CC=CC=C1)NC1=NC=CC=C1 Chemical compound CC=1SC2=C(N=1)C=CC(=C2)C1=C(NC=2N(C1=O)N=C(C=2C1=CC=CC=C1)C1=CC=CC=C1)NC1=NC=CC=C1 ZTNQNZDNHUAVEI-UHFFFAOYSA-N 0.000 title description 3
- 229940125535 MAT2A inhibitor Drugs 0.000 title description 3
- 150000001875 compounds Chemical class 0.000 claims abstract description 275
- 238000000034 method Methods 0.000 claims abstract description 56
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 42
- 238000011282 treatment Methods 0.000 claims abstract description 24
- 150000003839 salts Chemical class 0.000 claims description 108
- 125000000217 alkyl group Chemical group 0.000 claims description 75
- -1 alkoxycycloalkyl Chemical group 0.000 claims description 74
- 125000000623 heterocyclic group Chemical group 0.000 claims description 51
- 125000003118 aryl group Chemical group 0.000 claims description 48
- 229910052739 hydrogen Inorganic materials 0.000 claims description 41
- 239000001257 hydrogen Substances 0.000 claims description 41
- 125000000753 cycloalkyl group Chemical group 0.000 claims description 40
- 206010028980 Neoplasm Diseases 0.000 claims description 37
- 125000001072 heteroaryl group Chemical group 0.000 claims description 34
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 32
- 230000002829 reductive effect Effects 0.000 claims description 32
- 201000011510 cancer Diseases 0.000 claims description 30
- 125000003545 alkoxy group Chemical group 0.000 claims description 28
- 125000000304 alkynyl group Chemical group 0.000 claims description 26
- 125000003342 alkenyl group Chemical group 0.000 claims description 25
- 201000010099 disease Diseases 0.000 claims description 25
- 102100034187 S-methyl-5'-thioadenosine phosphorylase Human genes 0.000 claims description 22
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 21
- 101710136206 S-methyl-5'-thioadenosine phosphorylase Proteins 0.000 claims description 21
- 150000002431 hydrogen Chemical class 0.000 claims description 20
- 239000003814 drug Substances 0.000 claims description 19
- 125000004404 heteroalkyl group Chemical group 0.000 claims description 15
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 12
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 12
- 125000004093 cyano group Chemical group *C#N 0.000 claims description 11
- 125000001188 haloalkyl group Chemical group 0.000 claims description 11
- 229910052736 halogen Inorganic materials 0.000 claims description 11
- 150000002367 halogens Chemical class 0.000 claims description 11
- 125000004183 alkoxy alkyl group Chemical group 0.000 claims description 10
- 206010003571 Astrocytoma Diseases 0.000 claims description 9
- 206010005003 Bladder cancer Diseases 0.000 claims description 9
- 206010006187 Breast cancer Diseases 0.000 claims description 9
- 208000026310 Breast neoplasm Diseases 0.000 claims description 9
- 206010014733 Endometrial cancer Diseases 0.000 claims description 9
- 206010014759 Endometrial neoplasm Diseases 0.000 claims description 9
- 208000032612 Glial tumor Diseases 0.000 claims description 9
- 206010018338 Glioma Diseases 0.000 claims description 9
- 206010027406 Mesothelioma Diseases 0.000 claims description 9
- 208000015914 Non-Hodgkin lymphomas Diseases 0.000 claims description 9
- 206010033128 Ovarian cancer Diseases 0.000 claims description 9
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 9
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 9
- 206010039491 Sarcoma Diseases 0.000 claims description 9
- 208000021712 Soft tissue sarcoma Diseases 0.000 claims description 9
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 9
- 201000010536 head and neck cancer Diseases 0.000 claims description 9
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 9
- 208000032839 leukemia Diseases 0.000 claims description 9
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 9
- 201000001441 melanoma Diseases 0.000 claims description 9
- 201000009500 myxoid chondrosarcoma Diseases 0.000 claims description 9
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims description 9
- 201000008968 osteosarcoma Diseases 0.000 claims description 9
- 201000002528 pancreatic cancer Diseases 0.000 claims description 9
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 9
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 claims description 9
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 9
- 125000004448 alkyl carbonyl group Chemical group 0.000 claims description 7
- 125000003917 carbamoyl group Chemical group [H]N([H])C(*)=O 0.000 claims description 7
- 125000001316 cycloalkyl alkyl group Chemical group 0.000 claims description 7
- 238000004519 manufacturing process Methods 0.000 claims description 7
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 6
- 239000000460 chlorine Substances 0.000 claims description 6
- 101150102751 mtap gene Proteins 0.000 claims description 6
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 claims description 5
- 229910052801 chlorine Inorganic materials 0.000 claims description 5
- 230000002950 deficient Effects 0.000 claims description 5
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 5
- 125000002252 acyl group Chemical group 0.000 claims description 4
- 238000012217 deletion Methods 0.000 claims description 4
- 230000037430 deletion Effects 0.000 claims description 4
- 125000002768 hydroxyalkyl group Chemical group 0.000 claims description 4
- 230000001404 mediated effect Effects 0.000 claims description 4
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 4
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 claims description 4
- 125000000956 methoxy group Chemical group [H]C([H])([H])O* 0.000 claims description 3
- 125000004043 oxo group Chemical group O=* 0.000 claims description 3
- 125000004356 hydroxy functional group Chemical group O* 0.000 claims 4
- 125000001475 halogen functional group Chemical group 0.000 claims 1
- 239000003112 inhibitor Substances 0.000 abstract description 15
- 101000947881 Homo sapiens S-adenosylmethionine synthase isoform type-2 Proteins 0.000 abstract description 13
- 102100035947 S-adenosylmethionine synthase isoform type-2 Human genes 0.000 abstract description 11
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 153
- 238000006243 chemical reaction Methods 0.000 description 95
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 89
- 239000012043 crude product Substances 0.000 description 87
- 235000002639 sodium chloride Nutrition 0.000 description 83
- 125000004432 carbon atom Chemical group C* 0.000 description 78
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 62
- 239000000203 mixture Substances 0.000 description 62
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 47
- 238000004440 column chromatography Methods 0.000 description 46
- 239000012074 organic phase Substances 0.000 description 44
- PMZURENOXWZQFD-UHFFFAOYSA-L Sodium Sulfate Chemical compound [Na+].[Na+].[O-]S([O-])(=O)=O PMZURENOXWZQFD-UHFFFAOYSA-L 0.000 description 42
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 39
- 239000007787 solid Substances 0.000 description 38
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 36
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 36
- 239000000243 solution Substances 0.000 description 35
- 239000002904 solvent Substances 0.000 description 35
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 33
- 229910052757 nitrogen Inorganic materials 0.000 description 32
- 239000000543 intermediate Substances 0.000 description 28
- HPALAKNZSZLMCH-UHFFFAOYSA-M sodium;chloride;hydrate Chemical class O.[Na+].[Cl-] HPALAKNZSZLMCH-UHFFFAOYSA-M 0.000 description 28
- 239000003795 chemical substances by application Substances 0.000 description 27
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 26
- 210000004027 cell Anatomy 0.000 description 26
- 125000001424 substituent group Chemical group 0.000 description 24
- IAZDPXIOMUYVGZ-WFGJKAKNSA-N Dimethyl sulfoxide Chemical compound [2H]C([2H])([2H])S(=O)C([2H])([2H])[2H] IAZDPXIOMUYVGZ-WFGJKAKNSA-N 0.000 description 22
- JGFZNNIVVJXRND-UHFFFAOYSA-N N,N-Diisopropylethylamine (DIPEA) Chemical compound CCN(C(C)C)C(C)C JGFZNNIVVJXRND-UHFFFAOYSA-N 0.000 description 21
- 238000005160 1H NMR spectroscopy Methods 0.000 description 20
- 229940002612 prodrug Drugs 0.000 description 20
- 239000000651 prodrug Substances 0.000 description 20
- 239000000843 powder Substances 0.000 description 19
- 239000011541 reaction mixture Substances 0.000 description 18
- 230000000694 effects Effects 0.000 description 17
- 238000004128 high performance liquid chromatography Methods 0.000 description 16
- YXFVVABEGXRONW-UHFFFAOYSA-N Toluene Chemical compound CC1=CC=CC=C1 YXFVVABEGXRONW-UHFFFAOYSA-N 0.000 description 15
- 238000001914 filtration Methods 0.000 description 14
- 238000009472 formulation Methods 0.000 description 14
- 238000003756 stirring Methods 0.000 description 14
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 13
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 13
- VHYFNPMBLIVWCW-UHFFFAOYSA-N 4-Dimethylaminopyridine Chemical compound CN(C)C1=CC=NC=C1 VHYFNPMBLIVWCW-UHFFFAOYSA-N 0.000 description 12
- ZMXDDKWLCZADIW-UHFFFAOYSA-N N,N-Dimethylformamide Chemical compound CN(C)C=O ZMXDDKWLCZADIW-UHFFFAOYSA-N 0.000 description 12
- 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 11
- 239000002253 acid Substances 0.000 description 11
- 238000000605 extraction Methods 0.000 description 11
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 11
- 239000002207 metabolite Substances 0.000 description 11
- 229910052938 sodium sulfate Inorganic materials 0.000 description 11
- 235000011152 sodium sulphate Nutrition 0.000 description 11
- RYHBNJHYFVUHQT-UHFFFAOYSA-N 1,4-Dioxane Chemical compound C1COCCO1 RYHBNJHYFVUHQT-UHFFFAOYSA-N 0.000 description 10
- 229910052799 carbon Inorganic materials 0.000 description 10
- 229940079593 drug Drugs 0.000 description 10
- 239000003921 oil Substances 0.000 description 10
- 235000019198 oils Nutrition 0.000 description 10
- CTSLXHKWHWQRSH-UHFFFAOYSA-N oxalyl chloride Chemical compound ClC(=O)C(Cl)=O CTSLXHKWHWQRSH-UHFFFAOYSA-N 0.000 description 10
- 238000000746 purification Methods 0.000 description 10
- 238000003786 synthesis reaction Methods 0.000 description 10
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 9
- MEFKEPWMEQBLKI-AIRLBKTGSA-N S-adenosyl-L-methioninate Chemical compound O[C@@H]1[C@H](O)[C@@H](C[S+](CC[C@H](N)C([O-])=O)C)O[C@H]1N1C2=NC=NC(N)=C2N=C1 MEFKEPWMEQBLKI-AIRLBKTGSA-N 0.000 description 9
- 239000004480 active ingredient Substances 0.000 description 9
- 239000013543 active substance Substances 0.000 description 9
- 125000004429 atom Chemical group 0.000 description 9
- 239000003153 chemical reaction reagent Substances 0.000 description 9
- 230000005764 inhibitory process Effects 0.000 description 9
- 125000006239 protecting group Chemical group 0.000 description 9
- 229920006395 saturated elastomer Polymers 0.000 description 9
- 238000012360 testing method Methods 0.000 description 9
- ROSDSFDQCJNGOL-UHFFFAOYSA-N Dimethylamine Chemical compound CNC ROSDSFDQCJNGOL-UHFFFAOYSA-N 0.000 description 8
- 108010007784 Methionine adenosyltransferase Proteins 0.000 description 8
- 239000000872 buffer Substances 0.000 description 8
- 238000001514 detection method Methods 0.000 description 8
- 239000002609 medium Substances 0.000 description 8
- 125000002950 monocyclic group Chemical group 0.000 description 8
- 125000003367 polycyclic group Chemical group 0.000 description 8
- 238000002360 preparation method Methods 0.000 description 8
- 239000003826 tablet Substances 0.000 description 8
- 230000001225 therapeutic effect Effects 0.000 description 8
- WUUGFSXJNOTRMR-IOSLPCCCSA-N 5'-S-methyl-5'-thioadenosine Chemical compound O[C@@H]1[C@H](O)[C@@H](CSC)O[C@H]1N1C2=NC=NC(N)=C2N=C1 WUUGFSXJNOTRMR-IOSLPCCCSA-N 0.000 description 7
- NLXLAEXVIDQMFP-UHFFFAOYSA-N Ammonia chloride Chemical compound [NH4+].[Cl-] NLXLAEXVIDQMFP-UHFFFAOYSA-N 0.000 description 7
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 7
- 102000007357 Methionine adenosyltransferase Human genes 0.000 description 7
- 208000035475 disorder Diseases 0.000 description 7
- 239000005457 ice water Substances 0.000 description 7
- 239000012299 nitrogen atmosphere Substances 0.000 description 7
- 239000012453 solvate Substances 0.000 description 7
- 239000000725 suspension Substances 0.000 description 7
- 102000004190 Enzymes Human genes 0.000 description 6
- 108090000790 Enzymes Proteins 0.000 description 6
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 6
- 239000002775 capsule Substances 0.000 description 6
- 238000002648 combination therapy Methods 0.000 description 6
- 239000003085 diluting agent Substances 0.000 description 6
- 150000002148 esters Chemical class 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000000463 material Substances 0.000 description 6
- 239000000126 substance Substances 0.000 description 6
- 229910052717 sulfur Inorganic materials 0.000 description 6
- 239000011593 sulfur Substances 0.000 description 6
- 208000024891 symptom Diseases 0.000 description 6
- SCYULBFZEHDVBN-UHFFFAOYSA-N 1,1-Dichloroethane Chemical compound CC(Cl)Cl SCYULBFZEHDVBN-UHFFFAOYSA-N 0.000 description 5
- WSLDOOZREJYCGB-UHFFFAOYSA-N 1,2-Dichloroethane Chemical compound ClCCCl WSLDOOZREJYCGB-UHFFFAOYSA-N 0.000 description 5
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 5
- JAPYIBBSTJFDAK-UHFFFAOYSA-N 2,4,6-tri(propan-2-yl)benzenesulfonyl chloride Chemical compound CC(C)C1=CC(C(C)C)=C(S(Cl)(=O)=O)C(C(C)C)=C1 JAPYIBBSTJFDAK-UHFFFAOYSA-N 0.000 description 5
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 5
- 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 5
- 150000007513 acids Chemical class 0.000 description 5
- 239000012298 atmosphere Substances 0.000 description 5
- 239000007859 condensation product Substances 0.000 description 5
- 235000014113 dietary fatty acids Nutrition 0.000 description 5
- 239000002270 dispersing agent Substances 0.000 description 5
- 229960004679 doxorubicin Drugs 0.000 description 5
- 238000001704 evaporation Methods 0.000 description 5
- 230000008020 evaporation Effects 0.000 description 5
- 239000000194 fatty acid Substances 0.000 description 5
- 229930195729 fatty acid Natural products 0.000 description 5
- 150000004665 fatty acids Chemical class 0.000 description 5
- 239000003550 marker Substances 0.000 description 5
- 238000006467 substitution reaction Methods 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- 239000000375 suspending agent Substances 0.000 description 5
- WORJRXHJTUTINR-UHFFFAOYSA-N 1,4-dioxane;hydron;chloride Chemical compound Cl.C1COCCO1 WORJRXHJTUTINR-UHFFFAOYSA-N 0.000 description 4
- GQHTUMJGOHRCHB-UHFFFAOYSA-N 2,3,4,6,7,8,9,10-octahydropyrimido[1,2-a]azepine Chemical compound C1CCCCN2CCCN=C21 GQHTUMJGOHRCHB-UHFFFAOYSA-N 0.000 description 4
- BPXKZEMBEZGUAH-UHFFFAOYSA-N 2-(chloromethoxy)ethyl-trimethylsilane Chemical compound C[Si](C)(C)CCOCCl BPXKZEMBEZGUAH-UHFFFAOYSA-N 0.000 description 4
- HZAXFHJVJLSVMW-UHFFFAOYSA-N 2-Aminoethan-1-ol Chemical compound NCCO HZAXFHJVJLSVMW-UHFFFAOYSA-N 0.000 description 4
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 4
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 4
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 4
- IAYPIBMASNFSPL-UHFFFAOYSA-N Ethylene oxide Chemical compound C1CO1 IAYPIBMASNFSPL-UHFFFAOYSA-N 0.000 description 4
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 4
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 4
- 150000001412 amines Chemical class 0.000 description 4
- 239000002246 antineoplastic agent Substances 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 4
- 125000000499 benzofuranyl group Chemical group O1C(=CC2=C1C=CC=C2)* 0.000 description 4
- 239000013078 crystal Substances 0.000 description 4
- 125000004122 cyclic group Chemical group 0.000 description 4
- 229940127089 cytotoxic agent Drugs 0.000 description 4
- 239000002552 dosage form Substances 0.000 description 4
- 125000000524 functional group Chemical group 0.000 description 4
- 125000005842 heteroatom Chemical group 0.000 description 4
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 4
- 125000005647 linker group Chemical group 0.000 description 4
- 239000007788 liquid Substances 0.000 description 4
- 230000002503 metabolic effect Effects 0.000 description 4
- 229960004452 methionine Drugs 0.000 description 4
- 230000007935 neutral effect Effects 0.000 description 4
- 231100000252 nontoxic Toxicity 0.000 description 4
- 230000003000 nontoxic effect Effects 0.000 description 4
- 230000003287 optical effect Effects 0.000 description 4
- 239000012044 organic layer Substances 0.000 description 4
- 229910052760 oxygen Inorganic materials 0.000 description 4
- 239000001301 oxygen Substances 0.000 description 4
- 230000036961 partial effect Effects 0.000 description 4
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 4
- 239000003755 preservative agent Substances 0.000 description 4
- 125000004076 pyridyl group Chemical group 0.000 description 4
- 125000006413 ring segment Chemical group 0.000 description 4
- 230000019491 signal transduction Effects 0.000 description 4
- CESUXLKAADQNTB-UHFFFAOYSA-N tert-butanesulfinamide Chemical compound CC(C)(C)S(N)=O CESUXLKAADQNTB-UHFFFAOYSA-N 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- WYWHKKSPHMUBEB-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 4
- 238000011269 treatment regimen Methods 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- 239000000080 wetting agent Substances 0.000 description 4
- BFPMCZWKUSUMKE-UHFFFAOYSA-N 1-cyclopropyl-n-methylmethanamine Chemical compound CNCC1CC1 BFPMCZWKUSUMKE-UHFFFAOYSA-N 0.000 description 3
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 3
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 3
- 239000004475 Arginine Substances 0.000 description 3
- 235000003351 Brassica cretica Nutrition 0.000 description 3
- 235000003343 Brassica rupestris Nutrition 0.000 description 3
- 241000219193 Brassicaceae Species 0.000 description 3
- 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 3
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 3
- OVBPIULPVIDEAO-LBPRGKRZSA-N Folic acid Natural products C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 102000037982 Immune checkpoint proteins Human genes 0.000 description 3
- 108091008036 Immune checkpoint proteins Proteins 0.000 description 3
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 3
- 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 3
- 241000124008 Mammalia Species 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 241000699670 Mus sp. Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 102100034607 Protein arginine N-methyltransferase 5 Human genes 0.000 description 3
- 101710084427 Protein arginine N-methyltransferase 5 Proteins 0.000 description 3
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 3
- 238000002835 absorbance Methods 0.000 description 3
- 238000010521 absorption reaction Methods 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 235000019270 ammonium chloride Nutrition 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 239000008346 aqueous phase Substances 0.000 description 3
- 239000007864 aqueous solution Substances 0.000 description 3
- 239000007900 aqueous suspension Substances 0.000 description 3
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 230000003833 cell viability Effects 0.000 description 3
- 229960004630 chlorambucil Drugs 0.000 description 3
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 238000007405 data analysis Methods 0.000 description 3
- 238000009510 drug design Methods 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 230000002255 enzymatic effect Effects 0.000 description 3
- XGZNHFPFJRZBBT-UHFFFAOYSA-N ethanol;titanium Chemical compound [Ti].CCO.CCO.CCO.CCO XGZNHFPFJRZBBT-UHFFFAOYSA-N 0.000 description 3
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 3
- 239000000706 filtrate Substances 0.000 description 3
- 229960002949 fluorouracil Drugs 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 150000002430 hydrocarbons Chemical group 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 229940043355 kinase inhibitor Drugs 0.000 description 3
- GFIJNRVAKGFPGQ-LIJARHBVSA-N leuprolide Chemical compound CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 GFIJNRVAKGFPGQ-LIJARHBVSA-N 0.000 description 3
- 229960004338 leuprorelin Drugs 0.000 description 3
- 229940057995 liquid paraffin Drugs 0.000 description 3
- 229940107698 malachite green Drugs 0.000 description 3
- FDZZZRQASAIRJF-UHFFFAOYSA-M malachite green Chemical compound [Cl-].C1=CC(N(C)C)=CC=C1C(C=1C=CC=CC=1)=C1C=CC(=[N+](C)C)C=C1 FDZZZRQASAIRJF-UHFFFAOYSA-M 0.000 description 3
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 3
- 229960000485 methotrexate Drugs 0.000 description 3
- 239000003094 microcapsule Substances 0.000 description 3
- 229960004857 mitomycin Drugs 0.000 description 3
- 238000002156 mixing Methods 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 239000001788 mono and diglycerides of fatty acids Substances 0.000 description 3
- 235000010460 mustard Nutrition 0.000 description 3
- 239000004006 olive oil Substances 0.000 description 3
- 235000008390 olive oil Nutrition 0.000 description 3
- 239000012071 phase Substances 0.000 description 3
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 3
- 238000004237 preparative chromatography Methods 0.000 description 3
- GZUITABIAKMVPG-UHFFFAOYSA-N raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 3
- 229960004622 raloxifene Drugs 0.000 description 3
- 239000002994 raw material Substances 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 238000003860 storage Methods 0.000 description 3
- 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 3
- 229960004964 temozolomide Drugs 0.000 description 3
- 125000003039 tetrahydroisoquinolinyl group Chemical group C1(NCCC2=CC=CC=C12)* 0.000 description 3
- 125000000147 tetrahydroquinolinyl group Chemical group N1(CCCC2=CC=CC=C12)* 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 210000001519 tissue Anatomy 0.000 description 3
- JMXKSZRRTHPKDL-UHFFFAOYSA-N titanium(IV) ethoxide Substances [Ti+4].CC[O-].CC[O-].CC[O-].CC[O-] JMXKSZRRTHPKDL-UHFFFAOYSA-N 0.000 description 3
- PUPZLCDOIYMWBV-UHFFFAOYSA-N (+/-)-1,3-Butanediol Chemical compound CC(O)CCO PUPZLCDOIYMWBV-UHFFFAOYSA-N 0.000 description 2
- QYYZXEPEVBXNNA-QGZVFWFLSA-N (1R)-2-acetyl-N-[4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan-2-yl)phenyl]-5-methylsulfonyl-1,3-dihydroisoindole-1-carboxamide Chemical compound C(C)(=O)N1[C@H](C2=CC=C(C=C2C1)S(=O)(=O)C)C(=O)NC1=CC=C(C=C1)C(C(F)(F)F)(C(F)(F)F)O QYYZXEPEVBXNNA-QGZVFWFLSA-N 0.000 description 2
- IEXUMDBQLIVNHZ-YOUGDJEHSA-N (8s,11r,13r,14s,17s)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one Chemical compound C1=CC(N(C)C)=CC=C1[C@@H]1C2=C3CCC(=O)C=C3CC[C@H]2[C@H](CC[C@]2(O)CCCO)[C@@]2(C)C1 IEXUMDBQLIVNHZ-YOUGDJEHSA-N 0.000 description 2
- IAKHMKGGTNLKSZ-INIZCTEOSA-N (S)-colchicine Chemical compound C1([C@@H](NC(C)=O)CC2)=CC(=O)C(OC)=CC=C1C1=C2C=C(OC)C(OC)=C1OC IAKHMKGGTNLKSZ-INIZCTEOSA-N 0.000 description 2
- ZORQXIQZAOLNGE-UHFFFAOYSA-N 1,1-difluorocyclohexane Chemical compound FC1(F)CCCCC1 ZORQXIQZAOLNGE-UHFFFAOYSA-N 0.000 description 2
- RKDVKSZUMVYZHH-UHFFFAOYSA-N 1,4-dioxane-2,5-dione Chemical compound O=C1COC(=O)CO1 RKDVKSZUMVYZHH-UHFFFAOYSA-N 0.000 description 2
- 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 2
- SXGZJKUKBWWHRA-UHFFFAOYSA-N 2-(N-morpholiniumyl)ethanesulfonate Chemical compound [O-]S(=O)(=O)CC[NH+]1CCOCC1 SXGZJKUKBWWHRA-UHFFFAOYSA-N 0.000 description 2
- HNXQXTQTPAJEJL-UHFFFAOYSA-N 2-aminopteridin-4-ol Chemical compound C1=CN=C2NC(N)=NC(=O)C2=N1 HNXQXTQTPAJEJL-UHFFFAOYSA-N 0.000 description 2
- IZHVBANLECCAGF-UHFFFAOYSA-N 2-hydroxy-3-(octadecanoyloxy)propyl octadecanoate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)COC(=O)CCCCCCCCCCCCCCCCC IZHVBANLECCAGF-UHFFFAOYSA-N 0.000 description 2
- DVLFYONBTKHTER-UHFFFAOYSA-N 3-(N-morpholino)propanesulfonic acid Chemical compound OS(=O)(=O)CCCN1CCOCC1 DVLFYONBTKHTER-UHFFFAOYSA-N 0.000 description 2
- NMUSYJAQQFHJEW-KVTDHHQDSA-N 5-azacytidine Chemical compound O=C1N=C(N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NMUSYJAQQFHJEW-KVTDHHQDSA-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
- 239000005660 Abamectin Substances 0.000 description 2
- 108010012934 Albumin-Bound Paclitaxel Proteins 0.000 description 2
- 235000003911 Arachis Nutrition 0.000 description 2
- 244000105624 Arachis hypogaea Species 0.000 description 2
- 241000416162 Astragalus gummifer Species 0.000 description 2
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 2
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 2
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 2
- 108010092160 Dactinomycin Proteins 0.000 description 2
- QUSNBJAOOMFDIB-UHFFFAOYSA-N Ethylamine Chemical compound CCN QUSNBJAOOMFDIB-UHFFFAOYSA-N 0.000 description 2
- PXGOKWXKJXAPGV-UHFFFAOYSA-N Fluorine Chemical compound FF PXGOKWXKJXAPGV-UHFFFAOYSA-N 0.000 description 2
- IAJILQKETJEXLJ-UHFFFAOYSA-N Galacturonsaeure Natural products O=CC(O)C(O)C(O)C(O)C(O)=O IAJILQKETJEXLJ-UHFFFAOYSA-N 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 description 2
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 description 2
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 description 2
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 2
- FFEARJCKVFRZRR-UHFFFAOYSA-N L-Methionine Natural products CSCCC(N)C(O)=O FFEARJCKVFRZRR-UHFFFAOYSA-N 0.000 description 2
- 229930195722 L-methionine Natural products 0.000 description 2
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 2
- 240000007472 Leucaena leucocephala Species 0.000 description 2
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 2
- YNAVUWVOSKDBBP-UHFFFAOYSA-N Morpholine Chemical compound C1COCCN1 YNAVUWVOSKDBBP-UHFFFAOYSA-N 0.000 description 2
- OVBPIULPVIDEAO-UHFFFAOYSA-N N-Pteroyl-L-glutaminsaeure Natural products C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-UHFFFAOYSA-N 0.000 description 2
- YNLCVAQJIKOXER-UHFFFAOYSA-N N-[tris(hydroxymethyl)methyl]-3-aminopropanesulfonic acid Chemical compound OCC(CO)(CO)NCCCS(O)(=O)=O YNLCVAQJIKOXER-UHFFFAOYSA-N 0.000 description 2
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N N-debenzoyl-N-(tert-butoxycarbonyl)-10-deacetyltaxol Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical group [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 2
- 108091000080 Phosphotransferase Proteins 0.000 description 2
- GLUUGHFHXGJENI-UHFFFAOYSA-N Piperazine Chemical compound C1CNCCN1 GLUUGHFHXGJENI-UHFFFAOYSA-N 0.000 description 2
- NQRYJNQNLNOLGT-UHFFFAOYSA-N Piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- KAESVJOAVNADME-UHFFFAOYSA-N Pyrrole Chemical compound C=1C=CNC=1 KAESVJOAVNADME-UHFFFAOYSA-N 0.000 description 2
- SMWDFEZZVXVKRB-UHFFFAOYSA-N Quinoline Chemical compound N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 2
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 108091008874 T cell receptors Proteins 0.000 description 2
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 2
- 210000001744 T-lymphocyte Anatomy 0.000 description 2
- 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 2
- MUMGGOZAMZWBJJ-DYKIIFRCSA-N Testostosterone Chemical compound O=C1CC[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 MUMGGOZAMZWBJJ-DYKIIFRCSA-N 0.000 description 2
- FOCVUCIESVLUNU-UHFFFAOYSA-N Thiotepa Chemical compound C1CN1P(N1CC1)(=S)N1CC1 FOCVUCIESVLUNU-UHFFFAOYSA-N 0.000 description 2
- 229920001615 Tragacanth Polymers 0.000 description 2
- FYAMXEPQQLNQDM-UHFFFAOYSA-N Tris(1-aziridinyl)phosphine oxide Chemical compound C1CN1P(N1CC1)(=O)N1CC1 FYAMXEPQQLNQDM-UHFFFAOYSA-N 0.000 description 2
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 235000011054 acetic acid Nutrition 0.000 description 2
- 230000002378 acidificating effect Effects 0.000 description 2
- 229930183665 actinomycin Natural products 0.000 description 2
- 229960005305 adenosine Drugs 0.000 description 2
- 125000002947 alkylene group Chemical group 0.000 description 2
- 235000001014 amino acid Nutrition 0.000 description 2
- 150000001413 amino acids Chemical class 0.000 description 2
- 125000003277 amino group Chemical group 0.000 description 2
- 239000003708 ampul Substances 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 229940088710 antibiotic agent Drugs 0.000 description 2
- 235000010323 ascorbic acid Nutrition 0.000 description 2
- 239000011668 ascorbic acid Substances 0.000 description 2
- 229960005070 ascorbic acid Drugs 0.000 description 2
- 239000012131 assay buffer Substances 0.000 description 2
- 229960002756 azacitidine Drugs 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 125000003785 benzimidazolyl group Chemical group N1=C(NC2=C1C=CC=C2)* 0.000 description 2
- 125000001164 benzothiazolyl group Chemical group S1C(=NC2=C1C=CC=C2)* 0.000 description 2
- 125000004196 benzothienyl group Chemical group S1C(=CC2=C1C=CC=C2)* 0.000 description 2
- RROBIDXNTUAHFW-UHFFFAOYSA-N benzotriazol-1-yloxy-tris(dimethylamino)phosphanium Chemical compound C1=CC=C2N(O[P+](N(C)C)(N(C)C)N(C)C)N=NC2=C1 RROBIDXNTUAHFW-UHFFFAOYSA-N 0.000 description 2
- 239000013060 biological fluid Substances 0.000 description 2
- 239000007975 buffered saline Substances 0.000 description 2
- YSHOWEKUVWPFNR-UHFFFAOYSA-N burgess reagent Chemical compound CC[N+](CC)(CC)S(=O)(=O)N=C([O-])OC YSHOWEKUVWPFNR-UHFFFAOYSA-N 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical compound CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- 229910000019 calcium carbonate Inorganic materials 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 229910000389 calcium phosphate Inorganic materials 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- 125000000609 carbazolyl group Chemical group C1(=CC=CC=2C3=CC=CC=C3NC12)* 0.000 description 2
- 125000002837 carbocyclic group Chemical group 0.000 description 2
- 239000001569 carbon dioxide Substances 0.000 description 2
- 229910002092 carbon dioxide Inorganic materials 0.000 description 2
- 239000011203 carbon fibre reinforced carbon Substances 0.000 description 2
- 239000001768 carboxy methyl cellulose Substances 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 229960004316 cisplatin Drugs 0.000 description 2
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 2
- 238000013270 controlled release Methods 0.000 description 2
- 238000002425 crystallisation Methods 0.000 description 2
- 230000008025 crystallization Effects 0.000 description 2
- 125000006165 cyclic alkyl group Chemical group 0.000 description 2
- 229960004397 cyclophosphamide Drugs 0.000 description 2
- 230000016396 cytokine production Effects 0.000 description 2
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 2
- 239000000824 cytostatic agent Substances 0.000 description 2
- 230000001085 cytostatic effect Effects 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
- 238000006731 degradation reaction Methods 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 238000010511 deprotection reaction Methods 0.000 description 2
- 239000000032 diagnostic agent Substances 0.000 description 2
- 229940039227 diagnostic agent Drugs 0.000 description 2
- HPNMFZURTQLUMO-UHFFFAOYSA-N diethylamine Chemical compound CCNCC HPNMFZURTQLUMO-UHFFFAOYSA-N 0.000 description 2
- SGDINNZGYDHHKM-UHFFFAOYSA-N dilithium;trimethylsilylazanide Chemical compound [Li+].[Li+].C[Si](C)(C)[NH-].C[Si](C)(C)[NH-] SGDINNZGYDHHKM-UHFFFAOYSA-N 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- ZUOUZKKEUPVFJK-UHFFFAOYSA-N diphenyl Chemical compound C1=CC=CC=C1C1=CC=CC=C1 ZUOUZKKEUPVFJK-UHFFFAOYSA-N 0.000 description 2
- 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 2
- 229960003668 docetaxel Drugs 0.000 description 2
- 238000012377 drug delivery Methods 0.000 description 2
- VLCYCQAOQCDTCN-UHFFFAOYSA-N eflornithine Chemical compound NCCCC(N)(C(F)F)C(O)=O VLCYCQAOQCDTCN-UHFFFAOYSA-N 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 238000006911 enzymatic reaction Methods 0.000 description 2
- UFNVPOGXISZXJD-JBQZKEIOSA-N eribulin Chemical compound C([C@H]1CC[C@@H]2O[C@@H]3[C@H]4O[C@@H]5C[C@](O[C@H]4[C@H]2O1)(O[C@@H]53)CC[C@@H]1O[C@H](C(C1)=C)CC1)C(=O)C[C@@H]2[C@@H](OC)[C@@H](C[C@H](O)CN)O[C@H]2C[C@@H]2C(=C)[C@H](C)C[C@H]1O2 UFNVPOGXISZXJD-JBQZKEIOSA-N 0.000 description 2
- 238000003810 ethyl acetate extraction Methods 0.000 description 2
- 239000000796 flavoring agent Substances 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 239000011737 fluorine Substances 0.000 description 2
- 229910052731 fluorine Inorganic materials 0.000 description 2
- 229960000304 folic acid Drugs 0.000 description 2
- 235000019152 folic acid Nutrition 0.000 description 2
- 239000011724 folic acid Substances 0.000 description 2
- 235000013355 food flavoring agent Nutrition 0.000 description 2
- 235000003599 food sweetener Nutrition 0.000 description 2
- 230000008014 freezing Effects 0.000 description 2
- 238000007710 freezing Methods 0.000 description 2
- 125000002541 furyl group Chemical group 0.000 description 2
- CHPZKNULDCNCBW-UHFFFAOYSA-N gallium nitrate Chemical compound [Ga+3].[O-][N+]([O-])=O.[O-][N+]([O-])=O.[O-][N+]([O-])=O CHPZKNULDCNCBW-UHFFFAOYSA-N 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 239000007903 gelatin capsule Substances 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 229940075507 glyceryl monostearate Drugs 0.000 description 2
- KWIUHFFTVRNATP-UHFFFAOYSA-N glycine betaine Chemical compound C[N+](C)(C)CC([O-])=O KWIUHFFTVRNATP-UHFFFAOYSA-N 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 229940093915 gynecological organic acid Drugs 0.000 description 2
- 125000005843 halogen group Chemical group 0.000 description 2
- 238000010438 heat treatment Methods 0.000 description 2
- BXWNKGSJHAJOGX-UHFFFAOYSA-N hexadecan-1-ol Chemical compound CCCCCCCCCCCCCCCCO BXWNKGSJHAJOGX-UHFFFAOYSA-N 0.000 description 2
- VKYKSIONXSXAKP-UHFFFAOYSA-N hexamethylenetetramine Chemical compound C1N(C2)CN3CN1CN2C3 VKYKSIONXSXAKP-UHFFFAOYSA-N 0.000 description 2
- 229940125697 hormonal agent Drugs 0.000 description 2
- XGIHQYAWBCFNPY-AZOCGYLKSA-N hydrabamine Chemical compound C([C@@H]12)CC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC[C@@]1(C)CNCCNC[C@@]1(C)[C@@H]2CCC3=CC(C(C)C)=CC=C3[C@@]2(C)CCC1 XGIHQYAWBCFNPY-AZOCGYLKSA-N 0.000 description 2
- 150000004677 hydrates Chemical class 0.000 description 2
- 239000000017 hydrogel Substances 0.000 description 2
- 125000004435 hydrogen atom Chemical group [H]* 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
- 125000002883 imidazolyl group Chemical group 0.000 description 2
- 210000000987 immune system Anatomy 0.000 description 2
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 2
- 125000003453 indazolyl group Chemical group N1N=C(C2=C1C=CC=C2)* 0.000 description 2
- 125000003406 indolizinyl group Chemical group C=1(C=CN2C=CC=CC12)* 0.000 description 2
- 125000001041 indolyl group Chemical group 0.000 description 2
- 239000012442 inert solvent Substances 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 229910052742 iron Inorganic materials 0.000 description 2
- 125000000904 isoindolyl group Chemical group C=1(NC=C2C=CC=CC12)* 0.000 description 2
- 125000002183 isoquinolinyl group Chemical group C1(=NC=CC2=CC=CC=C12)* 0.000 description 2
- FABUFPQFXZVHFB-PVYNADRNSA-N ixabepilone Chemical compound C/C([C@@H]1C[C@@H]2O[C@]2(C)CCC[C@@H]([C@@H]([C@@H](C)C(=O)C(C)(C)[C@@H](O)CC(=O)N1)O)C)=C\C1=CSC(C)=N1 FABUFPQFXZVHFB-PVYNADRNSA-N 0.000 description 2
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 2
- 235000010445 lecithin Nutrition 0.000 description 2
- 239000000787 lecithin Substances 0.000 description 2
- 229940067606 lecithin Drugs 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- YNESATAKKCNGOF-UHFFFAOYSA-N lithium bis(trimethylsilyl)amide Chemical compound [Li+].C[Si](C)(C)[N-][Si](C)(C)C YNESATAKKCNGOF-UHFFFAOYSA-N 0.000 description 2
- 229960002247 lomustine Drugs 0.000 description 2
- 239000007937 lozenge Substances 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- WPBNNNQJVZRUHP-UHFFFAOYSA-L manganese(2+);methyl n-[[2-(methoxycarbonylcarbamothioylamino)phenyl]carbamothioyl]carbamate;n-[2-(sulfidocarbothioylamino)ethyl]carbamodithioate Chemical compound [Mn+2].[S-]C(=S)NCCNC([S-])=S.COC(=O)NC(=S)NC1=CC=CC=C1NC(=S)NC(=O)OC WPBNNNQJVZRUHP-UHFFFAOYSA-L 0.000 description 2
- 230000010534 mechanism of action Effects 0.000 description 2
- 229960001428 mercaptopurine Drugs 0.000 description 2
- 230000004060 metabolic process Effects 0.000 description 2
- OKKJLVBELUTLKV-VMNATFBRSA-N methanol-d1 Chemical compound [2H]OC OKKJLVBELUTLKV-VMNATFBRSA-N 0.000 description 2
- 229930182817 methionine Natural products 0.000 description 2
- 229920000609 methyl cellulose Polymers 0.000 description 2
- 235000010981 methylcellulose Nutrition 0.000 description 2
- 239000001923 methylcellulose Substances 0.000 description 2
- LXCFILQKKLGQFO-UHFFFAOYSA-N methylparaben Chemical compound COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 2
- 239000000693 micelle Substances 0.000 description 2
- 239000011325 microbead Substances 0.000 description 2
- 239000002480 mineral oil Substances 0.000 description 2
- 235000010446 mineral oil Nutrition 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
- 229960001156 mitoxantrone Drugs 0.000 description 2
- 229940042880 natural phospholipid Drugs 0.000 description 2
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 2
- 229960003301 nivolumab Drugs 0.000 description 2
- 239000000346 nonvolatile oil Substances 0.000 description 2
- 229950011093 onapristone Drugs 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 150000007524 organic acids Chemical class 0.000 description 2
- 235000005985 organic acids Nutrition 0.000 description 2
- 125000002971 oxazolyl group Chemical group 0.000 description 2
- 125000002255 pentenyl group Chemical group C(=CCCC)* 0.000 description 2
- 230000000737 periodic effect Effects 0.000 description 2
- 230000003285 pharmacodynamic effect Effects 0.000 description 2
- 125000004934 phenanthridinyl group Chemical group C1(=CC=CC2=NC=C3C=CC=CC3=C12)* 0.000 description 2
- 125000001791 phenazinyl group Chemical group C1(=CC=CC2=NC3=CC=CC=C3N=C12)* 0.000 description 2
- 125000001484 phenothiazinyl group Chemical group C1(=CC=CC=2SC3=CC=CC=C3NC12)* 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 229910052698 phosphorus Inorganic materials 0.000 description 2
- 239000011574 phosphorus Chemical group 0.000 description 2
- 102000020233 phosphotransferase Human genes 0.000 description 2
- 230000004962 physiological condition Effects 0.000 description 2
- 125000003386 piperidinyl group Chemical group 0.000 description 2
- 229910052697 platinum Inorganic materials 0.000 description 2
- 229920001200 poly(ethylene-vinyl acetate) Polymers 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 2
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 2
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 2
- 239000011591 potassium Substances 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- BWHMMNNQKKPAPP-UHFFFAOYSA-L potassium carbonate Chemical compound [K+].[K+].[O-]C([O-])=O BWHMMNNQKKPAPP-UHFFFAOYSA-L 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 239000000047 product Substances 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 125000001042 pteridinyl group Chemical group N1=C(N=CC2=NC=CN=C12)* 0.000 description 2
- 150000003212 purines Chemical class 0.000 description 2
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 125000003373 pyrazinyl group Chemical group 0.000 description 2
- 125000003226 pyrazolyl group Chemical group 0.000 description 2
- 125000002098 pyridazinyl group Chemical group 0.000 description 2
- 125000000714 pyrimidinyl group Chemical group 0.000 description 2
- 125000000168 pyrrolyl group Chemical group 0.000 description 2
- 125000002294 quinazolinyl group Chemical group N1=C(N=CC2=CC=CC=C12)* 0.000 description 2
- 125000002943 quinolinyl group Chemical group N1=C(C=CC2=CC=CC=C12)* 0.000 description 2
- 125000001567 quinoxalinyl group Chemical group N1=C(C=NC2=CC=CC=C12)* 0.000 description 2
- 238000001959 radiotherapy Methods 0.000 description 2
- 239000000376 reactant Substances 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 235000011069 sorbitan monooleate Nutrition 0.000 description 2
- 239000001593 sorbitan monooleate Substances 0.000 description 2
- 229940035049 sorbitan monooleate Drugs 0.000 description 2
- 238000009987 spinning Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 239000003765 sweetening agent Substances 0.000 description 2
- 230000002195 synergetic effect Effects 0.000 description 2
- DYHSDKLCOJIUFX-UHFFFAOYSA-N tert-butoxycarbonyl anhydride Chemical compound CC(C)(C)OC(=O)OC(=O)OC(C)(C)C DYHSDKLCOJIUFX-UHFFFAOYSA-N 0.000 description 2
- FPGGTKZVZWFYPV-UHFFFAOYSA-M tetrabutylammonium fluoride Chemical compound [F-].CCCC[N+](CCCC)(CCCC)CCCC FPGGTKZVZWFYPV-UHFFFAOYSA-M 0.000 description 2
- YAPQBXQYLJRXSA-UHFFFAOYSA-N theobromine Chemical compound CN1C(=O)NC(=O)C2=C1N=CN2C YAPQBXQYLJRXSA-UHFFFAOYSA-N 0.000 description 2
- 230000004797 therapeutic response Effects 0.000 description 2
- 125000000335 thiazolyl group Chemical group 0.000 description 2
- 125000001544 thienyl group Chemical group 0.000 description 2
- 238000004809 thin layer chromatography Methods 0.000 description 2
- 229960001196 thiotepa Drugs 0.000 description 2
- 229960003087 tioguanine Drugs 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- 125000001425 triazolyl group Chemical group 0.000 description 2
- 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 2
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 2
- RIOQSEWOXXDEQQ-UHFFFAOYSA-N triphenylphosphine Chemical compound C1=CC=CC=C1P(C=1C=CC=CC=1)C1=CC=CC=C1 RIOQSEWOXXDEQQ-UHFFFAOYSA-N 0.000 description 2
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 230000004614 tumor growth Effects 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- 125000000391 vinyl group Chemical group [H]C([*])=C([H])[H] 0.000 description 2
- 229920002554 vinyl polymer Polymers 0.000 description 2
- 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 1
- VXAGJYXIHQKJIP-REOHCLBHSA-N (2S)-2-azido-3-hydroxypropanoic acid Chemical compound [N+](=[N-])=N[C@@H](CO)C(=O)O VXAGJYXIHQKJIP-REOHCLBHSA-N 0.000 description 1
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2r,3r,4s,5r,6s)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2s,3r,4s,5r,6r)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2r,3r,4s,5r,6r)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 1
- LMGGOGHEVZMZCU-FGJMKEJPSA-N (2s,4s)-4-[(2r,4s,5s,6s)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-2,5,7,12-tetrahydroxy-6,11-dioxo-3,4-dihydro-1h-tetracene-2-carboxylic acid 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=C(O)C=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(O)=O)C1 LMGGOGHEVZMZCU-FGJMKEJPSA-N 0.000 description 1
- CGMTUJFWROPELF-YPAAEMCBSA-N (3E,5S)-5-[(2S)-butan-2-yl]-3-(1-hydroxyethylidene)pyrrolidine-2,4-dione Chemical compound CC[C@H](C)[C@@H]1NC(=O)\C(=C(/C)O)C1=O CGMTUJFWROPELF-YPAAEMCBSA-N 0.000 description 1
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 1
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 1
- LKJPYSCBVHEWIU-KRWDZBQOSA-N (R)-bicalutamide Chemical compound C([C@@](O)(C)C(=O)NC=1C=C(C(C#N)=CC=1)C(F)(F)F)S(=O)(=O)C1=CC=C(F)C=C1 LKJPYSCBVHEWIU-KRWDZBQOSA-N 0.000 description 1
- VOJUXHHACRXLTD-UHFFFAOYSA-N 1,4-dihydroxy-2-naphthoic acid Chemical compound C1=CC=CC2=C(O)C(C(=O)O)=CC(O)=C21 VOJUXHHACRXLTD-UHFFFAOYSA-N 0.000 description 1
- ONBQEOIKXPHGMB-VBSBHUPXSA-N 1-[2-[(2s,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]oxy-4,6-dihydroxyphenyl]-3-(4-hydroxyphenyl)propan-1-one Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1OC1=CC(O)=CC(O)=C1C(=O)CCC1=CC=C(O)C=C1 ONBQEOIKXPHGMB-VBSBHUPXSA-N 0.000 description 1
- 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 1
- DEEULBIVHZVMHX-UHFFFAOYSA-N 1-nitroacridine Chemical compound C1=CC=C2C=C3C([N+](=O)[O-])=CC=CC3=NC2=C1 DEEULBIVHZVMHX-UHFFFAOYSA-N 0.000 description 1
- 125000000530 1-propynyl group Chemical group [H]C([H])([H])C#C* 0.000 description 1
- 125000001462 1-pyrrolyl group Chemical group [*]N1C([H])=C([H])C([H])=C1[H] 0.000 description 1
- AVRPFRMDMNDIDH-UHFFFAOYSA-N 1h-quinazolin-2-one Chemical class C1=CC=CC2=NC(O)=NC=C21 AVRPFRMDMNDIDH-UHFFFAOYSA-N 0.000 description 1
- BTOTXLJHDSNXMW-POYBYMJQSA-N 2,3-dideoxyuridine Chemical compound O1[C@H](CO)CC[C@@H]1N1C(=O)NC(=O)C=C1 BTOTXLJHDSNXMW-POYBYMJQSA-N 0.000 description 1
- MSWZFWKMSRAUBD-IVMDWMLBSA-N 2-amino-2-deoxy-D-glucopyranose Chemical compound N[C@H]1C(O)O[C@H](CO)[C@@H](O)[C@@H]1O MSWZFWKMSRAUBD-IVMDWMLBSA-N 0.000 description 1
- BFSVOASYOCHEOV-UHFFFAOYSA-N 2-diethylaminoethanol Chemical compound CCN(CC)CCO BFSVOASYOCHEOV-UHFFFAOYSA-N 0.000 description 1
- 229940013085 2-diethylaminoethanol Drugs 0.000 description 1
- KOHBEDRJXKOYHL-UHFFFAOYSA-N 2-methoxy-n-methylethanamine Chemical compound CNCCOC KOHBEDRJXKOYHL-UHFFFAOYSA-N 0.000 description 1
- 125000004398 2-methyl-2-butyl group Chemical group CC(C)(CC)* 0.000 description 1
- 125000004918 2-methyl-2-pentyl group Chemical group CC(C)(CCC)* 0.000 description 1
- 125000004493 2-methylbut-1-yl group Chemical group CC(C*)CC 0.000 description 1
- CTRPRMNBTVRDFH-UHFFFAOYSA-N 2-n-methyl-1,3,5-triazine-2,4,6-triamine Chemical class CNC1=NC(N)=NC(N)=N1 CTRPRMNBTVRDFH-UHFFFAOYSA-N 0.000 description 1
- 125000003903 2-propenyl group Chemical group [H]C([*])([H])C([H])=C([H])[H] 0.000 description 1
- 125000001494 2-propynyl group Chemical group [H]C#CC([H])([H])* 0.000 description 1
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 1
- 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 1
- UNIJBMUBHBAUET-UHFFFAOYSA-N 3-(methylamino)propanenitrile Chemical compound CNCCC#N UNIJBMUBHBAUET-UHFFFAOYSA-N 0.000 description 1
- HAEQAUJYNHQVHV-UHFFFAOYSA-N 3-[4-(aminomethyl)-6-(trifluoromethyl)pyridin-2-yl]oxy-N-phenylbenzamide Chemical compound NCC1=CC(=NC(=C1)C(F)(F)F)OC=1C=C(C(=O)NC2=CC=CC=C2)C=CC=1 HAEQAUJYNHQVHV-UHFFFAOYSA-N 0.000 description 1
- HGWUUOXXAIISDB-UHFFFAOYSA-N 3-azabicyclo[3.1.0]hexane Chemical compound C1NCC2CC21 HGWUUOXXAIISDB-UHFFFAOYSA-N 0.000 description 1
- 125000004919 3-methyl-2-pentyl group Chemical group CC(C(C)*)CC 0.000 description 1
- 125000004921 3-methyl-3-pentyl group Chemical group CC(CC)(CC)* 0.000 description 1
- 125000001397 3-pyrrolyl group Chemical group [H]N1C([H])=C([*])C([H])=C1[H] 0.000 description 1
- 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 1
- DODQJNMQWMSYGS-QPLCGJKRSA-N 4-[(z)-1-[4-[2-(dimethylamino)ethoxy]phenyl]-1-phenylbut-1-en-2-yl]phenol Chemical compound C=1C=C(O)C=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 DODQJNMQWMSYGS-QPLCGJKRSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- ISXNTFNVBYPXMO-UHFFFAOYSA-N 4-chloro-2-fluorobenzamide Chemical compound NC(=O)C1=CC=C(Cl)C=C1F ISXNTFNVBYPXMO-UHFFFAOYSA-N 0.000 description 1
- HVCNXQOWACZAFN-UHFFFAOYSA-N 4-ethylmorpholine Chemical compound CCN1CCOCC1 HVCNXQOWACZAFN-UHFFFAOYSA-N 0.000 description 1
- HLOFWGGVFLUZMZ-UHFFFAOYSA-N 4-hydroxy-4-(6-methoxynaphthalen-2-yl)butan-2-one Chemical compound C1=C(C(O)CC(C)=O)C=CC2=CC(OC)=CC=C21 HLOFWGGVFLUZMZ-UHFFFAOYSA-N 0.000 description 1
- 125000004920 4-methyl-2-pentyl group Chemical group CC(CC(C)*)C 0.000 description 1
- 125000002471 4H-quinolizinyl group Chemical group C=1(C=CCN2C=CC=CC12)* 0.000 description 1
- 108010034457 5'-methylthioadenosine phosphorylase Proteins 0.000 description 1
- 102100022464 5'-nucleotidase Human genes 0.000 description 1
- NMUSYJAQQFHJEW-UHFFFAOYSA-N 5-Azacytidine Natural products O=C1N=C(N)N=CN1C1C(O)C(O)C(CO)O1 NMUSYJAQQFHJEW-UHFFFAOYSA-N 0.000 description 1
- FHIDNBAQOFJWCA-UAKXSSHOSA-N 5-fluorouridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 FHIDNBAQOFJWCA-UAKXSSHOSA-N 0.000 description 1
- 125000006043 5-hexenyl group Chemical group 0.000 description 1
- IBSREHMXUMOFBB-JFUDTMANSA-N 5u8924t11h Chemical compound O1[C@@H](C)[C@H](O)[C@@H](OC)C[C@@H]1O[C@@H]1[C@@H](OC)C[C@H](O[C@@H]2C(=C/C[C@@H]3C[C@@H](C[C@@]4(O3)C=C[C@H](C)[C@@H](C(C)C)O4)OC(=O)[C@@H]3C=C(C)[C@@H](O)[C@H]4OC\C([C@@]34O)=C/C=C/[C@@H]2C)/C)O[C@H]1C.C1=C[C@H](C)[C@@H]([C@@H](C)CC)O[C@]11O[C@H](C\C=C(C)\[C@@H](O[C@@H]2O[C@@H](C)[C@H](O[C@@H]3O[C@@H](C)[C@H](O)[C@@H](OC)C3)[C@@H](OC)C2)[C@@H](C)\C=C\C=C/2[C@]3([C@H](C(=O)O4)C=C(C)[C@@H](O)[C@H]3OC\2)O)C[C@H]4C1 IBSREHMXUMOFBB-JFUDTMANSA-N 0.000 description 1
- WYXSYVWAUAUWLD-SHUUEZRQSA-N 6-azauridine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C=N1 WYXSYVWAUAUWLD-SHUUEZRQSA-N 0.000 description 1
- YCWQAMGASJSUIP-YFKPBYRVSA-N 6-diazo-5-oxo-L-norleucine Chemical compound OC(=O)[C@@H](N)CCC(=O)C=[N+]=[N-] YCWQAMGASJSUIP-YFKPBYRVSA-N 0.000 description 1
- 229960005538 6-diazo-5-oxo-L-norleucine Drugs 0.000 description 1
- 102100023990 60S ribosomal protein L17 Human genes 0.000 description 1
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical compound [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 1
- 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 1
- DGGKXQQCVPAUEA-UHFFFAOYSA-N 8-azabicyclo[3.2.1]octane Chemical compound C1CCC2CCC1N2 DGGKXQQCVPAUEA-UHFFFAOYSA-N 0.000 description 1
- ZGXJTSGNIOSYLO-UHFFFAOYSA-N 88755TAZ87 Chemical compound NCC(=O)CCC(O)=O ZGXJTSGNIOSYLO-UHFFFAOYSA-N 0.000 description 1
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 1
- 230000035502 ADME Effects 0.000 description 1
- 244000215068 Acacia senegal Species 0.000 description 1
- 235000006491 Acacia senegal Nutrition 0.000 description 1
- DLFVBJFMPXGRIB-UHFFFAOYSA-N Acetamide Chemical compound CC(N)=O DLFVBJFMPXGRIB-UHFFFAOYSA-N 0.000 description 1
- OIRDTQYFTABQOQ-KQYNXXCUSA-N Adenosine Natural products C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 1
- 206010067484 Adverse reaction Diseases 0.000 description 1
- 229930195730 Aflatoxin Natural products 0.000 description 1
- XWIYFDMXXLINPU-UHFFFAOYSA-N Aflatoxin G Chemical compound O=C1OCCC2=C1C(=O)OC1=C2C(OC)=CC2=C1C1C=COC1O2 XWIYFDMXXLINPU-UHFFFAOYSA-N 0.000 description 1
- 235000001674 Agaricus brunnescens Nutrition 0.000 description 1
- 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 1
- CEIZFXOZIQNICU-UHFFFAOYSA-N Alternaria alternata Crofton-weed toxin Natural products CCC(C)C1NC(=O)C(C(C)=O)=C1O CEIZFXOZIQNICU-UHFFFAOYSA-N 0.000 description 1
- 239000005995 Aluminium silicate Substances 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 102000004452 Arginase Human genes 0.000 description 1
- 108700024123 Arginases Proteins 0.000 description 1
- 102100029361 Aromatase Human genes 0.000 description 1
- 108010078554 Aromatase Proteins 0.000 description 1
- NOWKCMXCCJGMRR-UHFFFAOYSA-N Aziridine Chemical compound C1CN1 NOWKCMXCCJGMRR-UHFFFAOYSA-N 0.000 description 1
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 1
- 108010074708 B7-H1 Antigen Proteins 0.000 description 1
- 102000008096 B7-H1 Antigen Human genes 0.000 description 1
- 238000011725 BALB/c mouse Methods 0.000 description 1
- 239000012664 BCL-2-inhibitor Substances 0.000 description 1
- 229940123711 Bcl2 inhibitor Drugs 0.000 description 1
- VGGGPCQERPFHOB-MCIONIFRSA-N Bestatin Chemical compound CC(C)C[C@H](C(O)=O)NC(=O)[C@@H](O)[C@H](N)CC1=CC=CC=C1 VGGGPCQERPFHOB-MCIONIFRSA-N 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- WKBOTKDWSSQWDR-UHFFFAOYSA-N Bromine atom Chemical compound [Br] WKBOTKDWSSQWDR-UHFFFAOYSA-N 0.000 description 1
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Natural products CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 description 1
- 102100038078 CD276 antigen Human genes 0.000 description 1
- 101710185679 CD276 antigen Proteins 0.000 description 1
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 101100434927 Caenorhabditis elegans prmt-5 gene Proteins 0.000 description 1
- 101100314454 Caenorhabditis elegans tra-1 gene Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 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 1
- 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 1
- 241000283707 Capra Species 0.000 description 1
- OKTJSMMVPCPJKN-NJFSPNSNSA-N Carbon-14 Chemical compound [14C] OKTJSMMVPCPJKN-NJFSPNSNSA-N 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- AOCCBINRVIKJHY-UHFFFAOYSA-N Carmofur Chemical compound CCCCCCNC(=O)N1C=C(F)C(=O)NC1=O AOCCBINRVIKJHY-UHFFFAOYSA-N 0.000 description 1
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 1
- 108091007741 Chimeric antigen receptor T cells Proteins 0.000 description 1
- JWBOIMRXGHLCPP-UHFFFAOYSA-N Chloditan Chemical compound C=1C=CC=C(Cl)C=1C(C(Cl)Cl)C1=CC=C(Cl)C=C1 JWBOIMRXGHLCPP-UHFFFAOYSA-N 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 229910021595 Copper(I) iodide Inorganic materials 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 108010009392 Cyclin-Dependent Kinase Inhibitor p16 Proteins 0.000 description 1
- 102000009508 Cyclin-Dependent Kinase Inhibitor p16 Human genes 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- WEAHRLBPCANXCN-UHFFFAOYSA-N Daunomycin Natural products CCC1(O)CC(OC2CC(N)C(O)C(C)O2)c3cc4C(=O)c5c(OC)cccc5C(=O)c4c(O)c3C1 WEAHRLBPCANXCN-UHFFFAOYSA-N 0.000 description 1
- YZCKVEUIGOORGS-OUBTZVSYSA-N Deuterium Chemical compound [2H] YZCKVEUIGOORGS-OUBTZVSYSA-N 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
- 102100029722 Ectonucleoside triphosphate diphosphohydrolase 1 Human genes 0.000 description 1
- 102000002322 Egg Proteins Human genes 0.000 description 1
- 108010000912 Egg Proteins Proteins 0.000 description 1
- SAMRUMKYXPVKPA-VFKOLLTISA-N Enocitabine Chemical compound O=C1N=C(NC(=O)CCCCCCCCCCCCCCCCCCCCC)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 SAMRUMKYXPVKPA-VFKOLLTISA-N 0.000 description 1
- 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 1
- 241000283086 Equidae Species 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- CWYNVVGOOAEACU-UHFFFAOYSA-N Fe2+ Chemical compound [Fe+2] CWYNVVGOOAEACU-UHFFFAOYSA-N 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- BLCLNMBMMGCOAS-URPVMXJPSA-N Goserelin Chemical compound C([C@@H](C(=O)N[C@H](COC(C)(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N1[C@@H](CCC1)C(=O)NNC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 BLCLNMBMMGCOAS-URPVMXJPSA-N 0.000 description 1
- 108010069236 Goserelin Proteins 0.000 description 1
- 239000007818 Grignard reagent Substances 0.000 description 1
- 229920000084 Gum arabic Polymers 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 1
- 101000678236 Homo sapiens 5'-nucleotidase Proteins 0.000 description 1
- 101000864344 Homo sapiens B- and T-lymphocyte attenuator Proteins 0.000 description 1
- 101001012447 Homo sapiens Ectonucleoside triphosphate diphosphohydrolase 1 Proteins 0.000 description 1
- 101001068133 Homo sapiens Hepatitis A virus cellular receptor 2 Proteins 0.000 description 1
- 101001037256 Homo sapiens Indoleamine 2,3-dioxygenase 1 Proteins 0.000 description 1
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 description 1
- 101000596234 Homo sapiens T-cell surface protein tactile Proteins 0.000 description 1
- 101000892398 Homo sapiens Tryptophan 2,3-dioxygenase Proteins 0.000 description 1
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 1
- VSNHCAURESNICA-UHFFFAOYSA-N Hydroxyurea Chemical compound NC(=O)NO VSNHCAURESNICA-UHFFFAOYSA-N 0.000 description 1
- 238000004566 IR spectroscopy Methods 0.000 description 1
- MPBVHIBUJCELCL-UHFFFAOYSA-N Ibandronate Chemical compound CCCCCN(C)CCC(O)(P(O)(O)=O)P(O)(O)=O MPBVHIBUJCELCL-UHFFFAOYSA-N 0.000 description 1
- 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 1
- 102000037978 Immune checkpoint receptors Human genes 0.000 description 1
- 108091008028 Immune checkpoint receptors Proteins 0.000 description 1
- 102100040061 Indoleamine 2,3-dioxygenase 1 Human genes 0.000 description 1
- 238000012695 Interfacial polymerization Methods 0.000 description 1
- LPHGQDQBBGAPDZ-UHFFFAOYSA-N Isocaffeine Natural products CN1C(=O)N(C)C(=O)C2=C1N(C)C=N2 LPHGQDQBBGAPDZ-UHFFFAOYSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 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
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 1
- 102000017578 LAG3 Human genes 0.000 description 1
- 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 1
- 108010000817 Leuprolide Proteins 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 101150022373 MAT2A gene Proteins 0.000 description 1
- 239000007993 MOPS buffer Substances 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- VFKZTMPDYBFSTM-KVTDHHQDSA-N Mitobronitol Chemical compound BrC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CBr VFKZTMPDYBFSTM-KVTDHHQDSA-N 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 101600097262 Monodelphis domestica Cyclin-dependent kinase inhibitor 2A (isoform 1) Proteins 0.000 description 1
- 150000007945 N-acyl ureas Chemical class 0.000 description 1
- UEEJHVSXFDXPFK-UHFFFAOYSA-N N-dimethylaminoethanol Chemical compound CN(C)CCO UEEJHVSXFDXPFK-UHFFFAOYSA-N 0.000 description 1
- HTLZVHNRZJPSMI-UHFFFAOYSA-N N-ethylpiperidine Chemical compound CCN1CCCCC1 HTLZVHNRZJPSMI-UHFFFAOYSA-N 0.000 description 1
- MBBZMMPHUWSWHV-BDVNFPICSA-N N-methylglucamine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 1
- 150000001204 N-oxides Chemical class 0.000 description 1
- OPFJDXRVMFKJJO-ZHHKINOHSA-N N-{[3-(2-benzamido-4-methyl-1,3-thiazol-5-yl)-pyrazol-5-yl]carbonyl}-G-dR-G-dD-dD-dD-NH2 Chemical compound S1C(C=2NN=C(C=2)C(=O)NCC(=O)N[C@H](CCCN=C(N)N)C(=O)NCC(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CC(O)=O)C(=O)N[C@H](CC(O)=O)C(N)=O)=C(C)N=C1NC(=O)C1=CC=CC=C1 OPFJDXRVMFKJJO-ZHHKINOHSA-N 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- UFWIBTONFRDIAS-UHFFFAOYSA-N Naphthalene Chemical compound C1=CC=CC2=CC=CC=C21 UFWIBTONFRDIAS-UHFFFAOYSA-N 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 1
- 229930187135 Olivomycin Natural products 0.000 description 1
- 229930012538 Paclitaxel Natural products 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 102000017343 Phosphatidylinositol kinases Human genes 0.000 description 1
- 108050005377 Phosphatidylinositol kinases Proteins 0.000 description 1
- 102000009097 Phosphorylases Human genes 0.000 description 1
- 108010073135 Phosphorylases Proteins 0.000 description 1
- KMSKQZKKOZQFFG-HSUXVGOQSA-N Pirarubicin Chemical compound O([C@H]1[C@@H](N)C[C@@H](O[C@H]1C)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]1CCCCO1 KMSKQZKKOZQFFG-HSUXVGOQSA-N 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 229920002732 Polyanhydride Polymers 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 229920002565 Polyethylene Glycol 400 Polymers 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920000954 Polyglycolide Polymers 0.000 description 1
- 229920002685 Polyoxyl 35CastorOil Polymers 0.000 description 1
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 1
- XBDQKXXYIPTUBI-UHFFFAOYSA-M Propionate Chemical compound CCC([O-])=O XBDQKXXYIPTUBI-UHFFFAOYSA-M 0.000 description 1
- 102000007327 Protamines Human genes 0.000 description 1
- 108010007568 Protamines Proteins 0.000 description 1
- 102000003708 Protein arginine N-methyltransferase Human genes 0.000 description 1
- 108020000912 Protein arginine N-methyltransferase Proteins 0.000 description 1
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 1
- 101710100968 Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 1
- 102100026115 S-adenosylmethionine synthase isoform type-1 Human genes 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 101100215487 Sus scrofa ADRA2A gene Proteins 0.000 description 1
- 102100035268 T-cell surface protein tactile Human genes 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 229940123237 Taxane Drugs 0.000 description 1
- CGMTUJFWROPELF-UHFFFAOYSA-N Tenuazonic acid Natural products CCC(C)C1NC(=O)C(=C(C)/O)C1=O CGMTUJFWROPELF-UHFFFAOYSA-N 0.000 description 1
- GUGOEEXESWIERI-UHFFFAOYSA-N Terfenadine Chemical compound C1=CC(C(C)(C)C)=CC=C1C(O)CCCN1CCC(C(O)(C=2C=CC=CC=2)C=2C=CC=CC=2)CC1 GUGOEEXESWIERI-UHFFFAOYSA-N 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- YZCKVEUIGOORGS-NJFSPNSNSA-N Tritium Chemical compound [3H] YZCKVEUIGOORGS-NJFSPNSNSA-N 0.000 description 1
- 102100040653 Tryptophan 2,3-dioxygenase Human genes 0.000 description 1
- 241000609666 Tuber aestivum Species 0.000 description 1
- 102000044209 Tumor Suppressor Genes Human genes 0.000 description 1
- 108700025716 Tumor Suppressor Genes Proteins 0.000 description 1
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 1
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 1
- 108010079206 V-Set Domain-Containing T-Cell Activation Inhibitor 1 Proteins 0.000 description 1
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 1
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 1
- 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 1
- 238000002441 X-ray diffraction Methods 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 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 1
- IFJUINDAXYAPTO-UUBSBJJBSA-N [(8r,9s,13s,14s,17s)-17-[2-[4-[4-[bis(2-chloroethyl)amino]phenyl]butanoyloxy]acetyl]oxy-13-methyl-6,7,8,9,11,12,14,15,16,17-decahydrocyclopenta[a]phenanthren-3-yl] benzoate Chemical compound C([C@@H]1[C@@H](C2=CC=3)CC[C@]4([C@H]1CC[C@@H]4OC(=O)COC(=O)CCCC=1C=CC(=CC=1)N(CCCl)CCCl)C)CC2=CC=3OC(=O)C1=CC=CC=C1 IFJUINDAXYAPTO-UUBSBJJBSA-N 0.000 description 1
- IHGLINDYFMDHJG-UHFFFAOYSA-N [2-(4-methoxyphenyl)-3,4-dihydronaphthalen-1-yl]-[4-(2-pyrrolidin-1-ylethoxy)phenyl]methanone Chemical compound C1=CC(OC)=CC=C1C(CCC1=CC=CC=C11)=C1C(=O)C(C=C1)=CC=C1OCCN1CCCC1 IHGLINDYFMDHJG-UHFFFAOYSA-N 0.000 description 1
- REAYFGLASQTHKB-UHFFFAOYSA-N [2-[3-(1H-pyrazol-4-yl)phenoxy]-6-(trifluoromethyl)pyridin-4-yl]methanamine Chemical compound N1N=CC(=C1)C=1C=C(OC2=NC(=CC(=C2)CN)C(F)(F)F)C=CC=1 REAYFGLASQTHKB-UHFFFAOYSA-N 0.000 description 1
- USDJGQLNFPZEON-UHFFFAOYSA-N [[4,6-bis(hydroxymethylamino)-1,3,5-triazin-2-yl]amino]methanol Chemical compound OCNC1=NC(NCO)=NC(NCO)=N1 USDJGQLNFPZEON-UHFFFAOYSA-N 0.000 description 1
- 229950008167 abamectin Drugs 0.000 description 1
- 229960004103 abiraterone acetate Drugs 0.000 description 1
- UVIQSJCZCSLXRZ-UBUQANBQSA-N abiraterone acetate Chemical compound C([C@@H]1[C@]2(C)CC[C@@H]3[C@@]4(C)CC[C@@H](CC4=CC[C@H]31)OC(=O)C)C=C2C1=CC=CN=C1 UVIQSJCZCSLXRZ-UBUQANBQSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 229940028652 abraxane Drugs 0.000 description 1
- 235000010489 acacia gum Nutrition 0.000 description 1
- 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 1
- 125000000641 acridinyl group Chemical group C1(=CC=CC2=NC3=CC=CC=C3C=C12)* 0.000 description 1
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Chemical compound 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 1
- 230000009471 action Effects 0.000 description 1
- 125000005073 adamantyl group Chemical group C12(CC3CC(CC(C1)C3)C2)* 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 229960001570 ademetionine Drugs 0.000 description 1
- 239000000674 adrenergic antagonist Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 230000006838 adverse reaction Effects 0.000 description 1
- 239000005409 aflatoxin Substances 0.000 description 1
- 125000003158 alcohol group Chemical group 0.000 description 1
- IAJILQKETJEXLJ-RSJOWCBRSA-N aldehydo-D-galacturonic acid Chemical compound O=C[C@H](O)[C@@H](O)[C@@H](O)[C@H](O)C(O)=O IAJILQKETJEXLJ-RSJOWCBRSA-N 0.000 description 1
- IAJILQKETJEXLJ-QTBDOELSSA-N aldehydo-D-glucuronic acid Chemical compound O=C[C@H](O)[C@@H](O)[C@H](O)[C@H](O)C(O)=O IAJILQKETJEXLJ-QTBDOELSSA-N 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- 125000002723 alicyclic group Chemical group 0.000 description 1
- 150000004703 alkoxides Chemical class 0.000 description 1
- 229940045714 alkyl sulfonate alkylating agent Drugs 0.000 description 1
- 150000008052 alkyl sulfonates Chemical class 0.000 description 1
- 229940100198 alkylating agent Drugs 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- SHGAZHPCJJPHSC-YCNIQYBTSA-N all-trans-retinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-YCNIQYBTSA-N 0.000 description 1
- HSFWRNGVRCDJHI-UHFFFAOYSA-N alpha-acetylene Natural products C#C HSFWRNGVRCDJHI-UHFFFAOYSA-N 0.000 description 1
- 229960000473 altretamine Drugs 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 235000012211 aluminium silicate Nutrition 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- ROBVIMPUHSLWNV-UHFFFAOYSA-N aminoglutethimide Chemical compound C=1C=C(N)C=CC=1C1(CC)CCC(=O)NC1=O ROBVIMPUHSLWNV-UHFFFAOYSA-N 0.000 description 1
- 229960003437 aminoglutethimide Drugs 0.000 description 1
- 229960002749 aminolevulinic acid Drugs 0.000 description 1
- 229960003896 aminopterin Drugs 0.000 description 1
- XKMRRTOUMJRJIA-UHFFFAOYSA-N ammonia nh3 Chemical compound N.N XKMRRTOUMJRJIA-UHFFFAOYSA-N 0.000 description 1
- 229960001220 amsacrine Drugs 0.000 description 1
- XCPGHVQEEXUHNC-UHFFFAOYSA-N amsacrine Chemical compound COC1=CC(NS(C)(=O)=O)=CC=C1NC1=C(C=CC=C2)C2=NC2=CC=CC=C12 XCPGHVQEEXUHNC-UHFFFAOYSA-N 0.000 description 1
- 239000003098 androgen Substances 0.000 description 1
- 229940030486 androgens Drugs 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 125000002178 anthracenyl group Chemical group C1(=CC=CC2=CC3=CC=CC=C3C=C12)* 0.000 description 1
- 230000002280 anti-androgenic effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 229940046836 anti-estrogen Drugs 0.000 description 1
- 230000001833 anti-estrogenic effect Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 230000001028 anti-proliverative effect Effects 0.000 description 1
- 239000000051 antiandrogen Substances 0.000 description 1
- 229940030495 antiandrogen sex hormone and modulator of the genital system Drugs 0.000 description 1
- 210000000612 antigen-presenting cell Anatomy 0.000 description 1
- 239000013059 antihormonal agent Substances 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 229940045687 antimetabolites folic acid analogs Drugs 0.000 description 1
- 229940045719 antineoplastic alkylating agent nitrosoureas Drugs 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- HJBWBFZLDZWPHF-UHFFFAOYSA-N apalutamide Chemical compound C1=C(F)C(C(=O)NC)=CC=C1N1C2(CCC2)C(=O)N(C=2C=C(C(C#N)=NC=2)C(F)(F)F)C1=S HJBWBFZLDZWPHF-UHFFFAOYSA-N 0.000 description 1
- 229950007511 apalutamide Drugs 0.000 description 1
- 229960003121 arginine Drugs 0.000 description 1
- 150000001483 arginine derivatives Chemical class 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 229940090047 auto-injector Drugs 0.000 description 1
- 230000005784 autoimmunity Effects 0.000 description 1
- 229940120638 avastin Drugs 0.000 description 1
- RRZXIRBKKLTSOM-XPNPUAGNSA-N avermectin B1a Chemical compound C1=C[C@H](C)[C@@H]([C@@H](C)CC)O[C@]11O[C@H](C\C=C(C)\[C@@H](O[C@@H]2O[C@@H](C)[C@H](O[C@@H]3O[C@@H](C)[C@H](O)[C@@H](OC)C3)[C@@H](OC)C2)[C@@H](C)\C=C\C=C/2[C@]3([C@H](C(=O)O4)C=C(C)[C@@H](O)[C@H]3OC\2)O)C[C@H]4C1 RRZXIRBKKLTSOM-XPNPUAGNSA-N 0.000 description 1
- 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 1
- 150000001541 aziridines Chemical class 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 235000013871 bee wax Nutrition 0.000 description 1
- 239000012166 beeswax Substances 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid group Chemical group C(C1=CC=CC=C1)(=O)O WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- MSWZFWKMSRAUBD-UHFFFAOYSA-N beta-D-galactosamine Natural products NC1C(O)OC(CO)C(O)C1O MSWZFWKMSRAUBD-UHFFFAOYSA-N 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 229960003237 betaine Drugs 0.000 description 1
- 229960000397 bevacizumab Drugs 0.000 description 1
- 229960000997 bicalutamide Drugs 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 239000004305 biphenyl Substances 0.000 description 1
- 235000010290 biphenyl Nutrition 0.000 description 1
- YNHIGQDRGKUECZ-UHFFFAOYSA-L bis(triphenylphosphine)palladium(ii) dichloride Chemical compound [Cl-].[Cl-].[Pd+2].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 YNHIGQDRGKUECZ-UHFFFAOYSA-L 0.000 description 1
- 229960001561 bleomycin Drugs 0.000 description 1
- 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 1
- 230000037396 body weight Effects 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- GDTBXPJZTBHREO-UHFFFAOYSA-N bromine Substances BrBr GDTBXPJZTBHREO-UHFFFAOYSA-N 0.000 description 1
- 229910052794 bromium Inorganic materials 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- DQXBYHZEEUGOBF-UHFFFAOYSA-N but-3-enoic acid;ethene Chemical compound C=C.OC(=O)CC=C DQXBYHZEEUGOBF-UHFFFAOYSA-N 0.000 description 1
- 235000019437 butane-1,3-diol Nutrition 0.000 description 1
- KDYFGRWQOYBRFD-NUQCWPJISA-N butanedioic acid Chemical compound O[14C](=O)CC[14C](O)=O KDYFGRWQOYBRFD-NUQCWPJISA-N 0.000 description 1
- 125000004369 butenyl group Chemical group C(=CCC)* 0.000 description 1
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- VJEONQKOZGKCAK-UHFFFAOYSA-N caffeine Natural products CN1C(=O)N(C)C(=O)C2=C1C=CN2C VJEONQKOZGKCAK-UHFFFAOYSA-N 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 235000010216 calcium carbonate Nutrition 0.000 description 1
- HXCHCVDVKSCDHU-LULTVBGHSA-N calicheamicin Chemical compound C1[C@H](OC)[C@@H](NCC)CO[C@H]1O[C@H]1[C@H](O[C@@H]2C\3=C(NC(=O)OC)C(=O)C[C@](C/3=C/CSSSC)(O)C#C\C=C/C#C2)O[C@H](C)[C@@H](NO[C@@H]2O[C@H](C)[C@@H](SC(=O)C=3C(=C(OC)C(O[C@H]4[C@@H]([C@H](OC)[C@@H](O)[C@H](C)O4)O)=C(I)C=3C)OC)[C@@H](O)C2)[C@@H]1O HXCHCVDVKSCDHU-LULTVBGHSA-N 0.000 description 1
- 229930195731 calicheamicin Natural products 0.000 description 1
- 229960004117 capecitabine Drugs 0.000 description 1
- 150000004657 carbamic acid derivatives Chemical class 0.000 description 1
- 150000001721 carbon Chemical group 0.000 description 1
- 150000004649 carbonic acid derivatives Chemical class 0.000 description 1
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- XREUEWVEMYWFFA-CSKJXFQVSA-N carminomycin 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=C(O)C=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XREUEWVEMYWFFA-CSKJXFQVSA-N 0.000 description 1
- 229930188550 carminomycin Natural products 0.000 description 1
- XREUEWVEMYWFFA-UHFFFAOYSA-N carminomycin I Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=C(O)C=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XREUEWVEMYWFFA-UHFFFAOYSA-N 0.000 description 1
- 229960003261 carmofur Drugs 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 229950001725 carubicin Drugs 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 238000001516 cell proliferation assay Methods 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 238000012054 celltiter-glo Methods 0.000 description 1
- 229960000541 cetyl alcohol Drugs 0.000 description 1
- 229910052729 chemical element Inorganic materials 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical compound C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- ZYVSOIYQKUDENJ-WKSBCEQHSA-N chromomycin A3 Chemical compound O([C@@H]1C[C@@H](O[C@H](C)[C@@H]1OC(C)=O)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](OC(C)=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](OC)[C@@H](C)O1 ZYVSOIYQKUDENJ-WKSBCEQHSA-N 0.000 description 1
- 229960004912 cilastatin Drugs 0.000 description 1
- DHSUYTOATWAVLW-WFVMDLQDSA-N cilastatin Chemical compound CC1(C)C[C@@H]1C(=O)N\C(=C/CCCCSC[C@H](N)C(O)=O)C(O)=O DHSUYTOATWAVLW-WFVMDLQDSA-N 0.000 description 1
- 125000000259 cinnolinyl group Chemical group N1=NC(=CC2=CC=CC=C12)* 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000005354 coacervation Methods 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 239000003240 coconut oil Substances 0.000 description 1
- 235000019864 coconut oil Nutrition 0.000 description 1
- 229960001338 colchicine Drugs 0.000 description 1
- 238000001246 colloidal dispersion Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 229940126543 compound 14 Drugs 0.000 description 1
- 229940126142 compound 16 Drugs 0.000 description 1
- 229940125782 compound 2 Drugs 0.000 description 1
- 229940125810 compound 20 Drugs 0.000 description 1
- 229940126086 compound 21 Drugs 0.000 description 1
- 229940126214 compound 3 Drugs 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 229920001577 copolymer Polymers 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- LSXDOTMGLUJQCM-UHFFFAOYSA-M copper(i) iodide Chemical compound I[Cu] LSXDOTMGLUJQCM-UHFFFAOYSA-M 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- 125000001995 cyclobutyl group Chemical group [H]C1([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000000582 cycloheptyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000003678 cyclohexadienyl group Chemical group C1(=CC=CCC1)* 0.000 description 1
- 125000000113 cyclohexyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C1([H])[H] 0.000 description 1
- 125000006547 cyclononyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 125000000640 cyclooctyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 125000001511 cyclopentyl group Chemical group [H]C1([H])C([H])([H])C([H])([H])C([H])(*)C1([H])[H] 0.000 description 1
- 125000001559 cyclopropyl group Chemical group [H]C1([H])C([H])([H])C1([H])* 0.000 description 1
- IGSKHXTUVXSOMB-UHFFFAOYSA-N cyclopropylmethanamine Chemical compound NCC1CC1 IGSKHXTUVXSOMB-UHFFFAOYSA-N 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 229940104302 cytosine Drugs 0.000 description 1
- 229960003901 dacarbazine Drugs 0.000 description 1
- 229960000975 daunorubicin Drugs 0.000 description 1
- 230000006240 deamidation Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 125000002704 decyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 238000007257 deesterification reaction Methods 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 239000008380 degradant Substances 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 230000002074 deregulated effect Effects 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 239000003599 detergent Substances 0.000 description 1
- 229910052805 deuterium Inorganic materials 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 150000004985 diamines Chemical class 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 125000001028 difluoromethyl group Chemical group [H]C(F)(F)* 0.000 description 1
- 125000005045 dihydroisoquinolinyl group Chemical group C1(NC=CC2=CC=CC=C12)* 0.000 description 1
- 125000005044 dihydroquinolinyl group Chemical group N1(CC=CC2=CC=CC=C12)* 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 239000003534 dna topoisomerase inhibitor Substances 0.000 description 1
- 229940115080 doxil Drugs 0.000 description 1
- 239000008298 dragée Substances 0.000 description 1
- 238000001035 drying Methods 0.000 description 1
- 229960002759 eflornithine Drugs 0.000 description 1
- 210000003278 egg shell Anatomy 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 229950011487 enocitabine Drugs 0.000 description 1
- WXCXUHSOUPDCQV-UHFFFAOYSA-N enzalutamide Chemical compound C1=C(F)C(C(=O)NC)=CC=C1N1C(C)(C)C(=O)N(C=2C=C(C(C#N)=CC=2)C(F)(F)F)C1=S WXCXUHSOUPDCQV-UHFFFAOYSA-N 0.000 description 1
- 229960004671 enzalutamide Drugs 0.000 description 1
- 230000008995 epigenetic change Effects 0.000 description 1
- 229960001904 epirubicin Drugs 0.000 description 1
- 229960003649 eribulin Drugs 0.000 description 1
- 125000004185 ester group Chemical group 0.000 description 1
- 230000032050 esterification Effects 0.000 description 1
- 238000005886 esterification reaction Methods 0.000 description 1
- FRPJXPJMRWBBIH-RBRWEJTLSA-N estramustine Chemical compound ClCCN(CCCl)C(=O)OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 FRPJXPJMRWBBIH-RBRWEJTLSA-N 0.000 description 1
- 229960001842 estramustine Drugs 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- 239000005038 ethylene vinyl acetate Substances 0.000 description 1
- 125000005469 ethylenyl group Chemical group 0.000 description 1
- 125000002534 ethynyl group Chemical group [H]C#C* 0.000 description 1
- 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 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 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 1
- 229960000390 fludarabine Drugs 0.000 description 1
- 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 1
- 125000003983 fluorenyl group Chemical group C1(=CC=CC=2C3=CC=CC=C3CC12)* 0.000 description 1
- 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 1
- 229960002074 flutamide Drugs 0.000 description 1
- MKXKFYHWDHIYRV-UHFFFAOYSA-N flutamide Chemical compound CC(C)C(=O)NC1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 MKXKFYHWDHIYRV-UHFFFAOYSA-N 0.000 description 1
- 150000002224 folic acids Chemical class 0.000 description 1
- 229960004783 fotemustine Drugs 0.000 description 1
- YAKWPXVTIGTRJH-UHFFFAOYSA-N fotemustine Chemical compound CCOP(=O)(OCC)C(C)NC(=O)N(CCCl)N=O YAKWPXVTIGTRJH-UHFFFAOYSA-N 0.000 description 1
- 229940044658 gallium nitrate Drugs 0.000 description 1
- 229940083124 ganglion-blocking antiadrenergic secondary and tertiary amines Drugs 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 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 1
- 229910052732 germanium Inorganic materials 0.000 description 1
- GNPVGFCGXDBREM-UHFFFAOYSA-N germanium atom Chemical compound [Ge] GNPVGFCGXDBREM-UHFFFAOYSA-N 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 229940080856 gleevec Drugs 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 229960002442 glucosamine Drugs 0.000 description 1
- 229940097043 glucuronic acid Drugs 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 229940074045 glyceryl distearate Drugs 0.000 description 1
- 229940075529 glyceryl stearate Drugs 0.000 description 1
- 229930182470 glycoside Natural products 0.000 description 1
- 229960002913 goserelin Drugs 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- JAXFJECJQZDFJS-XHEPKHHKSA-N gtpl8555 Chemical compound OC(=O)C[C@H](N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N1CCC[C@@H]1C(=O)N[C@H](B1O[C@@]2(C)[C@H]3C[C@H](C3(C)C)C[C@H]2O1)CCC1=CC=C(F)C=C1 JAXFJECJQZDFJS-XHEPKHHKSA-N 0.000 description 1
- 229940118951 halaven Drugs 0.000 description 1
- 239000007902 hard capsule Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 125000003187 heptyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 229940022353 herceptin Drugs 0.000 description 1
- 239000004312 hexamethylene tetramine Substances 0.000 description 1
- 235000010299 hexamethylene tetramine Nutrition 0.000 description 1
- UUVWYPNAQBNQJQ-UHFFFAOYSA-N hexamethylmelamine Chemical compound CN(C)C1=NC(N(C)C)=NC(N(C)C)=N1 UUVWYPNAQBNQJQ-UHFFFAOYSA-N 0.000 description 1
- 125000004051 hexyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- XMBWDFGMSWQBCA-UHFFFAOYSA-N hydrogen iodide Chemical compound I XMBWDFGMSWQBCA-UHFFFAOYSA-N 0.000 description 1
- 229940071870 hydroiodic acid Drugs 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 229960001330 hydroxycarbamide Drugs 0.000 description 1
- 229920003063 hydroxymethyl cellulose Polymers 0.000 description 1
- 229940031574 hydroxymethyl cellulose Drugs 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- 229940015872 ibandronate Drugs 0.000 description 1
- YLMAHDNUQAMNNX-UHFFFAOYSA-N imatinib methanesulfonate Chemical compound CS(O)(=O)=O.C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 YLMAHDNUQAMNNX-UHFFFAOYSA-N 0.000 description 1
- 125000002962 imidazol-1-yl group Chemical group [*]N1C([H])=NC([H])=C1[H] 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 229940121354 immunomodulator Drugs 0.000 description 1
- 239000007943 implant Substances 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 125000003392 indanyl group Chemical group C1(CCC2=CC=CC=C12)* 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 238000002329 infrared spectrum Methods 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 229940102223 injectable solution Drugs 0.000 description 1
- 229940102213 injectable suspension Drugs 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- PNDPGZBMCMUPRI-UHFFFAOYSA-N iodine Chemical compound II PNDPGZBMCMUPRI-UHFFFAOYSA-N 0.000 description 1
- 239000011630 iodine Substances 0.000 description 1
- 229910052740 iodine Inorganic materials 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 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 1
- 229960004768 irinotecan Drugs 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 125000001977 isobenzofuranyl group Chemical group C=1(OC=C2C=CC=CC12)* 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- 238000006317 isomerization reaction Methods 0.000 description 1
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 1
- 125000001972 isopentyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000003253 isopropoxy group Chemical group [H]C([H])([H])C([H])(O*)C([H])([H])[H] 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- 229960002014 ixabepilone Drugs 0.000 description 1
- 229940111707 ixempra Drugs 0.000 description 1
- 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 1
- 239000004310 lactic acid Substances 0.000 description 1
- 235000014655 lactic acid Nutrition 0.000 description 1
- JJTUDXZGHPGLLC-UHFFFAOYSA-N lactide Chemical compound CC1OC(=O)C(C)OC1=O JJTUDXZGHPGLLC-UHFFFAOYSA-N 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 229960003881 letrozole Drugs 0.000 description 1
- 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 1
- 239000003446 ligand Substances 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 1
- 239000007791 liquid phase Substances 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 229960003538 lonidamine Drugs 0.000 description 1
- WDRYRZXSPDWGEB-UHFFFAOYSA-N lonidamine Chemical compound C12=CC=CC=C2C(C(=O)O)=NN1CC1=CC=C(Cl)C=C1Cl WDRYRZXSPDWGEB-UHFFFAOYSA-N 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 108010078259 luprolide acetate gel depot Proteins 0.000 description 1
- 239000008176 lyophilized powder Substances 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 231100000682 maximum tolerated dose Toxicity 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 229960004961 mechlorethamine Drugs 0.000 description 1
- QZIQJVCYUQZDIR-UHFFFAOYSA-N mechlorethamine hydrochloride Chemical compound Cl.ClCCN(C)CCCl QZIQJVCYUQZDIR-UHFFFAOYSA-N 0.000 description 1
- 229960002868 mechlorethamine hydrochloride Drugs 0.000 description 1
- 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 1
- 229960001924 melphalan Drugs 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 230000004066 metabolic change Effects 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 229960002216 methylparaben Drugs 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 239000004005 microsphere Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 229960005485 mitobronitol Drugs 0.000 description 1
- 229960003539 mitoguazone Drugs 0.000 description 1
- MXWHMTNPTTVWDM-NXOFHUPFSA-N mitoguazone Chemical compound NC(N)=N\N=C(/C)\C=N\N=C(N)N MXWHMTNPTTVWDM-NXOFHUPFSA-N 0.000 description 1
- VFKZTMPDYBFSTM-GUCUJZIJSA-N mitolactol Chemical compound BrC[C@H](O)[C@@H](O)[C@@H](O)[C@H](O)CBr VFKZTMPDYBFSTM-GUCUJZIJSA-N 0.000 description 1
- 229950010913 mitolactol Drugs 0.000 description 1
- 229960000350 mitotane Drugs 0.000 description 1
- INAXVFBXDYWQFN-XHSDSOJGSA-N morphinan Chemical group C1C2=CC=CC=C2[C@]23CCCC[C@H]3[C@@H]1NCC2 INAXVFBXDYWQFN-XHSDSOJGSA-N 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- ACTNHJDHMQSOGL-UHFFFAOYSA-N n',n'-dibenzylethane-1,2-diamine Chemical compound C=1C=CC=CC=1CN(CCN)CC1=CC=CC=C1 ACTNHJDHMQSOGL-UHFFFAOYSA-N 0.000 description 1
- HVOYZOQVDYHUPF-UHFFFAOYSA-N n,n',n'-trimethylethane-1,2-diamine Chemical compound CNCCN(C)C HVOYZOQVDYHUPF-UHFFFAOYSA-N 0.000 description 1
- KRKPYFLIYNGWTE-UHFFFAOYSA-N n,o-dimethylhydroxylamine Chemical compound CNOC KRKPYFLIYNGWTE-UHFFFAOYSA-N 0.000 description 1
- UFVHVURXVBHPDA-UHFFFAOYSA-N n-(dichloromethyl)-n-ethylethanamine Chemical compound CCN(CC)C(Cl)Cl UFVHVURXVBHPDA-UHFFFAOYSA-N 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 1
- AZXFWSPGAJVTRL-UHFFFAOYSA-N n-methylcyclobutanamine;hydrochloride Chemical compound Cl.CNC1CCC1 AZXFWSPGAJVTRL-UHFFFAOYSA-N 0.000 description 1
- VEBLEROFGPOMPB-UHFFFAOYSA-N n-methylcyclopropanamine Chemical compound CNC1CC1 VEBLEROFGPOMPB-UHFFFAOYSA-N 0.000 description 1
- JUIXJPRSYHSLHK-UHFFFAOYSA-N n-methyloxetan-3-amine Chemical compound CNC1COC1 JUIXJPRSYHSLHK-UHFFFAOYSA-N 0.000 description 1
- IOXXVNYDGIXMIP-UHFFFAOYSA-N n-methylprop-2-en-1-amine Chemical compound CNCC=C IOXXVNYDGIXMIP-UHFFFAOYSA-N 0.000 description 1
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000003506 n-propoxy group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])O* 0.000 description 1
- 239000002088 nanocapsule Substances 0.000 description 1
- 125000005893 naphthalimidyl group Chemical group 0.000 description 1
- 125000001624 naphthyl group Chemical group 0.000 description 1
- 125000004593 naphthyridinyl group Chemical group N1=C(C=CC2=CC=CN=C12)* 0.000 description 1
- 229920001206 natural gum Polymers 0.000 description 1
- 229960002653 nilutamide Drugs 0.000 description 1
- XWXYUMMDTVBTOU-UHFFFAOYSA-N nilutamide Chemical compound O=C1C(C)(C)NC(=O)N1C1=CC=C([N+]([O-])=O)C(C(F)(F)F)=C1 XWXYUMMDTVBTOU-UHFFFAOYSA-N 0.000 description 1
- 229960001420 nimustine Drugs 0.000 description 1
- VFEDRRNHLBGPNN-UHFFFAOYSA-N nimustine Chemical compound CC1=NC=C(CNC(=O)N(CCCl)N=O)C(N)=N1 VFEDRRNHLBGPNN-UHFFFAOYSA-N 0.000 description 1
- 125000004433 nitrogen atom Chemical group N* 0.000 description 1
- 231100000344 non-irritating Toxicity 0.000 description 1
- 125000001400 nonyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 125000002868 norbornyl group Chemical group C12(CCC(CC1)C2)* 0.000 description 1
- 239000007764 o/w emulsion Substances 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- OIPZNTLJVJGRCI-UHFFFAOYSA-M octadecanoyloxyaluminum;dihydrate Chemical compound O.O.CCCCCCCCCCCCCCCCCC(=O)O[Al] OIPZNTLJVJGRCI-UHFFFAOYSA-M 0.000 description 1
- 125000002347 octyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- CZDBNBLGZNWKMC-MWQNXGTOSA-N olivomycin 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=1)O[C@H]1O[C@@H](C)[C@H](O)[C@@H](OC2O[C@@H](C)[C@H](O)[C@@H](O)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@H](O)[C@H](OC)[C@H](C)O1 CZDBNBLGZNWKMC-MWQNXGTOSA-N 0.000 description 1
- 229950005848 olivomycin Drugs 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 125000001715 oxadiazolyl group Chemical group 0.000 description 1
- DWAFYCQODLXJNR-BNTLRKBRSA-L oxaliplatin Chemical compound O1C(=O)C(=O)O[Pt]11N[C@@H]2CCCC[C@H]2N1 DWAFYCQODLXJNR-BNTLRKBRSA-L 0.000 description 1
- 229960001756 oxaliplatin Drugs 0.000 description 1
- 125000003566 oxetanyl group Chemical group 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- QUANRIQJNFHVEU-UHFFFAOYSA-N oxirane;propane-1,2,3-triol Chemical compound C1CO1.OCC(O)CO QUANRIQJNFHVEU-UHFFFAOYSA-N 0.000 description 1
- 125000004430 oxygen atom Chemical group O* 0.000 description 1
- 238000012856 packing Methods 0.000 description 1
- 229960001592 paclitaxel Drugs 0.000 description 1
- AHJRHEGDXFFMBM-UHFFFAOYSA-N palbociclib Chemical compound N1=C2N(C3CCCC3)C(=O)C(C(=O)C)=C(C)C2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 AHJRHEGDXFFMBM-UHFFFAOYSA-N 0.000 description 1
- 229960004390 palbociclib Drugs 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 229940046159 pegylated liposomal doxorubicin Drugs 0.000 description 1
- 229960002621 pembrolizumab Drugs 0.000 description 1
- 229960005079 pemetrexed Drugs 0.000 description 1
- QOFFJEBXNKRSPX-ZDUSSCGKSA-N pemetrexed Chemical compound C1=N[C]2NC(N)=NC(=O)C2=C1CCC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 QOFFJEBXNKRSPX-ZDUSSCGKSA-N 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- JLFNLZLINWHATN-UHFFFAOYSA-N pentaethylene glycol Chemical compound OCCOCCOCCOCCOCCO JLFNLZLINWHATN-UHFFFAOYSA-N 0.000 description 1
- 125000003538 pentan-3-yl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])C([H])([H])[H] 0.000 description 1
- 125000001147 pentyl group Chemical group C(CCCC)* 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 125000001644 phenoxazinyl group Chemical group C1(=CC=CC=2OC3=CC=CC=C3NC12)* 0.000 description 1
- WVDDGKGOMKODPV-ZQBYOMGUSA-N phenyl(114C)methanol Chemical compound O[14CH2]C1=CC=CC=C1 WVDDGKGOMKODPV-ZQBYOMGUSA-N 0.000 description 1
- ANRQGKOBLBYXFM-UHFFFAOYSA-M phenylmagnesium bromide Chemical compound Br[Mg]C1=CC=CC=C1 ANRQGKOBLBYXFM-UHFFFAOYSA-M 0.000 description 1
- PTMHPRAIXMAOOB-UHFFFAOYSA-N phosphoramidic acid Chemical compound NP(O)(O)=O PTMHPRAIXMAOOB-UHFFFAOYSA-N 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 125000004592 phthalazinyl group Chemical group C1(=NN=CC2=CC=CC=C12)* 0.000 description 1
- 125000005545 phthalimidyl group Chemical group 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 230000001766 physiological effect Effects 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 229960003634 pimozide Drugs 0.000 description 1
- YVUQSNJEYSNKRX-UHFFFAOYSA-N pimozide Chemical compound C1=CC(F)=CC=C1C(C=1C=CC(F)=CC=1)CCCN1CCC(N2C(NC3=CC=CC=C32)=O)CC1 YVUQSNJEYSNKRX-UHFFFAOYSA-N 0.000 description 1
- 125000004193 piperazinyl group Chemical group 0.000 description 1
- NUKCGLDCWQXYOQ-UHFFFAOYSA-N piposulfan Chemical compound CS(=O)(=O)OCCC(=O)N1CCN(C(=O)CCOS(C)(=O)=O)CC1 NUKCGLDCWQXYOQ-UHFFFAOYSA-N 0.000 description 1
- 229950001100 piposulfan Drugs 0.000 description 1
- 229960001221 pirarubicin Drugs 0.000 description 1
- 239000002798 polar solvent Substances 0.000 description 1
- 229920001308 poly(aminoacid) Polymers 0.000 description 1
- 229920000747 poly(lactic acid) Polymers 0.000 description 1
- 229920003229 poly(methyl methacrylate) Polymers 0.000 description 1
- 229920000768 polyamine Polymers 0.000 description 1
- 229920000728 polyester Polymers 0.000 description 1
- 239000008389 polyethoxylated castor oil Substances 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 239000004633 polyglycolic acid Substances 0.000 description 1
- 239000004926 polymethyl methacrylate Substances 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 150000004804 polysaccharides Chemical class 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 238000002600 positron emission tomography Methods 0.000 description 1
- 229910000027 potassium carbonate Inorganic materials 0.000 description 1
- LPNYRYFBWFDTMA-UHFFFAOYSA-N potassium tert-butoxide Chemical compound [K+].CC(C)(C)[O-] LPNYRYFBWFDTMA-UHFFFAOYSA-N 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 1
- 229960004618 prednisone Drugs 0.000 description 1
- 150000003141 primary amines Chemical class 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 125000004368 propenyl group Chemical group C(=CC)* 0.000 description 1
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical compound CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 1
- 229950008679 protamine sulfate Drugs 0.000 description 1
- 235000018102 proteins Nutrition 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- 125000005494 pyridonyl group Chemical group 0.000 description 1
- 150000003230 pyrimidines Chemical class 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- SBYHFKPVCBCYGV-UHFFFAOYSA-N quinuclidine Chemical compound C1CC2CCN1CC2 SBYHFKPVCBCYGV-UHFFFAOYSA-N 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 1
- 230000035484 reaction time Effects 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 230000008707 rearrangement Effects 0.000 description 1
- 108091006082 receptor inhibitors Proteins 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 238000001953 recrystallisation Methods 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 238000006722 reduction reaction Methods 0.000 description 1
- 238000005057 refrigeration Methods 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 230000008261 resistance mechanism Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 229930002330 retinoic acid Natural products 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 238000007363 ring formation reaction Methods 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 238000010898 silica gel chromatography Methods 0.000 description 1
- 229960002930 sirolimus Drugs 0.000 description 1
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- WBHQBSYUUJJSRZ-UHFFFAOYSA-M sodium bisulfate Chemical compound [Na+].OS([O-])(=O)=O WBHQBSYUUJJSRZ-UHFFFAOYSA-M 0.000 description 1
- 229910000342 sodium bisulfate Inorganic materials 0.000 description 1
- 229910001467 sodium calcium phosphate Inorganic materials 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 239000012312 sodium hydride Substances 0.000 description 1
- 229910000104 sodium hydride Inorganic materials 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 239000007901 soft capsule Substances 0.000 description 1
- 239000002195 soluble material Substances 0.000 description 1
- 238000003797 solvolysis reaction Methods 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 125000003003 spiro group Chemical group 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 229960001052 streptozocin Drugs 0.000 description 1
- 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 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 238000000967 suction filtration Methods 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 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 1
- 229960001278 teniposide Drugs 0.000 description 1
- 229960000351 terfenadine Drugs 0.000 description 1
- ISIJQEHRDSCQIU-UHFFFAOYSA-N tert-butyl 2,7-diazaspiro[4.5]decane-7-carboxylate Chemical compound C1N(C(=O)OC(C)(C)C)CCCC11CNCC1 ISIJQEHRDSCQIU-UHFFFAOYSA-N 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- JHLVEBNWCCKSGY-UHFFFAOYSA-N tert-butyl n-methylcarbamate Chemical compound CNC(=O)OC(C)(C)C JHLVEBNWCCKSGY-UHFFFAOYSA-N 0.000 description 1
- 229960003604 testosterone Drugs 0.000 description 1
- 125000003718 tetrahydrofuranyl group Chemical group 0.000 description 1
- 125000001712 tetrahydronaphthyl group Chemical group C1(CCCC2=CC=CC=C12)* 0.000 description 1
- 125000005958 tetrahydrothienyl group Chemical group 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 229960004559 theobromine Drugs 0.000 description 1
- 125000001113 thiadiazolyl group Chemical group 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 150000003573 thiols Chemical group 0.000 description 1
- 238000003354 tissue distribution assay Methods 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 229940044693 topoisomerase inhibitor Drugs 0.000 description 1
- XFCLJVABOIYOMF-QPLCGJKRSA-N toremifene Chemical compound C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 XFCLJVABOIYOMF-QPLCGJKRSA-N 0.000 description 1
- 229960005026 toremifene Drugs 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 108700012359 toxins Proteins 0.000 description 1
- 235000010487 tragacanth Nutrition 0.000 description 1
- 239000000196 tragacanth Substances 0.000 description 1
- 229940116362 tragacanth Drugs 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 238000000844 transformation Methods 0.000 description 1
- 229960000575 trastuzumab Drugs 0.000 description 1
- IUCJMVBFZDHPDX-UHFFFAOYSA-N tretamine Chemical compound C1CN1C1=NC(N2CC2)=NC(N2CC2)=N1 IUCJMVBFZDHPDX-UHFFFAOYSA-N 0.000 description 1
- 229950001353 tretamine Drugs 0.000 description 1
- 229960001727 tretinoin Drugs 0.000 description 1
- 125000004306 triazinyl group Chemical group 0.000 description 1
- WARKYKQCOXTIAO-UHFFFAOYSA-N tributyl(2-ethoxyethenyl)stannane Chemical compound CCCC[Sn](CCCC)(CCCC)\C=C/OCC WARKYKQCOXTIAO-UHFFFAOYSA-N 0.000 description 1
- 125000003866 trichloromethyl group Chemical group ClC(Cl)(Cl)* 0.000 description 1
- IMNIMPAHZVJRPE-UHFFFAOYSA-N triethylenediamine Chemical compound C1CN2CCN1CC2 IMNIMPAHZVJRPE-UHFFFAOYSA-N 0.000 description 1
- KVJXBPDAXMEYOA-CXANFOAXSA-N trilostane Chemical compound OC1=C(C#N)C[C@]2(C)[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CC[C@@]32O[C@@H]31 KVJXBPDAXMEYOA-CXANFOAXSA-N 0.000 description 1
- 229960001670 trilostane Drugs 0.000 description 1
- CWMFRHBXRUITQE-UHFFFAOYSA-N trimethylsilylacetylene Chemical compound C[Si](C)(C)C#C CWMFRHBXRUITQE-UHFFFAOYSA-N 0.000 description 1
- NOYPYLRCIDNJJB-UHFFFAOYSA-N trimetrexate Chemical compound COC1=C(OC)C(OC)=CC(NCC=2C(=C3C(N)=NC(N)=NC3=CC=2)C)=C1 NOYPYLRCIDNJJB-UHFFFAOYSA-N 0.000 description 1
- 229960001099 trimetrexate Drugs 0.000 description 1
- 229950000212 trioxifene Drugs 0.000 description 1
- YFTHZRPMJXBUME-UHFFFAOYSA-N tripropylamine Chemical compound CCCN(CCC)CCC YFTHZRPMJXBUME-UHFFFAOYSA-N 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- 229910052722 tritium Inorganic materials 0.000 description 1
- 229960000281 trometamol Drugs 0.000 description 1
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 1
- 229950009811 ubenimex Drugs 0.000 description 1
- 229940035893 uracil Drugs 0.000 description 1
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 1
- LLYYNOVSVPBRGV-MVNKZKPCSA-N valnemulin Chemical compound CC(C)[C@@H](N)C(=O)NCC(C)(C)SCC(=O)O[C@@H]1C[C@@](C)(C=C)[C@@H](O)[C@H](C)[C@@]23CC[C@@H](C)[C@]1(C)[C@@H]2C(=O)CC3 LLYYNOVSVPBRGV-MVNKZKPCSA-N 0.000 description 1
- 229950008166 valnemulin Drugs 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- 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 1
- 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 1
- 229960004528 vincristine Drugs 0.000 description 1
- 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 1
- 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 1
- 229960004355 vindesine Drugs 0.000 description 1
- GBABOYUKABKIAF-GHYRFKGUSA-N vinorelbine Chemical compound C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC GBABOYUKABKIAF-GHYRFKGUSA-N 0.000 description 1
- 229960002066 vinorelbine Drugs 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 150000003722 vitamin derivatives Chemical class 0.000 description 1
- 238000003260 vortexing Methods 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000001993 wax Substances 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- FBTUMDXHSRTGRV-ALTNURHMSA-N zorubicin 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)=N\NC(=O)C=1C=CC=CC=1)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 FBTUMDXHSRTGRV-ALTNURHMSA-N 0.000 description 1
- 229960000641 zorubicin Drugs 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/02—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
- C07D403/06—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Oncology (AREA)
- Hematology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present disclosure relates to novel compounds useful as MAT2A inhibitors, pharmaceutical compositions comprising these compounds and methods of treatment by administration of these compounds or the pharmaceutical compositions.
Description
Technical Field
The present disclosure relates generally to novel compounds useful as MAT2A inhibitors as well as pharmaceutical compositions comprising these compounds and methods of treatment by administration of these compounds or the pharmaceutical compositions.
Background
Methionine Adenosyltransferase (MAT), also known as S-adenosylmethionine synthetase, is an enzyme that catalyzes the synthesis of S-adenosylmethionine (SAM or AdoMet) by methionine and ATP. Non-liver endogenous SAM is synthesized by methionine adenosine transferase IIA (MAT 2A) enzyme. In organisms, the expression and activity of DNA, RNA and proteins can be regulated by methylation modification, thereby controlling the growth, differentiation and death of cells. Thus, intracellular SAM levels will be tightly regulated.
The protein arginine N-methyltransferase (PRMT 5) is a methylase that utilizes SAM as a methyl donor, SAM being necessary for PRMT5 activity, while PRMT5 is competitively inhibited by 5' Methylthioadenosine (MTA). During the initiation of the methi-adenosine phosphorylase (methylthioadenosine phosphorylase, MTAP), the MTA level of the cells remained low. MTAP is located near cyclin-dependent kinase inhibitor 2A (CDKN 2A) tumor suppressor genes, and is co-deleted with CDKN2A in about 15% of all cancers. Since MTAP is the only enzyme currently known to catalyze the degradation of MTA, it is believed that the loss of MTAP results in the accumulation of MTA in cancer cells. The absence of MTAP results in the accumulation of MTA, making MTAP-deficient cells (MTAP-DELETED CELLS) more dependent on SAM production and thus on MAT2A activity than MTAP-expressing cells.
Experiments show that after MAT2A gene is knocked out, the symmetrical arginine dimethyl marker (SYMMETRIC ARGININE dimethylation mark, SDMA) of the HCT116 isogenic pair is changed differently, and the SDMA level of MTAP deletion cells is obviously lower than that of MTAP wild type cells. Under the background of MTAP deficiency cells and metabolic change in a high MTA environment, inhibition of SAM level reduction caused by MAT2A activity can inhibit PRMT5 activity, so that tumor cell death is selectively induced, and a synthetic lethal effect is achieved.
With the elucidation of the mechanism of action of MAT2A protein in recent years, MAT2A inhibitors have begun to be developed continuously. However, no MAT2A inhibitor has been approved for clinical use so far, and thus, development of a highly potent inhibitor is necessary to bring new therapeutic means to related tumor patients in the future.
Disclosure of Invention
The present disclosure provides certain 2-oxoquinazoline derivatives that are inhibitors of methionine adenosyltransferase 2A (MAT 2A). Also disclosed herein are pharmaceutical compositions comprising such compounds and methods of treating diseases treatable by inhibition of MAT2A, such as cancer, including cancers characterized by reduced or inactive methicillin-p-phosphorylase (MTAP) activity.
In a first aspect, the present disclosure provides a compound of formula (I), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
wherein:
w is CR 4 or N;
x is CR 5 or N;
y is CR 6 or N;
z is CR 7 or N;
v is CR 8 or N;
R 4、R5、R6、R7 are each independently selected from the group consisting of: hydrogen, alkyl, alkylcarbonyl, hydroxy, halogen, haloalkyl, cyano, alkynyl and-NR aRb;
R 8 is selected from the group consisting of: hydrogen, alkyl, and hydroxy;
R 1 is selected from the group consisting of: alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl and-NR cRd;
R 2 is selected from the group consisting of: hydrogen, alkyl, alkoxy, heteroalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of alkyl, alkoxy, heteroalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is optionally substituted with one or more R e;
R 3 is selected from the group consisting of: heterocyclyl, aryl, and heteroaryl, wherein each of heterocyclyl, aryl, and heteroaryl is optionally substituted with one or more R e;
R a and R b are each independently selected from hydrogen or alkyl;
r c is selected from the group consisting of: hydrogen, alkyl, and cycloalkyl;
R d is selected from the group consisting of: alkyl, alkoxy, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally substituted with one or more R e;
Each R e is independently selected from the group consisting of: oxo, cyano, halogen, hydroxy, acyl, -NR fRg、-OC(O)NRfRg, carbamoyl, carboxy, alkyl, alkenyl, alkynyl, alkoxy, alkoxyalkyl, alkoxycycloalkyl, cycloalkylalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
Each R f is independently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, heteroaryl, wherein each of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, and heteroaryl is optionally substituted with one or more groups independently selected from cyano, halo, hydroxy, or amino; and
Each R g is independently hydrogen or C 1-3 alkyl.
In one embodiment, the compound or stereoisomer, tautomer, or pharmaceutically acceptable salt thereof has a structure represented by the following formula (Ia) or (Ib):
in one embodiment, the R 1 is-NR cRd.
In one embodiment, the R c is hydrogen or alkyl.
In one embodiment, the R c is methyl.
In one embodiment, the R d is selected from the group consisting of: alkyl, alkoxy, and cycloalkyl, wherein each of alkyl, alkoxy, and cycloalkyl is optionally substituted with one or more R e.
In a preferred embodiment, each R e is independently selected from the group consisting of: cyano, hydroxy, -NR fRg, carbamoyl, alkyl, alkenyl, alkoxy, alkoxyalkyl, alkoxycycloalkyl, cycloalkylalkyl, cycloalkyl and heterocyclyl.
In one embodiment, the R d is selected from the group consisting of: methyl group,
And methoxy.
In one embodiment, the R 2 is selected from the group consisting of: hydrogen, alkyl, and aryl.
In one embodiment, the R 2 is methyl.
In one embodiment, the R 2 is phenyl.
In one embodiment, the R 3 is heterocyclyl.
In one embodiment, the R 3 is a five membered heterocyclyl.
In one embodiment, the R 3 is selected from
In one embodiment, the R 4 is hydrogen.
In one embodiment, the R 5 is hydrogen.
In one embodiment, the R 6 is selected from the group consisting of: alkylcarbonyl, halogen, haloalkyl, alkynyl and-NR aRb.
In one embodiment, the R 6 is selected from the group consisting of: chlorine, trifluoromethyl,
In one embodiment, the R 7 is hydrogen.
In one embodiment, the R 8 is hydrogen.
In one embodiment, the compound has a formula selected from the group consisting of:
In a second aspect, the present disclosure provides a pharmaceutical composition comprising a compound as described in the first aspect above, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
In a third aspect, the present disclosure provides a method of treating a MAT 2A-mediated disease in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound as described in the first aspect above, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition as described in the second aspect above.
In one embodiment, the disease in the third aspect above is cancer.
In a fourth aspect, the present disclosure provides a method of treating MTAP-deficient cancers in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound as described in the first aspect above, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition as described in the second aspect above.
In a fifth aspect, the present disclosure provides a method for treating cancer in a patient, wherein the cancer is characterized by reduced or non-expression of MTAP gene, a deletion of MTAP gene, or reduced function of MTAP protein, the method comprising administering to the subject a therapeutically effective amount of a compound as described in the first aspect above, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition as described in the second aspect above.
In a sixth aspect, the present disclosure provides the use of a compound as described in the first aspect above, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition as described in the second aspect above, in the manufacture of a medicament for the treatment of cancer.
In one embodiment, the cancer in the third or fourth or fifth or sixth aspect above is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
Detailed Description
The definition of specific functional groups and chemical terms is described in more detail below. For purposes of this disclosure, chemical elements are identified according to the periodic Table of elements (Periodic Table of THE ELEMENTS), CAS version, handbook of physics and chemistry (CHEMISTRY AND PHYSICS), 75 th edition, inner cover, and specific functional groups are generally defined as described herein. In addition, the general principles of organic chemistry and specific functional moieties and reactivities are described in the following references: organic chemistry (Organic Chemistry), thomas Sorrell, 2 nd edition, university science book press (University Science Books), assailitot, 2006; smith and March, mach' S ADVANCED Organic Chemistry, 6 th edition, john wili father company (John Wiley & Sons, inc.), new york, 2007; larock, integrated organic transformation (Comprehensive OrganicTransformations), 3 rd edition, VCH Press (VCH Publishers, inc.), new York, 2018; carruthers, some modern methods of organic synthesis (Some Modern Methods of OrganicSynthesis), 4 th edition, cambridge university Press (Cambridge University Press), cambridge, 2004; the entire contents of each of the references are incorporated herein by reference.
Throughout this disclosure, linking substituents are described. It is particularly desirable that each linking substituent includes both the forward and reverse forms of the linking substituent. For example, -NR (CR 'R') -includes both-NR (CR 'R') -and- (CR 'R') NR-. In the case where a linking group is explicitly required for a structure, the Markush variable (Markush variable) listed for the group is understood to be the linking group. For example, if the structure requires a linking group and the markush group definition of the variables lists "alkyl", it is understood that "alkyl" means a linking alkylene.
Where a bond to a substituent is shown to intersect a bond connecting two atoms in a ring, the substituent may be bonded to any atom in the ring. Where a substituent is listed, but it is not specified through which atom the substituent is bonded to the remainder of a given formula compound, the substituent may be bonded through any atom in the formula. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
Where any variable (e.g., R i) occurs more than one time in any component or formula of a compound, its definition at each occurrence is independent of its definition at every other occurrence. Thus, for example, if a group is shown to be substituted with 0 to 2R i moieties, then the group may optionally be substituted with up to two R i moieties, and R i is independently selected at each occurrence from the definition of R i. Moreover, combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
As used herein, the term "C i-j" indicates a range of numbers of carbon atoms, where i and j are integers, and the range of numbers of carbon atoms includes the endpoints (i.e., i and j) and each integer point therebetween, and where j is greater than i. For example, C 1-6 indicates a range of one to six carbon atoms, including one carbon atom, two carbon atoms, three carbon atoms, four carbon atoms, five carbon atoms, and six carbon atoms. In some embodiments, the term "C 1-12" indicates 1 to 12 carbon atoms, especially 1 to 10 carbon atoms, especially 1 to 8 carbon atoms, especially 1 to 6 carbon atoms, especially 1 to 5 carbon atoms, especially 1 to 4 carbon atoms, especially 1 to 3 carbon atoms or especially 1 to 2 carbon atoms.
As used herein, the term "acyl" refers to-C (=o) -R, wherein R is a substituent such as hydrogen, alkyl, cycloalkyl, aryl, or heterocyclyl, wherein alkyl, cycloalkyl, aryl, and heterocyclyl are as defined herein.
As used herein, the term "alkyl", whether used as part of another term or independently, refers to a saturated straight or branched chain hydrocarbon group that may optionally be independently substituted with one or more substituents described below. The term "C i-j alkyl" refers to an alkyl group having from i to i carbon atoms. In some embodiments, the alkyl groups contain 1 to 10 carbon atoms. In some embodiments, the alkyl groups contain 1 to 9 carbon atoms. In some embodiments, the alkyl group contains 1 to 8 carbon atoms, 1 to 7 carbon atoms, 1 to 6 carbon atoms, 1 to 5 carbon atoms, 1 to 4 carbon atoms, 1 to 3 carbon atoms, or 1 to 2 carbon atoms. Examples of "C 1-10 alkyl" include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, and decyl. Examples of "C 1-6 alkyl" are methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-pentyl, 3-pentyl, 2-methyl-2-butyl, 3-methyl-1-butyl, 2-methyl-1-butyl, 1-hexyl, 2-hexyl, 3-hexyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 3-methyl-3-pentyl, 2, 3-dimethyl-2-butyl, 3-dimethyl-2-butyl and the like.
As used herein, the term "alkylcarbonyl" refers to a group of formula R 'C (O) -, wherein R' is alkyl as defined above.
As used herein, the term "alkenyl", whether used as part of another term or independently, refers to a straight or branched chain hydrocarbon group having at least one carbon-carbon double bond that may be optionally independently substituted with one or more substituents described herein and includes groups having a "cis" orientation and a "trans" orientation or alternatively an "E" orientation and a "Z" orientation. In some embodiments, alkenyl groups contain 2 to 12 carbon atoms. In some embodiments, alkenyl groups contain 2 to 11 carbon atoms. In some embodiments, alkenyl groups contain 2 to 11 carbon atoms, 2 to 10 carbon atoms, 2 to 9 carbon atoms, 2 to 8 carbon atoms, 2 to 7 carbon atoms, 2 to 6 carbon atoms, 2 to 5 carbon atoms, 2 to 4 carbon atoms, 2 to 3 carbon atoms, and in some embodiments, alkenyl groups contain 2 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (ethylenyl or vinyl), propenyl (allyl), butenyl, pentenyl, 1-methyl-2-buten-1-yl, 5-hexenyl, and the like.
As used herein, the term "alkynyl", whether used as part of another term or independently, refers to a straight or branched chain hydrocarbon group having at least one carbon-carbon triple bond that may be optionally independently substituted with one or more substituents described herein. In some embodiments, alkenyl groups contain 2 to 12 carbon atoms. In some embodiments, alkynyl groups contain 2 to 11 carbon atoms. In some embodiments, alkynyl groups contain 2 to 11 carbon atoms, 2 to 10 carbon atoms, 2 to 9 carbon atoms, 2 to 8 carbon atoms, 2 to 7 carbon atoms, 2 to 6 carbon atoms, 2 to 5 carbon atoms, 2 to 4 carbon atoms, 2 to 3 carbon atoms, and in some embodiments, alkynyl groups contain 2 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl, 1-propynyl, 2-propynyl, and the like.
As used herein, the term "alkoxy", whether used as part of another term or independently, refers to an alkyl group, as previously defined, attached to the parent molecule through an oxygen atom. The term "C i-j alkoxy" means that the alkyl portion of the alkoxy group has from i to i carbon atoms. In some embodiments, the alkoxy groups contain 1 to 10 carbon atoms. In some embodiments, the alkoxy groups contain 1 to 9 carbon atoms. In some embodiments, the alkoxy group contains 1 to 8 carbon atoms, 1 to 7 carbon atoms, 1 to 6 carbon atoms, 1 to 5 carbon atoms, 1 to 4 carbon atoms, 1 to 3 carbon atoms, or 1 to 2 carbon atoms. Examples of "C 1-6 alkoxy" include, but are not limited to, methoxy, ethoxy, propoxy (e.g., n-propoxy and isopropoxy), t-butoxy, neopentyloxy, n-hexyloxy, and the like.
As used herein, the term "alkoxyalkyl" refers to a group of formula-R "OR ', wherein R' and R" are independently alkyl as defined above.
As used herein, the term "amino" refers to the-NH 2 group. The amino group may also be substituted with one or more groups such as alkyl, aryl, carbonyl, or other amino groups.
As used herein, the term "aryl", whether used as part of another term or independently, refers to mono-and polycyclic ring systems having a total of from 5 to 20 ring members, wherein at least one ring in the system is aromatic, and wherein each ring in the system contains from 3 to 12 ring members. Examples of "aryl" include, but are not limited to, phenyl, biphenyl, naphthyl, anthracenyl, and the like, which may bear one or more substituents. As used herein, the term "aryl" also includes groups in which an aromatic ring is fused to one or more additional rings. In the case of a polycyclic system, only one ring need be aromatic (e.g., 2, 3-indoline), but all rings may be aromatic (e.g., quinoline). The second ring may also be fused or bridged. Examples of polycyclic aryl groups include, but are not limited to, benzofuranyl, indanyl, phthalimidyl, naphthalimidyl, phenanthridinyl, tetrahydronaphthyl, and the like. Aryl groups may be substituted at one or more ring positions with substituents as described above.
As used herein, the term "carbamoyl" refers to-C (O) NH 2.
As used herein, the term "carboxy" refers to-COOH.
As used herein, the term "cycloalkyl", whether used as part of another term or independently, refers to monovalent non-aromatic saturated or partially unsaturated monocyclic and polycyclic systems wherein all ring atoms are carbon and the system contains at least three ring-forming carbon atoms. In some embodiments, cycloalkyl groups may contain 3 to 12 ring-forming carbon atoms, 3 to 10 ring-forming carbon atoms, 3 to 9 ring-forming carbon atoms, 3 to 8 ring-forming carbon atoms, 3 to 7 ring-forming carbon atoms, 3 to 6 ring-forming carbon atoms, 3 to 5 ring-forming carbon atoms, 4 to 12 ring-forming carbon atoms, 4 to 10 ring-forming carbon atoms, 4 to 9 ring-forming carbon atoms, 4 to 8 ring-forming carbon atoms, 4 to 7 ring-forming carbon atoms, 4 to 6 ring-forming carbon atoms, 4 to 5 ring-forming carbon atoms. Cycloalkyl groups may be saturated or partially unsaturated. Cycloalkyl groups may be substituted. In some embodiments, cycloalkyl groups may be saturated cyclic alkyl groups. In some embodiments, cycloalkyl groups may be partially unsaturated cyclic alkyl groups containing at least one double or triple bond in their ring system. In some embodiments, cycloalkyl groups may be monocyclic or polycyclic. Examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, 1-cyclohex-3-enyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl and cyclododecyl. Examples of polycyclic cycloalkyl groups include, but are not limited to, adamantyl, norbornyl, fluorenyl, spiro-pentadienyl, spiro [3.6] -decyl, bicyclo [1, 1] pentenyl, bicyclo [2, 1] heptenyl, and the like.
As used herein, the term "cycloalkylalkyl" refers to a radical of formula-R 'R ", wherein R' is alkyl as defined above, and R" is cycloalkyl as defined above.
As used herein, the term "alkoxycycloalkyl" refers to a group of formula-R 'OR "wherein R' is alkyl as defined above and R" is cycloalkyl as defined above.
As used herein, the term "cyano" refers to-CN.
As used herein, the term "halogen" refers to an atom selected from fluorine, chlorine, bromine and iodine.
As used herein, the term "haloalkyl" refers to an alkyl group as defined above substituted with one or more halogens as defined above. Examples of haloalkyl include, but are not limited to, trifluoromethyl, difluoromethyl, trichloromethyl, 2-trifluoroethyl, 1, 2-difluoroethyl, 3-bromo-2-fluoropropyl, 1, 2-dibromoethyl and the like.
As used herein, the term "heteroatom" refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur as well as any quaternized form of basic nitrogen (including N-oxides).
As used herein, the term "heteroaryl", whether used as part of another term or independently, refers to an aryl group having one or more heteroatoms in addition to carbon atoms. Heteroaryl groups may be monocyclic. Examples of monocyclic heteroaryl groups include, but are not limited to, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, benzofuranyl, and pteridinyl. Heteroaryl also includes polycyclic groups in which the heteroaryl ring is fused to one or more aryl, alicyclic, or heterocyclic rings, wherein the linking group or point of attachment is on the heteroaryl ring. Examples of polycyclic heteroaryl groups include, but are not limited to, indolyl, isoindolyl, benzothienyl, benzofuranyl, benzo [1,3] dioxolyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, dihydroquinolinyl, dihydroisoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
As used herein, the term "heterocyclyl" refers to a saturated or partially unsaturated carbocyclic group in which one or more ring atoms are heteroatoms independently selected from oxygen, sulfur, nitrogen, phosphorus, and the like, the remaining ring atoms being carbon, wherein one or more ring atoms may be optionally independently substituted with one or more substituents. In some embodiments, the heterocyclyl is a saturated heterocyclyl. In some embodiments, a heterocyclyl is a partially unsaturated heterocyclyl having one or more double bonds in its ring system. In some embodiments, the heterocyclyl may contain any oxidized form of carbon, nitrogen or sulfur and any quaternized form of basic nitrogen. "heterocyclyl" also includes groups in which the heterocyclyl is fused to a saturated, partially unsaturated, or fully unsaturated (i.e., aromatic) carbocyclic or heterocyclic ring. The heterocyclic group may be carbon-linked or nitrogen-linked, where possible. In some embodiments, the heterocycle is carbon-linked. In some embodiments, the heterocycle is nitrogen-linked. For example, the groups derived from pyrrole may be pyrrol-1-yl (nitrogen-linked) or pyrrol-3-yl (carbon-linked). Furthermore, the group derived from imidazole may be imidazol-1-yl (nitrogen linked) or imidazol-3-yl (carbon linked).
In some embodiments, the term "3-to 12-membered heterocyclyl" refers to a 3-to 12-membered saturated or partially unsaturated monocyclic or polycyclic heterocyclic ring system having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Fused, spiro, and bridged ring systems are also included within the scope of this definition. Examples of monocyclic heterocyclyl groups include, but are not limited to, oxetanyl, 1-dioxothietanylpyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrrolyl, furanyl, thienyl, pyrazolyl, imidazolyl, triazolyl, oxazolyl, thiazolyl, piperidinyl, piperazinyl, piperidinyl, morpholinyl, pyridinyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, pyridonyl, pyrimidinonyl, pyrazinonyl, pyrimidinonyl, pyridazinonyl, pyrrolidinyl, triazinonyl, and the like. Examples of fused heterocyclic groups include, but are not limited to, phenyl condensed rings or pyridyl condensed rings, such as quinolinyl, isoquinolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, quinoxalinyl, quinolizinyl, quinazolinyl, azaindolizinyl, pteridinyl, chroenyl, isochroenyl, indolyl, isoindolyl, indolizinyl, indazolyl, purinyl, benzofuranyl, isobenzofuranyl, benzimidazolyl, benzothienyl, benzothiazolyl, carbazolyl, phenazinyl, phenothiazinyl, phenanthridinyl, imidazo [1,2-a ] pyridinyl, [1,2,4] triazolo [4,3-a ] pyridinyl, [1,2,3] triazolo [4,3-a ] pyridinyl, and the like. Examples of spiroheterocyclyl groups include, but are not limited to, spiropyranyl, spirooxazinyl, and the like. Examples of bridged heterocyclyl groups include, but are not limited to, morphinan, hexamethylenetetramine, 3-aza-bicyclo [3.1.0] hexane, 8-aza-bicyclo [3.2.1] octane, 1-aza-bicyclo [2.2.2] octane, 1, 4-diazabicyclo [2.2.2] octane (DABCO), and the like.
As used herein, the term "hydroxy" refers to-OH.
As used herein, the term "hydroxyalkyl" refers to an alkyl group as defined above substituted with one or more hydroxyl groups.
As used herein, the term "oxo" refers to an =o substituent.
As used herein, the term "partially unsaturated" refers to a group that includes at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aromatic (i.e., fully unsaturated) moieties.
As used herein, the term "substituted", whether preceded by the term "optional", means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. It is to be understood that "substitution" or "substituted" includes implicit preconditions that such substitution is consistent with the permissible valences of the substituted atoms, and that the substitution results in stable or chemically feasible compounds, e.g., compounds that do not spontaneously undergo transformations such as rearrangement, cyclization, elimination, and the like. Unless otherwise indicated, an "optionally substituted" group may have the appropriate substituent at each substitutable position of the group, and where more than one position in any given structure may be substituted with more than one substituent selected from the specified group, the substituents may be the same or different at each position. It will be appreciated by those skilled in the art that the substituents themselves may be substituted, if appropriate. Unless specifically stated as "unsubstituted," references to chemical moieties herein are to be understood as including substituted variants. For example, reference to an "aryl" group or moiety implicitly includes both substituted and unsubstituted variants.
As used herein, the term "about" is intended to define a modified numerical value, thereby representing the value as a variable within an error range. When a particular range of error is not recited (e.g., standard deviation of the average values given in a chart or data table), the term "about" is understood to mean that a range of 10%, preferably 5%, is encompassed by the term "about" and is included.
As used herein, the term "disease" is intended to be generally synonymous with the terms "disorder," "disorder," and "condition" (as in medical conditions), interchangeably used, as they each reflect abnormal conditions of the human or animal body or a portion thereof that impair normal function, often manifested as distinguishing signs and symptoms, and resulting in reduced life span or reduced quality of life of the human or animal.
"Patient" is generally synonymous with the term "subject" and as used herein includes all mammals (including humans). Examples of patients include humans, livestock (e.g., cattle, goats, sheep, pigs, and rabbits), and companion animals (e.g., dogs, cats, rabbits, and horses). Preferably, the patient is a human.
As used herein, "in need of treatment" refers to a determination made by a physician or other caregiver that a subject is in need of or will benefit from treatment. The determination is made based on a variety of factors within the expertise of the physician or caregiver.
"Administering," "administering," and the like, when used in, for example, a patient, cell, tissue, organ, or biological fluid, refers to contacting a compound of formula (I), a pharmaceutical composition or diagnostic agent comprising the same, with a subject, cell, tissue, organ, or biological fluid. In the case of cells, administration includes contact of the agent with the cells (e.g., in vitro or ex vivo), as well as contact of the agent with a fluid, wherein the fluid is in contact with the cells.
As used herein, a "therapeutically effective amount" refers to an amount of a compound of formula (I), formula (Ia), formula (Ib), or a sub-embodiment described herein and/or a pharmaceutically acceptable salt thereof, which when administered alone or as part of a pharmaceutical composition and in a single dose or as part of a series of doses to a patient for treatment of a disease, is sufficient to affect treatment of such disease. The "therapeutically effective amount" will vary depending on the compound, the disease and its severity, the age, weight, etc., of the mammal to be treated. The therapeutically effective amount can be determined by measuring the relevant physiological effects and can be adjusted in connection with dosing regimens, diagnostic analysis of the subject's condition, and the like. For example, measuring the serum level of a compound of formula (I) (or a metabolite thereof) at a particular time after administration may indicate whether a therapeutically effective amount has been used.
"Treating" or "treatment" of a disease includes:
(1) Preventing a disease, i.e., preventing the clinical symptoms of the disease from developing in a mammal that may be exposed to or susceptible to the disease but has not experienced or exhibited symptoms of the disease;
(2) Inhibiting the disease, i.e., preventing or alleviating the progression of the disease or its clinical symptoms; or alternatively
(3) Remit the disease, i.e., cause regression of the disease or its clinical symptoms.
"Inhibit", "decrease" or any variation of these terms with respect to MAT2A includes any measurable decrease or complete inhibition to achieve the desired result. For example, the decrease in MAT2A activity can be reduced by about, up to about, or at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or more, or any derivable range thereof, as compared to its normal activity.
Compounds of formula (I)
The present disclosure provides novel compounds of formula (I), formula (Ia) or formula (Ib), or stereoisomers, tautomers or pharmaceutically acceptable salts thereof, pharmaceutical compositions containing the compounds or stereoisomers, tautomers or pharmaceutically acceptable salts thereof, and various uses of the disclosed compounds.
In a first aspect, the present disclosure provides a compound of formula (I), or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
wherein:
w is CR 4 or N;
x is CR 5 or N;
y is CR 6 or N;
z is CR 7 or N;
v is CR 8 or N;
R 4、R5、R6、R7 are each independently selected from the group consisting of: hydrogen, alkyl, alkylcarbonyl, hydroxy, halogen, haloalkyl, cyano, alkynyl and-NR aRb;
R 8 is selected from the group consisting of: hydrogen, alkyl, and hydroxy;
R 1 is selected from the group consisting of: alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl and-NR cRd;
R 2 is selected from the group consisting of: hydrogen, alkyl, alkoxy, heteroalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of alkyl, alkoxy, heteroalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is optionally substituted with one or more R e;
R 3 is selected from the group consisting of: heterocyclyl, aryl, and heteroaryl, wherein each of heterocyclyl, aryl, and heteroaryl is optionally substituted with one or more R e;
R a and R b are each independently selected from hydrogen or alkyl;
r c is selected from the group consisting of: hydrogen, alkyl, and cycloalkyl;
R d is selected from the group consisting of: alkyl, alkoxy, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally substituted with one or more R e;
Each R e is independently selected from the group consisting of: oxo, cyano, halogen, hydroxy, acyl, -NR fRg、-OC(O)NRfRg, carbamoyl, carboxy, alkyl, alkenyl, alkynyl, alkoxy, alkoxyalkyl, alkoxycycloalkyl, cycloalkylalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
Each R f is independently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, heteroaryl, wherein each of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, and heteroaryl is optionally substituted with one or more groups independently selected from cyano, halo, hydroxy, or amino; and
Each R g is independently hydrogen or C 1-3 alkyl.
In one embodiment, the compound or stereoisomer, tautomer, or pharmaceutically acceptable salt thereof has a structure represented by the following formula (Ia) or (Ib):
in one embodiment, the R 1 is-NR cRd.
In one embodiment, the R c is hydrogen or alkyl.
In one embodiment, the R c is methyl.
In one embodiment, the R d is selected from the group consisting of: alkyl, alkoxy, and cycloalkyl, wherein each of alkyl, alkoxy, and cycloalkyl is optionally substituted with one or more R e.
In a preferred embodiment, each R e is independently selected from the group consisting of: cyano, hydroxy, -NR fRg, carbamoyl, alkyl, alkenyl, alkoxy, alkoxyalkyl, alkoxycycloalkyl, cycloalkylalkyl, cycloalkyl and heterocyclyl.
In one embodiment, the R d is selected from the group consisting of: methyl group,
And methoxy.
In one embodiment, the R 2 is selected from the group consisting of: hydrogen, alkyl, and aryl.
In one embodiment, the R 2 is methyl.
In one embodiment, the R 2 is phenyl.
In one embodiment, the R 3 is heterocyclyl.
In one embodiment, the R 3 is a five membered heterocyclyl.
In one embodiment, the R 3 is selected from
In one embodiment, the R 4 is hydrogen.
In one embodiment, the R 5 is hydrogen.
In one embodiment, the R 6 is selected from the group consisting of: alkylcarbonyl, halogen, haloalkyl, alkynyl and-NR aRb.
In one embodiment, the R 6 is selected from the group consisting of: chlorine, trifluoromethyl,
In one embodiment, the R 7 is hydrogen.
In one embodiment, the R 8 is hydrogen.
In one embodiment, the compound has a formula selected from the group consisting of:
The compounds provided herein are described with reference to the general formula and specific compounds. Furthermore, the compounds of the present disclosure may exist in a variety of different forms or derivatives, including but not limited to prodrugs, soft drugs, active metabolic derivatives (active metabolites) and pharmaceutically acceptable salts thereof, all of which are within the scope of the present disclosure.
As used herein, the term "prodrug" refers to a compound or a pharmaceutically acceptable salt thereof that upon metabolism under physiological conditions or conversion by solvolysis yields the desired active compound. Prodrugs include, but are not limited to, esters, amides, carbamates, carbonates, ureides, solvates or hydrates of the active compounds. Typically, prodrugs are inactive or less active than the active compound, but may provide one or more advantageous handling, administration, and/or metabolic properties. For example, some prodrugs are esters of the active compound; during metabolic breakdown, the ester groups are cleaved to yield the active drug. In addition, some prodrugs are enzymatically activated to produce the active compound or compounds that produce the active compound upon further chemical reaction. The prodrug may be developed from a prodrug form to an active form in a single step, or may have one or more intermediate forms that may or may not be active themselves. The preparation and use of prodrugs is discussed in the following references: higuchi and v.stilla, "Pro-drug as novel delivery system (Pro-drugs as Novel DELIVERY SYSTEMS)", volume 14 of the a.c.s. seminar Series (a.c. symposium Series), bioreversible carrier in drug design (BioreversibleCarriers in Drug Design), editors Edward b.roche, american pharmaceutical society (AmericanPharmaceutical Association) and pegamon Press, 1987. Prodrug: challenge and return (Prodrugs: CHALLENGES AND REWARDS), edit V.Stella, R.Borchardt, M.Hageman, R.Oliyai, H.Maag, J.Tilley, new York schopleger press (SPRINGER VERLAG NEW York), 2007, all of which are hereby incorporated by reference in their entirety.
As used herein, the term "soft drug" refers to a compound that exerts a pharmacological effect but breaks down into inactive metabolite degradants such that the activity time is limited. See, for example, soft medicine: principles and methods of safe drug design (Soft drugs: PRINCIPLES AND methods for THE DESIGN of safe drugs), nicholas Bodor, drug research review (MEDICINAL RESEARCH REVIEWS), volume 4, stage 4, 449-469, 1984, which references are hereby incorporated by reference in their entirety.
As used herein, the term "metabolite", e.g., an active metabolite, overlaps with the prodrug as described above. Such metabolites are therefore pharmacologically active compounds, or compounds that are further metabolized to pharmacologically active compounds, which are derivatives produced by metabolic processes in the subject. For example, such metabolites may result from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, etc. of the administered compound or salt or prodrug. Wherein the active metabolite is such a pharmacologically active derivative compound. For prodrugs, the prodrug compounds are generally inactive or less active than the metabolite. For active metabolites, the parent compound may be an active compound or may be an inactive prodrug.
Prodrugs and active metabolites may be identified using conventional techniques known in the art. See, for example, bertolini et al, 1997, journal of pharmaceutical chemistry (J Med Chem) 40:2011-2016; shan et al, journal of pharmaceutical science (JPharm Sci) 86:756-757; bagshawe,1995, drug Dev Res 34:220-230; wermuth, supra.
As used herein, the term "pharmaceutically acceptable" means that the substance or composition is chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the subject being treated.
As used herein, the term "pharmaceutically acceptable salt" is meant to include salts of the active compounds prepared with relatively non-toxic acids or bases, depending on the particular substituents found on the compounds described herein. When the compounds disclosed herein contain relatively acidic functional groups, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base (neat or in a suitable inert solvent). Examples of salts derived from pharmaceutically acceptable inorganic bases include aluminum, ammonium, calcium, copper, iron, ferrous, lithium, magnesium, trivalent manganese, divalent manganese, potassium, sodium, zinc, and the like. Salts derived from pharmaceutically acceptable organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally occurring amines and the like, such as arginine, betaine, caffeine, choline, N' -dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucosamine, histidine, hydrabamine (hydrabamine), isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like. When the compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid (neat or in a suitable inert solvent). Examples of pharmaceutically acceptable acid addition salts include salts derived from inorganic acids such as hydrochloric, hydrobromic, nitric, carbonic, monohydrocarbonic, phosphoric, monohydrophosphoric, dihydrogenphosphoric, sulfuric, monohydrosulfuric, hydroiodic or phosphorous acid and the like, and salts derived from relatively non-toxic organic acids such as acetic, propionic, isobutyric, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic and the like. Also included are salts of amino acids, such as arginine salts and the like, and salts of organic acids, such as glucuronic acid or galacturonic acid and the like (see, e.g., berge, s.m. and the like, "pharmaceutically acceptable salts", journal of pharmaceutical science, 1977, 66, pages 1-19). Certain specific compounds of the present invention contain both basic and acidic functionalities, which allow the compounds to be converted into base or acid addition salts.
The neutral form of the compound may be regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties (e.g., solubility in polar solvents), but for the purposes of the present invention these salts are equivalent to the parent form of the compound.
The present disclosure also includes protected derivatives of the compounds of the present disclosure. For example, when the compounds of the present disclosure contain groups such as hydroxyl, carboxyl, thiol, or any group containing one or more nitrogen atoms, these groups may be protected by suitable protecting groups. A complete list of suitable protecting groups can be found in T.W.Greene, protecting groups in organic synthesis, 5 th edition, john Wiley father-son Co., ltd (2014), the disclosure of which is incorporated herein by reference in its entirety. Protected derivatives of the compounds of the present disclosure may be prepared by methods well known in the art.
The compounds of the present disclosure may exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention.
As used herein, the term "solvate" or "solvated form" refers to a solvent addition form containing a stoichiometric or non-stoichiometric amount of solvent. Some compounds tend to trap a fixed molar ratio of solvent molecules in the crystalline solid state, forming solvates. If the solvent is water, the solvate formed is a hydrate; and if the solvent is an alcohol, the solvate formed is an alkoxide. Hydrates are formed by combining one or more water molecules with one molecule of a substance that holds it as a molecular state of H 2 O. Examples of solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
The compounds of the present disclosure may exist in a variety of crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to fall within the scope of the present disclosure.
As used herein, the terms "crystalline form," "polymorphic form," and "polymorph" are used interchangeably and refer to a crystalline structure of a compound (or a salt or solvate thereof) that can crystallize in a different crystal packing arrangement, all of which have the same elemental composition. Different crystal forms typically have different X-ray diffraction patterns, infrared spectra, melting points, densities, hardness, crystal shapes, optical and electrical properties, stability and solubility. Recrystallization solvent, crystallization rate, storage temperature, and other factors may dominate one crystal form. Polymorphs of a compound can be prepared by crystallization under different conditions.
The compounds of the present disclosure may have asymmetric carbon atoms (optical centers) or double bonds; racemates, diastereomers, geometric isomers, regioisomers and individual isomers (e.g., individual enantiomers) are all intended to be included within the scope of the present invention. When a stereochemical description is shown, it refers to a compound in which one isomer is present and substantially free of the other isomer. By "substantially free" of the other isomer is meant that the ratio of the two isomers is at least 80/20, more preferably 90/10, or 95/5 or more. In some embodiments, one of the isomers will be present in an amount of at least 99%.
Those skilled in the art will appreciate that the compounds of the present disclosure may exist in different tautomeric forms, and that all such forms are contemplated as falling within the scope of the present disclosure. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that can be converted to each other by a low energy barrier. The existence and concentration of the isomeric forms will depend on the environment in which the compound is located and may vary depending, for example, on whether the compound is solid or in an organic or aqueous solution. For example, proton tautomers (also known as proton-metamorphosing tautomers) include interconversions by proton transfer, such as keto-enol, amide-imide, lactam-lactam, imine-enamine isomerisation, and cyclic forms where a proton may occupy two or more positions of a heterocyclic ring system. Valence tautomers include interconversions by recombination of some of the bond-forming electrons. Tautomers may be in equilibrium or sterically locked into one form by appropriate substitution. Unless otherwise indicated, compounds of the present disclosure identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms.
The compounds of the present disclosure may also contain unnatural amounts of isotopes on one or more of the atoms that make up such compounds. Unnatural amounts of isotopes can be defined as amounts found in nature of the atoms in question to amounts of 100%. Only when one or more isotopically enriched atoms are present. Exemplary isotopes that can be incorporated into compounds of the invention, e.g., compounds of formula (I), formula (Ia), formula (Ib) (and any of the embodiments thereof disclosed herein, including specific compounds), include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, e.g., 2H、3H、11C、13C、14C、13N、15N、15O、17O、18O、32P、33P、35S、18F、36Cl、123I and 125 I, respectively. Isotopically-labeled compounds (e.g., those labeled with 3 H and 14 C) are useful in compound or substrate tissue distribution assays. Tritium (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are useful for their ease of preparation and detectability. In addition, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). In some embodiments, in the compounds disclosed herein that are included in table 1 below, one or more hydrogen atoms are replaced with 2 H or 3 H, or one or more carbon atoms are replaced with 13 C-or 14 C-enriched carbon. Positron emitting isotopes (e.g., 15O、13N、11 C and 15 F) can be used in Positron Emission Tomography (PET) studies to examine occupancy of substrate receptors. Isotopically-labeled compounds can generally be prepared following procedures analogous to those disclosed in the schemes or examples herein by substituting an isotopically-labeled reagent for a non-isotopically-labeled reagent.
Representative compounds of the present disclosure are listed in table 1 below:
TABLE 1
Note that: compounds 8 and 9 in the above table are chiral isomers and Peak 1 and 2 are named according to the order of the peaks.
Synthesis of Compounds
The compounds provided herein may be prepared using any known organic synthesis technique and may be synthesized according to any of a number of possible synthetic routes.
The reactions for preparing the compounds of the present disclosure may be carried out in suitable solvents that may be readily selected by those skilled in the art of organic synthesis. Suitable solvents may be substantially unreactive with the starting materials (reactants), intermediates or products at the temperature at which the reaction is carried out, for example, a temperature which may range from the freezing temperature of the solvent to the boiling temperature of the solvent. A given reaction may be carried out in one solvent or a mixture of more than one solvent. Depending on the particular reaction step, the appropriate solvent for the particular reaction step may be selected by one skilled in the art.
The preparation of the compounds of the present disclosure may involve the protection and deprotection of various chemical groups. The need for protection and deprotection and the choice of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in the following references: T.W.Greene and P.GM.Wuts, protecting group in organic Synthesis (Protective Groups in Organic Synthesis), 3 rd edition, john Willi parent, new York (1999); kocienski, protecting group (Protecting Groups), george Tami Press (GeorgThieme Verlag), 2003; and Peter g.m. wuts, greene' sProtective Groups in Organic Synthesis, 5th edition, wiley,2014, all of which are incorporated herein by reference in their entirety.
The reaction may be monitored according to any suitable method known in the art. For example, product formation may be monitored by spectroscopic means such as nuclear magnetic resonance spectroscopy (e.g., 1H or 13C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographic means such as High Performance Liquid Chromatography (HPLC), liquid chromatography-mass spectrometry (LCMS), or Thin Layer Chromatography (TLC). The compounds can be purified by a variety of methods including High Performance Liquid Chromatography (HPLC) ("preparative LC-MS purification: improved compound specific method optimization" Karl f. Blom, brian Glass, RICHARD SPARKS, andrew p. Combos journal of combinatorial chemistry (j. Combi. Chem.) "2004,6 (6), 874-883, which is incorporated herein by reference in its entirety) and normal phase silica gel chromatography.
Pharmaceutical composition
In another aspect, a pharmaceutical composition is provided comprising one or more molecules or compounds of the present disclosure or a pharmaceutically acceptable salt thereof.
In another aspect, a pharmaceutical composition is provided comprising one or more molecules or compounds of the present disclosure, or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
The compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or pharmaceutically acceptable salts thereof may be in the form of compositions suitable for administration to a subject. Typically, such compositions are pharmaceutical compositions comprising a compound of formula (I), (Ia), (Ib) or a sub-embodiment thereof as described herein or a pharmaceutically acceptable salt thereof and one or more pharmaceutically or physiologically acceptable excipients. In certain embodiments, a compound of formula (I), (Ia), (Ib) or a sub-embodiment thereof, or a pharmaceutically acceptable salt thereof, as described herein, is present in a therapeutically effective amount. Pharmaceutical compositions can be used in the methods disclosed herein; thus, for example, the pharmaceutical compositions can be administered to a subject ex vivo or in vivo to practice the methods of treatment and uses described herein.
The pharmaceutical composition may be formulated to be compatible with the intended method or route of administration; exemplary routes of administration are set forth herein. Furthermore, the pharmaceutical compositions may be used in combination with other therapeutically active agents or compounds described herein to treat diseases, disorders, and conditions contemplated by the present invention.
Pharmaceutical compositions comprising an active ingredient (e.g., a compound of formula (I), (Ia), (Ib) or a sub-embodiment thereof, as described herein, a pharmaceutically acceptable salt thereof) may be in a form suitable for oral use, such as tablets, capsules, dragees (troches), lozenges (lozenges), aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups, solutions, microbeads or elixirs. Pharmaceutical compositions for oral use may be prepared according to any method known in the art for manufacturing pharmaceutical compositions, and such compositions may contain one or more agents, such as sweeteners, flavoring agents, coloring agents and preservatives to provide pharmaceutically elegant and palatable preparations. Tablets, capsules, and the like contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets, capsules, and the like. These excipients may be, for example, diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
Tablets, capsules and the like suitable for oral administration may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. The tablets may also be coated by techniques known in the art to form osmotic therapeutic tablets for controlled release. Additional agents include biodegradable or biocompatible particles or polymeric materials such as polyesters, polyamino acids, hydrogels, polyvinylpyrrolidone, polyanhydrides, polyglycolic acid, ethylene vinyl acetate, methylcellulose, carboxymethylcellulose, protamine sulfate, or copolymers of lactide and glycolide, polylactides and glycolide, or ethylene vinyl acetate copolymers to control delivery of the applied composition. For example, the oral agents may be embedded in microcapsules prepared by coacervation techniques or by interfacial polymerization (by using hydroxymethyl cellulose or gelatin-microcapsules or poly (methyl methacrylate) microcapsules, respectively), or in colloidal drug delivery systems. Colloidal dispersion systems include macromolecular complexes, nanocapsules, microspheres, microbeads, and lipid-based systems, including oil-in-water emulsions, micelles, mixed micelles, and liposomes. Methods of preparing the above formulations are known in the art.
Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example calcium carbonate, calcium phosphate, kaolin or microcrystalline cellulose, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
The aqueous suspension comprises the active substance in admixture with excipients which are suitable for the manufacture thereof. These excipients may be suspending agents, for example sodium carboxymethyl cellulose, methyl cellulose, (hydroxypropyl) methyl cellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersants or wetting agents, for example natural phospholipids (e.g. lecithin), or condensation products of alkylene oxides with fatty acids (e.g. polyoxyethylene stearate), or condensation products of ethylene oxide with long chain aliphatic alcohols (e.g. heptadecaethyleneoxycetyl alcohol), or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitols (e.g. polyoxyethylene sorbitol monooleate), or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitols (e.g. polyethylene sorbitan monooleate). The aqueous suspension may also contain one or more preservatives.
Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. Oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweeteners and flavoring agents such as those described above may be added to provide a palatable oral preparation.
Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are described in this Wen Zhongju.
The pharmaceutical composition may also be in the form of an oil-in-water emulsion. The oily phase may be a vegetable oil (for example olive oil or arachis oil), or a mineral oil (for example liquid paraffin) or a mixture thereof. Suitable emulsifying agents may be natural gums, for example acacia or tragacanth; natural phospholipids, such as soy, lecithin, and fatty acid derived or partial esters; hexitoanhydrides such as sorbitan monooleate; and condensation products of partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate.
The pharmaceutical compositions generally comprise a therapeutically effective amount of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof as described herein, and one or more pharmaceutically acceptable excipients. Suitable pharmaceutically acceptable excipients include, but are not limited to, antioxidants (e.g., ascorbic acid and sodium bisulfate), preservatives (e.g., benzyl alcohol, methyl paraben, ethyl or n-propyl paraben), emulsifiers, suspending agents, dispersants, solvents, fillers, compatibilizers, detergents, buffers, vehicles, diluents and/or adjuvants. For example, a suitable vehicle may be a physiological saline solution or a citrate buffered saline, possibly supplemented with other substances common in pharmaceutical compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. Those skilled in the art will readily recognize a variety of buffers that may be used in the pharmaceutical compositions and dosage forms contemplated herein. Typical buffers include, but are not limited to, pharmaceutically acceptable weak acids, weak bases, or mixtures thereof. For example, the buffer component may be a water-soluble material such as phosphoric acid, tartaric acid, lactic acid, succinic acid, citric acid, acetic acid, ascorbic acid, aspartic acid, glutamic acid, and salts thereof. Acceptable buffers include, for example, tris buffer, N- (2-hydroxyethyl) piperazine-N' - (2-ethanesulfonic acid) (HEPES), 2- (N-morpholino) ethanesulfonic acid (MES), 2- (N-morpholino) ethanesulfonic acid sodium salt (MES), 3- (N-morpholino) propanesulfonic acid (MOPS), and N-Tris [ hydroxymethyl ] methyl-3-aminopropanesulfonic acid (TAPS).
After the pharmaceutical composition is formulated, it may be stored in sterile vials in the form of a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder. Such formulations may be stored in a ready-to-use form, a lyophilized form that requires reconstitution prior to use, a liquid form that requires dilution prior to use, or other acceptable form. In some embodiments, the pharmaceutical composition is provided in a disposable container (e.g., a disposable vial, ampoule, syringe, or auto-injector (similar to, for example)), while a multi-use container (e.g., a multi-use vial) is provided in other embodiments.
The formulation may also include a carrier to protect the composition from rapid degradation or clearance from the body, such as controlled release formulations, including liposomes, hydrogels, prodrugs, and microencapsulated delivery systems. For example, a time delay material (e.g., glyceryl monostearate or glyceryl stearate) may be employed alone or with a wax. Any drug delivery device may be used to deliver the compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof, including implants (e.g. implantable pumps) and catheter systems, slow injection pumps and devices, all of which are well known to the skilled person.
Depot injections, typically administered subcutaneously or intramuscularly, may also be used to release the compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof over a period of time. Depot injections are typically solid or oil based and typically include at least one of the formulation ingredients described herein. One of ordinary skill in the art is familiar with possible formulations and uses of depot injections.
The pharmaceutical composition may be in the form of a sterile injectable aqueous or oleaginous suspension. The suspensions may be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents as mentioned herein. 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. Acceptable diluents, solvents and dispersion media that may be used include water, ringer's solution, isotonic sodium chloride solution, cremophor EL TM (basf, pasiponi, new jersey) or Phosphate Buffered Saline (PBS), ethanol, polyols (e.g., glycerol, propylene glycol and liquid polyethylene glycol) and suitable mixtures thereof. 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. Prolonged absorption of a particular injectable formulations can be brought about by the inclusion of agents which delay absorption, for example, aluminum monostearate or gelatin.
The compounds of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein may also be administered in the form of suppositories for rectal administration or sprays for nasal or inhalation. Suppositories may be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include, but are not limited to, cocoa butter and polyethylene glycols.
Methods of treating diseases
In another aspect, the present disclosure provides a method of treating a MAT 2A-mediated disease in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound provided herein or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein.
In some embodiments, the disease in the method of treating a MAT 2A-mediated disease in a patient is cancer, e.g., selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
In another aspect, the present disclosure provides a method of treating MTAP-deficient cancer in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound provided herein, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein.
In some embodiments, the cancer in the method of treating MTAP-deficient cancer in a patient is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
In yet another aspect, the present disclosure provides a method for treating cancer in a patient, wherein the cancer is characterized by reduced or no expression of MTAP gene, a deletion of MTAP gene, or reduced function of MTAP protein, comprising administering to the subject a therapeutically effective amount of a compound provided herein, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition provided herein.
In some embodiments, the cancer in the method for treating cancer in a patient is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
Pharmaceutical use
In another aspect, the present disclosure provides the use of a compound described herein, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein, for the manufacture of a medicament for the treatment of cancer.
In some embodiments, the cancer is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
Route of administration
The compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof and compositions comprising them may be administered in any suitable manner. Suitable routes of administration include oral, parenteral (e.g., intramuscular, intravenous, subcutaneous (e.g., injection or implantation), intraperitoneal, intracisternal, intra-articular, intraperitoneal, intracerebral (intraparenchymal) and intraventricular), nasal, vaginal, sublingual, intraocular, rectal, topical (e.g., transdermal), buccal, and inhalation. Depot injections, usually administered subcutaneously or intramuscularly, may also be used to administer the compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof over a period of time. Particular embodiments of the present invention contemplate oral administration.
Combination therapy
The present invention contemplates the use of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof as described herein in combination with one or more active therapeutic agents (e.g. chemotherapeutic agents) or other prophylactic or therapeutic means (e.g. radiation). In such combination therapies, the various active agents often have different complementary mechanisms of action. Such combination therapies may be particularly beneficial by reducing the dosage of one or more agents thereby reducing or eliminating the adverse effects associated with the one or more agents. Furthermore, such combination therapies may have synergistic therapeutic or prophylactic effects on potential diseases, disorders or conditions.
As used herein, "combination" is meant to include therapies that can be administered singly, separately, e.g., therapies formulated separately for administration (e.g., as provided in a kit), as well as therapies that can be co-administered in a single formulation (i.e., a "co-formulation").
In certain embodiments, a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein is administered or applied sequentially, e.g., one agent is administered prior to one or more other agents. In other embodiments, a compound of formula (I), (Ia), (Ib) or a sub-embodiment described herein, or a salt thereof, is administered simultaneously, e.g., two or more agents are administered simultaneously or about simultaneously; the two or more agents may be present in two or more separate formulations or combined into a single formulation (i.e., a co-formulation). Whether two or more agents are administered sequentially or simultaneously, they are considered to be administered in combination for purposes of this disclosure.
In this case, the compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof may be used in combination with at least one other (active) agent in any suitable manner. In one embodiment, treatment with at least one active agent and at least one compound of formula (I), (Ia), (Ib) or sub-embodiment or salt thereof described herein is maintained for a period of time. In another embodiment, the treatment of at least one active agent is reduced or discontinued (e.g., when the subject is stable), and the treatment of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein is performed in a constant dosage regimen. In another embodiment, the treatment of at least one active agent is reduced or discontinued (e.g., when the subject is stable), while the treatment of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein is reduced (e.g., lower dose, less frequent dosing, or a shorter treatment regimen). In yet another embodiment, the treatment of at least one active agent is reduced or discontinued (e.g., when the subject is stable), and the treatment of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein is increased (e.g., higher doses, more frequent dosing, or longer treatment regimen). In yet another embodiment, treatment of at least one active agent is maintained, and treatment of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein is reduced or discontinued (e.g., lower doses, less frequent dosing, or a shorter treatment regimen). In yet another embodiment, the treatment of at least one active agent and a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein is reduced or discontinued (e.g., lower doses, less frequent dosing, or a shorter treatment regimen).
The present disclosure provides methods of treating cancer with a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof, as described herein, and at least one additional therapeutic or diagnostic agent.
In some embodiments, a compound of formula (I), (Ia), (Ib) or a fruiting embodiment or salt thereof described herein is administered in combination with at least one additional therapeutic agent selected from temozolomide, pemetrexed, pegylated liposomal doxorubicin (Doxil), eribulin (Halaven, sea Le Wei), ixabepilone (Ixempra), protein-bound paclitaxel (Abraxane), oxaliplatin, irinotecan, valnemulin (bcl 2 inhibitor), 5-azacytidine, anti-CD 20 therapeutic agents such as rituximab and obin You Tuozhu mab, hormonal agents (anatolzole, ai Kexi mad (exemestand), letrozole, norrad, leuprorelin acetate (lupon eligard CDK)), 4/6 inhibitors, palbociclib, abberamide, CPI (avermectin, cimepruno Li Shan-resistance (Cemiplimab-rwlc), and bevacizumab.
In certain embodiments, the present disclosure provides methods of treating cancer comprising administering a compound of formula (I), (Ia), (Ib) or a sub-embodiment described herein, or a salt thereof, in combination with a Signal Transduction Inhibitor (STI) to achieve additive or synergistic inhibition of tumor growth. As used herein, the term "signal transduction inhibitor" refers to an agent that selectively inhibits one or more steps in a signal pathway. Examples of Signal Transduction Inhibitors (STI) useful in the methods described herein include, but are not limited to: (i) bcr/abl kinase inhibitors (e.g., gleevec); (ii) Epidermal Growth Factor (EGF) receptor inhibitors, including kinase inhibitors and antibodies; (iii) Inhibitors of her-2/neu receptor (e.g., herceptin); (iv) Inhibitors of Akt family kinases or Akt pathways (e.g., rapamycin); (v) Cell cycle kinase inhibitors (e.g., fraapine); (vi) inhibitors of phosphatidylinositol kinase. Agents involved in immunomodulation may also be used in combination with one or more compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof to inhibit tumor growth in cancer patients.
In certain embodiments, the present disclosure provides methods of treating cancer comprising administering a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein in combination with a chemotherapeutic agent. Examples of chemotherapeutic agents include, but are not limited to, alkylating agents, such as thiotepa and cyclophosphamide; alkyl sulfonates such as busulfan, imperoshu, piposulfan; aziridines, such as benzodopa (benzodopa), carboquinone, methodol (meturedopa), and Wu Leiduo bar (uredopa); ethyleneimine and methyl melamines (METHYLAMELAMINE) include altretamine, triethylenemelamine, triethylenephosphoramide (trietylenephosphoramide), triethylenephosphoramide (triethylenethiophosphaoramide), and trimethylol melamine; nitrogen mustards, such as chlorambucil, napthalene mustards, chlorflufosamide (cholophosphamide), estramustine, ifosfamide, dichloromethyl diethylamine, mechlorethamine hydrochloride, melphalan, new enbixing, chlorambucil cholesterol, prednisone mustards, qu Luolin amine, uracil mustards; nitrosoureas such as carmustine, pimozide, fotemustine, lomustine, nimustine, and ramustine; antibiotics, such as doxorubicin, actinomycin, aflatoxin, diazoserine, bleomycin, C actinomycin, calicheamicin, karabin (carabicin), carminomycin, carcinomycin, chromomycin, D actinomycin, daunorubicin, ditubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, elubicin, nordaunorubicin (idambicin), marrubicin, mitomycin, mycophenolic acid, nolamycin, olivomycin, pelomycin, bofevernix Luo Mei (potfiromycin), puromycin, tri-iron doxorubicin, rodubicin, streptozocin, truffle, ubenimex, cilastatin, zorubicin; antimetabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as, for example, dimethyl folic acid, methotrexate, pterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thioadenine, thioguanine; pyrimidine analogs such as ambcitabine, azacytidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, deoxyfluorouridine, enocitabine, fluorouridine, 5-FU; androgens, such as carbosterone, drotasone propionate, cyclothioandrostane, emasculan, and testosterone; anti-adrenergic agents such as aminoglutethimide, mitotane, trilostane; folic acid supplements such as She Wansuan; acetoglucurolactone; aldehyde phosphoramidate glycoside; aminolevulinic acid; amsacrine; bei Labu shake (bestrabucil); a specific group; idatroxas; ground phosphoramide (defofamine); colchicine; deaquinone; efluoroornithine (elfornithine); eli acetoamide; eggshell robust; gallium nitrate; hydroxyurea; mushroom polysaccharide; lonidamine; mitoguazone; mitoxantrone; mo Pai darol; diamine nitroacridine; prastatin; egg ammonia nitrogen mustard; pirarubicin; podophylloic acid; 2-ethyl hydrazide; procarbazine; carrying out a process of preparing the raw materials; sugar (sizofran) of the west adjuvant; germanium spiroamine; tenuazonic acid; triiminoquinone; 2,2',2 "-trichlorotriethylamine; uratam; vindesine; dacarbazine; mannitol; dibromomannitol; dibromodulcitol; pipobromine; adding cytosine; arabinocytidine (Ara-C); cyclophosphamide; thiotepa; taxanes, such as paclitaxel and docetaxel; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum and platinum coordination complexes such as cisplatin and carboplatin; vinblastine; trastuzumab; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; north vitamin; nux An Tuo; teniposide; daunomycin; aminopterin; proper roda; ibandronate; CPT11; topoisomerase inhibitors; difluoromethyl ornithine (DMFO); retinoic acid; lamycin; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. In particular embodiments, the compounds of the present disclosure are co-administered with a cytostatic compound selected from cisplatin, doxorubicin, paclitaxel (taxol), docetaxel, and mitomycin C. In a particular embodiment, the cytostatic compound is doxorubicin.
Chemotherapeutic agents also include anti-hormonal agents, such as antiestrogens, including, for example, tamoxifen, raloxifene, aromatase inhibiting 4 (5) -imidazole, 4-hydroxy tamoxifen, trawoxifene (trioxifene), raloxifene (keoxifene), onapristone (onapristone), and toremifene; antiandrogens, such as flutamide, nilutamide, bicalutamide, enzalutamide, apalutamide, abiraterone acetate, leuprorelin, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. In certain embodiments, the combination therapy comprises administration of a hormone or related hormonal agent.
The present disclosure also contemplates the use of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof as described herein in combination with an immune checkpoint inhibitor. The large number of gene and epigenetic changes that characterize all cancers provide a diverse set of antigens that the immune system can use to distinguish tumor cells from their normal counterparts. In the case of T cells, the final magnitude (e.g., level of cytokine production or proliferation) and quality (e.g., type of immune response produced, e.g., pattern of cytokine production) of the response initiated by T Cell Receptor (TCR) antigen recognition is regulated by a balance between co-stimulatory and inhibitory signals (immune checkpoints). Under normal physiological conditions, immune checkpoints are critical for preventing autoimmunity (i.e., maintaining self-tolerance) and protecting tissue from damage when the immune system reacts to pathogen infection. Expression of immune checkpoint proteins may be deregulated by tumors as an important immune resistance mechanism. Examples of immune checkpoint inhibitors include, but are not limited to, CTLA-4, PD-1, PD-L1, BTLA, TIM3, LAG3, OX40, 41BB, VISTA, CD96, TGF beta, CD73, CD39, A2AR, A2BR, IDO1, TDO2, arginase, B7-H3, B7-H4. Cell-based anti-cancer immunomodulators are also contemplated herein. Examples of such modulators include, but are not limited to, chimeric antigen receptor T cells, tumor infiltrating T cells, and dendritic cells.
The present disclosure contemplates the use of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein in combination with an immune checkpoint receptor and a ligand inhibitor as described above (e.g., ipilimumab, abamectin, nivolumab, pembrolizumab, atuzumab, nivolumab, and divalizumab).
Other therapeutic modalities that may be combined with the compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof include radiation therapy, monoclonal antibodies to tumor antigens, complexes of monoclonal antibodies and toxins, T cell adjuvants, bone marrow transplants or antigen presenting cells (e.g., dendritic cell therapy).
The present disclosure contemplates the use of a compound of formula (I), (Ia), (Ib) or an embodiment or salt thereof described herein, alone or in combination with radiation therapy and/or Temozolomide (TMZ), avastin or lomustine, for the treatment of glioblastoma.
The present disclosure includes pharmaceutically acceptable salts, acids, or derivatives of any of the above.
Administration of drugs
The amount of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein that can be administered to a subject depends, for example, on the target of administration (e.g., resolution desired); age, weight, sex, and health and physical condition of the subject to whom the formulation is administered; route of administration; and the nature of the disease, disorder, condition or symptom. The dosage regimen may also take into account the presence, nature and extent of any side effects associated with the administered agent. Effective dosages and dosage regimens can be readily determined from, for example, safety and dose escalation assays, in vivo studies (e.g., animal models), and other methods known to those of skill in the art.
Generally the dosing parameters prescribe that the dose should be less than the amount likely to produce irreversible toxicity to the subject (maximum tolerated dose (MTD)) and not less than the amount required to produce a measurable effect on the subject. Such amounts are determined by, for example, pharmacokinetic and pharmacodynamic parameters associated with ADME, taking into account the route of administration and other factors.
An effective dose (EFFECTIVE DOSE, ED) is a dose or amount of an agent that produces a therapeutic response or desired effect in a portion of the subject taking the agent. A "median effective dose" or ED 50 of an agent refers to the dose or amount of the agent that produces a therapeutic response or desired effect in 50% of the population to whom the agent is administered. Although ED 50 is generally used as a reasonable predictor of the efficacy of an agent, it is not necessarily the appropriate dosage for a clinician to consider all relevant factors. Thus, in some cases, the effective amount is greater than the calculated ED 50, while in still other cases, the effective amount is less than the calculated ED 50, and in still other cases, the effective amount is the same as the calculated ED 50.
Furthermore, an effective dose of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein may be an amount that produces a desired result relative to a healthy subject when administered to the subject in one or more doses. For example, for a subject experiencing a particular disorder, an effective dose may be a dose that improves a diagnostic parameter, measure, marker, etc., of the disorder by at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or greater than 90%, where 100% is defined as the diagnostic parameter, measure, marker, etc., exhibited by a normal subject.
In certain embodiments, a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein may be administered (e.g., orally) one or more times per day at a dosage level of about 0.01mg/kg to about 50mg/kg, or about 1mg/kg to about 25mg/kg of the subject's body weight per day to achieve the desired therapeutic effect.
For administration of oral medicaments, the compositions may be provided in the form of tablets, capsules and the like, containing from 1.0 to 1000 mg of the active ingredient, in particular 1.0、3.0、5.0、10.0、15.0、20.0、25.0、50.0、75.0、100.0、150.0、200.0、250.0、300.0、400.0、500.0、600.0、750.0、800.0、900.0 and 1.00.0 mg of the active ingredient.
In certain embodiments, the dosages of the compounds of formula (I), (Ia), (Ib) or sub-embodiments or salts thereof described herein are contained in a "unit dosage form". The phrase "unit dosage form" refers to physically discrete units, each unit containing a predetermined amount of a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein, alone or in combination with one or more additional agents, sufficient to produce the desired effect. It will be appreciated that the parameters of the unit dosage form will depend on the particular agent and the effect to be achieved.
Kit for detecting a substance in a sample
The invention also contemplates kits comprising compounds of formula (I), (Ia), (Ib), or sub-embodiments described herein, or salts thereof, and pharmaceutical compositions thereof. As described below, the kit is generally in the form of a physical structure containing the various components and may be used, for example, to carry out the above-described methods.
The kit may comprise one or more compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof (provided, for example, in a sterile container), which may be in the form of a pharmaceutical composition suitable for administration to a subject. The compounds of formula (I), (Ia), (Ib) or sub-embodiments described herein or salts thereof may be provided in a form ready for use (e.g. tablet or capsule) or in a form that is required, for example, to be reconstituted or diluted (e.g. powder) prior to administration. When a compound of formula (I), (Ia), (Ib) or a sub-embodiment or salt thereof described herein is in a form that requires reconstitution or dilution by a user, the kit may further include diluents (e.g., sterile water), buffers, pharmaceutically acceptable excipients, etc., packaged together or separately with the compound of formula (I), (Ia), (Ib) or sub-embodiment or salt thereof described herein. When combination therapies are contemplated, the kit may comprise several agents alone, or they may already be combined in the kit. Each component of the kit may be enclosed in a separate container, and all of the various containers may be in a single package. The kit of the present invention may be designed for proper maintenance of the conditions (e.g., refrigeration or freezing) necessary for the components contained therein.
The kit may comprise a label or package insert that includes therein identifying information of the components and their instructions for use (e.g., dosage parameters, clinical pharmacology of the active ingredient(s), including mechanism of action, pharmacokinetics and pharmacodynamics, adverse reactions, contraindications, etc.). The label or insert may include manufacturer information such as lot number and expiration date. The label or package insert may be, for example, integrated into the physical structure containing the components, contained within the physical structure alone, or attached to the components of the kit (e.g., ampoule, tube, or vial).
The label or insert may additionally include or be incorporated into a computer-readable medium, such as a magnetic disk (e.g., hard disk, card, memory disk), optical disk, such as CD-or DVD-ROM/RAM, DVD, MP, magnetic tape, or electronic storage medium (e.g., RAM and ROM), or a combination thereof, such as magnetic/optical storage medium, flash memory medium, or memory card. In some embodiments, no actual instructions are present in the kit, but a means is provided to obtain instructions from a remote source, such as through the internet.
Synthetic examples
The following examples and references (intermediates) are intended to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the invention and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the following experiments, or all experiments that can be performed, are performed. It should be understood that the exemplary descriptions written at the present time are not necessarily made, but rather that these descriptions may be made to generate data or the like as described herein. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.), but some experimental errors and deviations should be accounted for.
Example 1
First step
Reaction 1-1 (10 g,90.8 mmol), N, N-diisopropylethylamine (23 g,181 mmol) was dissolved in N, N-dimethylformamide (100 mL), 2- (trimethylsilyl) ethoxymethyl chloride (18.2 g,109 mmol) was added at 0deg.C, and after the addition was completed, the temperature was raised to 50deg.C for 16h. After the completion of the reaction, water (400 mL), ethyl acetate extraction (100 mL. Times.3), organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated, and the crude product was purified by column chromatography to give intermediate 1-2 (15 g, yield 68.8%).
Second step
Intermediate 1-2 (12 g,50.4 mmol), t-butylsulfinamide (9.2 g,75.6 mmol) was dissolved in tetrahydrofuran (300 mL), tetraethyltitanate (46 g,201.6 mmol) was added and stirred for 16h at 70℃under nitrogen. After the reaction was completed, cooled to room temperature, ethyl acetate (150 mL) was added, poured into ice water (300 mL), stirred for 15min, the solid was filtered off, the filtrate was extracted by liquid-liquid separation, the aqueous phase was extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated, and the crude product was purified by column chromatography to give intermediate 1-3 (12.5 g, yield 73.1%).
Third step
Intermediate 1-3 (12.5 g,36.4 mmol) was dissolved in tetrahydrofuran (120 mL) and lithium tri-sec-butylborohydride (54.7 mL,54.6 mmol) was added dropwise at-50℃under nitrogen. After the completion of the dropwise addition, the reaction was naturally warmed to room temperature and quenched for 1h, saturated aqueous ammonium chloride (50 mL) was added dropwise under ice bath, the reaction was quenched with stirring for 10min, extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated, and the crude product was purified by column chromatography to give intermediate 1-4 (9.6 g, yield 76.4%).
Fourth step
Intermediate 1-4 (5 g,14.5 mmol) was dissolved in 1, 4-dioxane (50 mL), hydrochloric acid-dioxane solution (50 mL) was added, reacted at room temperature for 1h, the solvent was removed by concentration under reduced pressure, dichloroethane (50 mL) was added to the residue, triethylamine (4 mL,29.0 mmol) was added to give crude IM-01, which was carried forward without purification.
Fifth step
Compounds 1-5 (2.5 g,14.45 mmol) were dissolved in 1, 2-dichloroethane (25 mL) and oxalyl chloride (1.65 mL,19.51 mmol) was slowly added at room temperature. Stirring at 55℃for 1h, then heating to 85℃and stirring for 16h. The reaction system was cooled to 0℃and IM-01 (3.5 g,13.00 mmol) was added dropwise thereto, followed by stirring at room temperature for 2 hours. Water (20 mL) was added to the reaction system, extraction was performed with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, and concentrated by filtration. The crude product was purified by column chromatography to give compound 1-6 (2.11 g, yield 33.2%) as a yellow powder.
Sixth step
Compounds 1-6 (2.11 g,4.79 mmol) were dissolved in toluene (20 mL) and nitrogen blanketed, lithium bis trimethylsilylamide (50 mL,7.19 mmol) was added dropwise at 0deg.C and stirred at 80deg.C for 16h. Water (20 mL) was added to the reaction system, extraction was performed with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate (50 g), and concentrated by filtration. The crude product was purified by column chromatography to give compound 1-7 (1.56 g, yield 93.0%) as a white powder.
Seventh step
Compounds 1-7 (150 mg,0.36 mmol) were dissolved in anhydrous acetonitrile (5 mL) and 4-dimethylaminopyridine (88 mg,0.72 mmol), triethylamine (0.25 mL,1.80 mmol) and 2,4, 6-triisopropylbenzenesulfonyl chloride (545 mg,1.80 mmol) were added, respectively, at room temperature. And (3) stirring for 4 hours at 80 ℃ under the protection of nitrogen to obtain an IM-02 crude product solution, wherein the solution is directly used for the next step without purification.
Eighth step
Compound IM-02 was added 3-methylaminooxetane (622.2 mg,7.14 mmol) at room temperature and stirred for 2h at room temperature. Water (20 mL) was added to the reaction system, extraction was performed with ethyl acetate (10 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate (5 g), and concentrated by filtration. The crude product was purified by column chromatography to give 1-8 (50 mg, yield 27.6%) as pale yellow solid.
Ninth step
Compounds 1-8 (50 mg,0.10 mmol) were dissolved in dichloromethane (1 mL) and trifluoroacetic acid (1 mL) was added dropwise at room temperature and stirred at room temperature for 1h. The solvent was removed by concentration under reduced pressure, dichloromethane (5 mL) was added, the system was basified with N, N-diisopropylethylamine to ph=8 and concentrated under reduced pressure to give a residue. The crude product was purified by high performance liquid chromatography to give 1 (5.0 mg, yield 13.4%) as a white solid.
1H NMR(400MHz,DMSO-d6)δ12.09(s,1H),8.05(d,J=8.4Hz,1H),7.62(s,1H),7.53(s,1H),7.28(d,J=8.6Hz,1H),7.18(s,2H),6.39(s,1H),4.54(s,1H),4.11(dd,J=13.0,6.7Hz,1H),3.91(d,J=6.8Hz,1H),3.47(s,1H),2.87~2.77(m,1H),2.34(s,3H),1.81(d,J=7.2Hz,3H).
MS calcd for C17H20ClN5O3[M+H]+378.2.
Example 2
First step
The compound IM-02 was added to 3-methylaminopropionitrile (293 mg,3.4 mmol) at room temperature, and the reaction mixture was stirred at room temperature for 2h. Extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give compound 2-1 (40 mg, yield 47.1%) as a transparent oil.
Second step
Compound 2-1 (40 mg,0.082 mmol) was dissolved in dichloromethane (1 mL) and trifluoroacetic acid (1 mL) was added dropwise at room temperature. The reaction solution was stirred at room temperature for 1h, and the obtained reaction solution was directly concentrated, and the crude product was purified by preparative chromatography to give white solid 2 (15 mg, yield 51.7%).
1H NMR(400MHz,DMSO)δ12.01(s,1H),7.84(d,J=8.8Hz,1H),7.58(s,1H),7.44(s,2H),7.12–7.02(m,2H),6.89(s,1H),6.21(q,J=7.0Hz,1H),3.88–3.73(m,2H),3.24(s,3H),2.58–2.52(m,2H),1.73(d,J=7.2Hz,3H).
MS calculated for C17H19O2ClN6[M+H]+374.123,found 375.60.
Example 3
First step
The compound IM-02 (130 mg,0.19 mmol) was added N, N, N' -trimethylethylenediamine (387 mg,3.79 mmol) at room temperature. The resulting mixture was stirred at room temperature under an atmosphere of N 2 for 2h. The mixture was extracted with ethyl acetate (50 mL. Times.3). The organic phases were combined and dried over sodium sulfate, filtered and concentrated. The mixture was concentrated to give the crude product. The crude product was purified by column chromatography to give compound 3-1 (34 mg, yield 35.6%) as a transparent oil.
Second step
Compound 3-1 (34 mg,0.07 mmol) was dissolved in dichloromethane (1 mL), and trifluoroacetic acid (1 mL) was added dropwise at room temperature. The resulting mixture was stirred at room temperature under an atmosphere of N 2 for 1h. Evaporation of the solvent gave the crude product. The crude product was purified by column chromatography to give compound 3 (10.1 mg, yield 40.1%) as a pale yellow powder.
1H NMR(400MHz,DMSO)δ12.00(s,1H),7.90(d,J=8.8Hz,1H),7.57(s,1H),7.48(s,1H),7.12–7.02(m,2H),6.20(q,J=7.0Hz,1H),3.79–3.65(m,2H),3.25(s,3H),2.60(t,J=6.5Hz,2H),2.18(s,6H),1.72(d,J=7.2Hz,3H).
MS calcd for C18H23ClN6O[M+H]+375.16,found 375.70.
Example 4
First step
Compound IM-02 (130 mg,0.19 mmol) was added N- (2-methoxyethyl) methylamine (387 mg,3.79 mmol) at room temperature. The resulting mixture was stirred at room temperature under an atmosphere of N 2 for 2h. The mixture was extracted with ethyl acetate (50 mL. Times.3). The organic phases were combined and dried over sodium sulfate, filtered and concentrated. The mixture was concentrated to give the crude product. The crude product was purified by column chromatography to give 4-1 (26 mg, yield 28.0%) as a transparent oil.
Second step
Compound 4-1 (26 mg,0.05 mmol) was dissolved in dichloromethane (1 mL), and trifluoroacetic acid (1 mL) was added dropwise at room temperature. The resulting mixture was stirred at room temperature under an atmosphere of N 2 for 1h. Spin-drying the solvent to give the crude product. The crude product was purified by column chromatography to give compound 4 (10.9 mg, yield 56.9%) as a pale yellow powder.
1H NMR(400MHz,DMSO)δ11.99(s,1H),7.94(d,J=8.7Hz,1H),7.57(s,1H),7.50(s,1H),7.08(d,J=9.8Hz,2H),6.20(q,J=6.9Hz,1H),3.83–3.76(m,2H),3.67(t,J=5.4Hz,2H),3.29(s,3H),3.26(s,3H),1.73(d,J=7.1Hz,3H).
MS calcd for C17H20ClN5O2[M+H]+362.13,found 362.50.
Example 5
Compound 2 (200 mg,0.53 mmol) was dissolved in tetrahydrofuran (1 mL) and Burgess reagent (191 mg,1.07 mmol) was added under an ice-water bath. The reaction was stirred at room temperature overnight. The reaction mixture was diluted with ethyl acetate, followed by addition of water and extraction with ethyl acetate (15 mL. Times.3). The combined organic phases were dried over sodium sulfate, filtered and concentrated. The crude product was purified by preparative chromatography to give 5 as a white solid (15 mg, yield 7.9%).
1H NMR(400MHz,DMSO)δ12.00(s,1H),7.89(d,J=8.8Hz,1H),7.58(s,1H),7.53(d,J=1.9Hz,1H),7.11(dd,J=8.8,2.0Hz,1H),7.07(s,1H),6.23(q,J=6.9Hz,1H),3.88(ddd,J=27.3,13.7,6.8Hz,2H),3.33(s,3H),2.99(d,J=6.8Hz,2H),1.73(d,J=7.2Hz,3H).
MS calculated for C17H17OClN6[M+H]+357.12,found 375.00.
Example 6
First step
The compound IM-02 was added to N-methyl ring Ding Jiaan (178.0 mg,1.8 mmol) at room temperature and stirred at room temperature for 2h. Water (20 mL) was added to the reaction system, extraction was performed with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and concentrated by filtration. The crude product was purified by column chromatography to give 6-1 (100 mg, yield 56.2%) as a pale yellow solid.
Second step
Compound 6-1 (60 mg,0.13 mmol) was dissolved in dichloromethane (1 mL), trifluoroacetic acid (1 mL) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1h. The solvent was removed by concentration under reduced pressure, dichloromethane (5 mL) was added, the system was basified with N, N-diisopropylethylamine to ph=8 and concentrated under reduced pressure to give a residue. The crude product was purified by high performance liquid chromatography to give 6 as a white solid (20.0 mg, yield 27.0%).
1H NMR(400MHz,DMSO-d6)δ11.98(d,J=36.9Hz,1H),7.84(d,J=8.8Hz,1H),7.61(s,1H),7.52(d,J=1.9Hz,1H),7.11(dd,J=8.9,2.1Hz,2H),6.23(q,J=7.0Hz,1H),3.80(dd,J=13.6,7.5Hz,1H),3.68(dd,J=13.5,7.0Hz,1H),3.24(d,J=8.7Hz,3H),2.79(dt,J=15.3,7.7Hz,1H),2.11~2.00(m,2H),1.95~1.76(m,4H),1.75(d,J=7.1Hz,3H).
MS calcd for C19H22ClN5O[M+H]+372.0.
Example 7
First step
Reactant 7-1 (8.7 g,90.8 mmol), N, N-diisopropylethylamine (23 g,181.0 mmol) was dissolved in N, N-dimethylformamide (100 mL), 2- (trimethylsilyl) ethoxymethyl chloride (18.2 g,109.0 mmol) was added at 0deg.C, and after the addition was completed, the temperature was raised to 50deg.C for 16h. After the completion of the reaction, water (400 mL), ethyl acetate extraction (100 mL. Times.3), organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated to give intermediate 7-2 (4.1 g, yield 68.8%) as a crude product by column chromatography.
Second step
Intermediate 7-2 (11.4 g,50.4 mmol), t-butylsulfinamide (9.2 g,75.6 mmol) was dissolved in tetrahydrofuran (300 mL), tetraethyltitanate (46 g,201.6 mmol) was added and stirred for 16h at 70℃under nitrogen. After the reaction was completed, cooled to room temperature, ethyl acetate (150 mL) was added, poured into ice water (300 mL), stirred for 15min, the solid was filtered off, the filtrate was extracted by liquid-phase extraction, the aqueous phase was extracted with ethyl acetate (100 ml×3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate (50 g), filtered and concentrated, and the crude product was purified by column chromatography to give intermediate 7-3 (12.1 g, yield 73.1%).
Third step
Intermediate 7-3 (12.1 g,36.4 mmol) was dissolved in tetrahydrofuran (120 mL) and lithium tri-sec-butylborohydride (54.7 mL,54.6 mmol) was added dropwise at-50℃under nitrogen. After the completion of the dropwise addition, the reaction was naturally warmed to room temperature and quenched for 1h, saturated aqueous ammonium chloride (50 mL) was added dropwise under ice bath, the reaction was quenched with stirring for 10min, extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated, and the crude product was purified by column chromatography to give intermediate 7-4 (9.2 g, yield 76.4%).
Fourth step
Intermediate 7-4 (4.8 g,14.5 mmol) was dissolved in 1, 4-dioxane (50 mL), hydrochloric acid-dioxane solution (50 mL) was added, reacted at room temperature for 1h, and the solvent was removed by concentration under reduced pressure to give a residue. Dichloroethane (50 mL) and triethylamine (4 mL,29.0 mmol) were then added, the pH was adjusted to 8, and the residue 7-5 was obtained after concentration and carried forward without purification.
Fifth step
Compounds 1-5 (2.5 g,14.45 mmol) were dissolved in 1, 2-dichloroethane (25 mL) and oxalyl chloride (1.65 mL,19.51 mmol) was slowly added at room temperature. Stirring at 55℃for 1h, then heating to 85℃and stirring for 16h. The reaction system was cooled to 0℃and 7-5 (3.4 g,13.00 mmol) was added dropwise thereto at 0℃and stirred at room temperature for 2 hours. To the reaction system was added 20mL of water, extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give compound 7-7 (2.11 g, yield 33.0%) as a yellow powder.
Sixth step
Compound 7-7 (2.24 g,4.79 mmol) was dissolved in toluene (20 mL), and lithium bis (trimethylsilylamide) (50 mL,7.19 mmol) was added dropwise at 0deg.C under nitrogen and stirred at 80deg.C for 16h. Water (20 mL) was added to the reaction system, extraction was performed with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and concentrated by filtration. The crude product was purified by column chromatography to give compound 7-8 (1.56 g, yield 80.0%) as a white powder.
Seventh step
Compound 7-8 (146 mg,0.36 mmol) was dissolved in anhydrous acetonitrile (5 mL) and 4-dimethylaminopyridine (88 mg,0.72 mmol), triethylamine (0.25 mL,1.80 mmol) and 2,4, 6-triisopropylbenzenesulfonyl chloride (545 mg,1.80 mmol) were added, respectively, at room temperature. The reaction solution was stirred at 80℃for 4h under nitrogen protection and was used directly in the next step without treatment.
Eighth step
Compounds 7-9 1-cyclopropyl-N-methyl methylamine (607.91 mg,7.14 mmol) was added at room temperature and stirred for 2h at room temperature. Water (20 mL) was added to the reaction system, extraction was performed with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and concentrated by filtration. The crude product was purified by column chromatography to give 7-10 (100 mg, yield 58.7%) as a pale yellow solid.
Ninth step
Compounds 7-10 (100 mg,0.21 mmol) were dissolved in dichloromethane (1 mL) and trifluoroacetic acid (1 mL) was added dropwise at room temperature and stirred at room temperature for 1h. The solvent was removed by concentration under reduced pressure, dichloromethane (5 mL) was added, the system was basified with N, N-diisopropylethylamine to ph=8 and concentrated under reduced pressure to give a residue. The crude product was purified by high performance liquid chromatography to give 7 as a white solid (43 mg, yield 58.0%).
1H NMR(400MHz,DMSO-d6)δ11.91(d,J=42.6Hz,1H),7.95(d,J=8.8Hz,1H),7.79(d,J=1.7Hz,1H),7.59(s,1H),7.20(dd,J=8.8,1.8Hz,1H),6.99(s,1H),5.16(s,2H),3.55(d,J=6.7Hz,2H),1.21(t,J=5.9Hz,1H),0.67~0.49(m,2H),0.32(q,J=4.9Hz,2H).
MS calcd for C17H18ClN5O[M+H]+344.3.
Examples 8&9
First step
1-Cyclopropyl-N-methyl methylamine (607.91 mg,7.14 mmol) was added to the compound IM-02 at room temperature, and the mixture was stirred at room temperature for 2h. Water (20 mL) was added to the reaction system, extraction was performed with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and concentrated by filtration. The crude product was purified by column chromatography to give 8-1 (100 mg, yield 57.5%) as a pale yellow solid.
Second step
Compound 8-1 (100 mg,0.21 mmol) was dissolved in dichloromethane (1 mL), trifluoroacetic acid (1 mL) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1h. The solvent was removed by concentration under reduced pressure, dichloromethane (5 mL) was added, the system was basified with N, N-diisopropylethylamine to ph=8 and concentrated under reduced pressure to give a residue. The crude product was purified by high performance liquid chromatography to give 8-2 as a white solid (43 mg, yield 58.0%).
Third step
Chiral resolution of compound 8-2 (43 mg) with 0.1% diethyl amine in the mobile phase gave compound 8 (22 mg) and compound 9 (14 mg) with ee >98%.
1H NMR(400MHz,DMSO-d6)δ11.99(d,J=32.5Hz,1H),7.89(d,J=8.7Hz,1H),7.61(s,1H),7.52(s,1H),7.12(d,J=10.6Hz,2H),6.23(q,J=6.9Hz,1H),3.65~3.44(m,2H),3.33(s,3H),1.75(d,J=7.2Hz,3H),1.20(d,J=6.6Hz,1H),0.57(q,J=5.2Hz,2H),0.32(d,J=4.2Hz,2H).
MS calcd for C18H20ClN5O[M+H]+358.2.
Example 10
First step
Compound 10-1 (2.0 g,7.8 mmol) was dissolved in tetrahydrofuran (20 mL) and sodium hydride (625 mg,15.6 mmol) was added at 0deg.C. The reaction solution was stirred at room temperature for 1h. Tert-butyl methylcarbamate (2.1 g,15.6 mmol) was then added and stirred at room temperature for 2h. Ethyl acetate was added thereto, water was added thereto, and extraction was performed with ethyl acetate (30 ml×3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give 10-2 (1.0 g, yield 57.1%) as a colorless oily compound.
Second step
Compound 10-2 (1.0 g,4.4 mmol) was dissolved in dichloromethane (20 mL) and 1, 4-dioxane (4M, 5 mL) was added at 0deg.C. The reaction solution was stirred at room temperature for 1h. Stirring for 1h at room temperature. Direct concentration gave 10-3 (550 mg) as a white solid.
Third step
Compound 10-3 (400 mg,3.6 mmol) was added IM-02 at room temperature and stirred at room temperature for 2h. The reaction mixture was diluted with ethyl acetate, and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give 10-4 (120 mg, yield 65%) as a transparent oily compound.
Fourth step
Compound 10-4 (120 mg,0.23 mmol) was dissolved in dichloromethane (1 mL) and trifluoroacetic acid (1 mL) was added dropwise at room temperature. The reaction solution was stirred at room temperature for 1h, and the obtained reaction solution was directly concentrated, and the crude product was prepared by high performance liquid chromatography to give 10 (30 mg, yield 33.7%) as a white solid.
1H NMR(400MHz,DMSO)δ12.01(s,1H),7.92(d,J=8.7Hz,1H),7.54(d,J=29.9Hz,2H),7.08(d,J=10.1Hz,2H),6.21(q,J=7.1Hz,1H),3.77(d,J=4.2Hz,4H),3.24(s,3H),1.73(d,J=7.1Hz,3H),0.44(dd,J=9.0,6.7Hz,4H).
MS calculated for C17H18ClN5O[M+H]+388.15,found 388.80.
Example 11
First step
Raw material 11-1 (10.0 g,90.8 mmol) was dissolved in N, N-dimethylformamide (100 mL), N-diisopropylethylamine (23.5 g,181.6 mmol) and 2- (trimethylsilyl) ethoxymethyl chloride (24.2 g,145 mmol) were added sequentially to the above reaction system, and the reaction was heated to 50℃for 16h. The reaction was cooled to room temperature, diluted with ethyl acetate, extracted with water (400 mL), three times with ethyl acetate (100 mL. Times.3), and the combined organic phases were washed with saturated brine (400 mL), dried, filtered, concentrated, and the crude product purified by column chromatography to give 11-2 (13.0 g, yield 55.3%) as a white solid.
Second step
Intermediate 11-2 (13.0 g,57.5 mmol) was dissolved in tetrahydrofuran (300 mL), and tert-butylsulfinamide (10.4 g,86.2 mmol) and Ti (OEt) 4 (52.5 g,230.0 mmol) were added sequentially, and the reaction was stirred under nitrogen at 70deg.C for 16h. After the reaction was completed, the reaction mixture was cooled to room temperature, ethyl acetate (200 mL) was added, the mixture was poured into ice water (400 mL) and stirred for 15min, the filtrate was extracted with ethyl acetate (200 mL. Times.3), the combined organic layers were washed with saturated brine (400 mL), dried, filtered and concentrated, and the crude product was purified by column chromatography to give intermediate 11-3 (14.5 g, yield 80.3%).
Third step
Intermediate 11-3 (14.5 g,46.2 mmol) was dissolved in tetrahydrofuran (150 mL) and methyl Grignard reagent (55.4 mL,55.4 mmol) was slowly added dropwise to the reaction solution at 0deg.C under nitrogen. Then the reaction system was naturally warmed to room temperature, stirred for 3 hours, after the reaction was completed, the reaction was quenched with saturated aqueous ammonium chloride solution (100 mL) at 0 ℃, extracted three times (100 ml×3) with ethyl acetate after stirring for 10 minutes, the combined organic layers were washed with saturated brine (300 mL), dried over anhydrous sodium sulfate, filtered, concentrated, and the crude product was purified by column chromatography to give intermediate 11-4 (11.2 g, 77.1%).
Fourth step
Intermediate 11-4 (5.0 g,14.5 mmol) was dissolved in 1, 4-dioxane (50 mL), 1, 4-dioxane solution (50 mL) was added and reacted at room temperature for 1h, after the reaction was completed, the obtained hydrochloride was concentrated, dichloroethane (50 mL) was added, triethylamine (4 mL,29.0 mmol) was added under ice-water bath, and free 11-5 (3.6 g, crude product) was concentrated, and the next step was directly fed without purification.
Fifth step
4-Chloro-2-fluorobenzamide (2.0 g,11.6 mmol) was dissolved in anhydrous 1, 2-dichloroethane (20 mL) and oxalyl chloride (1.6 mL,12.7 mmol) was slowly added at room temperature. The reaction system was stirred at 80℃for 16h under an N 2 atmosphere. The reaction mixture was cooled to room temperature, and after concentration, the crude product was dissolved in anhydrous 1, 2-dichloroethane (20 mL), to which 11-5 (3.0 g,12.7 mmol) was added dropwise at 0℃and stirred at room temperature for 2h. After completion of the reaction, the reaction mixture was poured into water and extracted with methylene chloride (100 mL. Times.3). The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product, which was purified by column chromatography to give 11-6 (546 mg, 11.8%) as a white solid.
Sixth step
Compound 11-6 (546 mg,1.8 mmol) was dissolved in toluene (20 mL) and potassium bistrimethylsilylamino (9.0 mL,9.0 mmol) was added dropwise at 0deg.C. The reaction solution was stirred at 80℃for 16h under an atmosphere of N 2. The reaction was cooled to room temperature, and the reaction mixture was poured into a saturated ammonium chloride (50 mL) solution and extracted with ethyl acetate (50 mL. Times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give 11-7 (264 mg, 48.3%).
Seventh step
Compound 11-7 (264 mg,0.91 mmol) was dissolved in dichloromethane (3.0 mL), 4-dimethylaminopyridine (22 mg,0.18 mmol), triethylamine (0.18 mL,1.82 mmol) and di-tert-butyl dicarbonate (294 mg,1.37 mmol) were added separately at room temperature, and the reaction was stirred at room temperature for 2h. After the reaction was completed, the reaction mixture was poured into water (50 mL) and extracted with methylene chloride (20 mL. Times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give 11-8 (271mg, 76.3%).
Eighth step
Compound 11-8 (97 mg,0.24 mmol) was dissolved in anhydrous acetonitrile (3.0 mL) and 4-dimethylaminopyridine (58.6 mg,0.48 mmol), triethylamine (0.07 mL,0.72 mmol) and 2,4, 6-triisopropylbenzenesulfonyl chloride (362 mg,1.2 mmol) were added, respectively, at room temperature. The reaction mixture was stirred at 80℃for 4h under N 2 atmosphere and the reaction was used directly in the next step.
Ninth step
1-Cyclopropyl-N-methyl methylamine (204 mg,2.4 mmol) was added to the reaction solution at room temperature, and stirred at room temperature for 2 hours. After the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted with methylene chloride (20 mL. Times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give 11-9 (54 mg, 39.4%).
Tenth step
Compound 11-9 (54 mg,0.12 mmol) was dissolved in dioxane (2 mL), and hydrochloric acid-dioxane solution (4M, 2 mL) was added dropwise at room temperature. The reaction solution was stirred at room temperature for 1h, after completion of the reaction, the crude product was concentrated to give 11 (13.6 mg, yield 31.1%) as a white solid by purification through high performance liquid chromatography.
1H NMR(400MHz,MeOD)δ7.92(d,J=8.6Hz,1H),7.63(s,1H),7.24(s,1H),7.13(d,J=8.5Hz,1H),6.41(q,J=7.1Hz,1H),6.23(s,1H),3.73–3.58(m,2H),3.42(s,3H),1.88(d,J=7.2Hz,3H),1.37–1.17(m,1H),0.62(q,J=5.6Hz,2H),0.35(q,J=4.9Hz,2H).
MS calculated for C18H20ClN5O[M+H]+358.14,found 358.00.
Example 12
First step
Compound 12-1 (150 mg,1.07 mmol) was added at room temperature to intermediate IM-02 and stirred at room temperature for 2h. The reaction mixture was diluted with ethyl acetate, and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were dried over sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give 12-2 (55 mg, 42.3%) as a clear oil.
Second step
Compound 12-2 (55 mg,0.10 mmol) was dissolved in dichloromethane (1 mL), and trifluoroacetic acid (1 mL) was added dropwise at room temperature. The reaction was stirred at room temperature for 1h to give a direct concentration of the reaction solution, and the crude product was purified by preparative chromatography to give 12 (20 mg, 47.9%) as a white solid.
1H NMR(400MHz,DMSO)δ8.21(s,1H),7.91(d,J=8.8Hz,1H),7.52(s,1H),7.36(s,1H),7.18–7.12(m,1H),6.22–6.05(m,1H),4.67–4.56(m,2H),3.20(s,3H),3.10(d,J=7.3Hz,1H),2.43(s,3H),2.17(s,3H),1.76(d,J=7.0Hz,3H).
MS calculated for C20H21O2ClN6[M+H]+413.14,found 412.70
Example 13
First step
N-allylmethylamine (245 mg,3.5 mmol) was added to the intermediate IM-02 reaction solution at room temperature, and stirred at room temperature for 2h. After the reaction was completed, the reaction mixture was poured into water (20 mL) and extracted with methylene chloride (20 mL. Times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated. The crude product was purified by column chromatography to give 13-1 (14 mg, 23.5%).
Second step
Compound 13-1 (14 mg,0.03 mmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (2 mL) was added dropwise at room temperature. The reaction solution was stirred at room temperature for 2 hours, and after completion of the reaction, the crude product was concentrated to give 13 (3.9 mg, 38.6%) as a white solid by purification through high performance liquid chromatography.
1H NMR(400MHz,MeOD)δ7.84(d,J=8.8Hz,1H),7.57(s,1H),7.32(s,1H),7.10(s,1H),7.06(dd,J=8.8,1.4Hz,1H),6.26(q,J=7.0Hz,1H),6.11–5.97(m,1H),5.39–5.34(m,2H),4.35–4.30(m,1H),4.22–4.17(m,1H),3.22(s,3H),1.83(d,J=7.1Hz,3H).
MS calculated for C17H18OClN5[M+H]+343.12,found 343.00.
Example 14
First step
Compound 14-1 (15 g,156 mmol), potassium carbonate (43 g,311 mmol) was dissolved in acetonitrile (150 mL), 2- (trimethylsilyl) ethoxymethyl chloride (28.6 g,171.6 mmol) was added, and the reaction was completed at 50℃for 16h. After the completion of the reaction, water (500 mL) was added, extraction was performed three times with ethyl acetate (3X 500 mL), and the organic layer was washed with saturated brine (400 mL), dried and concentrated, and the crude product was purified by column chromatography to give intermediate 14-2 (11 g, 31.4%).
Second step
Compound 14-2 (3 g,13.3 mmol), t-butylsulfinamide (2.4 g,19.8 mmol) was dissolved in tetrahydrofuran (60 mL), tetraethyltitanate (15 g,52.8 mmol) was added and the reaction stirred under nitrogen at 70deg.C for 16h. After the reaction was cooled to room temperature, ethyl acetate (40 mL) was added, poured into ice water (50 mL), stirred for 15min, the solid was filtered off, the organic phase was separated, the aqueous phase was extracted with ethyl acetate (2X 20 mL), the organic phase was combined with saturated brine (30 mL) and dried and concentrated, and the crude product was purified by column chromatography to give intermediate 14-3 (3.1 g, 71%).
Third step
Compound 14-3 (2.5 g,7.59 mmol) was dissolved in tetrahydrofuran (40 mL), and phenylmagnesium bromide (16 mL,16 mmol) was added dropwise to the reaction solution at 0deg.C under nitrogen. After the reaction was completed, the reaction system was warmed to room temperature and quenched with water (15 mL) dropwise in ice bath, stirred for 10min, extracted with ethyl acetate (2X 50 mL), the combined organic layers were washed with saturated brine (30 mL), dried and concentrated, and the crude product was purified by column chromatography to give intermediate 14-4 (3.1 g, 83.6%).
Fourth step
Compound 14-4 (2 g,4.9 mmol) was dissolved in 1, 4-dioxane (20 mL), and after the addition of hydrochloric acid-dioxane solution (10 mL), the reaction was carried out at room temperature for 16h, after the completion of the reaction, the solvent was dried by spinning, the hydrochloride was added to dichloroethane (30 mL), triethylamine (4 mL,29.0 mmol) was added under ice bath, the solvent was dried by spinning to give the free amine, and the next step was directly fed without purification.
Fifth step
Raw material 14-6 (1 g,4.8 mmol) was dissolved in N, N-dimethylformamide (25 mL), 2- (7-azabenzotriazol) -N, N, N ', N' -tetramethylurea hexafluorophosphate (2.74 g,7.2 mmol), amine chloride (0.77 g,14.4 mmol) and N, N-diisopropylethylamine (1.86 g,14.4 mmol) were added, and after completion of the reaction, water (100 mL) was added, and after stirring for 10min, suction filtration was performed, and the cake was dried by water washing to give compound 14-7 (0.6 g, 60.3%) as a white powder.
Sixth step
Compound 14-7 (1 g,4.83 mmol) was dissolved in anhydrous 1, 2-dichloroethane (15 mL) and oxalyl chloride (800 mg,6.28 mmol) was slowly added at room temperature. The resulting mixture was stirred under nitrogen at 55℃for 1 hour and then at 85℃for 16 hours. The mixture was stirred at 0℃for 2 hours at room temperature by adding intermediate 14-5 (1.46 g,4.83 mmol). LCMS indicated complete reaction. The mixture was quenched with ice water (15 mL) and extracted with ethyl acetate (3×20 mL). The organic phases were combined and dried over sodium sulfate, filtered and concentrated to give the crude product. The crude product was purified by column chromatography to give compound 14-8 (750 mg, 33.2%) as a pale yellow powder.
Seventh step
Compound 14-8 (630 mg,1.53 mmol) was dissolved in toluene (12 mL) and lithium bis trimethylsilylamide (4.58 mL,4.58 mmol) was added dropwise at 0deg.C. The resulting mixture was stirred under nitrogen at 80℃for 16 hours. LCMS indicated complete reaction. The mixture was quenched with water (10 mL) and extracted with ethyl acetate (3X 10 mL). The organic phases were combined and dried over sodium sulfate, filtered and concentrated to give the crude product. The crude product was purified by column chromatography to give compound 14-9 (580 mg, 73.5%) as a pale yellow powder.
Eighth step
Compound 14-9 (560 mg,1.08 mmol) was dissolved in anhydrous acetonitrile (10 mL) and benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (1.92 g,4.34 mmol), 1, 8-diazabicyclo [5.4.0] undec-7-ene (660 mg,4.34 mmol) and a solution of dimethylamine in tetrahydrofuran (21.6 mL,43.2 mmol) were added, respectively, at room temperature. The resulting mixture was stirred at room temperature under sealed conditions for 16 hours, and the reaction mixture was quenched with water (10 mL) and extracted with ethyl acetate (3X 10 mL). The organic phases were combined and dried over sodium sulfate, filtered and concentrated to give the crude product. The crude product was purified by column chromatography to give compound 14-10 (160 mg, 27.2%) as a pale yellow solid.
Ninth step
Compound 14-10 (25 mg,0.05 mmol) was dissolved in dichloromethane (1 mL) and trifluoroacetic acid (1 mL) was added dropwise at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. LCMS indicated complete reaction and evaporation of solvent gave the crude product. The crude product was purified by high performance liquid chromatography to give compound 14 (10 mg, 52.6%) as a white powder.
1H NMR(400MHz,DMSO-d6)δ12.13(s,1H),8.07(d,J=8.5Hz,1H),7.97(s,1H),7.68(s,1H),7.63(s,1H),7.29(d,J=7.4Hz,3H),7.24(d,J=6.7Hz,1H),7.20(d,J=7.7Hz,2H),7.12(s,1H),3.29(s,6H).
MS calcd for C21H18OF3N5[M+H]+414.15,found 414.2.
Example 15
First step
N-methylcyclopropylamine (508.0 mg,7.14 mmol) was added to the compound IM-02 at room temperature, and the mixture was stirred at room temperature for 2h. The reaction mixture was quenched with 20mL of water, extracted with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate (5 g), and concentrated by filtration. The crude product was purified by column chromatography to give 15-1 (150 mg, yield 90.0%) as a pale yellow solid.
Second step
Compound 15-1 (150 mg,0.10 mmol) was dissolved in dichloromethane (1 mL), trifluoroacetic acid (1 mL) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour. The solvent was removed by concentration under reduced pressure, dichloromethane (5 mL) was added, the system was basified with N, N-diisopropylethylamine to ph=8 and concentrated under reduced pressure to give a residue. The crude product was purified by high performance liquid chromatography to give 15 (40 mg, 30.3% yield) as a white solid.
1H NMR(400MHz,DMSO-d6)δ12.02(s,1H),8.29(d,J=8.8Hz,1H),7.61(s,1H),7.51(s,1H),7.09(d,J=8.8Hz,2H),6.29(q,J=7.0Hz,1H),3.43(s,1H),3.19(s,3H),1.75(d,J=7.2Hz,3H),0.86(d,J=5.9Hz,2H),0.65(dd,J=10.4,3.8Hz,1H),0.53(s,1H).
MS calcd for C17H18ClN5O[M+H]+344.2.
Example 16
First step
A solution of compound IM-02 (244 mg,0.36 mmol) in acetonitrile was added N-methylcyclobutylamine hydrochloride (870 mg,7.2 mmol) and N, N-diisopropylethylamine (923 mg,7.2 mmol) at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. LCMS indicated complete reaction. The mixture was extracted with ethyl acetate (3X 50 mL). The organic phases were combined and dried over sodium sulfate, filtered and concentrated to give the crude product. The crude product was purified by prep. to give compound 16-1 (150 mg, 85.0%) as a pale yellow solid.
Second step
Compound 16-1 (200 mg,0.41 mmol) was dissolved in dichloromethane (2 mL), and trifluoroacetic acid (2 mL) was added dropwise at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. LCMS indicated complete reaction and evaporation of solvent gave the crude product. The crude product was purified by high performance liquid chromatography to give compound 16 (40 mg, 27.3%) as a white powder.
1H NMR(400MHz,DMSO-d6)δ12.00(s,1H),7.70(d,J=8.7Hz,1H),7.53(d,J=32.3Hz,2H),7.20–7.01(m,2H),6.26–6.16(m,1H),4.55(p,J=8.6Hz,1H),3.11(s,3H),2.36–2.14(m,4H),1.79–1.52(m,5H).
MS calcd for C18H20OClN5[M+H]+358.14,found 358.3.
Example 17
First step
Compound IM-02 was added methoxymethyl amine (436.2 mg,7.14 mmol) at room temperature and stirred at room temperature for 2h. To the reaction system was added 20mL of water, extracted with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate (5 g), and concentrated by filtration. The crude product was purified by column chromatography to give 17-1 (50 mg, yield 30.3%) as a pale yellow solid.
Second step
Compound 17-1 (50 mg,0.11 mmol) was dissolved in dichloromethane (1 mL), trifluoroacetic acid (1 mL) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour. The solvent was removed by concentration under reduced pressure, DCM (5 mL) was added, the system was basified with N, N-diisopropylethylamine to ph=8 and concentrated under reduced pressure to give a residue. The crude product was purified by high performance liquid chromatography to give 17 (2.0 mg, yield 5.6%) as a white solid.
1H NMR(400MHz,DMSO-d6)δ12.05(s,1H),8.38(d,J=8.8Hz,
1H),7.60(d,J=11.9Hz,2H),7.19(d,J=8.8Hz,1H),7.12(s,1H),6.42(d,J=7.1Hz,1H),3.80(s,3H),3.44(s,3H),1.78(d,J=7.2Hz,3H).
MS calcd for C15H16ClN5O2[M+H]+334.2.
Example 18
First step
Compound 18-1 (2.5 g,14.45 mmol) was dissolved in dichloroethane (25 mL) and oxalyl chloride (1.65 mL,19.51 mmol) was slowly added at room temperature. After stirring at 55℃for 1 hour, the temperature was raised to 85℃and stirred for 16 hours. IM-01 (3.5 g,13.00 mmol) was added dropwise thereto at 0deg.C and stirred at room temperature for 2 hours. To the reaction system was added 20mL of water, extracted with ethyl acetate (100 mL. Times.3), the organic phases were combined, washed with saturated brine (100 mL), dried over anhydrous sodium sulfate (50 g), and concentrated by filtration. The crude product was purified by column chromatography to give compound 18-2 (2.11 g, yield 33.2%) as a yellow powder.
Second step
Compound 18-2 (2.11 g,4.79 mmol) was dissolved in toluene (20 mL), and lithium bis (trimethylsilylamide) (50 mL,7.19 mmol) was added dropwise at 0deg.C under nitrogen and stirred at 80deg.C for 16h. To the reaction system was added 20mL of water, extracted with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate (50 g), and concentrated by filtration. The crude product was purified by column chromatography to give compound IM-03 (1.56 g, yield 93.0%) as a white powder.
Third step
Compound IM-03 (400 mg,0.88 mmol) was dissolved in anhydrous dioxane (5 mL) and tributyl (2-ethoxyvinyl) tin (480 mg,1.44 mmol), triethylamine (180 mg,1.80 mmol) and tribenzylideneacetone dipalladium (54 mg,0.08 mmol) were added at room temperature, respectively. Under the protection of nitrogen, stirring for 16 hours at 80 ℃, adding 2N hydrochloric acid aqueous solution after the reaction is finished, and stirring for 1 hour. To the reaction system was added 10mL of water, extracted with ethyl acetate (10 mL. Times.3), the organic phases were combined, washed with saturated brine (20 mL), dried over anhydrous sodium sulfate (5 g), and concentrated by filtration. The crude product was purified by column chromatography to give 18-4 (110 mg, yield 29.1%) as a yellow oil.
Fourth step
Compound 18-4 (150 mg,0.36 mmol) was dissolved in anhydrous acetonitrile (5 mL) and 4-dimethylaminopyridine (88 mg,0.72 mmol), triethylamine (0.25 mL,1.80 mmol) and 2,4, 6-triisopropylbenzenesulfonyl chloride (545 mg,1.80 mmol) were added, respectively, at room temperature. The reaction solution was concentrated under nitrogen protection at 80℃for 4 hours to give 18-5 crude, which was used directly in the next step without purification.
Compound 18-5A solution of dimethylamine in tetrahydrofuran (7.1 mL,7.14 mmol) was added at room temperature and stirred at room temperature for 2h. To the reaction system was added 20mL of water, extracted with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate (5 g), and concentrated by filtration. The crude product was purified by column chromatography to give 18-6 (90 mg, yield 76.9%) as a pale yellow solid.
Sixth step
Compound 18-6 (90 mg,0.20 mmol) was dissolved in dichloromethane (1 mL), trifluoroacetic acid (1 mL) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour. The solvent was removed by concentration under reduced pressure. The crude product was purified by high performance liquid chromatography to give white solid 18 (7.1 mg, yield 11.0%).
1H NMR(400MHz,DMSO-d6)δ9.00(s,1H),8.11(d,J=8.4Hz,1H),7.81(s,1H),7.76~7.66(m,2H),6.27(d,J=5.3Hz,1H),3.30(s,6H),2.60(s,3H),1.84(d,J=6.8Hz,3H).
MS calcd for C17H19N5O2[M+H]+326.2.
Example 19
First step
Cyclopropylmethylamine (241 mg,3.4 mmol) was added to compound IM-02 at room temperature, and the mixture was stirred at room temperature for 2h. To the reaction system was added 20mL of water, extracted with ethyl acetate (20 mL. Times.3), the organic phases were combined, washed with saturated brine (50 mL), dried over anhydrous sodium sulfate (5 g), and concentrated by filtration. The crude product was purified by column chromatography to give compound 19-1 (20 mg, 25%) as a clear oil.
Second step
Compound 19-1 (20 mg,0.038 mmol) was dissolved in dichloromethane (1 mL), and trifluoroacetic acid (1 mL) was added dropwise at room temperature. The reaction solution was stirred at room temperature for 1 hour, and the obtained reaction solution was directly concentrated, and the crude product was purified by high performance liquid chromatography to obtain white solid 19 (12 mg, 80.5%).
1H NMR(400MHz,DMSO)δ11.99(s,1H),8.44(s,1H),8.09(d,J=8.6Hz,1H),7.56(s,1H),7.40(s,1H),7.15(d,J=8.0Hz,1H),7.05(s,1H),6.45(q,J=6.8Hz,1H),1.71(d,J=7.2Hz,3H),1.21–1.11(m,1H),0.51–0.44(m,2H),0.31–0.25(m,2H).
MS calculated for C17H18ClN5O[M+H]+344.12,found 344.30.
Example 20
First step
Compound IM-03 (210 mg,0.45 mmol), trimethylethynyl silicon (70 mg,0.71 mmol) was dissolved in dioxane (5 mL), and dichloro-bis (triphenylphosphine) palladium (35 mg,0.05 mmol), cuprous iodide (21 mg,0.11 mmol), triphenylphosphine (28 mg,0.11 mmol) and triethylamine (5 mL) were added, respectively. The resulting mixture was nitrogen-substituted three times and reacted at 90℃for 16 hours. LCMS indicated complete reaction. The reaction mixture was added with water (10 mL) and extracted with ethyl acetate (3X 10 mL). The organic phases were combined and dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product. The crude product is directly fed to the next step without purification.
Second step
Compound 20-1 (217 mg,0.45 mmol) was dissolved in tetrabutylammonium fluoride in tetrahydrofuran (6 mL,6 mmol), stirred at room temperature for 2 hours, and the reaction mixture was concentrated to give a crude product. The crude product was purified by column chromatography to give compound 20-2 (115 mg, 62.3%) as a pale yellow solid.
Third step
Compound 20-2 (85 mg,0.21 mmol) was dissolved in anhydrous acetonitrile (5 mL) and 4-dimethylaminopyridine (45 mg,0.37 mmol), triethylamine (95 mg,0.94 mmol) and 2,4, 6-triisopropylbenzenesulfonyl chloride (300 mg,1.0 mmol) were added, respectively, at room temperature. The resulting mixture was stirred at 80℃for 4 hours under nitrogen protection and the reaction solution was used directly in the next step without treatment.
Fourth step
Compound 20-3 (142 mg,0.21 mmol) was dissolved in anhydrous acetonitrile (5 mL) and dimethylamine (2.1 mL,4.2 mmol) was added at room temperature. The resulting mixture was stirred at room temperature under nitrogen for 2 hours. LCMS indicated complete reaction. The mixture was added with water (10 mL) and extracted with ethyl acetate (3X 10 mL). The organic phases were combined and dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product. The crude product was purified by column chromatography to give compound 20-4 (52 mg, 56.7%) as a pale yellow solid.
Fifth step
Compound 20-4 (63 mg,0.41 mmol) was dissolved in dichloromethane (1.5 mL) and trifluoroacetic acid (1.5 mL) was added dropwise at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. LCMS indicated complete reaction and evaporation of solvent gave the crude product. The crude product was purified by high performance liquid chromatography to give compound 20 (20 mg, 45.2%) as a white powder.
1H NMR(400MHz,DMSO-d6)δ11.98(s,1H),7.85(d,J=8.4Hz,1H),7.52(d,J=33.7Hz,2H),7.09(d,J=8.5Hz,1H),7.04(s,1H),6.21(d,J=7.2Hz,1H),4.35(s,1H),3.22(s,6H),1.72(d,J=7.2Hz,3H).
MS calcd for C17H17ON5[M+H]+308.14,found 308.0.
Example 21
Compound IM-03 (110 mg,0.24 mmol) was dissolved in dioxane (10 mL), and tribenzylidene acetone dipalladium (46 mg,0.05 mmol), 2- (dicyclohexylphosphine) 3, 6-dimethoxy-2, 4, 6-triisopropyl-1, 1' -biphenyl (54 mg,0.1 mmol), potassium t-butoxide (38 mg,0.34 mmol) and ethylamine (0.5 mL,0.5 mmol) were added, respectively. The resulting mixture was nitrogen-substituted three times and reacted at 100℃for 16 hours. LCMS indicated complete reaction. The reaction mixture was added with water (10 mL) and extracted with ethyl acetate (3X 10 mL). The organic phases were combined and dried over sodium sulfate, filtered and concentrated to give the crude product. The crude product was purified by column chromatography to give compound 21-1 (67 mg, 65.9%) as a yellow powder.
Second step
Compound 21-1 (65 mg,0.15 mmol) was dissolved in anhydrous acetonitrile (10 mL) and benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (265 mg,0.6 mmol), 1, 8-diazabicyclo [5.4.0] undec-7-ene (91 mg,0.6 mmol) and dimethylamine (3 mL,6 mmol) were added separately at room temperature. The resulting mixture was stirred at room temperature under sealed conditions for 16 hours, and the reaction mixture was extracted with ethyl acetate (3X 10 mL) by adding water (10 mL). The organic phases were combined and dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude product. The crude product was purified by prep. to give compound 21-2 (45 mg, 65.3%) as a pale yellow solid.
Third step
Compound 21-2 (40 mg,0.41 mmol) was dissolved in dichloromethane (1.5 mL) and trifluoroacetic acid (1.5 mL) was added dropwise at room temperature. The resulting mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. LCMS indicated complete reaction and evaporation of solvent gave the crude product. The crude product was purified by prep. to give compound 21 (13 mg, 45.5%) as a white powder.
1H NMR(400MHz,DMSO-d6)δ11.92(s,1H),7.56–7.40(m,2H),6.94(d,J=27.9Hz,1H),6.55–6.38(m,1H),6.37–6.23(m,3H),3.13(d,J=8.7Hz,6H),2.87(q,2H),1.72(dd,J=7.2,4.5Hz,3H),1.03(t,J=7.1Hz,3H).
MS calcd for C17H22ON6[M+H]+327.19,found 327.3.
The control compounds of the test examples of the present invention were prepared as described in reference patent WO2021252678, the structure of which is shown below:
Biological examples
Test example 1: enzymatic Activity assay
The malachite green fluorescence test described below was used to determine the ability of compounds of the present disclosure to inhibit MAT2a enzyme.
Experimental materials:
enzyme: MAT2A
HMAT2A:50nM, sipulex (Cepter), 10mg/mL (234. Mu.M), amino acids 1-395 reaction time: 1 hour
L-methionine substrate: alfa elsha (ALFA AESAR)
ATP substrate: alfa elsha
Malachite green detection reagent: milibo Sigma (Millipore Sigma)
Detection buffer: 50mM Hepes,pH 7.5;50mM KCl;10mM MgCl 2; 10mM DTT.
Temperature: 25 DEG C
The steps are as follows:
mu.L of DMSO or 3 Xfinal concentration of test compound in DMSO is transferred to an appropriate well of a microtiter plate, which is centrifuged at 1000rpm for 1 minute. mu.L of 3 Xfinal concentration of MAT2A enzyme in assay buffer or assay buffer alone was transferred to the appropriate wells, which were centrifuged at 1000rpm for 1 min. The well plate was incubated at room temperature for 15 minutes, and then 5. Mu.L of a 3 Xmixture of L-methionine and ATP substrate in detection buffer was transferred to all test wells. The well plate was centrifuged at 1000rpm for 1 minute and then incubated at room temperature for 1 hour. To all test wells, 5. Mu.L of malachite green detection reagent was added and the well plate was centrifuged at 1000rpm for 1min and incubated at room temperature for 30 min. The absorbance of the well plate at 620nm was read on a microplate reader (INFINITE M1000,1000). A high control with high absorbance (DMSO) represents no inhibition of the enzymatic reaction, while a low control with low absorbance (no enzyme) represents complete inhibition of the enzymatic reaction. After the reaction was completed, 10. Mu.L of the kinase detection reagent was added to each well, and after 30 minutes of reaction at 25℃the chemiluminescence was read using a PERKINELMER NIVO-multiple-tag analyzer, and the integration time was 0.5 seconds.
Data analysis:
The raw data is converted to inhibition rate using the equation (Sample-Min)/(Max-Min) ×100%, and the value of IC 50 can be obtained by curve fitting four parameters (log (inhibitor) vs. response-Variable slope mode in GRAPHPAD PRISM).
IC 50 for the compounds in table 1 above is disclosed in table 2 below:
TABLE 2 results of enzymatic Activity assay of Compounds
Numbering of compounds | IC50(nM) | Numbering of compounds | IC50(nM) |
1 | 9323 | 12 | >1000 |
2 | 290.6 | 13 | 50.26 |
3 | 865.5 | 14 | >1000 |
4 | 80 | 15 | 604 |
5 | 77.67 | 16 | 71.7 |
6 | 98 | 17 | 77.67 |
7 | 61.35 | 18 | >1000 |
8 | 89.56 | 19 | 105.9 |
9 | 63.19 | 20 | 68.04 |
10 | 58.91 | 21 | >1000 |
11 | 49.79 | Control compounds | 90.57 |
Experimental results show that the compound has good inhibition activity on MAT2A enzyme and is superior to a control compound.
Test example 2: in vitro cell proliferation assay
Experimental materials:
McCoy's 5A medium, purchased from VivaCell, penicillin/streptomycin antibiotics from source cultures and fetal bovine serum from Gibco. HCT116 MTAP KO cell line was purchased from Horizon. CellTiter-Glo (cell viability chemiluminescent detection reagent) reagent was purchased from Promega.
The experimental method comprises the following steps:
HCT116 MTAP KO cell antiproliferation assay:
Cells were seeded in white 384-well plates, 40 μl of cell suspension containing 500 HCT116 MTAP KO cells per well. Cell plates were placed in a carbon dioxide incubator overnight for culture. The test compounds were diluted 3-fold with a row gun to the 9 th concentration, i.e. from 200. Mu.M to 0.03. Mu.M, and a double multiplex well experiment was set up. 78. Mu.L of medium was added to the intermediate plate, and 2. Mu.L of the gradient diluted compound per well was transferred to the intermediate plate at the corresponding position, and after mixing, 10. Mu.L of the gradient diluted compound per well was transferred to the cell plate. The concentration of compound transferred into the cell plate ranged from 1. Mu.M to 0.15nM. The cell plates were placed in a carbon dioxide incubator for 6 days. A cell plate was also prepared and the signal value read on the day of dosing as the maximum value (Max value in the following equation) was used in the data analysis. To this plate, 10. Mu.L of cell viability chemiluminescent detection reagent was added per well and incubated at room temperature for 10 minutes to stabilize the luminescent signal. Multiple marker analyzer readings were used. After incubation of the cell plates with the compounds, 10 μl of cell viability chemiluminescent detection reagent per well was added to the cell plates and incubated at room temperature for 10 minutes to stabilize the luminescent signal. Multiple marker analyzer readings were used.
Data analysis:
The raw data is converted to inhibition rate using equation (Sample-Min)/(Max-Min) ×100%, and the value of IC 50 can be obtained by curve fitting four parameters (obtained in "log (inhibitor) vs. response-Variable slope" mode in GRAPHPAD PRISM).
IC 50 for some of the compounds in table 1 above is disclosed in table 3 below:
table 3 results of in vitro cell proliferation experiments with Compounds
Test example 3: pharmacokinetic studies in mice
18 Male BALB/c mice were used, the dose was 10mg/kg, the administration route was gastric lavage, the vehicle was 5% DMSO+45% PEG400+50% aqueous solution, and the blood collection time points were 5, 15, 30 minutes and 1,2, 4, 6, 8, 24 hours after administration. After blood sample collection, placing the blood sample in a marked ice bath centrifuge tube, and rapidly centrifuging to separate out plasma, wherein the centrifugation conditions are as follows: 4000 rpm, 10min, 4 deg.C, and plasma at-60 deg.C. After thawing the slurry sample at room temperature, 50. Mu.L of the internal standard (200 ng/mL, acetonitrile, terfenadine) was added, and after vortexing and mixing for 1min, centrifugation was performed for 10min at 4℃under 15400 g. Taking supernatant for LC-MS/MS quantitative analysis, and outputting data such as original map, concentration, accuracy and the like by using analysis 1.6.3 software for main pharmacokinetic parameters.
The pharmacokinetic parameters of compound 8 and the control compound in table 1 above are disclosed in table 4 below:
TABLE 4 essential pharmacokinetic parameters in mice
Experimental results show that the compound of the invention shows excellent pharmacokinetic properties in mice and is superior to control compounds.
The foregoing description is considered as illustrative only of the principles of the disclosure. In addition, since numerous modifications and variations will be readily apparent to those skilled in the art, it is not desired to limit the invention to the exact construction and process shown and described above. Accordingly, all suitable modifications and equivalents may be resorted to as falling within the scope of the invention as defined by the appended claims.
Claims (31)
1. A compound of formula (I) or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof:
wherein:
w is CR 4 or N;
x is CR 5 or N;
y is CR 6 or N;
z is CR 7 or N;
v is CR 8 or N;
R 4、R5、R6、R7 are each independently selected from the group consisting of: hydrogen, alkyl, alkylcarbonyl, hydroxy, halogen, haloalkyl, cyano, alkynyl and-NR aRb;
R 8 is selected from the group consisting of: hydrogen, alkyl, and hydroxy;
R 1 is selected from the group consisting of: alkyl, haloalkyl, hydroxyalkyl, alkoxyalkyl and-NR cRd;
R 2 is selected from the group consisting of: hydrogen, alkyl, alkoxy, heteroalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of alkyl, alkoxy, heteroalkyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl is optionally substituted with one or more R e;
R 3 is selected from the group consisting of: heterocyclyl, aryl, and heteroaryl, wherein each of heterocyclyl, aryl, and heteroaryl is optionally substituted with one or more R e;
R a and R b are each independently selected from hydrogen or alkyl;
r c is selected from the group consisting of: hydrogen, alkyl, and cycloalkyl;
R d is selected from the group consisting of: alkyl, alkoxy, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, cycloalkyl, heterocyclyl, aryl, and heteroaryl, wherein each of alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl, heteroaryl is optionally substituted with one or more R e;
Each R e is independently selected from the group consisting of: oxo, cyano, halogen, hydroxy, acyl, -NR fRg、-OC(O)NRfRg, carbamoyl, carboxy, alkyl, alkenyl, alkynyl, alkoxy, alkoxyalkyl, alkoxycycloalkyl, cycloalkylalkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl;
Each R f is independently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, heteroaryl, wherein each of alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, and heteroaryl is optionally substituted with one or more groups independently selected from cyano, halo, hydroxy, or amino; and
Each R g is independently hydrogen or C 1-3 alkyl.
2. The compound of claim 1, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, having a structure according to formula (Ia) or (Ib) below:
3. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 1 is-NR cRd.
4. The compound of claim 3, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R c is hydrogen or alkyl.
5. The compound of claim 4, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R c is methyl.
6. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R d is selected from the group consisting of: alkyl, alkoxy, and cycloalkyl, wherein each of alkyl, alkoxy, and cycloalkyl is optionally substituted with one or more R e.
7. The compound of claim 6, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein each R e is independently selected from the group consisting of: cyano, hydroxy, -NR fRg, carbamoyl, alkyl, alkenyl, alkoxy, alkoxyalkyl, alkoxycycloalkyl, cycloalkylalkyl, cycloalkyl and heterocyclyl.
8. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R d is selected from the group consisting of: methyl group, And methoxy.
9. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 2 is selected from the group consisting of: hydrogen, alkyl, and aryl.
10. The compound of claim 9, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 2 is methyl.
11. The compound of claim 9, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 2 is phenyl.
12. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 3 is heterocyclyl.
13. The compound of claim 12, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 3 is five membered heterocyclyl.
14. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 3 is selected from the group consisting of
15. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 4 is hydrogen.
16. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 5 is hydrogen.
17. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 6 is selected from the group consisting of: alkylcarbonyl, halogen, haloalkyl, alkynyl and-NR aRb.
18. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 6 is selected from the group consisting of: chlorine, trifluoromethyl,
19. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 7 is hydrogen.
20. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein R 8 is hydrogen.
21. The compound of claim 1 or 2, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, wherein the compound has a formula selected from the group consisting of:
22. A pharmaceutical composition comprising a compound according to any one of claims 1 to 21, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
23. A method of treating a MAT 2A-mediated disease in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound of any one of claims 1 to 21, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 22.
24. The method of claim 23, wherein the disease is cancer.
25. The method of claim 24, wherein the cancer is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
26. A method of treating MTAP-deficient cancer in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound of any one of claims 1 to 21, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 22.
27. The method of claim 26, wherein the cancer is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
28. A method for treating cancer in a patient, wherein the cancer is characterized by reduced or no expression of the MTAP gene, a deletion of the MTAP gene, or reduced function of the MTAP protein, the method comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1 to 21, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 22.
29. The method of claim 28, wherein the cancer is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
30. Use of a compound according to any one of claims 1 to 21, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition according to claim 22, in the manufacture of a medicament for the treatment of cancer.
31. The method of claim 30, wherein the cancer is selected from the group consisting of: leukemia, glioma, melanoma, pancreatic cancer, non-small cell lung cancer, bladder cancer, astrocytoma, osteosarcoma, head and neck cancer, myxoid chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, non-hodgkin's lymphoma, and mesothelioma.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202211569257.XA CN118164966A (en) | 2022-12-08 | 2022-12-08 | MAT2A inhibitor and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202211569257.XA CN118164966A (en) | 2022-12-08 | 2022-12-08 | MAT2A inhibitor and application thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN118164966A true CN118164966A (en) | 2024-06-11 |
Family
ID=91349065
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202211569257.XA Pending CN118164966A (en) | 2022-12-08 | 2022-12-08 | MAT2A inhibitor and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN118164966A (en) |
-
2022
- 2022-12-08 CN CN202211569257.XA patent/CN118164966A/en active Pending
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10370363B2 (en) | Inhibitors of CXCR2 | |
ES2883285T3 (en) | Tlr7 / 8 antagonists and their uses | |
WO2021129824A1 (en) | New-type k-ras g12c inhibitor | |
KR101749192B1 (en) | Triazine, pyrimidine and pyridine analogs and their use as therapeutic agents and diagnostic probes | |
ES2751602T3 (en) | Tricyclic diaza and triaza compounds substituted with (1-fluoro-cyclohex-1-yl) -ethyl as indole-amine-2,3-dioxygenase (ido) antagonists for the treatment of cancer | |
JP2023500395A (en) | Salts and Crystal Forms of PD-1/PD-L1 Inhibitors | |
ES2931316T3 (en) | Salt form for EZH2 inhibition | |
TW202115059A (en) | Salts of a pd-1/pd-l1 inhibitor | |
UA121104C2 (en) | Heterocyclic compounds and uses thereof | |
CA2905993A1 (en) | Substituted 4-amino-pyrimidinyl-2-amino-phenyl derivatives and pharmaceutical compositions thereof for use as jak2 and alk2 inhibitors | |
JP2021524457A (en) | Condensed pyrimidine derivative as an A2A / A2B inhibitor | |
JP2018505166A (en) | Pyridazinone macrocycles and their use as IRAK inhibitors | |
CN105153122A (en) | [(indole-3-yl)pyrimidine-2-yl]aminophenylpropyl-2-eneamide derivative and its salt, preparation method of derivative, and application of derivative and salt | |
JP7384536B2 (en) | Quinazoline compounds and their preparation, use and pharmaceutical compositions | |
UA114894C2 (en) | Inhibitors of nedd8-activating enzyme | |
WO2019158070A1 (en) | A2a and/or a2b receptor antagonist | |
CN110914277A (en) | Imidazo [1,2-b ] pyrimido [4,5-d ] pyridazin-5 (6H) -one compounds and application thereof | |
WO2021252678A1 (en) | Heteroaryl alkylene substituted 2-oxoquinazoline derivatives as methionine adenosyltransferase 2a inhibitors | |
CN104557871B (en) | Arylmorpholine compounds with spiro substituents as well as preparation method and use thereof | |
WO2021252680A1 (en) | 4-arylquinazoline derivatives as methionine adenosyltransferase 2a inhibitors | |
CN104557913B (en) | Pyridopyrimidine compounds as well as preparation method and application thereof | |
TW202216701A (en) | Atr inhibitors and uses thereof | |
AU2018359413B2 (en) | Kinase inhibitors for the treatment of central and peripheral nervous system disorders | |
CN110857304B (en) | Trk inhibitor, preparation method and application thereof | |
CN118164966A (en) | MAT2A inhibitor and application thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |