NZ756084B2 - Sustained release dosage forms for a jak1 inhibitor - Google Patents
Sustained release dosage forms for a jak1 inhibitor Download PDFInfo
- Publication number
- NZ756084B2 NZ756084B2 NZ756084A NZ75608414A NZ756084B2 NZ 756084 B2 NZ756084 B2 NZ 756084B2 NZ 756084 A NZ756084 A NZ 756084A NZ 75608414 A NZ75608414 A NZ 75608414A NZ 756084 B2 NZ756084 B2 NZ 756084B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- pyrrolo
- sustained release
- isonicotinoyl
- acetonitrile
- fluoro
- Prior art date
Links
- 230000002459 sustained Effects 0.000 title claims abstract description 159
- 239000002552 dosage form Substances 0.000 title description 231
- 230000002401 inhibitory effect Effects 0.000 title description 53
- 239000003112 inhibitor Substances 0.000 title description 26
- 101700034277 JAK1 Proteins 0.000 title description 4
- 239000011780 sodium chloride Substances 0.000 claims abstract description 101
- 150000003839 salts Chemical class 0.000 claims abstract description 100
- 239000000203 mixture Substances 0.000 claims abstract description 93
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 claims abstract description 39
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 claims abstract description 39
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims abstract description 23
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 claims abstract description 22
- 229960003943 hypromellose Drugs 0.000 claims abstract description 20
- 229920000168 Microcrystalline cellulose Polymers 0.000 claims abstract description 17
- 235000019813 microcrystalline cellulose Nutrition 0.000 claims abstract description 17
- 239000008108 microcrystalline cellulose Substances 0.000 claims abstract description 13
- 229940016286 microcrystalline cellulose Drugs 0.000 claims abstract description 13
- WSVLPVUVIUVCRA-RJMJUYIDSA-N (2R,3R,4S,5R,6S)-2-(hydroxymethyl)-6-[(2R,3S,4R,5R)-4,5,6-trihydroxy-2-(hydroxymethyl)oxan-3-yl]oxyoxane-3,4,5-triol;hydrate Chemical compound O.O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)OC(O)[C@H](O)[C@H]1O WSVLPVUVIUVCRA-RJMJUYIDSA-N 0.000 claims abstract description 12
- 229960001021 Lactose Monohydrate Drugs 0.000 claims abstract description 12
- 239000012458 free base Substances 0.000 claims description 73
- 235000012054 meals Nutrition 0.000 claims description 59
- 230000036470 plasma concentration Effects 0.000 claims description 55
- 230000037242 Cmax Effects 0.000 claims description 49
- -1 {1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H- pyrrolo[2,3-d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile Chemical compound 0.000 claims description 26
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 claims description 22
- 235000019359 magnesium stearate Nutrition 0.000 claims description 11
- KTBSXLIQKWEBRB-UHFFFAOYSA-N 2-[1-[1-[3-fluoro-2-(trifluoromethyl)pyridine-4-carbonyl]piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]azetidin-3-yl]acetonitrile Chemical compound C1=CN=C(C(F)(F)F)C(F)=C1C(=O)N1CCC(N2CC(CC#N)(C2)N2N=CC(=C2)C=2C=3C=CNC=3N=CN=2)CC1 KTBSXLIQKWEBRB-UHFFFAOYSA-N 0.000 abstract 1
- WEVYAHXRMPXWCK-UHFFFAOYSA-N acetonitrile Substances CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 99
- 150000001875 compounds Chemical class 0.000 description 82
- 125000004567 azetidin-3-yl group Chemical group N1CC(C1)* 0.000 description 74
- 201000010099 disease Diseases 0.000 description 65
- 125000003386 piperidinyl group Chemical group 0.000 description 64
- 230000035492 administration Effects 0.000 description 57
- 239000003826 tablet Substances 0.000 description 55
- 125000002023 trifluoromethyl group Chemical group FC(F)(F)* 0.000 description 44
- 201000011510 cancer Diseases 0.000 description 35
- 230000000694 effects Effects 0.000 description 33
- 125000004482 piperidin-4-yl group Chemical group N1CCC(CC1)* 0.000 description 31
- 239000003814 drug Substances 0.000 description 25
- 210000004027 cells Anatomy 0.000 description 24
- 208000003476 Primary Myelofibrosis Diseases 0.000 description 21
- 239000003795 chemical substances by application Substances 0.000 description 21
- 206010028537 Myelofibrosis Diseases 0.000 description 19
- 200000000018 inflammatory disease Diseases 0.000 description 19
- 230000035852 Tmax Effects 0.000 description 17
- 210000000056 organs Anatomy 0.000 description 17
- 206010013774 Dry eye Diseases 0.000 description 16
- 210000002381 Plasma Anatomy 0.000 description 16
- 201000004681 psoriasis Diseases 0.000 description 16
- 206010003816 Autoimmune disease Diseases 0.000 description 15
- 206010028576 Myeloproliferative disease Diseases 0.000 description 15
- 230000035489 relative bioavailability Effects 0.000 description 15
- 206010052779 Transplant rejections Diseases 0.000 description 14
- 125000002393 azetidinyl group Chemical group 0.000 description 14
- 235000013305 food Nutrition 0.000 description 14
- 230000002829 reduced Effects 0.000 description 14
- 102100019516 JAK2 Human genes 0.000 description 13
- 101700016050 JAK2 Proteins 0.000 description 13
- 239000002775 capsule Substances 0.000 description 13
- 239000008187 granular material Substances 0.000 description 13
- 150000001278 adipic acid derivatives Chemical class 0.000 description 12
- 201000009596 autoimmune hypersensitivity disease Diseases 0.000 description 12
- 238000009472 formulation Methods 0.000 description 12
- 208000007502 Anemia Diseases 0.000 description 11
- 235000019886 MethocelTM Nutrition 0.000 description 11
- 201000010816 bone resorption disease Diseases 0.000 description 11
- 230000002757 inflammatory Effects 0.000 description 11
- 102000004127 Cytokines Human genes 0.000 description 10
- 108090000695 Cytokines Proteins 0.000 description 10
- 102000001253 Protein Kinases Human genes 0.000 description 10
- 102100019388 SOAT1 Human genes 0.000 description 10
- 101700025022 SOAT1 Proteins 0.000 description 10
- 101710028406 satA Proteins 0.000 description 10
- 102000001554 Hemoglobins Human genes 0.000 description 9
- 108010054147 Hemoglobins Proteins 0.000 description 9
- 230000011664 signaling Effects 0.000 description 9
- 230000000699 topical Effects 0.000 description 9
- 206010003246 Arthritis Diseases 0.000 description 8
- 229960003957 Dexamethasone Drugs 0.000 description 8
- UREBDLICKHMUKA-CXSFZGCWSA-N Dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 8
- 208000002047 Essential Thrombocythemia Diseases 0.000 description 8
- 230000037361 pathway Effects 0.000 description 8
- 230000004044 response Effects 0.000 description 8
- 230000001225 therapeutic Effects 0.000 description 8
- 230000036912 Bioavailability Effects 0.000 description 7
- 208000006386 Bone Resorption Diseases 0.000 description 7
- 230000036499 Half live Effects 0.000 description 7
- 206010061218 Inflammation Diseases 0.000 description 7
- 206010028980 Neoplasm Diseases 0.000 description 7
- 108091000081 Phosphotransferases Proteins 0.000 description 7
- 206010035226 Plasma cell myeloma Diseases 0.000 description 7
- 208000008696 Polycythemia Vera Diseases 0.000 description 7
- 241000283984 Rodentia Species 0.000 description 7
- 230000001363 autoimmune Effects 0.000 description 7
- 230000035514 bioavailability Effects 0.000 description 7
- 230000024279 bone resorption Effects 0.000 description 7
- 230000001965 increased Effects 0.000 description 7
- 230000004054 inflammatory process Effects 0.000 description 7
- 150000002500 ions Chemical class 0.000 description 7
- 239000000463 material Substances 0.000 description 7
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 7
- 239000008213 purified water Substances 0.000 description 7
- 102000015617 Janus Kinases Human genes 0.000 description 6
- 108010024121 Janus Kinases Proteins 0.000 description 6
- 102100017873 PTK2 Human genes 0.000 description 6
- 206010039073 Rheumatoid arthritis Diseases 0.000 description 6
- 210000001744 T-Lymphocytes Anatomy 0.000 description 6
- 206010046851 Uveitis Diseases 0.000 description 6
- 238000004166 bioassay Methods 0.000 description 6
- 230000002354 daily Effects 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- NQRYJNQNLNOLGT-UHFFFAOYSA-N piperidine Chemical compound C1CCNCC1 NQRYJNQNLNOLGT-UHFFFAOYSA-N 0.000 description 6
- 239000000902 placebo Substances 0.000 description 6
- 229940068196 placebo Drugs 0.000 description 6
- 230000035755 proliferation Effects 0.000 description 6
- 206010012442 Dermatitis contact Diseases 0.000 description 5
- 210000001508 Eye Anatomy 0.000 description 5
- 206010020751 Hypersensitivity Diseases 0.000 description 5
- 101700007593 JAK3 Proteins 0.000 description 5
- 102100019518 JAK3 Human genes 0.000 description 5
- 230000036917 MEAN CMAX Effects 0.000 description 5
- 229920000881 Modified starch Polymers 0.000 description 5
- UIIMBOGNXHQVGW-UHFFFAOYSA-M NaHCO3 Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 5
- 241000700159 Rattus Species 0.000 description 5
- 210000003491 Skin Anatomy 0.000 description 5
- 210000001138 Tears Anatomy 0.000 description 5
- 238000010521 absorption reaction Methods 0.000 description 5
- 230000004913 activation Effects 0.000 description 5
- WNLRTRBMVRJNCN-UHFFFAOYSA-L adipate(2-) Chemical class [O-]C(=O)CCCCC([O-])=O WNLRTRBMVRJNCN-UHFFFAOYSA-L 0.000 description 5
- 235000010980 cellulose Nutrition 0.000 description 5
- 229920002678 cellulose Polymers 0.000 description 5
- 230000001419 dependent Effects 0.000 description 5
- 231100000080 dermatitis contact Toxicity 0.000 description 5
- 229940079593 drugs Drugs 0.000 description 5
- 230000014509 gene expression Effects 0.000 description 5
- 238000005469 granulation Methods 0.000 description 5
- 230000003179 granulation Effects 0.000 description 5
- 239000003102 growth factor Substances 0.000 description 5
- 230000000051 modifying Effects 0.000 description 5
- 201000009251 multiple myeloma Diseases 0.000 description 5
- 239000008177 pharmaceutical agent Substances 0.000 description 5
- 210000002345 respiratory system Anatomy 0.000 description 5
- 230000001629 suppression Effects 0.000 description 5
- UCSJYZPVAKXKNQ-HZYVHMACSA-N 1-[(1S,2R,3R,4S,5R,6R)-3-carbamimidamido-6-{[(2R,3R,4R,5S)-3-{[(2S,3S,4S,5R,6S)-4,5-dihydroxy-6-(hydroxymethyl)-3-(methylamino)oxan-2-yl]oxy}-4-formyl-4-hydroxy-5-methyloxolan-2-yl]oxy}-2,4,5-trihydroxycyclohexyl]guanidine 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 4
- ZKHQWZAMYRWXGA-KQYNXXCUSA-N Adenosine triphosphate Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](COP(O)(=O)OP(O)(=O)OP(O)(O)=O)[C@@H](O)[C@H]1O ZKHQWZAMYRWXGA-KQYNXXCUSA-N 0.000 description 4
- 208000006673 Asthma Diseases 0.000 description 4
- 206010010741 Conjunctivitis Diseases 0.000 description 4
- 208000010247 Contact Dermatitis Diseases 0.000 description 4
- 101710009074 FLT3 Proteins 0.000 description 4
- 206010021972 Inflammatory bowel disease Diseases 0.000 description 4
- 241001465754 Metazoa Species 0.000 description 4
- 208000009525 Myocarditis Diseases 0.000 description 4
- 206010070834 Sensitisation Diseases 0.000 description 4
- QIQXTHQIDYTFRH-UHFFFAOYSA-N Stearic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 4
- 206010043554 Thrombocytopenia Diseases 0.000 description 4
- 239000002253 acid Substances 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 239000000427 antigen Substances 0.000 description 4
- 108091007172 antigens Proteins 0.000 description 4
- 102000038129 antigens Human genes 0.000 description 4
- 239000001913 cellulose Substances 0.000 description 4
- 230000001684 chronic Effects 0.000 description 4
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 4
- 238000002156 mixing Methods 0.000 description 4
- 230000000275 pharmacokinetic Effects 0.000 description 4
- 102000004169 proteins and genes Human genes 0.000 description 4
- 108090000623 proteins and genes Proteins 0.000 description 4
- 230000008313 sensitization Effects 0.000 description 4
- 230000001235 sensitizing Effects 0.000 description 4
- 231100000486 side effect Toxicity 0.000 description 4
- 230000004083 survival Effects 0.000 description 4
- 229960001350 tofacitinib Drugs 0.000 description 4
- UJLAWZDWDVHWOW-YPMHNXCESA-N tofacitinib Chemical compound C[C@@H]1CCN(C(=O)CC#N)C[C@@H]1N(C)C1=NC=NC2=C1C=CN2 UJLAWZDWDVHWOW-YPMHNXCESA-N 0.000 description 4
- PMATZTZNYRCHOR-CGLBZJNRSA-N (3S,6S,9S,12R,15S,18S,21S,24S,30S,33S)-30-ethyl-33-[(E,1R,2R)-1-hydroxy-2-methylhex-4-enyl]-1,4,7,10,12,15,19,25,28-nonamethyl-6,9,18,24-tetrakis(2-methylpropyl)-3,21-di(propan-2-yl)-1,4,7,10,13,16,19,22,25,28,31-undecazacyclotritriacontane-2,5,8,11,14,17 Chemical group CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 3
- LOTKRQAVGJMPNV-UHFFFAOYSA-N 1-Fluoro-2,4-dinitrobenzene Chemical compound [O-][N+](=O)C1=CC=C(F)C([N+]([O-])=O)=C1 LOTKRQAVGJMPNV-UHFFFAOYSA-N 0.000 description 3
- CZPWVGJYEJSRLH-UHFFFAOYSA-N 289-95-2 Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 3
- 101700006234 AKT1 Proteins 0.000 description 3
- 230000035533 AUC Effects 0.000 description 3
- HONIICLYMWZJFZ-UHFFFAOYSA-N Azetidine Chemical compound C1CNC1 HONIICLYMWZJFZ-UHFFFAOYSA-N 0.000 description 3
- 210000001772 Blood Platelets Anatomy 0.000 description 3
- 241000700199 Cavia porcellus Species 0.000 description 3
- 108010036949 Cyclosporine Proteins 0.000 description 3
- 208000010201 Exanthema Diseases 0.000 description 3
- 102100019517 JAK1 Human genes 0.000 description 3
- 102000008986 Janus Human genes 0.000 description 3
- 108050000950 Janus Proteins 0.000 description 3
- 239000002144 L01XE18 - Ruxolitinib Substances 0.000 description 3
- 229960001375 Lactose Drugs 0.000 description 3
- GUBGYTABKSRVRQ-UUNJERMWSA-N Lactose Natural products O([C@@H]1[C@H](O)[C@H](O)[C@H](O)O[C@@H]1CO)[C@H]1[C@@H](O)[C@@H](O)[C@H](O)[C@H](CO)O1 GUBGYTABKSRVRQ-UUNJERMWSA-N 0.000 description 3
- 206010025135 Lupus erythematosus Diseases 0.000 description 3
- 229940057948 Magnesium stearate Drugs 0.000 description 3
- 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 3
- 229920003091 Methocel™ Polymers 0.000 description 3
- 229920003094 Methocel™ K4M Polymers 0.000 description 3
- 210000003819 Peripheral blood mononuclear cell Anatomy 0.000 description 3
- 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 3
- 108060006633 Protein Kinases Proteins 0.000 description 3
- 206010037844 Rash Diseases 0.000 description 3
- 206010039083 Rhinitis Diseases 0.000 description 3
- 208000006641 Skin Disease Diseases 0.000 description 3
- 229920002397 Thermoplastic olefin Polymers 0.000 description 3
- 239000004012 Tofacitinib Substances 0.000 description 3
- 102100013671 ZHX2 Human genes 0.000 description 3
- XEKOWRVHYACXOJ-UHFFFAOYSA-N acetic acid ethyl ester Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 3
- 108090001123 antibodies Proteins 0.000 description 3
- 102000004965 antibodies Human genes 0.000 description 3
- 201000008937 atopic dermatitis Diseases 0.000 description 3
- 239000002585 base Substances 0.000 description 3
- 230000037182 bone density Effects 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 230000000973 chemotherapeutic Effects 0.000 description 3
- 239000003246 corticosteroid Substances 0.000 description 3
- 230000003247 decreasing Effects 0.000 description 3
- 201000004624 dermatitis Diseases 0.000 description 3
- 238000002868 homogeneous time resolved fluorescence Methods 0.000 description 3
- 230000028709 inflammatory response Effects 0.000 description 3
- 201000004614 iritis Diseases 0.000 description 3
- 239000008101 lactose Substances 0.000 description 3
- GUBGYTABKSRVRQ-XLOQQCSPSA-N 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 3
- 239000007788 liquid Substances 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 229960001924 melphalan Drugs 0.000 description 3
- 238000000034 method Methods 0.000 description 3
- 201000005962 mycosis fungoide Diseases 0.000 description 3
- 230000002071 myeloproliferative Effects 0.000 description 3
- 201000008482 osteoarthritis Diseases 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 238000003908 quality control method Methods 0.000 description 3
- HFNKQEVNSGCOJV-OAHLLOKOSA-N ruxolitinib Chemical compound C1([C@@H](CC#N)N2N=CC(=C2)C=2C=3C=CNC=3N=CN=2)CCCC1 HFNKQEVNSGCOJV-OAHLLOKOSA-N 0.000 description 3
- 229960000215 ruxolitinib Drugs 0.000 description 3
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- FFTVPQUHLQBXQZ-KVUCHLLUSA-N (4S,4aS,5aR,12aR)-4,7-bis(dimethylamino)-1,10,11,12a-tetrahydroxy-3,12-dioxo-4a,5,5a,6-tetrahydro-4H-tetracene-2-carboxamide Chemical compound C1C2=C(N(C)C)C=CC(O)=C2C(O)=C2[C@@H]1C[C@H]1[C@H](N(C)C)C(=O)C(C(N)=O)=C(O)[C@@]1(O)C2=O FFTVPQUHLQBXQZ-KVUCHLLUSA-N 0.000 description 2
- JSFATNQSLKRBCI-VAEKSGALSA-N 15-Hydroxyicosatetraenoic acid Chemical compound CCCCC[C@H](O)\C=C\C=C/C\C=C/C\C=C/CCCC(O)=O JSFATNQSLKRBCI-VAEKSGALSA-N 0.000 description 2
- VUKAUDKDFVSVFT-UHFFFAOYSA-N 2-[6-[4,5-bis(2-hydroxypropoxy)-2-(2-hydroxypropoxymethyl)-6-methoxyoxan-3-yl]oxy-4,5-dimethoxy-2-(methoxymethyl)oxan-3-yl]oxy-6-(hydroxymethyl)-5-methoxyoxane-3,4-diol Chemical compound COC1C(OC)C(OC2C(C(O)C(OC)C(CO)O2)O)C(COC)OC1OC1C(COCC(C)O)OC(OC)C(OCC(C)O)C1OCC(C)O VUKAUDKDFVSVFT-UHFFFAOYSA-N 0.000 description 2
- ZEYUSQVGRCPBPG-UHFFFAOYSA-N 4,5-dihydroxy-1,3-bis(hydroxymethyl)imidazolidin-2-one Chemical compound OCN1C(O)C(O)N(CO)C1=O ZEYUSQVGRCPBPG-UHFFFAOYSA-N 0.000 description 2
- AOJJSUZBOXZQNB-TZSSRYMLSA-N ADRIAMYCIN 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 2
- 206010000880 Acute myeloid leukaemia Diseases 0.000 description 2
- 206010004446 Benign prostatic hyperplasia Diseases 0.000 description 2
- 210000004369 Blood Anatomy 0.000 description 2
- 210000000988 Bone and Bones Anatomy 0.000 description 2
- 206010006451 Bronchitis Diseases 0.000 description 2
- 208000000594 Bullous Pemphigoid Diseases 0.000 description 2
- 229940095259 Butylated Hydroxytoluene Drugs 0.000 description 2
- 239000004322 Butylated hydroxytoluene Substances 0.000 description 2
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 2
- 206010006895 Cachexia Diseases 0.000 description 2
- 210000000845 Cartilage Anatomy 0.000 description 2
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 description 2
- 230000037250 Clearance Effects 0.000 description 2
- 206010009900 Colitis ulcerative Diseases 0.000 description 2
- 102000008186 Collagen Human genes 0.000 description 2
- 108010035532 Collagen Proteins 0.000 description 2
- 206010010099 Combined immunodeficiency Diseases 0.000 description 2
- 108010062580 Concanavalin A Proteins 0.000 description 2
- 206010011401 Crohn's disease Diseases 0.000 description 2
- 206010011715 Cyclitis Diseases 0.000 description 2
- 229940119017 Cyclosporine Drugs 0.000 description 2
- 208000006313 Delayed Hypersensitivity Diseases 0.000 description 2
- 206010012438 Dermatitis atopic Diseases 0.000 description 2
- 229960004679 Doxorubicin Drugs 0.000 description 2
- 102100016635 EPOR Human genes 0.000 description 2
- 108091007936 ERK family Proteins 0.000 description 2
- 206010015084 Episcleritis Diseases 0.000 description 2
- 229960001433 Erlotinib Drugs 0.000 description 2
- AAKJLRGGTJKAMG-UHFFFAOYSA-N Erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 2
- 108010075944 Erythropoietin Receptors Proteins 0.000 description 2
- 206010016256 Fatigue Diseases 0.000 description 2
- FEBLZLNTKCEFIT-VSXGLTOVSA-N Fluocinolone acetonide Chemical group C1([C@@H](F)C2)=CC(=O)C=C[C@]1(C)[C@]1(F)[C@@H]2[C@@H]2C[C@H]3OC(C)(C)O[C@@]3(C(=O)CO)[C@@]2(C)C[C@@H]1O FEBLZLNTKCEFIT-VSXGLTOVSA-N 0.000 description 2
- 108010091824 Focal Adhesion Kinase 1 Proteins 0.000 description 2
- 206010017758 Gastric cancer Diseases 0.000 description 2
- 206010051066 Gastrointestinal stromal tumour Diseases 0.000 description 2
- 210000004907 Glands Anatomy 0.000 description 2
- 241000701085 Human alphaherpesvirus 3 Species 0.000 description 2
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 2
- 241000701806 Human papillomavirus Species 0.000 description 2
- 206010048643 Hypereosinophilic syndrome Diseases 0.000 description 2
- 230000036938 INCREASE IN AUC Effects 0.000 description 2
- KTUFNOKKBVMGRW-UHFFFAOYSA-N Imatinib Chemical compound 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 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 description 2
- 210000000987 Immune System Anatomy 0.000 description 2
- 206010027665 Immune disorder Diseases 0.000 description 2
- 206010021425 Immune system disease Diseases 0.000 description 2
- 108060003951 Immunoglobulins Proteins 0.000 description 2
- 102000018358 Immunoglobulins Human genes 0.000 description 2
- 102000000588 Interleukin-2 Human genes 0.000 description 2
- 108010002350 Interleukin-2 Proteins 0.000 description 2
- 210000002510 Keratinocytes Anatomy 0.000 description 2
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 2
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 2
- 210000004561 Lacrimal Apparatus Anatomy 0.000 description 2
- 206010024324 Leukaemias Diseases 0.000 description 2
- 208000008456 Leukemia, Myelogenous, Chronic, BCR-ABL Positive Diseases 0.000 description 2
- 208000007046 Leukemia, Myeloid, Acute Diseases 0.000 description 2
- 208000000214 Leukemia, Myelomonocytic, Chronic Diseases 0.000 description 2
- 208000003543 Lymphoma, T-Cell, Cutaneous Diseases 0.000 description 2
- 210000003593 Megakaryocytes Anatomy 0.000 description 2
- 229960004023 Minocycline Drugs 0.000 description 2
- 206010028417 Myasthenia gravis Diseases 0.000 description 2
- 206010029149 Nephropathy Diseases 0.000 description 2
- 210000001331 Nose Anatomy 0.000 description 2
- 206010049088 Osteopenia Diseases 0.000 description 2
- 208000001132 Osteoporosis Diseases 0.000 description 2
- NMLMACJWHPHKGR-NCOIDOBVSA-N P(1),P(4)-bis(uridin-5'-yl) tetraphosphate Chemical compound N1([C@@H]2O[C@@H]([C@H]([C@H]2O)O)COP(O)(=O)OP(O)(=O)OP(O)(=O)OP(O)(=O)OC[C@@H]2[C@H]([C@H]([C@@H](O2)N2C(NC(=O)C=C2)=O)O)O)C=CC(=O)NC1=O NMLMACJWHPHKGR-NCOIDOBVSA-N 0.000 description 2
- 241000282322 Panthera Species 0.000 description 2
- 206010034277 Pemphigoid Diseases 0.000 description 2
- 241000233805 Phoenix Species 0.000 description 2
- 241001463913 Pinna Species 0.000 description 2
- 239000004372 Polyvinyl alcohol Substances 0.000 description 2
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 2
- 210000002307 Prostate Anatomy 0.000 description 2
- 239000007759 RPMI Media 1640 Substances 0.000 description 2
- QTTRZHGPGKRAFB-OOKHYKNYSA-N Rimexolone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CC)(C)[C@@]1(C)C[C@@H]2O QTTRZHGPGKRAFB-OOKHYKNYSA-N 0.000 description 2
- 206010039705 Scleritis Diseases 0.000 description 2
- 208000002491 Severe Combined Immunodeficiency Diseases 0.000 description 2
- 206010040767 Sjogren's syndrome Diseases 0.000 description 2
- 206010070835 Skin sensitisation Diseases 0.000 description 2
- 235000021355 Stearic acid Nutrition 0.000 description 2
- 229960005322 Streptomycin Drugs 0.000 description 2
- 229960004247 TOFACITINIB CITRATE Drugs 0.000 description 2
- 229940033134 Talc Drugs 0.000 description 2
- 229960003433 Thalidomide Drugs 0.000 description 2
- UEJJHQNACJXSKW-UHFFFAOYSA-N Thalidomide Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 2
- 208000005485 Thrombocytosis Diseases 0.000 description 2
- 108090000848 Ubiquitin Proteins 0.000 description 2
- 102400000757 Ubiquitin Human genes 0.000 description 2
- 229960004528 Vincristine Drugs 0.000 description 2
- 208000001756 Virus Disease Diseases 0.000 description 2
- 230000002378 acidificating Effects 0.000 description 2
- 201000005510 acute lymphocytic leukemia Diseases 0.000 description 2
- 230000002730 additional Effects 0.000 description 2
- 230000001058 adult Effects 0.000 description 2
- 201000005794 allergic hypersensitivity disease Diseases 0.000 description 2
- 230000003042 antagnostic Effects 0.000 description 2
- 230000003110 anti-inflammatory Effects 0.000 description 2
- 230000000259 anti-tumor Effects 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 239000000607 artificial tear Substances 0.000 description 2
- 230000027455 binding Effects 0.000 description 2
- 230000003115 biocidal Effects 0.000 description 2
- 230000015572 biosynthetic process Effects 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 230000010072 bone remodeling Effects 0.000 description 2
- 235000010354 butylated hydroxytoluene Nutrition 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 230000001413 cellular Effects 0.000 description 2
- 201000006934 chronic myeloid leukemia Diseases 0.000 description 2
- 201000010902 chronic myelomonocytic leukemia Diseases 0.000 description 2
- 229960001265 ciclosporin Drugs 0.000 description 2
- 230000035512 clearance Effects 0.000 description 2
- 239000011248 coating agent Substances 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 229920001436 collagen Polymers 0.000 description 2
- 229960005188 collagen Drugs 0.000 description 2
- 229940075614 colloidal silicon dioxide Drugs 0.000 description 2
- 201000007241 cutaneous T cell lymphoma Diseases 0.000 description 2
- PMATZTZNYRCHOR-UHFFFAOYSA-N cyclosporine A Chemical compound CCC1NC(=O)C(C(O)C(C)CC=CC)N(C)C(=O)C(C(C)C)N(C)C(=O)C(CC(C)C)N(C)C(=O)C(CC(C)C)N(C)C(=O)C(C)NC(=O)C(C)NC(=O)C(CC(C)C)N(C)C(=O)C(C(C)C)NC(=O)C(CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-UHFFFAOYSA-N 0.000 description 2
- 231100000406 dermatitis Toxicity 0.000 description 2
- 231100001000 dermatological side-effect Toxicity 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- RTZKZFJDLAIYFH-UHFFFAOYSA-N diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 2
- 229950003529 diquafosol Drugs 0.000 description 2
- 230000004064 dysfunction Effects 0.000 description 2
- 239000002158 endotoxin Substances 0.000 description 2
- 235000020828 fasting Nutrition 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 239000007888 film coating Substances 0.000 description 2
- 238000009501 film coating Methods 0.000 description 2
- 238000005755 formation reaction Methods 0.000 description 2
- 201000011243 gastrointestinal stromal tumor Diseases 0.000 description 2
- 201000005569 gout Diseases 0.000 description 2
- 239000001963 growth media Substances 0.000 description 2
- 230000002489 hematologic Effects 0.000 description 2
- 229960002411 imatinib Drugs 0.000 description 2
- 230000003053 immunization Effects 0.000 description 2
- 238000002649 immunization Methods 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 239000000314 lubricant Substances 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 230000001404 mediated Effects 0.000 description 2
- OKKJLVBELUTLKV-UHFFFAOYSA-N methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 2
- 150000007522 mineralic acids Chemical class 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 201000006417 multiple sclerosis Diseases 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 201000000050 myeloid neoplasm Diseases 0.000 description 2
- LRHPLDYGYMQRHN-UHFFFAOYSA-N n-butanol Chemical compound CCCCO LRHPLDYGYMQRHN-UHFFFAOYSA-N 0.000 description 2
- 230000003000 nontoxic Effects 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 230000036961 partial Effects 0.000 description 2
- 230000008506 pathogenesis Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 230000000865 phosphorylative Effects 0.000 description 2
- 230000001885 phytohemagglutinin Effects 0.000 description 2
- 229920003253 poly(benzobisoxazole) Polymers 0.000 description 2
- 229920002451 polyvinyl alcohol Polymers 0.000 description 2
- 235000019422 polyvinyl alcohol Nutrition 0.000 description 2
- 229960004618 prednisone Drugs 0.000 description 2
- 201000000306 sarcoidosis Diseases 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 201000009890 sinusitis Diseases 0.000 description 2
- 231100000046 skin rash Toxicity 0.000 description 2
- 231100000370 skin sensitisation Toxicity 0.000 description 2
- 235000017557 sodium bicarbonate Nutrition 0.000 description 2
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 239000008117 stearic acid Substances 0.000 description 2
- 150000003431 steroids Chemical class 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 201000011549 stomach cancer Diseases 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 239000007939 sustained release tablet Substances 0.000 description 2
- 239000000454 talc Substances 0.000 description 2
- 229910052623 talc Inorganic materials 0.000 description 2
- 230000017423 tissue regeneration Effects 0.000 description 2
- GWEVSGVZZGPLCZ-UHFFFAOYSA-N titan oxide Chemical compound O=[Ti]=O GWEVSGVZZGPLCZ-UHFFFAOYSA-N 0.000 description 2
- 230000002103 transcriptional Effects 0.000 description 2
- 201000006704 ulcerative colitis Diseases 0.000 description 2
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 2
- 238000005550 wet granulation Methods 0.000 description 2
- ZROHGHOFXNOHSO-BNTLRKBRSA-L (1R,2R)-cyclohexane-1,2-diamine;oxalate;platinum(2+) Chemical compound [H][N]([C@@H]1CCCC[C@H]1[N]1([H])[H])([H])[Pt]11OC(=O)C(=O)O1 ZROHGHOFXNOHSO-BNTLRKBRSA-L 0.000 description 1
- GXMBHQRROXQUJS-UHFFFAOYSA-N (2-hept-2-ynylsulfanylphenyl) acetate Chemical compound CCCCC#CCSC1=CC=CC=C1OC(C)=O GXMBHQRROXQUJS-UHFFFAOYSA-N 0.000 description 1
- PUSNGFYSTWMJSK-GSZQVNRLSA-N (2R,3R,4S,5R,6R)-2,3,4-trimethoxy-6-(methoxymethyl)-5-[(2S,3R,4S,5R,6R)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxyoxane;1-[[(2R,3R,4S,5R,6S)-3,4,5-tris(2-hydroxypropoxy)-6-[(2R,3R,4S,5R,6R)-4,5,6-tris(2-hydroxypropoxy)-2-(2-hydroxypropoxymethyl)oxan- 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](OC)O[C@@H]1COC.CC(O)CO[C@@H]1[C@@H](OCC(C)O)[C@H](OCC(C)O)[C@@H](COCC(O)C)O[C@H]1O[C@H]1[C@H](OCC(C)O)[C@@H](OCC(C)O)[C@H](OCC(C)O)O[C@@H]1COCC(C)O PUSNGFYSTWMJSK-GSZQVNRLSA-N 0.000 description 1
- HMLGSIZOMSVISS-ONJSNURVSA-N (7R)-7-[[(2Z)-2-(2-amino-1,3-thiazol-4-yl)-2-(2,2-dimethylpropanoyloxymethoxyimino)acetyl]amino]-3-ethenyl-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylic acid Chemical compound N([C@@H]1C(N2C(=C(C=C)CSC21)C(O)=O)=O)C(=O)\C(=N/OCOC(=O)C(C)(C)C)C1=CSC(N)=N1 HMLGSIZOMSVISS-ONJSNURVSA-N 0.000 description 1
- FLBAYUMRQUHISI-UHFFFAOYSA-N 1,8-naphthyridine Chemical compound N1=CC=CC2=CC=CN=C21 FLBAYUMRQUHISI-UHFFFAOYSA-N 0.000 description 1
- CNIIGCLFLJGOGP-UHFFFAOYSA-N 2-(naphthalen-1-ylmethyl)-4,5-dihydro-1H-imidazole Chemical compound C=1C=CC2=CC=CC=C2C=1CC1=NCCN1 CNIIGCLFLJGOGP-UHFFFAOYSA-N 0.000 description 1
- 125000006305 3-iodophenyl group Chemical group [H]C1=C([H])C(I)=C([H])C(*)=C1[H] 0.000 description 1
- HPLNQCPCUACXLM-PGUFJCEWSA-N 4-[4-[[2-(4-chlorophenyl)phenyl]methyl]piperazin-1-yl]-N-[4-[[(2R)-4-(dimethylamino)-1-phenylsulfanylbutan-2-yl]amino]-3-nitrophenyl]sulfonylbenzamide Chemical compound C([C@@H](CCN(C)C)NC=1C(=CC(=CC=1)S(=O)(=O)NC(=O)C=1C=CC(=CC=1)N1CCN(CC=2C(=CC=CC=2)C=2C=CC(Cl)=CC=2)CC1)[N+]([O-])=O)SC1=CC=CC=C1 HPLNQCPCUACXLM-PGUFJCEWSA-N 0.000 description 1
- GHASVSINZRGABV-UHFFFAOYSA-N 5-flurouricil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 1
- 229930000680 A04AD01 - Scopolamine Natural products 0.000 description 1
- 229960004308 ACETYLCYSTEINE Drugs 0.000 description 1
- 108060005293 AGA Proteins 0.000 description 1
- 229940116904 ANTIINFLAMMATORY THERAPEUTIC RADIOPHARMACEUTICALS Drugs 0.000 description 1
- 102100004323 ASPG Human genes 0.000 description 1
- 230000036680 AUC0-t Effects 0.000 description 1
- 210000001015 Abdomen Anatomy 0.000 description 1
- 208000009956 Adenocarcinoma Diseases 0.000 description 1
- 208000002029 Allergic Contact Dermatitis Diseases 0.000 description 1
- 206010027654 Allergic conditions Diseases 0.000 description 1
- 108009000283 Allograft Rejection Proteins 0.000 description 1
- 206010001897 Alzheimer's disease Diseases 0.000 description 1
- 240000006249 Ambrosia artemisiifolia Species 0.000 description 1
- 235000003129 Ambrosia artemisiifolia var elatior Nutrition 0.000 description 1
- 206010002556 Ankylosing spondylitis Diseases 0.000 description 1
- REYFJDPCWQRWAA-UHFFFAOYSA-N Antazoline Chemical compound N=1CCNC=1CN(C=1C=CC=CC=1)CC1=CC=CC=C1 REYFJDPCWQRWAA-UHFFFAOYSA-N 0.000 description 1
- 206010003267 Arthritis reactive Diseases 0.000 description 1
- 229960002756 Azacitidine Drugs 0.000 description 1
- 210000003719 B-Lymphocytes Anatomy 0.000 description 1
- 229960003071 Bacitracin Drugs 0.000 description 1
- 108010001478 Bacitracin Proteins 0.000 description 1
- 208000010217 Blepharitis Diseases 0.000 description 1
- 210000001185 Bone Marrow Anatomy 0.000 description 1
- GXJABQQUPOEUTA-RDJZCZTQSA-N Bortezomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-RDJZCZTQSA-N 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 206010006458 Bronchitis chronic Diseases 0.000 description 1
- 102100009787 CABIN1 Human genes 0.000 description 1
- 108010066057 CABIN1 Proteins 0.000 description 1
- 102100005170 CRLF2 Human genes 0.000 description 1
- 101700020064 CRLF2 Proteins 0.000 description 1
- 102100006400 CSF2 Human genes 0.000 description 1
- 229960004117 Capecitabine Drugs 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
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 206010007279 Carcinoid tumour of the gastrointestinal tract Diseases 0.000 description 1
- 206010007515 Cardiac arrest Diseases 0.000 description 1
- 206010007554 Cardiac failure Diseases 0.000 description 1
- 208000005024 Castleman Disease Diseases 0.000 description 1
- 108010022830 Cetuximab Proteins 0.000 description 1
- 102000019034 Chemokines Human genes 0.000 description 1
- 108010012236 Chemokines Proteins 0.000 description 1
- 229960005091 Chloramphenicol Drugs 0.000 description 1
- WIIZWVCIJKGZOK-RKDXNWHRSA-N Chloramphenicol Chemical compound ClC(Cl)C(=O)N[C@H](CO)[C@H](O)C1=CC=C([N+]([O-])=O)C=C1 WIIZWVCIJKGZOK-RKDXNWHRSA-N 0.000 description 1
- 208000007451 Chronic Bronchitis Diseases 0.000 description 1
- MYSWGUAQZAJSOK-UHFFFAOYSA-N Ciprofloxacin Chemical compound C12=CC(N3CCNCC3)=C(F)C=C2C(=O)C(C(=O)O)=CN1C1CC1 MYSWGUAQZAJSOK-UHFFFAOYSA-N 0.000 description 1
- 230000035785 Cl/F Effects 0.000 description 1
- 206010009887 Colitis Diseases 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 210000000795 Conjunctiva Anatomy 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 210000004087 Cornea Anatomy 0.000 description 1
- 229940064701 Corticosteroid nasal preparations for topical use Drugs 0.000 description 1
- 229960001334 Corticosteroids Drugs 0.000 description 1
- MFYSYFVPBJMHGN-ZPOLXVRWSA-N Cortisone Chemical compound O=C1CC[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 MFYSYFVPBJMHGN-ZPOLXVRWSA-N 0.000 description 1
- 206010011219 Costochondritis Diseases 0.000 description 1
- 229940009997 Cromolyn Drugs 0.000 description 1
- 229960004397 Cyclophosphamide Drugs 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- DYDCUQKUCUHJBH-UWTATZPHSA-N D-cycloserine Chemical compound N[C@@H]1CONC1=O DYDCUQKUCUHJBH-UWTATZPHSA-N 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N D-sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 230000000970 DNA cross-linking Effects 0.000 description 1
- XAUDJQYHKZQPEU-KVQBGUIXSA-N Decitabine Chemical compound O=C1N=C(N)N=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 XAUDJQYHKZQPEU-KVQBGUIXSA-N 0.000 description 1
- FMGSKLZLMKYGDP-USOAJAOKSA-N Dehydroepiandrosterone Chemical compound C1[C@@H](O)CC[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CC=C21 FMGSKLZLMKYGDP-USOAJAOKSA-N 0.000 description 1
- 206010012601 Diabetes mellitus Diseases 0.000 description 1
- 206010012689 Diabetic retinopathy Diseases 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- YVPYQUNUQOZFHG-UHFFFAOYSA-N Diatrizoic acid Chemical compound CC(=O)NC1=C(I)C(NC(C)=O)=C(I)C(C(O)=O)=C1I YVPYQUNUQOZFHG-UHFFFAOYSA-N 0.000 description 1
- 229960003722 Doxycycline Drugs 0.000 description 1
- XQTWDDCIUJNLTR-CVHRZJFOSA-N Doxycycline Chemical compound O.O=C1C2=C(O)C=CC=C2[C@H](C)[C@@H]2C1=C(O)[C@]1(O)C(=O)C(C(N)=O)=C(O)[C@@H](N(C)C)[C@@H]1[C@H]2O XQTWDDCIUJNLTR-CVHRZJFOSA-N 0.000 description 1
- 229940121647 EGFR inhibitors Drugs 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 229960002549 ENOXACIN Drugs 0.000 description 1
- 206010014025 Ear swelling Diseases 0.000 description 1
- IWCWQNVIUXZOMJ-MISYRCLQSA-N Ecabet Chemical compound OC(=O)[C@@](C)([C@@H]1CC2)CCC[C@]1(C)C1=C2C=C(C(C)C)C(S(O)(=O)=O)=C1 IWCWQNVIUXZOMJ-MISYRCLQSA-N 0.000 description 1
- 208000005679 Eczema Diseases 0.000 description 1
- IDYZIJYBMGIQMJ-UHFFFAOYSA-N Enoxacin Chemical compound N1=C2N(CC)C=C(C(O)=O)C(=O)C2=CC(F)=C1N1CCNCC1 IDYZIJYBMGIQMJ-UHFFFAOYSA-N 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 210000003743 Erythrocytes Anatomy 0.000 description 1
- 102000003951 Erythropoietin Human genes 0.000 description 1
- 108090000394 Erythropoietin Proteins 0.000 description 1
- 108010008165 Etanercept Proteins 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
- 229960005420 Etoposide Drugs 0.000 description 1
- 229940012356 Eye Drops Drugs 0.000 description 1
- 102100014139 FKBP1A Human genes 0.000 description 1
- 208000001640 Fibromyalgia Diseases 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 229920001917 Ficoll Polymers 0.000 description 1
- RFHAOTPXVQNOHP-UHFFFAOYSA-N Fluconazole Chemical compound C1=NC=NN1CC(C=1C(=CC(F)=CC=1)F)(O)CN1C=NC=N1 RFHAOTPXVQNOHP-UHFFFAOYSA-N 0.000 description 1
- XRECTZIEBJDKEO-UHFFFAOYSA-N Flucytosine Chemical compound NC1=NC(=O)NC=C1F XRECTZIEBJDKEO-UHFFFAOYSA-N 0.000 description 1
- UUOUOERPONYGOS-CLCRDYEYSA-N Fluocinolone Chemical compound O=C1C=C[C@]2(C)[C@@]3(F)[C@@H](O)C[C@](C)([C@@]([C@H](O)C4)(O)C(=O)CO)[C@@H]4[C@@H]3C[C@H](F)C2=C1 UUOUOERPONYGOS-CLCRDYEYSA-N 0.000 description 1
- 229960001347 Fluocinolone Acetonide Drugs 0.000 description 1
- FAOZLTXFLGPHNG-KNAQIMQKSA-N Fluorometholone Chemical compound C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@]2(F)[C@@H](O)C[C@]2(C)[C@@](O)(C(C)=O)CC[C@H]21 FAOZLTXFLGPHNG-KNAQIMQKSA-N 0.000 description 1
- SYTBZMRGLBWNTM-UHFFFAOYSA-N Flurbiprofen Chemical compound FC1=CC(C(C(O)=O)C)=CC=C1C1=CC=CC=C1 SYTBZMRGLBWNTM-UHFFFAOYSA-N 0.000 description 1
- 208000004262 Food Hypersensitivity Diseases 0.000 description 1
- 206010017533 Fungal infection Diseases 0.000 description 1
- ZPACYDRSPFRDHO-ROBAGEODSA-N Gefarnate Chemical compound CC(C)=CCC\C(C)=C\CC\C(C)=C\CCC(=O)OC\C=C(/C)CCC=C(C)C ZPACYDRSPFRDHO-ROBAGEODSA-N 0.000 description 1
- 229920001386 Gefarnate Polymers 0.000 description 1
- 108010010803 Gelatin Proteins 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
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 description 1
- 208000005017 Glioblastoma Diseases 0.000 description 1
- OKMWKBLSFKFYGZ-UHFFFAOYSA-N Glyceryl behenate Chemical compound CCCCCCCCCCCCCCCCCCCCCC(=O)OCC(O)CO OKMWKBLSFKFYGZ-UHFFFAOYSA-N 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 206010018634 Gouty arthritis Diseases 0.000 description 1
- 206010018651 Graft versus host disease Diseases 0.000 description 1
- 208000009329 Graft vs Host Disease Diseases 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 229940093912 Gynecological Sulfonamides Drugs 0.000 description 1
- 206010018987 Haemorrhage Diseases 0.000 description 1
- 210000003128 Head Anatomy 0.000 description 1
- 208000010496 Heart Arrest Diseases 0.000 description 1
- 206010019280 Heart failure Diseases 0.000 description 1
- 206010073069 Hepatic cancer Diseases 0.000 description 1
- 208000002672 Hepatitis B Diseases 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 229940088597 Hormone Drugs 0.000 description 1
- 208000006572 Human Influenza Diseases 0.000 description 1
- XXSMGPRMXLTPCZ-UHFFFAOYSA-N Hydroxychloroquine Chemical compound ClC1=CC=C2C(NC(C)CCCN(CCO)CC)=CC=NC2=C1 XXSMGPRMXLTPCZ-UHFFFAOYSA-N 0.000 description 1
- 229960004171 Hydroxychloroquine Drugs 0.000 description 1
- 241000257303 Hymenoptera Species 0.000 description 1
- STECJAGHUSJQJN-GAUPFVANSA-N Hyoscine Natural products C1([C@H](CO)C(=O)OC2C[C@@H]3N([C@H](C2)[C@@H]2[C@H]3O2)C)=CC=CC=C1 STECJAGHUSJQJN-GAUPFVANSA-N 0.000 description 1
- 229960003444 IMMUNOSUPPRESSANTS Drugs 0.000 description 1
- 206010022000 Influenza Diseases 0.000 description 1
- RCINICONZNJXQF-MZXODVADSA-N Intaxel 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 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
- 229940045773 Jakafi Drugs 0.000 description 1
- 208000009388 Job Syndrome Diseases 0.000 description 1
- SBUJHOSQTJFQJX-NOAMYHISSA-N Kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 1
- 208000007766 Kaposi Sarcoma Diseases 0.000 description 1
- OZWKMVRBQXNZKK-UHFFFAOYSA-N Ketorolac Chemical compound OC(=O)C1CCN2C1=CC=C2C(=O)C1=CC=CC=C1 OZWKMVRBQXNZKK-UHFFFAOYSA-N 0.000 description 1
- PWKSKIMOESPYIA-BYPYZUCNSA-N L-N-acetyl-Cysteine Chemical compound CC(=O)N[C@@H](CS)C(O)=O PWKSKIMOESPYIA-BYPYZUCNSA-N 0.000 description 1
- 102100006672 LACRT Human genes 0.000 description 1
- 101700077657 LACRT Proteins 0.000 description 1
- 235000010701 Lavanda vera Nutrition 0.000 description 1
- 240000002809 Lavandula angustifolia Species 0.000 description 1
- 235000003515 Lavandula officinalis Nutrition 0.000 description 1
- 210000000265 Leukocytes Anatomy 0.000 description 1
- ZCGOMHNNNFPNMX-YHYDXASRSA-N Levocabastinum Chemical compound C1([C@@]2(C(O)=O)CCN(C[C@H]2C)C2CCC(CC2)(C#N)C=2C=CC(F)=CC=2)=CC=CC=C1 ZCGOMHNNNFPNMX-YHYDXASRSA-N 0.000 description 1
- 229960004305 Lodoxamide Drugs 0.000 description 1
- RVGLGHVJXCETIO-UHFFFAOYSA-N Lodoxamide Chemical compound OC(=O)C(=O)NC1=CC(C#N)=CC(NC(=O)C(O)=O)=C1Cl RVGLGHVJXCETIO-UHFFFAOYSA-N 0.000 description 1
- ZEKZLJVOYLTDKK-UHFFFAOYSA-N Lomefloxacin Chemical compound FC1=C2N(CC)C=C(C(O)=O)C(=O)C2=CC(F)=C1N1CCNC(C)C1 ZEKZLJVOYLTDKK-UHFFFAOYSA-N 0.000 description 1
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 206010025650 Malignant melanoma Diseases 0.000 description 1
- 208000008585 Mastocytosis Diseases 0.000 description 1
- BYBLEWFAAKGYCD-UHFFFAOYSA-N Miconazole Chemical compound ClC1=CC(Cl)=CC=C1COC(C=1C(=CC(Cl)=CC=1)Cl)CN1C=NC=C1 BYBLEWFAAKGYCD-UHFFFAOYSA-N 0.000 description 1
- 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 1
- 229960001156 Mitoxantrone Drugs 0.000 description 1
- 102000001621 Mucoproteins Human genes 0.000 description 1
- 108010093825 Mucoproteins Proteins 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 229960000951 Mycophenolic Acid Drugs 0.000 description 1
- 206010028561 Myeloid metaplasia Diseases 0.000 description 1
- 125000000815 N-oxide group Chemical group 0.000 description 1
- 229960003255 Natamycin Drugs 0.000 description 1
- NCXMLFZGDNKEPB-FFPOYIOWSA-N Natamycin Chemical compound O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1/C=C/C=C/C=C/C=C/C[C@@H](C)OC(=O)/C=C/[C@H]2O[C@@H]2C[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 NCXMLFZGDNKEPB-FFPOYIOWSA-N 0.000 description 1
- 210000003739 Neck Anatomy 0.000 description 1
- 206010029113 Neovascularisation Diseases 0.000 description 1
- 241001182492 Nes Species 0.000 description 1
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Nitrumon Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 1
- 229960001180 Norfloxacin Drugs 0.000 description 1
- OGJPXUAPXNRGGI-UHFFFAOYSA-N Norfloxacin Chemical compound C1=C2N(CC)C=C(C(O)=O)C(=O)C2=CC(F)=C1N1CCNCC1 OGJPXUAPXNRGGI-UHFFFAOYSA-N 0.000 description 1
- 229940074726 OPHTHALMOLOGIC ANTIINFLAMMATORY AGENTS Drugs 0.000 description 1
- 208000004361 Obstructive Lung Disease Diseases 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 206010025310 Other lymphomas Diseases 0.000 description 1
- 229960000625 Oxytetracycline Drugs 0.000 description 1
- IWVCMVBTMGNXQD-PXOLEDIWSA-N Oxytetracycline Chemical compound C1=CC=C2[C@](O)(C)[C@H]3[C@H](O)[C@H]4[C@H](N(C)C)C(O)=C(C(N)=O)C(=O)[C@@]4(O)C(O)=C3C(=O)C2=C1O IWVCMVBTMGNXQD-PXOLEDIWSA-N 0.000 description 1
- 239000004100 Oxytetracycline Substances 0.000 description 1
- 101710018405 PTPN11 Proteins 0.000 description 1
- 102100017818 PTPN11 Human genes 0.000 description 1
- 229960001592 Paclitaxel Drugs 0.000 description 1
- 229940049954 Penicillin Drugs 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
- IJHNSHDBIRRJRN-UHFFFAOYSA-N Pheniramine Chemical compound C=1C=CC=NC=1C(CCN(C)C)C1=CC=CC=C1 IJHNSHDBIRRJRN-UHFFFAOYSA-N 0.000 description 1
- 229960001190 Pheniramine Drugs 0.000 description 1
- 102000029987 Phosphatidylethanolamine-binding protein Human genes 0.000 description 1
- 108091000043 Phosphatidylethanolamine-binding protein Proteins 0.000 description 1
- KASDHRXLYQOAKZ-XDSKOBMDSA-N Pimecrolimus Chemical compound C/C([C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@]2(O)O[C@@H]([C@H](C[C@H]2C)OC)[C@@H](OC)C[C@@H](C)C/C(C)=C/[C@H](C(C[C@H](O)[C@H]1C)=O)CC)=C\[C@@H]1CC[C@H](Cl)[C@H](OC)C1 KASDHRXLYQOAKZ-XDSKOBMDSA-N 0.000 description 1
- 240000003524 Pinus torreyana Species 0.000 description 1
- 235000006235 Pinus torreyana Nutrition 0.000 description 1
- 208000007452 Plasmacytoma Diseases 0.000 description 1
- 102000004179 Plasminogen activator inhibitor-2 Human genes 0.000 description 1
- 108090000614 Plasminogen activator inhibitor-2 Proteins 0.000 description 1
- 229920002565 Polyethylene Glycol 400 Polymers 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- OIGNJSKKLXVSLS-VWUMJDOOSA-N Prednisolone Chemical compound O=C1C=C[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OIGNJSKKLXVSLS-VWUMJDOOSA-N 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 206010064911 Pulmonary arterial hypertension Diseases 0.000 description 1
- 108010029812 Purinergic P2Y2 Receptors Proteins 0.000 description 1
- 102000001744 Purinergic P2Y2 Receptors Human genes 0.000 description 1
- 210000003324 RBC Anatomy 0.000 description 1
- JQXXHWHPUNPDRT-WLSIYKJHSA-N RIFAMPICIN Chemical compound O([C@](C1=O)(C)O/C=C/[C@@H]([C@H]([C@@H](OC(C)=O)[C@H](C)[C@H](O)[C@H](C)[C@@H](O)[C@@H](C)\C=C\C=C(C)/C(=O)NC=2C(O)=C3C([O-])=C4C)C)OC)C4=C1C3=C(O)C=2\C=N\N1CC[NH+](C)CC1 JQXXHWHPUNPDRT-WLSIYKJHSA-N 0.000 description 1
- 229960001487 RIMEXOLONE Drugs 0.000 description 1
- 230000025458 RNA interference Effects 0.000 description 1
- 102220420181 RPS19BP1 K15M Human genes 0.000 description 1
- 208000002574 Reactive Arthritis Diseases 0.000 description 1
- ALLWOAVDORUJLA-UHFFFAOYSA-N Rebamipida Chemical compound C=1C(=O)NC2=CC=CC=C2C=1CC(C(=O)O)NC(=O)C1=CC=C(Cl)C=C1 ALLWOAVDORUJLA-UHFFFAOYSA-N 0.000 description 1
- 206010038038 Rectal cancer Diseases 0.000 description 1
- 206010038389 Renal cancer Diseases 0.000 description 1
- 206010063837 Reperfusion injury Diseases 0.000 description 1
- 229940053174 Restasis Drugs 0.000 description 1
- 206010038910 Retinitis Diseases 0.000 description 1
- SHGAZHPCJJPHSC-YCNIQYBTSA-N 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
- 229940061341 Retisert Drugs 0.000 description 1
- 206010072736 Rheumatic disease Diseases 0.000 description 1
- 229940081190 Rifampin Drugs 0.000 description 1
- 108010001645 Rituximab Proteins 0.000 description 1
- XMSXOLDPMGMWTH-UHFFFAOYSA-N Rivoglitazone Chemical compound CN1C2=CC(OC)=CC=C2N=C1COC(C=C1)=CC=C1CC1SC(=O)NC1=O XMSXOLDPMGMWTH-UHFFFAOYSA-N 0.000 description 1
- 229950010764 Rivoglitazone Drugs 0.000 description 1
- 241000315672 SARS coronavirus Species 0.000 description 1
- 102100019667 STAT3 Human genes 0.000 description 1
- 108010017324 STAT3 Transcription Factor Proteins 0.000 description 1
- LSIXBBPOJBJQHN-UHFFFAOYSA-N Santene Chemical compound C1CC2C(C)=C(C)C1C2 LSIXBBPOJBJQHN-UHFFFAOYSA-N 0.000 description 1
- 206010039710 Scleroderma Diseases 0.000 description 1
- STECJAGHUSJQJN-FWXGHANASA-N Scopolamine Chemical compound C1([C@@H](CO)C(=O)O[C@H]2C[C@@H]3N([C@H](C2)[C@@H]2[C@H]3O2)C)=CC=CC=C1 STECJAGHUSJQJN-FWXGHANASA-N 0.000 description 1
- 206010040070 Septic shock Diseases 0.000 description 1
- 208000009359 Sezary Syndrome Diseases 0.000 description 1
- 201000006984 Sezary's disease Diseases 0.000 description 1
- 241000580858 Simian-Human immunodeficiency virus Species 0.000 description 1
- 206010040880 Skin irritation Diseases 0.000 description 1
- 229940010747 Sodium Hyaluronate Drugs 0.000 description 1
- 229920002385 Sodium hyaluronate Polymers 0.000 description 1
- 210000000952 Spleen Anatomy 0.000 description 1
- DCXXMTOCNZCJGO-UHFFFAOYSA-N Stearin Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(OC(=O)CCCCCCCCCCCCCCCCC)COC(=O)CCCCCCCCCCCCCCCCC DCXXMTOCNZCJGO-UHFFFAOYSA-N 0.000 description 1
- 229960004492 Suprofen Drugs 0.000 description 1
- MDKGKXOCJGEUJW-UHFFFAOYSA-N Suprofen Chemical compound C1=CC(C(C(O)=O)C)=CC=C1C(=O)C1=CC=CS1 MDKGKXOCJGEUJW-UHFFFAOYSA-N 0.000 description 1
- 102100008416 TSPYL2 Human genes 0.000 description 1
- 101710036648 TSPYL2 Proteins 0.000 description 1
- 101700057652 TYK2 Proteins 0.000 description 1
- 108010006877 Tacrolimus Binding Protein 1A Proteins 0.000 description 1
- NAVMQTYZDKMPEU-UHFFFAOYSA-N Targretin Chemical compound CC1=CC(C(CCC2(C)C)(C)C)=C2C=C1C(=C)C1=CC=C(C(O)=O)C=C1 NAVMQTYZDKMPEU-UHFFFAOYSA-N 0.000 description 1
- OFVLGDICTFRJMM-WESIUVDSSA-N Tetracycline Chemical group C1=CC=C2[C@](O)(C)[C@H]3C[C@H]4[C@H](N(C)C)C(O)=C(C(N)=O)C(=O)[C@@]4(O)C(O)=C3C(=O)C2=C1O OFVLGDICTFRJMM-WESIUVDSSA-N 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 229940040944 Tetracyclines Drugs 0.000 description 1
- 102000036902 Thrombopoietin Human genes 0.000 description 1
- 108010041111 Thrombopoietin Proteins 0.000 description 1
- 206010043709 Thyroid disease Diseases 0.000 description 1
- 206010043778 Thyroiditis Diseases 0.000 description 1
- 201000008908 Tietze's syndrome Diseases 0.000 description 1
- 229940035295 Ting Drugs 0.000 description 1
- GFNANZIMVAIWHM-OBYCQNJPSA-N Triamcinolone Chemical compound O=C1C=C[C@]2(C)[C@@]3(F)[C@@H](O)C[C@](C)([C@@]([C@H](O)C4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 GFNANZIMVAIWHM-OBYCQNJPSA-N 0.000 description 1
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Tris Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- WVPSKSLAZQPAKQ-CDMJZVDBSA-N Trovafloxacin Chemical compound C([C@H]1[C@@H]([C@H]1C1)N)N1C(C(=CC=1C(=O)C(C(O)=O)=C2)F)=NC=1N2C1=CC=C(F)C=C1F WVPSKSLAZQPAKQ-CDMJZVDBSA-N 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- NMUSYJAQQFHJEW-KVTDHHQDSA-N U-18,496 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 1
- 206010046799 Uterine leiomyosarcoma Diseases 0.000 description 1
- 229960003165 Vancomycin Drugs 0.000 description 1
- 108010059993 Vancomycin Proteins 0.000 description 1
- 229940055059 Vexol Drugs 0.000 description 1
- 206010047461 Viral infection Diseases 0.000 description 1
- 206010047571 Visual impairment Diseases 0.000 description 1
- 229960005289 Voclosporin Drugs 0.000 description 1
- BICRTLVBTLFLRD-PTWUADNWSA-N Voclosporin Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C=C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O BICRTLVBTLFLRD-PTWUADNWSA-N 0.000 description 1
- 229960000237 Vorinostat Drugs 0.000 description 1
- WAEXFXRVDQXREF-UHFFFAOYSA-N Vorinostat Chemical compound ONC(=O)CCCCCCC(=O)NC1=CC=CC=C1 WAEXFXRVDQXREF-UHFFFAOYSA-N 0.000 description 1
- 230000036835 Vz/F Effects 0.000 description 1
- 229940039916 Xeljanz Drugs 0.000 description 1
- YKPUWZUDDOIDPM-VURMDHGXSA-N Zucapsaicin Chemical compound COC1=CC(CNC(=O)CCCC\C=C/C(C)C)=CC=C1O YKPUWZUDDOIDPM-VURMDHGXSA-N 0.000 description 1
- GXJABQQUPOEUTA-NVXWUHKLSA-N [(1S)-3-methyl-1-[[(2R)-3-phenyl-2-(pyrazine-2-carbonylamino)propanoyl]amino]butyl]boronic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-NVXWUHKLSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 230000001154 acute Effects 0.000 description 1
- 230000000996 additive Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000000240 adjuvant Effects 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 239000003513 alkali Substances 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 229930002945 all-trans-retinaldehyde Natural products 0.000 description 1
- 230000000172 allergic Effects 0.000 description 1
- 230000001668 ameliorated Effects 0.000 description 1
- 230000003444 anaesthetic Effects 0.000 description 1
- 229960004238 anakinra Drugs 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 230000019552 anatomical structure morphogenesis Effects 0.000 description 1
- 239000004037 angiogenesis inhibitor Substances 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 235000003484 annual ragweed Nutrition 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 229960002469 antazoline Drugs 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000843 anti-fungal Effects 0.000 description 1
- 239000002260 anti-inflammatory agent Substances 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 230000000692 anti-sense Effects 0.000 description 1
- 230000000840 anti-viral Effects 0.000 description 1
- 239000000043 antiallergic agent Substances 0.000 description 1
- 238000009166 antihormone therapy Methods 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 230000001640 apoptogenic Effects 0.000 description 1
- 239000012131 assay buffer Substances 0.000 description 1
- 230000003416 augmentation Effects 0.000 description 1
- CLKOFPXJLQSYAH-ABRJDSQDSA-N bacitracin A Chemical compound C1SC([C@@H](N)[C@@H](C)CC)=N[C@@H]1C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](CCC(O)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]1C(=O)N[C@H](CCCN)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@H](CC=2C=CC=CC=2)C(=O)N[C@@H](CC=2N=CNC=2)C(=O)N[C@H](CC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)NCCCC1 CLKOFPXJLQSYAH-ABRJDSQDSA-N 0.000 description 1
- 229960000626 benzylpenicillin Drugs 0.000 description 1
- 229960002938 bexarotene Drugs 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 230000000740 bleeding Effects 0.000 description 1
- 231100000319 bleeding Toxicity 0.000 description 1
- 229960001467 bortezomib Drugs 0.000 description 1
- 235000006263 bur ragweed Nutrition 0.000 description 1
- 239000000480 calcium channel blocker Substances 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 230000003197 catalytic Effects 0.000 description 1
- 230000024881 catalytic activity Effects 0.000 description 1
- 108020001778 catalytic domains Proteins 0.000 description 1
- 230000001364 causal effect Effects 0.000 description 1
- 230000020411 cell activation Effects 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 238000001516 cell proliferation assay Methods 0.000 description 1
- 230000003833 cell viability Effects 0.000 description 1
- 238000003570 cell viability assay Methods 0.000 description 1
- 150000001780 cephalosporins Chemical class 0.000 description 1
- 229960005395 cetuximab Drugs 0.000 description 1
- 239000000064 cholinergic agonist Substances 0.000 description 1
- 229960003405 ciprofloxacin Drugs 0.000 description 1
- 229940108538 colistimethate Drugs 0.000 description 1
- IQWHCHZFYPIVRV-VLLYEMIKSA-I colistin A sodium methanesulfonate Chemical compound [Na+].[Na+].[Na+].[Na+].[Na+].CC[C@@H](C)CCCCC(=O)N[C@@H](CCNCS([O-])(=O)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCNCS([O-])(=O)=O)C(=O)N[C@H]1CCNC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCNCS([O-])(=O)=O)NC(=O)[C@H](CCNCS([O-])(=O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@H](CCNCS([O-])(=O)=O)NC1=O IQWHCHZFYPIVRV-VLLYEMIKSA-I 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 201000011231 colorectal cancer Diseases 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 235000003488 common ragweed Nutrition 0.000 description 1
- 201000009541 complex regional pain syndrome Diseases 0.000 description 1
- 230000002508 compound effect Effects 0.000 description 1
- 230000001010 compromised Effects 0.000 description 1
- 230000001268 conjugating Effects 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 229940099112 cornstarch Drugs 0.000 description 1
- 230000002596 correlated Effects 0.000 description 1
- 230000000875 corresponding Effects 0.000 description 1
- 229960004544 cortisone Drugs 0.000 description 1
- 229960000265 cromoglicic acid Drugs 0.000 description 1
- 239000003431 cross linking reagent Substances 0.000 description 1
- 229960003077 cycloserine Drugs 0.000 description 1
- 229960003603 decitabine Drugs 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 229940119350 dehydroepiandrosterone Drugs 0.000 description 1
- 230000001335 demethylating Effects 0.000 description 1
- 230000002074 deregulated Effects 0.000 description 1
- 230000001627 detrimental Effects 0.000 description 1
- 201000008286 diarrhea Diseases 0.000 description 1
- 229960001259 diclofenac Drugs 0.000 description 1
- DCOPUUMXTXDBNB-UHFFFAOYSA-N diclofenac Chemical compound OC(=O)CC1=CC=CC=C1NC1=C(Cl)C=CC=C1Cl DCOPUUMXTXDBNB-UHFFFAOYSA-N 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 201000009910 diseases by infectious agent Diseases 0.000 description 1
- 201000008325 diseases of cellular proliferation Diseases 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- VLARUOGDXDTHEH-UHFFFAOYSA-L disodium cromoglycate Chemical compound [Na+].[Na+].O1C(C([O-])=O)=CC(=O)C2=C1C=CC=C2OCC(O)COC1=CC=CC2=C1C(=O)C=C(C([O-])=O)O2 VLARUOGDXDTHEH-UHFFFAOYSA-L 0.000 description 1
- 239000006196 drop Substances 0.000 description 1
- 108010067071 duramycin Proteins 0.000 description 1
- 229950003246 ecabet Drugs 0.000 description 1
- 231100001003 eczema Toxicity 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N edta Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 229960000284 efalizumab Drugs 0.000 description 1
- 108010010371 efalizumab Proteins 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 210000002257 embryonic structures Anatomy 0.000 description 1
- 239000003974 emollient agent Substances 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 230000001973 epigenetic Effects 0.000 description 1
- 229940105423 erythropoietin Drugs 0.000 description 1
- 229960000403 etanercept Drugs 0.000 description 1
- 150000002170 ethers Chemical class 0.000 description 1
- HXTBSJOGZDYEIO-UHFFFAOYSA-N ethyl 2-[3-(cyanomethyl)indol-1-yl]acetate Chemical compound C1=CC=C2N(CC(=O)OCC)C=C(CC#N)C2=C1 HXTBSJOGZDYEIO-UHFFFAOYSA-N 0.000 description 1
- 230000005713 exacerbation Effects 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- 239000003172 expectorant agent Substances 0.000 description 1
- 239000003889 eye drop Substances 0.000 description 1
- 230000004761 fibrosis Effects 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 229960004884 fluconazole Drugs 0.000 description 1
- 229960004413 flucytosine Drugs 0.000 description 1
- 229940043075 fluocinolone Drugs 0.000 description 1
- 229960001048 fluorometholone Drugs 0.000 description 1
- 229960002390 flurbiprofen Drugs 0.000 description 1
- 235000020932 food allergy Nutrition 0.000 description 1
- WSFSSNUMVMOOMR-UHFFFAOYSA-N formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 1
- 201000003928 fungal infectious disease Diseases 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 229960003779 gefarnate Drugs 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 239000003862 glucocorticoid Substances 0.000 description 1
- 229940049654 glyceryl behenate Drugs 0.000 description 1
- LYCAIKOWRPUZTN-UHFFFAOYSA-N glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 230000003394 haemopoietic Effects 0.000 description 1
- 239000003481 heat shock protein 90 inhibitor Substances 0.000 description 1
- 230000011132 hemopoiesis Effects 0.000 description 1
- 239000003276 histone deacetylase inhibitor Substances 0.000 description 1
- 229940121372 histone deacetylase inhibitors Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 235000003642 hunger Nutrition 0.000 description 1
- 230000009610 hypersensitivity Effects 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000002055 immunohistochemical Effects 0.000 description 1
- 238000002991 immunohistochemical analysis Methods 0.000 description 1
- 230000001861 immunosuppresant Effects 0.000 description 1
- 230000001976 improved Effects 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000004941 influx Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000000977 initiatory Effects 0.000 description 1
- 230000000968 intestinal Effects 0.000 description 1
- 229940079867 intestinal antiinfectives Sulfonamides Drugs 0.000 description 1
- 229960004768 irinotecan Drugs 0.000 description 1
- 230000000366 juvenile Effects 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- 229960004752 ketorolac Drugs 0.000 description 1
- 201000010982 kidney cancer Diseases 0.000 description 1
- 238000000021 kinase assay Methods 0.000 description 1
- SFWLDKQAUHFCBS-WWXQEMPQSA-N lancovutide Chemical compound C([C@H]1C(=O)N[C@H](C(N[C@@H]2C(=O)N[C@H](C(=O)NCC(=O)N[C@@H](CC(N)=O)C(=O)N[C@H]3C(=O)N[C@@H](CCCCNC[C@H]4C(=O)N[C@@H](CC=5C=CC=CC=5)C(=O)NCC(=O)N5CCC[C@H]5C(=O)N[C@@H](CC=5C=CC=CC=5)C(=O)N[C@H]([C@@H](SC[C@H](NC(=O)[C@H](NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@@H](N)CSC3C)CSC2)C(=O)N4)C)C(=O)N1)C(O)=O)[C@@H](O)C(O)=O)=O)C(C)C)C1=CC=CC=C1 SFWLDKQAUHFCBS-WWXQEMPQSA-N 0.000 description 1
- 239000001102 lavandula vera Substances 0.000 description 1
- 235000018219 lavender Nutrition 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 230000000670 limiting Effects 0.000 description 1
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 1
- 201000007270 liver cancer Diseases 0.000 description 1
- 229960002422 lomefloxacin Drugs 0.000 description 1
- 229960003744 loteprednol etabonate Drugs 0.000 description 1
- DMKSVUSAATWOCU-HROMYWEYSA-N loteprednol etabonate Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@@](C(=O)OCCl)(OC(=O)OCC)[C@@]1(C)C[C@@H]2O DMKSVUSAATWOCU-HROMYWEYSA-N 0.000 description 1
- 235000004213 low-fat Nutrition 0.000 description 1
- 230000001050 lubricating Effects 0.000 description 1
- 201000005202 lung cancer Diseases 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 230000036244 malformation Effects 0.000 description 1
- 239000003628 mammalian target of rapamycin inhibitor Substances 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 239000002609 media Substances 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 230000003340 mental Effects 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 125000000250 methylamino group Chemical group [H]N(*)C([H])([H])[H] 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 229960002509 miconazole Drugs 0.000 description 1
- 230000000394 mitotic Effects 0.000 description 1
- 238000007479 molecular analysis Methods 0.000 description 1
- 150000004682 monohydrates Chemical class 0.000 description 1
- 239000000472 muscarinic agonist Substances 0.000 description 1
- 229940083353 mycophenolate sodium Drugs 0.000 description 1
- DOZYTHNHLLSNIK-JOKMOOFLSA-M mycophenolate sodium Chemical compound [Na+].OC1=C(C\C=C(/C)CCC([O-])=O)C(OC)=C(C)C2=C1C(=O)OC2 DOZYTHNHLLSNIK-JOKMOOFLSA-M 0.000 description 1
- 201000002481 myositis Diseases 0.000 description 1
- 229960005016 naphazoline Drugs 0.000 description 1
- 235000010298 natamycin Nutrition 0.000 description 1
- 239000004311 natamycin Substances 0.000 description 1
- 230000031990 negative regulation of inflammatory response Effects 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- ZBGPYVZLYBDXKO-HILBYHGXSA-N netilmycin Chemical compound O([C@@H]1[C@@H](N)C[C@H]([C@@H]([C@H]1O)O[C@@H]1[C@]([C@H](NC)[C@@H](O)CO1)(C)O)NCC)[C@H]1OC(CN)=CC[C@H]1N ZBGPYVZLYBDXKO-HILBYHGXSA-N 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 239000000041 non-steroidal anti-inflammatory agent Substances 0.000 description 1
- 239000012457 nonaqueous media Substances 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 102000006255 nuclear receptors Human genes 0.000 description 1
- 108020004017 nuclear receptors Proteins 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 229940005931 ophthalmologic Fluoroquinolone antiinfectives Drugs 0.000 description 1
- 229940005938 ophthalmologic antiinfectives Sulfonamides Drugs 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 229960001756 oxaliplatin Drugs 0.000 description 1
- 235000019366 oxytetracycline Nutrition 0.000 description 1
- 230000001717 pathogenic Effects 0.000 description 1
- 244000052769 pathogens Species 0.000 description 1
- 201000011152 pemphigus Diseases 0.000 description 1
- 201000001976 pemphigus vulgaris Diseases 0.000 description 1
- JLFNLZLINWHATN-UHFFFAOYSA-N pentaethylene glycol Chemical compound OCCOCCOCCOCCOCCO JLFNLZLINWHATN-UHFFFAOYSA-N 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 229960005330 pimecrolimus Drugs 0.000 description 1
- 101710039033 pkbA Proteins 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 238000011176 pooling Methods 0.000 description 1
- 238000010837 poor prognosis Methods 0.000 description 1
- 230000003334 potential Effects 0.000 description 1
- 229960002847 prasterone Drugs 0.000 description 1
- 229960005205 prednisolone Drugs 0.000 description 1
- 230000000770 pro-inflamatory Effects 0.000 description 1
- 230000002250 progressing Effects 0.000 description 1
- 230000002062 proliferating Effects 0.000 description 1
- 230000002035 prolonged Effects 0.000 description 1
- 230000001185 psoriatic Effects 0.000 description 1
- 125000000561 purinyl group Chemical group N1=C(N=C2N=CNC2=C1)* 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 235000009736 ragweed Nutrition 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 229950004535 rebamipide Drugs 0.000 description 1
- 239000002464 receptor antagonist Substances 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 201000001275 rectum cancer Diseases 0.000 description 1
- 231100001019 reduced numbers of red blood cells Toxicity 0.000 description 1
- 230000011514 reflex Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000241 respiratory Effects 0.000 description 1
- 200000000008 restenosis Diseases 0.000 description 1
- 230000002207 retinal Effects 0.000 description 1
- 235000020945 retinal Nutrition 0.000 description 1
- 239000011604 retinal Substances 0.000 description 1
- 230000002441 reversible Effects 0.000 description 1
- 101710004466 rgy Proteins 0.000 description 1
- 101710030364 rgy1 Proteins 0.000 description 1
- 101710030359 rgy2 Proteins 0.000 description 1
- 229960001225 rifampicin Drugs 0.000 description 1
- 229960004641 rituximab Drugs 0.000 description 1
- 229960002646 scopolamine Drugs 0.000 description 1
- 230000000580 secretagogue Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 239000000333 selective estrogen receptor modulator Substances 0.000 description 1
- 101700064492 senju Proteins 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 230000036303 septic shock Effects 0.000 description 1
- 201000003176 severe acute respiratory syndrome Diseases 0.000 description 1
- 201000000849 skin cancer Diseases 0.000 description 1
- 231100000475 skin irritation Toxicity 0.000 description 1
- 230000036556 skin irritation Effects 0.000 description 1
- 201000002314 small intestine cancer Diseases 0.000 description 1
- 229940045902 sodium stearyl fumarate Drugs 0.000 description 1
- MAKUBRYLFHZREJ-JWBQXVCJSA-M sodium;(2S,3S,4R,5R,6R)-3-[(2S,3R,5S,6R)-3-acetamido-5-hydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-4,5,6-trihydroxyoxane-2-carboxylate Chemical compound [Na+].CC(=O)N[C@@H]1C[C@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](C([O-])=O)O[C@@H](O)[C@H](O)[C@H]1O MAKUBRYLFHZREJ-JWBQXVCJSA-M 0.000 description 1
- STFSJTPVIIDAQX-LTRPLHCISA-M sodium;(E)-4-octadecoxy-4-oxobut-2-enoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCCOC(=O)\C=C\C([O-])=O STFSJTPVIIDAQX-LTRPLHCISA-M 0.000 description 1
- KISFEBPWFCGRGN-UHFFFAOYSA-M sodium;2-(2,4-dichlorophenoxy)ethyl sulfate Chemical compound [Na+].[O-]S(=O)(=O)OCCOC1=CC=C(Cl)C=C1Cl KISFEBPWFCGRGN-UHFFFAOYSA-M 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000010911 splenectomy Methods 0.000 description 1
- 230000037351 starvation Effects 0.000 description 1
- 230000003068 static Effects 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 230000003637 steroidlike Effects 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 150000003456 sulfonamides Chemical class 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000002889 sympathetic Effects 0.000 description 1
- 201000010874 syndrome Diseases 0.000 description 1
- 230000002195 synergetic Effects 0.000 description 1
- 229940041075 systemic Fluoroquinolone antibacterials Drugs 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 201000009594 systemic scleroderma Diseases 0.000 description 1
- 229930003347 taxol Natural products 0.000 description 1
- 230000004489 tear production Effects 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 201000002510 thyroid cancer Diseases 0.000 description 1
- 210000001519 tissues Anatomy 0.000 description 1
- 239000010936 titanium Substances 0.000 description 1
- 239000004408 titanium dioxide Substances 0.000 description 1
- SYIKUFDOYJFGBQ-YLAFAASESA-N tofacitinib citrate Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O.C[C@@H]1CCN(C(=O)CC#N)C[C@@H]1N(C)C1=NC=NC2=C1C=CN2 SYIKUFDOYJFGBQ-YLAFAASESA-N 0.000 description 1
- 229940026752 topical Sulfonamides Drugs 0.000 description 1
- 229940083878 topical for treatment of hemorrhoids and anal fissures Corticosteroids Drugs 0.000 description 1
- 230000002588 toxic Effects 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 102000003995 transcription factors Human genes 0.000 description 1
- 108090000464 transcription factors Proteins 0.000 description 1
- 229960005294 triamcinolone Drugs 0.000 description 1
- 229960000497 trovafloxacin Drugs 0.000 description 1
- 230000004565 tumor cell growth Effects 0.000 description 1
- 210000004881 tumor cells Anatomy 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 230000000381 tumorigenic Effects 0.000 description 1
- 231100000588 tumorigenic Toxicity 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- MYPYJXKWCTUITO-LYRMYLQWSA-O vancomycin(1+) Chemical compound O([C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1OC1=C2C=C3C=C1OC1=CC=C(C=C1Cl)[C@@H](O)[C@H](C(N[C@@H](CC(N)=O)C(=O)N[C@H]3C(=O)N[C@H]1C(=O)N[C@H](C(N[C@@H](C3=CC(O)=CC(O)=C3C=3C(O)=CC=C1C=3)C([O-])=O)=O)[C@H](O)C1=CC=C(C(=C1)Cl)O2)=O)NC(=O)[C@@H](CC(C)C)[NH2+]C)[C@H]1C[C@](C)([NH3+])[C@H](O)[C@H](C)O1 MYPYJXKWCTUITO-LYRMYLQWSA-O 0.000 description 1
- 230000017613 viral reproduction Effects 0.000 description 1
- 230000004304 visual acuity Effects 0.000 description 1
- 108010057559 voclosporin Proteins 0.000 description 1
- 230000035513 volume of distribution Effects 0.000 description 1
- 229960002860 zucapsaicin Drugs 0.000 description 1
- 150000003952 β-lactams Chemical class 0.000 description 1
Abstract
This invention relates to a sustained release composition, comprising: (i) {1-{1-[3-fluoro-2-(trifluoromethyl)isonicotinoyl]piperidin-4-yl}-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]azetidin-3-yl}acetonitrile, or a pharmaceutically acceptable salt thereof; (ii) a first hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of about 80 cP to about 120 cP; (iii) a second hypromellose, characterized by having an apparent viscosity at a concentration of 2% in water of about 3000 cP to about 5600 cP, wherein the composition comprises about 8% to about 20% of the first and second hypromelloses; (iv) about 16% to about 22% by weight of microcrystalline cellulose; and (v) about 45% to about 55% by weight of lactose monohydrate. acterized by having an apparent viscosity at a concentration of 2% in water of about 80 cP to about 120 cP; (iii) a second hypromellose, characterized by having an apparent viscosity at a concentration of 2% in water of about 3000 cP to about 5600 cP, wherein the composition comprises about 8% to about 20% of the first and second hypromelloses; (iv) about 16% to about 22% by weight of microcrystalline cellulose; and (v) about 45% to about 55% by weight of lactose monohydrate.
Description
SUSTAINED RELEASE DOSAGE FORMS FOR A JAK1 INHIBITOR
The present Application is a Divisional Application from New Zealand Patent
Application No. 717230. The entire disclosures of New Zealand Patent Application No.
717230 and its ponding ational Patent Application No. ,
are incorporated herein by reference. This Application claims the benefit of priority of
U.S. Prov. Appl. No. 61/863,325, filed August 7, 2013, and U.S. Prov. Appl. No.
61/913,066, filed December 6, 2013, each of which is incorporated herein by reference in
its entirety.
TECHNICAL FIELD
This application relates to a sustained release dosage form comprising {1-{1-[3-
fluoro(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-
d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile, or a pharmaceutically
acceptable salt thereof, and doses and methods related thereto.
BACKGROUND
Protein kinases (PKs) regulate diverse biological processes including cell
, survival, entiation, organ formation, morphogenesis, neovascularization,
tissue repair, and regeneration, among others. Protein kinases also play specialized
roles in a host of human diseases including cancer. Cytokines, low-molecular weight
polypeptides or glycoproteins, regulate many pathways involved in the host
inflammatory se to sepsis. Cytokines influence cell differentiation, proliferation
and activation, and can modulate both pro-inflammatory and anti-inflammatory
responses to allow the host to react appropriately to pathogens. Signaling of a wide
range of cytokines involves the Janus kinase family (JAKs) of protein tyrosine kinases
and Signal Transducers and Activators of Transcription (STATs). There are four
known mammalian JAKs: JAK1 (Janus -1), JAK2, JAK3 (also known as Janus
kinase, leukocyte; JAKL; and L-JAK), and TYK2 (protein-tyrosine kinase 2).
Cytokine-stimulated immune and inflammatory ses contribute to
pathogenesis of diseases: pathologies such as severe combined immunodeficiency
(SCID) arise from suppression of the immune system, while a ctive or
inappropriate immune/inflammatory response contributes to the ogy of
autoimmune es (e.g., asthma, systemic lupus matosus, thyroiditis
myocarditis), and illnesses such as scleroderma and osteoarthritis (Ortmann, R. A., T.
Cheng, et al. (2000) Arthritis Res 2(1): 16—32).
Deficiencies in expression of JAKs are associated with many disease states.
For e, — mice are runted at birth, fail to nurse, and die perinatally (Rodig,
S. J., M. A. Meraz, et al. (1998) Cell 93(3): 373—83). Jak2—/— mouse embryos are
anemic and die around day 12.5 postcoitum due to the absence of definitive
erythropoiesis.
The JAK/STAT pathway, and in particular all four JAKs, are believed to play
a role in the pathogenesis of asthmatic response, chronic obstructive pulmonary
disease, bronchitis, and other related inflammatory es of the lower respiratory
tract. Multiple cytokines that signal through JAKs have been linked to inflammatory
es/conditions of the upper respiratory tract, such as those affecting the nose and
s (e. g., rhinitis and sinusitis) whether classically allergic reactions or not. The
JAK/STAT pathway has also been implicated in inflammatory diseases/conditions of
the eye and chronic allergic responses.
Activation of JAK/STAT in s may occur by cytokine stimulation (e.g.
IL—6 or GM—CSF) or by a reduction in the endogenous suppressors of JAK signaling
such as SOCS (suppressor or cytokine signaling) or PIAS in inhibitor of
activated STAT) (Boudny, V., and Kovarik, J
., Neoplasm. —355, 2002).
Activation of STAT signaling, as well as other pathways downstream of JAKS (e.g.,
Akt), has been correlated with poor prognosis in many cancer types (Bowman, T., et
al. Oncogene 19:2474—2488, 2000). ed levels of circulating cytokines that
signal through JAK/STAT play a causal role in cachexia and/or chronic fatigue. As
such, JAK inhibition may be beneficial to cancer patients for reasons that extend
beyond potential anti-tumor activity.
JAK2 ne kinase can be beneficial for ts with myeloproliferative
disorders, e. g., polycythemia vera (PV), essential thrombocythemia (ET), myeloid
metaplasia with myelofibrosis (MMM) (Levin, et al, Cancer Cell, vol. 7, 2005: 387—
397). Inhibition of the JAK2V617F kinase decreases proliferation of hematopoietic
cells, suggesting JAK2 as a potential target for pharmacologic inhibition in patients
with PV, ET, and MMM.
Inhibition of the JAKs may benefit patients suffering from skin immune
disorders such as psoriasis, and skin ization. The maintenance of psoriasis is
believed to depend on a number of inflammatory cytokines in addition to various
chemokines and growth factors (JCI, 11321664—1675), many of which signal through
JAKs (Adv col. 2000;47:113—74).
Due to the usefulness of compounds which inhibit JAK in targeting
augmentation or suppression of the immune and inflammatory pathways (such as
immunosuppressive agents for organ transplants), as well as the treatment of
autoimmune es, diseases involving a hyperactive inflammatory response (e.g.,
eczema), allergies, cancer (e. g., prostate, leukemia, multiple myeloma), and some
immune ons (e.g., skin rash or contact dermatitis or diarrhea) caused by other
therapeutics, there is a need for improved formulations for administering JAK
kinases. The dosages forms described herein, as well as the doses and methods
described supra are directed toward this need and other ends.
SUMMARY
JAK inhibitors are described in US. Serial No. 13/043,986 (US
2011/0224190), filed March 9, 2011, which is incorporated herein by reference in its
entirety, including { l- { l - [3 (trifluoromethyl)isonicotinoyl]piperidinyl} -3 -
[4-(7H-pyrrolo[2,3 imidinyl)— 1 H-pyrazol- l -yl]azetidin—3 -yl} acetonitrile,
which is ed below as Formula I.
NC”\
N N
The present application provides, inter alia, sustained—release dosage forms
comprising about 25 mg to about 600 mg (e. g., 25 mg, 100 mg, 200 mg, 300 mg, or
600 mg) on a free base basis of {l— { l—[3—fluoro-2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof.
The present invention further provides one or more sustained release dosage
forms each comprising {1-{1-[3-fluoro(trifluoromethy1)isonicotinoyl]piperidin
yl} -3 -[4-(7H-pyrrolo [2,3 imidinyl)- l H-pyrazol- l -yl]azetidin
yl}acetonitrile, or a pharmaceutically able salt thereof; wherein said one or
more sustained e dosage forms together provide a once—daily oral dosage of
about 400 mg to about 600 mg on a free base basis of {l-{ l—[3 —fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— azol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient.
The present invention also provides a dose, comprising one or more sustained
release dosage forms each sing {1—{ l—[3—fluoro-2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—1H—pyrazol—l-yl]azetidin-3—yl}acetonitrile, or a pharmaceutically acceptable salt
thereof; n said dose provides a once—daily oral dosage of about 400 mg to about
600 mg on a free base basis of {l—{ 1-[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)-lH-pyrazol-l-yl]azetidin-3 -y1}acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient.
The present application further provides one or more sustained e dosage
forms as described herein, which together provide a once—daily oral dosage of about
600 mg on a free base basis of {l—{ 1—[3-fluoro—2-
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient.
The present application also es a dose comprising one or more sustained
e dosage forms as described herein, which together provide a once—daily oral
dosage of about 600 mg on a free base basis of {l—{l—[3-fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient.
The present application further provides methods of ng an autoimmune
disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption e, or organ transplant rejection in a t in need thereof,
comprising orally administering to said patient one or more sustained release dosage
forms as described herein.
The present application also provides methods of treating an autoimmune
e, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption e, or organ transplant rejection in a patient in need thereof,
sing orally administering to said t a once-daily dose of about 400 mg to
about 600 mg on a free base basis of {l— { 1-[3 —fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} itrile, or a pharmaceutically acceptable salt
thereof, wherein the dose comprises one or more sustained release dosage forms each
comprising { l- { l oro-2—(trifluoromethyl)isonicotinoyl]piperidinyl} [4-
(7H-pyrrolo[2,3 -d]pyrimidin-4—yl)— lH—pyrazol— l -yl]azetidiny1} acetonitrile, or a
pharmaceutically acceptable salt thereof.
The present application further provides methods of treating an autoimmune
disease, a cancer, a myeloproliferative er, an atory disease, a bone
resorption disease, or organ transplant rejection in a patient in need thereof,
comprising orally administering to said patient one or more sustained release dosage
as described herein.
The present application also provides methods of treating an autoimmune
disease, a cancer, a myeloproliferative disorder, an inflammatory e, a bone
resorption disease, or organ transplant rejection in a patient in need thereof, wherein
the method comprises orally administering to said patient the one or more ned
release dosage forms as a once—daily dosage of about 600 mg on a free base basis of
{ l- { l—[3 -fluoro(trifluoromethyl)isonicotinoy1]piperidiny1} -3 -[4-(7H—
pyrrolo [2,3 -d]pyrimidinyl)— l H—pyrazol- l -yl]azetidin-3 -yl} acetonitrile, or a
pharmaceutically acceptable salt thereof.
DESCRIPTION OF DRAWINGS
FIG. lA-C depicts plasma concentrations for the compound of Formula I
(Mean 1 SE) in healthy subjects receiving single doses of 300 mg IR capsules (1A:
Cohorts l-4, fasted), SRl, SR2, SR3, and SR4 tablets (2B: Cohorts l-4, fasted; and
2C: Cohorts 1—4, fed a high-fat meal).
—B depicts single—dose 300 mg SR3 PK profiles (Mean i SE) (2A:
Cohort 3, SR3, fasted versus high-fat meal; and 2B: Cohort 5, SR3, fasted versus
medium-fat meal).
depicts a comparison of PK profiles (mean i SE) between the 25 mg
and 100 mg SR3 s (treatment A vs C) and the food effect of a high—fat meal on
the 25 mg SR3 tablet (treatment B vs A).
depicts the percent change from baseline for hemoglobin for several
dosing regimens for ned release tablets versus placebo.
a) depicts the percentage of ts having a 2 50% reduction in total
symptom score (TSS) at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600
mg QD).
b) depicts the percent change in total symptom score (TSS) from
ne at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD).
a) depicts mean hemoglobin levels over time by dose cohort (100 mg
BID, 200 mg BID, and 600 mg QD).
b) depicts mean hemoglobin levels (g/dL) over time by dose cohort
(100 mg BID, 200 mg BID, and 600 mg QD) at 48 weeks.
c) depicts mean hemoglobin levels (g/dL) over time by dose cohort at
48 weeks as an average for three dose cohorts as compared to individuals dosed with
o or ruxolitinib.
DETAILED DESCRIPTION
The present application provides sustained—release dosage forms comprising
{ l- { l—[3 (trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-
pyrrolo[2,3-d]pyrimidin—4-yl)- l H—pyrazol- 1 etidin—3-yl} acetonitrile, or a
pharmaceutically acceptable salt thereof. In some embodiments, the present
application provides a sustained—release dosage form comprising about 25 mg to
about 600 mg on a free base basis of {1— { l-[3 -fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl} [4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof.
In some embodiments, the sustained—release dosage form comprises about 300
mg on a free base basis of {l—{ l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin-
4-yl} -3 - -pyrrolo[2,3 -d]pyrimidinyl)— lH-pyrazol- l -yl]azetidin—3 -
tonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the sustained—release dosage form comprises about 200
mg on a free base basis of {l- { l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin-
4-yl} -3 - —pyrrolo[2,3 -d]pyrimidin—4-yl)- lH-pyrazol- l -yl]azetidin—3 -
yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the sustained—release dosage form ses about 100
mg on a free base basis of {l- { luoro(trifluoromethyl)isonicotinoyl]piperidin-
4-yl} -3—[4-(7H—pyrrolo[2,3 -d]pyrimidin—4-yl)- 1 H—pyrazolyl]azetidin—3 —
yl}acetonitrile, or a pharmaceutically acceptable salt thereof.
In some embodiments, the sustained-release dosage form comprises about 300
mg on a free base basis of {l- { l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin-
4-y1} [4-(7H-pyrrolo[2,3 -d]pyrimidinyl)—lH-pyrazolyl]azetidin-3 -
tonitrile adipic acid salt.
In some embodiments, the sustained—release dosage form comprises about 200
mg on a free base basis of {l- { l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin-
4-yl} —3 - [4-(7H-pyrrolo[2,3 -d]pyrimidinyl)— l H—pyrazolyl]azetidin—3 -
yl}acetonitrile adipic acid salt.
In some embodiments, the sustained—release dosage form comprises about 100
mg on a free base basis of {l- { l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin-
4-yl} —3 - [4-(7H-pyrrolo[2,3 -d]pyrimidinyl)— 1 H—pyrazol- l -yl]azetidin—3 -
yl}acetonitrile adipic acid salt.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a mean peak plasma concentration (Cmax) of {1- { l-[3 -fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)—lH-pyrazol-l-yl]azetidinyl}acetonitrile of about 100 nM to about 1000 nM. As
used in this context, oral stration means that a single dose is administered to
the individual (in this case, 3 x 100 mg) and the PK ter is calculated from the
measurements of plasma concentration over time. In this context, the PK parameter
(in this case, Cmax) is being used to characterize the single sustained release dosage
form (i.e., the claims are directed to a single dosage form, not three dosage forms).
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a mean peak plasma concentration (Cmax) of {1-{1-[3-fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3—yl}acetonitrile of about 400 nM to about 700 nM.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a mean time to peak plasma concentration (Tmax) of {l- {l-[3-fluoro—2—
oromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)—lH—pyrazol—l—yl]azetidin—3-yl}acetonitrile of about 0.5 hours to about 3 hours.
In some embodiments of the sustained-release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a mean time to peak plasma concentration (Tmax) of {1- fluoro
oromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidin—3—yl}acetonitrile of at least 0.5 hours.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma
concentration ) of { l— { l—[3 -fluoro(trifluoromethyl)isonicotinoy1]piperidin
yl} -3 —[4-(7H—pyrrolo [2,3 -d]pyrimidin—4-yl)- l H—pyrazol- l -yl]azetidin-3—
yl}acetonitrile of about 5 to about 50.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted dual
provides a ratio of mean peak plasma tration (Cmax) to mean 12-hour plasma
concentration (Cizh) of {l—{l-[3—fluoro—2—(trifluoromethyl)isonicotinoyl]piperidin—4-
yl} -3 -[4-(7H-pyrrolo [2,3 -d]pyrimidinyl)- l H-pyrazol- l -yl]azetidin-3—
yl}acetonitrile of about 9 to about 40.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma
concentration (C1211) of {1— { 1—[3-fluoro—2—(trifluoromethy1)isonicotinoy1]piperidin-4—
yl} -3 H-pyrrolo [2,3 -d]pyrimidinyl)- l H—pyrazol— l -yl]azetidin
yl}acetonitrile of about 15 to about 30.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a mean half-life (ti/2) of {l—{ 1—[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—1H—pyrazol—1—y1]azetidin-3—yl}acetonitrile of about 5 hours to about 15 hours.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
es a mean half—life (ti/2) of [3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl} [4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)—lH—pyrazol—l—yl]azetidin—3-yl}acetonitrile of about 7 hours to about 12 hours.
In some ments of the sustained-release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a mean half—life (ti/2) of {1-{1-[3—fluoro-2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—1H-pyrazol-1—yl]azetidin—3—y1}acetonitrile of about 1 hour to about 20 hours.
In some ments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
provides a mean bioavailability (AUCO—oo) of {1—{1-[3-fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3—yl}acetonitrile of about 1000 nM*h to about 4000
nM*h.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to a fasted individual
es a mean bioavailability (AUCO-oo) of {1—{1-[3-fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—1H-pyrazoly1]azetidinyl}acetonitrile of about 1500 nM*h to about 3100
nM*h.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high—fat meal provides a mean peak plasma concentration (Cmax) of {1— { 1-[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidin-3—y1}acetonitrile of about 200 nM to about 2000 nM.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high-fat meal provides a mean peak plasma concentration (Cmax) of {1-{1—[3-fluoro—2—
oromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3—yl}acetonitrile of about 500 nM to about 1500 nM.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high-fat meal provides a mean time to peak plasma concentration (Tmax) of {l— {1-[3-
2-(trifluoromethyl)isonicotinoy1]piperidiny1}[4-(7H-pyrrolo[2,3-
midinyl)—lH-pyrazol-l-yl]azetidinyl}acetonitrile of about 1 hour to about
9 hours.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high-fat meal es a mean time to peak plasma concentration (Tmax) of {1- {1-[3-
fluoro—2-(trifluoromethyl)isonicotinoy1]piperidinyl}—3-[4-(7H—pyrrolo[2,3-
d]pyrimidin-4—yl)-lH-pyrazol-l-yl]azetidin-3 -y1}acetonitrile of at least 1.5 hours.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-
hour plasma concentration (Cizh) of { 1—{1-[3-fluoro—2-
oromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3—yl}acetonitrile of about 10 to about 70.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
at meal provides a ratio of mean peak plasma tration (Cmax) to mean 12-
hour plasma concentration (Cizh) of {1- {1-[3-fluoro
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—1H-pyrazoly1]azetidin—3—y1}acetonitrile of about 15 to about 50.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high-fat meal es a ratio of mean peak plasma concentration (Cmax) to mean 12-
hour plasma concentration (Cizh) of {l— {l-[3-fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)—lH-pyrazol-l-yl]azetidin-3—yl}acetonitrile of about 25 to about 45.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an dual after a
high-fat meal provides a mean half-life (ti/2) of {l— { l-[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—1H—pyrazol—1—yl]azetidin-3—yl}acetonitrile of about 1 hour to about 7 hours.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high-fat meal es a mean half-life (ti/2) of {l- { l-[3-fluoro—2—
(trifluoromethy1)isonicotinoy1]piperidinyl}[4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)—lH—pyrazol—l—yl]azetidin—3—yl}acetonitrile of about 2 hours to about 5 hours.
In some embodiments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high—fat meal provides a mean bioavailability (AUCO—oo) of {l- {1-[3 -fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)—lH-pyrazol-l-yl]azetidin-3—yl}acetonitrile of about 2000 nM*h to about 5000
nM*h.
In some ments of the sustained—release dosage form comprising about
100 mg, oral administration of three of said dosage forms to an individual after a
high-fat meal provides a mean bioavailability (AUCO—oo) of {l- {1-[3 -fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)—lH-pyrazol-l-yl]azetidinyl}acetonitrile of about 3000 nM*h to about 4000
nM*h.
In some embodiments, the t geometric mean ratio of the sustained
release dosage form relative to an immediate release dosage form for Cmax is about
% to about 30%, wherein one or more immediate release dosage forms and one or
more sustained release dosage forms are independently orally administered to fasted
individuals as a single dose, wherein the same size dose of {l—{ uoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidin-3—yl}acetonitrile, or a pharmaceutically acceptable salt,
is administered.
In some embodiments, the t ric mean ratio of the sustained
e dosage form relative to an immediate release dosage form for Cmax is about
% to about 30%, n one or more immediate release dosage forms and one or
more sustained release dosage forms are independently orally administered to fasted
individuals as a single dose, wherein the same size dose of {l-{ l—[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3—yl}acetonitrile, or a pharmaceutically acceptable salt,
is administered.
In some embodiments, the percent geometric mean ratio of the sustained
release dosage form relative to an immediate release dosage form for AUCO—oo is about
40% to about 55%, wherein one or more immediate release dosage forms and one or
more sustained release dosage forms are independently orally administered to fasted
individuals as a single dose, wherein the same size dose of {l—{ 1-[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidin-3—yl}acetonitrile, or a pharmaceutically acceptable salt,
is stered.
In some ments, the percent geometric mean ratio for Cmax of the
sustained release dosage form orally administered to an individual after a high-fat
meal ve to the sustained release dosage form orally stered to a fasted
individual is about 150% to about 250%.
In some embodiments, the percent geometric mean ratio for AUCO—oo of the
sustained release dosage form orally administered to an individual after a high-fat
meal relative to the sustained release dosage form orally administered to a fasted
dual is about 125% to about 170%.
In some embodiments, the sustained—release dosage forms of the ion
may include a sustained-release matrix former. Example sustained-release matrix
formers include cellulosic ethers such as hydroxypropyl methylcellulose (HPMC,
hypromellose) which is a high viscosity polymer, and methyl celluloses. Example
hydroxypropyl methylcelluloses include MethocelTM K15M, MethocelTM K4M,
MethocelTM KIOOLV, MethocelTM E3, MethocelTM E5, MethocelTM E6, MethocelTM
E15, MethocelTM E50, MethocelTM E10M, MethocelTM E4M, and MethocelTM E10M.
In some embodiments, the sustained release dosage form comprises one or more
hypromelloses. In some embodiments, the sustained release dosage form comprises a
first hypromellose characterized by having an apparent viscosity at a concentration of
2% in water of about 80 CF to about 120 cP and a second hypromellose characterized
by having an apparent viscosity at a concentration of 2% in water of about 3000 CF to
about 5600 CR In some embodiments, the sustained release dosage form comprises
about 8% to about 20% by weight of one or more hypromelloses. In some
embodiments, the sustained e dosage form comprises about 10% to about 15%
by weight of one or more hypromelloses.
In some embodiments, the sustained-release dosage forms of the invention can
further include one or more , glidants, disintegrants, binders, or ants as
ve ingredients. In some embodiments, the filler ses rystalline
cellulose, lactose monohydrate, or both. In some embodiments, the sustained release
dosage form comprises about 16% to about 22% by weight of microcrystalline
cellulose. In some ments, the sustained release dosage form comprises about
45% to about 55% by weight of lactose monohydrate,
In some embodiments, lubricants can be present in the dosage forms of the
invention in an amount of 0 to about 5% by weight. Non-limiting examples of
lubricants include magnesium stearate, stearic acid (stearin), hydrogenated oil,
polyethylene glycol, sodium stearyl fumarate, and glyceryl behenate. In some
ments, the formulations include magnesium stearate, stearic acid, or both. In
some embodiments, the sustained release dosage form comprises about 0.3% to about
0.7% by weight of magnesium stearate.
In some ments, glidants may be present in the dosage forms. In some
embodiments, glidants can be present in the dosage forms of the invention in an
amount of 0 to about 5% by weight. Non-limiting examples of glidants include talc,
colloidal silicon dioxide, and cornstarch. In some embodiments, the glidant is
colloidal silicon dioxide.
In some embodiments, film—coating agents can be present in an amount of 0 to
about 5% by . miting rative examples of film-coating agents
include hypromellose or polyvinyl alcohol based coating with titanium dioxide, talc
and optionally colorants available in several commercially ble te coating
systems.
In some embodiments, the ned release dosage form comprises
pregelatinized starch.
In some embodiments, the sustained release dosage form is a .
In some embodiments, the sustained release dosage form is prepared by
process comprising wet granulation.
In some embodiments, the sustained release dosage form comprises one or
more excipients independently selected from hypromelloses and microcrystalline
celluloses.
In some embodiments, the sustained release dosage form comprises one or
more ents ndently selected from hypromelloses, microcrystalline
celluloses, magnesium stearate, lactose, and lactose monohydrate.
In some embodiments, the sustained release dosage form ses one or
more excipients independently selected from hypromelloses, microcrystalline
celluloses, magnesium stearate, lactose, lactose monohydrate, and pregelatinized
starch.
The present invention further provides one or more sustained e dosage
forms each comprising {1-{ l—[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin
yl} -3 -[4-(7H-pyrrolo[2,3-d]pyrimidin-4—yl)- l H—pyrazol— l -yl]azetidin-3—
tonitrile, or a pharmaceutically acceptable salt thereof; wherein said one or
more sustained release dosage forms er provide a once—daily oral dosage of
about 400 mg to about 600 mg on a free base basis of {l—{ l—[3 —fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient.
The present invention also provides a dose, comprising one or more sustained
release dosage forms each comprising {1-{1-[3-fluoro
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)— lH-pyrazol-l-y1]azetidin—3 —y1} acetonitrile, or a pharmaceutically able salt
thereof; wherein said dose provides a once—daily oral dosage of about 400 mg to about
600 mg on a free base basis of {1—{1—[3-fluoro—2-
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— azol-l-yl]azetidin-3 —yl} acetonitrile, or a ceutically acceptable salt
thereof, to a patient.
The present application further provides one or more sustained release dosage
forms as described herein, which together provide a once—daily oral dosage of about
600 mg on a free base basis of {l—{ uoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient.
The present application further provides one or more sustained release dosage
forms as described herein, which together provide a once-daily oral dosage of about
500 mg on a free base basis of {l— {l—[3-fluoro—2—
(trifluoromethy1)isonicotinoyl]piperidiny1}[4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a t.
The present application further provides one or more sustained release dosage
forms as described herein, which together e a once—daily oral dosage of about
400 mg on a free base basis of {1—{1—[3—fluoro-2—
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a ceutically acceptable salt
thereof, to a patient.
In some embodiments, the one or more sustained release dosage forms are six
dosage forms of about 100 mg on a free base basis of {1—{1—[3-fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—1H—pyrazol—l-yl]azetidin-3 —yl}acetonitrile, or a pharmaceutically acceptable salt
thereof, are provided. In some embodiments, the one or more sustained release
dosage forms are three dosage forms of about 200 mg on a free base basis of {1-{1-
[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} -3 - [4-(7H-pyrrolo [2,3-
d]pyrimidin—4—yl)—1H—pyrazol—1-yl]azetidin—3—y1}acetonitrile, or a pharmaceutically
acceptable salt thereof, are ed. In some embodiments, the one or more
sustained release dosage forms are two dosage forms of about 300 mg on a free base
basis of {l-{ l—[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-
pyrrolo [2,3 -d]pyrimidinyl)— l H—pyrazol- l -y1]azetidin-3 -yl}acetonitri1e, or a
pharmaceutically acceptable salt thereof, are provided. In some embodiments, the one
or more sustained e dosage forms is one dosage form of about 600 mg on a free
base basis of {l- { l—[3 —fluoro-2—(trifluoromethyl)isonicotinoyl]piperidin—4-yl} -3—[4-
(7H-pyrrolo[2,3 -d]pyrimidinyl)- lH—pyrazol— l etidinyl} acetonitrile, or a
pharmaceutically acceptable salt thereof, is provided.
The present application also provides a dose comprising one or more sustained
release dosage forms as described herein, which provide a once—daily oral dosage of
about 600 mg on a free base basis of {1— { l-[3 -fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl} -pyrrolo[2,3 -d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a ceutically acceptable salt
thereof, to a patient.
The present application also provides a dose comprising one or more sustained
e dosage forms as described herein, which provide a once—daily oral dosage of
about 500 mg on a free base basis of {l—{l-[3-fluoro
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— azol-l-yl]azetidin-3 —yl} itrile, or a pharmaceutically acceptable salt
thereof, to a patient.
The present application also provides a dose comprising one or more sustained
release dosage forms as described herein, which e a once—daily oral dosage of
about 400 mg on a free base basis of {1— { l—[3 —fluoro—2—
oromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient.
In some embodiments, the dose comprises six dosage forms of about 100 mg
on a free base basis of {1- {1-[3-fluoro(trifluoromethyl)isonicotinoy1]piperidin
yl} -3 -[4-(7H-pyrrolo [2,3 -d]pyrimidinyl)- l H-pyrazol- l -yl]azetidin
yl}acetonitrile, or a pharmaceutically able salt thereof. In some embodiments,
the dose comprises three dosage forms of about 200 mg on a free base basis of { l—{l—
[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidin-4—yl} -3 - [4—(7H—pyrrolo [2,3—
d]pyrimidin-4—yl)— lH-pyrazol-l-yl]azetidinyl} acetonitrile, or a ceutically
acceptable salt thereof. In some ments, the dose comprises two dosage forms
of about 300 mg on a free base basis of {l— {l—[3-fluoro—2—
oromethyl)isonicotinoyl]piperidiny1}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— lH—pyrazol—l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof. In some embodiments, the dose comprises one dosage form of about 600 mg
on a free base basis of {1— { l—[3—fluoro—2—(trifluoromethy1)isonicotinoy1]piperidin—4—
yl} -3 —[4-(7H-pyrrolo [2,3 -d]pyrimidinyl)- l H-pyrazol- l -yl]azetidin-3—
yl}acetonitrile, or a pharmaceutically acceptable salt thereof
The present application further provides a kit comprising one or more
sustained release dosage forms as bed herein, which er provide a once-
daily oral dosage of about 400 mg to about 600 mg on a free base basis of {l— { 1—[3—
fluoro(trifluoromethyl)isonicotinoyl]piperidiny1}[4-(7H-pyrrolo[2,3-
d]pyrimidin-4—yl)— lH-pyrazol-l-yl]azetidinyl} acetonitrile, or a pharmaceutically
acceptable salt thereof, to a patient. In some embodiments, the kit further ses
an instruction to administer the one or more sustained release dosage forms as a once—
daily dose of about 400 mg to about 600 mg on a free base basis of {1—{ l-[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof.
The present application further provides a kit comprising one or more
sustained release dosage forms as described herein, which together provide a once—
daily oral dosage of about 600 mg on a free base basis of {l— { l—[3—fluoro—2—
oromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} itrile, or a pharmaceutically acceptable salt
thereof, to a patient. In some embodiments, the kit further comprises an instruction to
administer the one or more sustained release dosage forms as a once—daily dose of
about 600 mg on a free base basis of {1-{1-[3-fluoro
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)— lH-pyrazol-l-y1]azetidin—3 —y1} acetonitrile, or a pharmaceutically acceptable salt
thereof.
The present application further provides a kit comprising one or more
sustained release dosage forms as described herein, which together provide a once—
daily oral dosage of about 500 mg on a free base basis of {l—{l—[3—fluoro-2—
oromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient. In some embodiments, the kit further comprises an instruction to
administer the one or more sustained release dosage forms as a once—daily dose of
about 600 mg on a free base basis of {1— { 1-[3 —2—
uoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3 —yl}acetonitrile, or a pharmaceutically acceptable salt
thereof.
The t application further provides a kit comprising one or more
sustained e dosage forms as described herein, which er provide a once—
daily oral dosage of about 400 mg on a free base basis of {1-{1-[3-fluoro
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin—3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, to a patient. In some embodiments, the kit further comprises an instruction to
administer the one or more sustained release dosage forms as a once—daily dose of
about 600 mg on a free base basis of {l— { l-[3 -fluoro—2-
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof.
In some embodiments, the kit ses six dosage forms of about 100 mg on
a free base basis of [3-fluoro—2-(trifluoromethyl)isonicotinoyl]piperidin-4—yl}—
3 H-pyrrolo[2,3 -d]pyrimidinyl)— lH-pyrazol- l -yl] azetidin-3 -yl} acetonitrile, or
a pharmaceutically acceptable salt thereof. In some embodiments, the kit comprises
three dosage forms of about 200 mg on a free base basis of {l—{ l—[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl} [4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof. In some embodiments, the kit comprises two dosage forms of about 300 mg
on a free base basis of {l- { l —[3 -2—(trifluoromethyl)isonicotinoyl]piperidin-4—
yl} -3 —[4-(7H—pyrrolo [2,3 -d]pyrimidin—4-yl)- l H—pyrazol- l -yl]azetidin-3—
yl}acetonitrile, or a pharmaceutically acceptable salt thereof. In some embodiments,
the kit comprises one dosage form of about 600 mg on a free base basis of {l- { 1-[3—
fluoro—2-(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H-pyrrolo[2,3-
d]pyrimidin-4—yl)— lH—pyrazol—l-yl]azetidinyl} acetonitrile, or a pharmaceutically
acceptable salt f.
As used , “sustained—release” is used as generally understood in the art
and refers to a formulation designed to slowly release the active ient into a
patient after oral administration.
As used herein, “dose” refers to the total amount of the compound of Formula
I orally administered to the individual or patient. The dose may be in a single dosage
form, or a plurality of dosage forms (e.g., a 600 mg dose may be one 600 mg dosage
form, two 300 mg dosage forms, three 200 mg dosage forms, six 100 mg dosage
forms, etc.). Hence, a dose can refer to a plurality of pills to be taken by a patient at
nearly simultaneously.
As used herein, “a fasted individual” means an individual who has fasted for at
least 10 hours prior to administration of the dose.
As used herein, "mean" when preceding a pharmacokinetic value (e. g. mean
Cmax) represents the arithmetic mean value of the pharmacokinetic value taken from a
population of patients unless otherwise specified.
As used herein, "Cmax" means the m observed plasma concentration.
As used herein, “C1211” refers to the plasma concentration measured at 12 hours
from administration.
As used herein, "Tmax" refers to the time at which the maximum blood plasma
concentration is observed.
As used herein, “Ti/2” refers to the time at which the plasma concentration is
half of the observed maximum.
As used herein, "AUC" refers to the area under the plasma concentration-time
curve which is a measure of total bioavailability.
As used herein, "AUCo-oo" refers to the area under the plasma tration-
time curve extrapolated to infinity.
As used herein, "AUCo-t" refers to the area under the plasma concentration-
time curve from time 0 to the last time point with a quantifiable plasma concentration,
y about 12—36 hours.
As used herein, "AUCoJ' refers to the area under the plasma tration-
time curve from time 0 to the time of the next dose.
As used herein, “Cl/F” refers to oral clearance.
The present invention also includes pharmaceutically acceptable salts of the
compounds described herein. As used herein, "pharmaceutically acceptable salts"
refers to derivatives of the disclosed compounds wherein the parent compound is
modified by converting an existing acid or base moiety to its salt form. Examples of
pharmaceutically acceptable salts include, but are not limited to, mineral or organic
acid salts of basic residues such as ; alkali or c salts of acidic residues
such as ylic acids; and the like. The pharmaceutically acceptable salts of the
present invention include the non-toxic salts of the parent compound formed, for
example, from non-toxic inorganic or organic acids. The pharmaceutically acceptable
salts of the present invention can be sized from the parent compound which
contains a basic or acidic moiety by tional chemical methods. Generally, such
salts can be prepared by reacting the free acid or base forms of these compounds with
a stoichiometric amount of the appropriate base or acid in water or in an organic
solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl
acetate, alcohols (e. g., methanol, l, opanol, or butanol) or acetonitrile
(ACN) are preferred. Lists of suitable salts are found in Remington ’s Pharmaceutical
Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418 and
Journal ofPharmaceutical Science, 66, 2 (1977), each of which is incorporated herein
by reference in its entirety. In some embodiments, the compounds described herein
include the N—oxide forms.
Methods
The present application further provides methods of treating an autoimmune
disease, a , a roliferative disorder, an inflammatory disease, a bone
resorption disease, or organ transplant rejection in a patient in need thereof,
comprising orally administering to said patient one or more ned release dosage
forms as described herein.
The t application also provides a method of treating an autoimmune
disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption e, or organ transplant rejection in a patient in need thereof,
comprising orally stering to said patient a aily dose of about 400 mg to
about 600 mg on a free base basis of {l- { 1-[3 —fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)—1H-pyrazolyl]azetidin-3—yl}acetonitrile, or a pharmaceutically acceptable salt
thereof, wherein the dose comprises one or more sustained release dosage forms each
comprising { 1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidinyl}[4-
rrolo[2,3 imidin-4—yl)- lH—pyrazol— 1 —yl]azetidiny1} acetonitrile, or a
pharmaceutically acceptable salt f. The present application further provides
a method of treating an autoimmune e, a cancer, a myeloproliferative disorder,
an inflammatory disease, a bone resorption disease, or organ transplant ion in a
patient in need thereof, comprising orally administering to said patient one or more
sustained release dosage as described .
The present application also provides a method of treating an autoimmune
disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption disease, or organ transplant rejection in a patient in need thereof, wherein
the method comprises orally administering to said patient the one or more sustained
release dosage forms as a once-daily dosage of about 600 mg on a free base basis of
{ 1-{1—[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} —3 -[4-(7H-
pyrrolo [2,3 -d]pyrimidinyl)— l H—pyrazol- l -yl]azetidin-3 -yl} acetonitrile, or a
pharmaceutically acceptable salt thereof.
The present application also provides a method of treating an autoimmune
disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption disease, or organ transplant rejection in a patient in need thereof, wherein
the method comprises orally administering to said patient the one or more sustained
release dosage forms as a once—daily dosage of about 500 mg on a free base basis of
{1-{1-[3 (trifluoromethyl)isonicotinoyl]piperidinyl}-3 -[4-(7H-
pyrrolo [2,3 imidinyl)- 1 H-pyrazol- l -yl]azetidin-3 -yl} acetonitrile, or a
pharmaceutically acceptable salt thereof.
The present application also provides a method of treating an autoimmune
disease, a cancer, a myeloproliferative er, an inflammatory disease, a bone
resorption disease, or organ transplant rejection in a patient in need f, wherein
the method comprises orally administering to said patient the one or more sustained
release dosage forms as a once—daily dosage of about 400 mg on a free base basis of
{ l- { l—[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H—
pyrrolo [2,3 -d]pyrimidinyl)- l H-pyrazol- l -yl]azetidin—3 -yl} acetonitrile, or a
ceutically acceptable salt thereof.
In some embodiments of the methods in the preceding three paragraphs, the
one or more sustained release dosage forms are six dosage forms of about 100 mg on
a free base basis of {1—{1—[3-fluoro—2-(trifluoromethyl)isonicotinoyl]piperidin-4—yl}—
3 -[4-(7H-pyrrolo[2,3 -d]pyrimidinyl)— lH-pyrazol- l -yl] azetidin-3 -yl} acetonitrile, or
a pharmaceutically acceptable salt thereof, are provided. In some embodiments of the
methods in the preceding three paragraphs, the one or more sustained release dosage
forms are three dosage forms of about 200 mg on a free base basis of {1-{1-[3 -
fluoromethyl)isonicotinoyl]piperidinyl} -3 H-pyrrolo[2,3 -d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin—3 —yl} acetonitrile, or a ceutically acceptable salt
f, are provided. In some embodiments of the methods in the preceding three
aphs, the one or more sustained release dosage forms are two dosage forms of
about 300 mg on a free base basis of {1— { l-[3 -fluoro—2-
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— lH—pyrazol—l-yl]azetidin-3 —yl} itrile, or a pharmaceutically acceptable salt
thereof, are provided. In some embodiments of the methods in the preceding three
paragraphs, the one or more sustained release dosage forms is one dosage form of
about 600 mg on a free base basis of {1— { l-[3 -fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, is provided.
In some embodiments, oral administration of one or more sustained release
dosage forms to a fasted individual provides a mean time to peak plasma
concentration (Tmax) of {l- { l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin
yl} —3 —[4-(7H—pyrrolo[2,3-d]pyrimidin—4—yl)—lH-pyrazolyl]azetidin—3—
yl}acetonitrileof about 0.5 hours to about 3 hours.
In some embodiments,oral administration of one or more sustained release
dosage forms to a fasted individual provides a mean time to peak plasma
concentration (Tmax) of {l-{l—[3-fluoro(trifluoromethyl)isonicotinoy1]piperidin
yl} -3 -[4-(7H-pyrrolo [2,3 imidinyl)- lH—pyrazol— l -yl]azetidin-3—
yl}acetonitrile of at least 0.5 hours.
In some embodiments,oral administration of one or more sustained e
dosage forms to a fasted individual provides a ratio of mean peak plasma
concentration (Cmax) to mean 12—hour plasma concentration (Cm) of {l—{ uoro—
fluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3—yl}acetonitrile of about 5 to about 50.
In some embodiments,oral administration of one or more sustained release
dosage forms to a fasted individual provides a ratio of mean peak plasma
concentration (Cmax) to mean 12-hour plasma tration (Cizh) of {l-{ l-[3-fluoro—
2-(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidin-3—yl}acetonitrile of about 9 to about 40.
In some ments,oral stration of one or more sustained release
dosage forms to a fasted individual provides a ratio of mean peak plasma
concentration (Cmax) to mean 12-hour plasma concentration (Cizh) of {l—{ l-[3-fluoro—
2-(trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidin-3—yl}acetonitrile of about 15 to about 30.
In some embodiments, oral administration of one or more sustained release
dosage forms to a fasted individual es a mean half-life (ha) of {l—{ l-[3-fluoro—
2-(trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)—lH—pyrazol—l-yl]azetidin-3—yl}acetonitrileof about 1 hour to about 20 hours.
In some embodiments, oral administration of one or more sustained e
dosage forms to an individual after a high-fat meal provides a mean time to peak
plasma concentration (Tmax) of {l-{ l-[3-fluoro-2—
(trifluoromethyl)isonicotinoyl]piperidiny1}[4-(7H-pyrrolo[2,3 -d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidinyl}acetonitrileof about 1 hour to about 9 hours.
In some embodiments, oral administration of one or more sustained release
dosage forms to an dual after a high-fat meal provides a mean time to peak
plasma concentration (Tmax) of {l-{ 1—[3-fluoro—2-
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)—lH-pyrazol-l-yl]azetidin-3—yl}acetonitrile of at least 1.5 hours.
In some embodiments, oral administration of one or more sustained release
dosage forms to an individual after a high-fat meal provides a ratio of mean peak
plasma concentration (Cmax) to mean 12—hour plasma concentration (Cizh) of {l- { l-[3-
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H-pyrrolo[2,3-
d]pyrimidin-4—yl)—lH-pyrazol-l—yl]azetidin-3 -yl}acetonitrile of about 10 to about 70.
In some embodiments, oral stration of one or more sustained release
dosage forms to an dual after a high-fat meal provides a ratio of mean peak
plasma concentration (Cmax) to mean 12-hour plasma concentration (C12h)0f {l—{ l-[3-
fluoro(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-
d]pyrimidinyl)-lH-pyrazol-l-yl]azetidin-3 -yl}acetonitrile of about 15 to about 50.
In some embodiments, oral administration of one or more sustained release
dosage forms to an dual after a at meal provides a ratio of mean peak
plasma concentration (Cmax) to mean 12-hour plasma concentration (C12h) of {1—{ l-[3—
fluoro—2-(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3-
d]pyrimidin-4—yl)-lH—pyrazol—l-yl]azetidin-3 —yl}acetonitrile of about 25 to about 45.
In some embodiments, oral administration of one or more sustained release
dosage forms to an individual after a high-fat meal provides a mean half-life (ti/2) of
{ l- { l —[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} —3 -[4-(7H-
o[2,3-d]pyrimidinyl)—lH—pyrazol-l-yl]azetidinyl}acetonitrile of about 1
hour to about 7 hours.
In some embodiments, oral administration of one or more sustained release
dosage forms to an dual after a high-fat meal provides a mean ife (he) of
{ l- { l—[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} -3 H-
pyrrolo[2,3-d]pyrimidin—4-yl)-lH—pyrazol-l-yl]azetidin—3—yl}acetonitrile of about 2
hours to about 5 hours.
In some embodiments, the one or more sustained release dosage forms are
each a tablet. In some embodiments, the one or more sustained release dosage forms
are prepared by process comprising wet granulation.
In some embodiments, the one or more sustained release dosage forms each
comprises one or more hypromelloses. In some embodiments, the one or more
sustained release dosage forms each comprises one or more excipients independently
selected from hypromelloses and rystalline celluloses. In some embodiments,
the one or more sustained release dosage forms each comprises one or more
ents independently selected from hypromelloses, microcrystalline celluloses,
magnesium stearate, lactose, and lactose monohydrate. In some embodiments, the
one or more sustained release dosage forms each comprises a first hypromellose
characterized by having an apparent viscosity at a concentration of 2% in water of
about 80 CF to about 120 cP and a second hypromellose characterized by having an
nt viscosity at a concentration of 2% in water of about 3000 CF to about 5600
In some embodiments, the one or more sustained e dosage forms each
comprises about 10% to about 15% by weight of one or more elloses. In some
embodiments, the one or more sustained release dosage forms each comprises about
16% to about 22% by weight of microcrystalline cellulose. In some embodiments, the
one or more sustained release dosage forms each comprises about 45% to about 55%
by weight of lactose monohydrate. In some embodiments, the one or more sustained
release dosage forms each comprises about 0.3% to about 0.7% by weight of
magnesium stearate.
In some embodiments, the t application provides a method of treating
myelofibrosis in a patient, comprising orally administering to said patient a once—daily
dose of about 400 mg to about 600 mg on a free base basis of {1- fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidin—4-yl}-3—[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— lH-pyrazol-l-yl]azetidin-3 -yl} itrile, or a pharmaceutically acceptable salt
thereof, wherein the dose comprises one or more sustained release dosage forms each
comprising { l- { l -[3-fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} [4-
(7H-pyrrolo[2,3 —d]pyrimidin-4—yl)-lH—pyrazol—l—yl]azetidiny1}acetonitrile, or a
pharmaceutically acceptable salt thereof; wherein the method results in a d total
symptom score (TSS) of said patient compared with baseline. In some ments,
the present application provides a method of treating myelofibrosis in a patient,
comprising orally administering to said patient the one or more sustained release
dosage forms as a once—daily dosage of about 600 mg on a free base basis of {l—{ l-
[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidin-4—yl} -3 - [4—(7H—pyrrolo [2,3—
d]pyrimidin-4—yl)—lH-pyrazolyl]azetidinyl}acetonitrile, or a pharmaceutically
acceptable salt thereof; wherein the method s in a reduced total symptom score
(TSS) of said patient compared with ne.
In some embodiments, the present application provides a method of treating
myeloflbrosis in a patient, comprising orally administering to said patient the one or
more sustained release dosage forms as a once—daily dosage of about 500 mg on a free
base basis of {l- { l—[3 -2—(trifluoromethy1)isonicotinoyl]piperidin—4-yl} -3—[4-
(7H-pyrrolo[2,3 -d]pyrimidinyl)- lH—pyrazol— l -yl]azetidinyl} acetonitrile, or a
pharmaceutically acceptable salt thereof; wherein the method s in a reduced total
symptom score (TSS) of said patient compared with baseline.
In some embodiments, the present ation provides a method of ng
myelofibrosis in a patient, comprising orally administering to said patient the one or
more sustained release dosage forms as a once-daily dosage of about 400 mg on a free
base basis of {l- { l—[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidin—4-yl} -3—[4-
(7H—pyrrolo[2,3 -d]pyrimidin—4-yl)-lH—pyrazol—l-yl]azetidin-3—y1}acetonitrile, or a
pharmaceutically able salt thereof; wherein the method results in a reduced total
symptom score (TSS) of said patient compared with baseline.
In some embodiments of the methods in the preceding three paragraphs, the
one or more sustained release dosage forms are six dosage forms of about 100 mg on
a free base basis of {1—{ l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin-4—yl} [4-(7H—pyrrolo[2,3 —d]pyrimidinyl)— lH—pyrazol- l -yl] azetidin-3 -yl} acetonitrile, or
a pharmaceutically acceptable salt thereof, are provided. In some ments of the
methods in the preceding three paragraphs, the one or more sustained release dosage
forms are three dosage forms of about 200 mg on a free base basis of {l- {l—[3 —fluoro—
2-(trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-pyrrolo[2,3 -d]pyrimidin
—pyrazol—l-yl]azetidin-3 etonitrile, or a pharmaceutically acceptable salt
thereof, are ed. In some embodiments of the methods in the preceding three
paragraphs, the one or more sustained release dosage forms are two dosage forms of
about 300 mg on a free base basis of {l- { l—[3—fluoro—2—
(trifluoromethy1)isonicotinoyl]piperidin-4—y1}—3-[4—(7H—pyrrolo[2,3 —d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, are provided. In some embodiments of the methods in the preceding three
paragraphs, the one or more sustained release dosage forms is one dosage form of
about 600 mg on a free base basis of {l— { l-[3 —fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin—4-
yl)— azol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically acceptable salt
thereof, is provided.
In some embodiments, “total symptom score (TS S)” refers to the TSS derived
from the modified Myelofibrosis Symptom Assessment Form (MFSAF) (e.g., v3.0)
onic diary as compared with baseline (baseline is the patient’s baseline TSS
before treatment). In some embodiments, myelofibrosis is primary myelofibrosis
(PMF), post-polycythemia vera MF, or post-essential thrombocythemia MP.
The present application also provides a method of treating an autoimmune
disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
tion disease, or organ transplant rejection in a patient in need thereof,
comprising orally administering to said patient a once-daily dose of about 400 mg to
about 600 mg on a free base basis of {l— { 1-[3 —fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— lH-pyrazol-l-yl]azetidin-3 —yl} acetonitrile, or a pharmaceutically able salt
thereof, wherein the dose comprises one or more sustained e dosage forms each
comprising { l- { l -[3-fluoro-2—(trifluoromethyl)isonicotinoyl]piperidin—4-yl} [4-
(7H-pyrrolo[2,3 imidin-4—yl)— azol— l -yl]azetidinyl} acetonitrile, or a
pharmaceutically acceptable salt thereof; wherein said method results in reduced
anemia.
The present application also provides a method of ng an autoimmune
e, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption disease, or organ transplant rejection in a patient in need thereof, n
the method comprises orally administering to said patient the one or more sustained
release dosage forms as a once—daily dosage of about 600 mg on a free base basis of
{l— { l -[3 -fluoro(trifluoromethy1)isonicotinoyl]piperidinyl}-3 -[4-(7H-
pyrrolo [2,3 -d]pyrimidinyl)- l H-pyrazol- l -yl]azetidin-3 -yl} acetonitrile, or a
pharmaceutically able salt thereof; wherein said method results in d
anemia.
The present application also provides a method of treating an autoimmune
disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption disease, or organ transplant rejection in a patient in need thereof, wherein
the method comprises orally administering to said t the one or more sustained
release dosage forms as a once-daily dosage of about 500 mg on a free base basis of
{ l- { l —[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} —3 -[4-(7H-
pyrrolo [2,3 -d]pyrimidinyl)— l H-pyrazol- l -yl]azetidin-3 -yl} acetonitrile, or a
pharmaceutically acceptable salt f; wherein said method results in reduced
anemia.
The present application also provides a method of treating an mune
disease, a cancer, a myeloproliferative disorder, an inflammatory disease, a bone
resorption disease, or organ tranSplant rejection in a patient in need thereof, wherein
the method comprises orally administering to said patient the one or more sustained
release dosage forms as a once-daily dosage of about 400 mg on a free base basis of
{ l- { l—[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-
pyrrolo [2,3 -d]pyrimidinyl)— l H—pyrazol- l -yl]azetidin-3 -yl} acetonitrile, or a
pharmaceutically acceptable salt thereof; wherein said method s in reduced
anemia. In some ments, the one or more ned release dosage forms are
six dosage forms of about 100 mg on a free base basis of {l—{ l—[3—fluoro—2—
(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-d]pyrimidin
yl)— lH—pyrazol—l-yl]azetidin-3 —yl} acetonitrile, or a ceutically acceptable salt
thereof, are provided. In some embodiments, the one or more sustained release
dosage forms are three dosage forms of about 200 mg on a free base basis of {l—{ l—
[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} -3 - [4—(7H-pyrrolo [2,3—
d]pyrimidin-4—yl)— lH-pyrazol-l—yl]azetidinyl} itrile, or a ceutically
acceptable salt thereof, are provided. In some embodiments, the one or more
sustained release dosage forms are two dosage forms of about 300 mg on a free base
basis of {l-{ l-[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidinyl} -3 -[4-(7H-
pyrrolo[2,3-d]pyrimidinyl)- l H-pyrazol- l -yl]azetidinyl} acetonitrile, or a
ceutically able salt thereof, are provided. In some embodiments, the one
or more ned release dosage forms is one dosage form of about 600 mg on a free
base basis of {l- { l—[3 -fluoro(trifluoromethyl)isonicotinoyl]piperidin—4-yl} -3—[4-
(7H—pyrrolo[2,3 -d]pyrimidin—4-yl)- lH—pyrazol— l -yl]azetidin-3—yl} acetonitrile, or a
pharmaceutically acceptable salt thereof, is provided.
Reduced anemia is relative to that experienced for a twice—daily dose of 200
mg on a free base basis of {l— { l-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin-
4-yl} —3 - [4-(7H-pyrrolo[2,3 -d]pyrimidinyl)— l H—pyrazol- l -yl]azetidin—3 -
yl}acetonitrile, or a pharmaceutically acceptable salt thereof, wherein the dose
comprises one or more ned release dosage forms each comprising {1— {1—[3-
fluoro—2-(trifluoromethyl)isonicotinoyl]piperidinyl}—3-[4-(7H—pyrrolo[2,3-
d]pyrimidin-4—yl)— lH-pyrazol—l-yl]azetidinyl} acetonitrile, or a ceutically
acceptable salt thereof.
The compound of Formula I is a JAK inhibitor. A JAKl selective inhibitor is
a compound that inhibits JAKl activity preferentially over other Janus kinases. JAKl
plays a central role in a number of cytokine and growth factor signaling pathways
that, when dysregulated, can result in or contribute to disease states. For example, IL-
6 levels are elevated in rheumatoid arthritis, a disease in which it has been suggested
to have detrimental effects (Fonesca, J.E. et al., munity Reviews, 8:53 8—42,
2009). Because IL-6 signals, at least in part, through JAKl, antagonizing IL-6
directly or ctly through JAKl inhibition is expected to provide clinical benefit
(Guschin, D., N., et al Embo J 14:1421, 1995; Smolen, J. S., et a1. Lancet 371:987,
2008). Moreover, in some cancers JAKl is mutated resulting in constitutive
undesirable tumor cell growth and survival (Mullighan CG, Proc Natl Acad Sci U S
A. 10629414—8, 2009; Flex E., et all Exp Med. 205:751—8, 2008). In other
autoimmune diseases and cancers elevated systemic levels of inflammatory nes
that te JAKl may also contribute to the disease and/or associated symptoms.
Therefore, patients with such diseases may benefit from JAKl inhibition. Selective
inhibitors of JAKl may be efficacious while avoiding unnecessary and potentially
undesirable effects of inhibiting other JAK kinases.
Selective inhibitors of JAKl, relative to other JAK kinases, may have multiple
therapeutic advantages over less selective inhibitors. With respect to selectivity
t JAK2, a number of important cytokines and growth factors signal through
JAK2 including, for example, erythropoietin (Epo) and thrombopoietin (Tpo)
(Parganas E, et al. Cell. 93:3 85—95, 1998). Epo is a key growth factor for red blood
cells production; hence a paucity of Epo-dependent signaling can result in reduced
numbers of red blood cells and anemia (Kaushansky K, NEJM 354:2034—45, 2006).
Tpo, another example of a JAK2-dependent growth factor, plays a l role in
controlling the proliferation and maturation of megakaryocytes — the cells from which
platelets are ed (Kaushansky K, NEJM 354:2034-45, 2006). As such, reduced
Tpo signaling would se megakaryocyte numbers (megakaryocytopenia) and
lower circulating platelet counts (thrombocytopenia). This can result in undesirable
and/or uncontrollable bleeding. Reduced inhibition of other JAKs, such as JAK3 and
Tyk2, may also be desirable as humans lacking functional version of these kinases
have been shown to suffer from numerous maladies such as severe—combined
immunodeficiency or hyperimmunoglobulin E syndrome (Minegishi, Y, et al.
Immunity 25:745-55, 2006; Macchi P, et al. Nature. 377:65-8, 1995). ore a
JAKl inhibitor with reduced y for other JAKs would have significant
advantages over a less-selective inhibitor with respect to reduced side effects
involving immune ssion, anemia and thrombocytopenia.
Another aspect of the present invention pertains to methods of treating a JAK-
associated disease or disorder in an individual (e. g., patient) by administering to the
individual in need of such treatment a sustained-release dosage form of the ion.
A JAK—associated disease can include any disease, disorder or condition that is
directly or indirectly linked to expression or activity of the JAK, ing
overexpression and/or abnormal activity . A JAK—associated disease can also
include any e, er or condition that can be prevented, ameliorated, or cured
by modulating JAK activity.
Examples of JAK—associated diseases include diseases involving the immune
system including, for example, organ transplant rejection (e. g., allograft rejection and
graft versus host disease).
Further examples of JAK—associated diseases include autoimmune diseases
such as multiple sclerosis, rheumatoid arthritis, juvenile tis, psoriatic tis,
type I diabetes, lupus, psoriasis, inflammatory bowel disease, ulcerative colitis,
Crohn’s disease, myasthenia gravis, immunoglobulin nephropathies, myocarditis,
autoimmune thyroid disorders, c obstructive pulmonary disease (COPD), and
the like. In some embodiments, the autoimmune disease is an autoimmune bullous
skin disorder such as pemphigus vulgaris (PV) or bullous pemphigoid (BP).
Further examples of JAK—associated diseases include allergic conditions such
as asthma, food allergies, eszematous itis, contact dermatitis, atopic dermatitis
(atropic ), and rhinitis. Further examples of JAK—associated diseases include
viral diseases such as Epstein Barr Virus (EBV), Hepatitis B, Hepatitis C, HIV,
HTLV l, Varicella-Zoster Virus (VZV) and Human Papilloma Virus (HPV).
Further es of sociated disease include diseases associated with
cartilage turnover, for example, gouty arthritis, septic or ious arthritis, reactive
arthritis, reflex sympathetic phy, algodystrophy, Tietze syndrome, costal
athropathy, osteoarthritis deformans endemica, Mseleni disease, Handigodu disease,
degeneration resulting from fibromyalgia, systemic lupus erythematosus, scleroderma,
or ankylosing spondylitis.
Further examples of JAK-associated disease include congenital cartilage
malformations, ing hereditary chrondrolysis, chrondrodysplasias, and
pseudochrondrodysplasias (e.g., ia, enotia, and metaphyseal
chrondrodysplasia).
Further examples of JAK—associated diseases or conditions include skin
disorders such as psoriasis (for e, psoriasis vulgaris), atopic dermatitis, skin
rash, skin irritation, skin sensitization (e. g., contact dermatitis or allergic contact
dermatitis). For e, n substances including some pharmaceuticals when
topically d can cause skin sensitization. In some embodiments, co—
stration or sequential administration of at least one JAK tor of the
invention together with the agent causing unwanted sensitization can be l in
treating such unwanted sensitization or dermatitis. In some embodiments, the skin
disorder is treated by topical administration of at least one JAK inhibitor of the
invention.
In further embodiments, the JAK—associated e is cancer including those
characterized by solid tumors (e. g., prostate cancer, renal cancer, hepatic cancer,
pancreatic , gastric cancer, breast cancer, lung cancer, cancers of the head and
neck, thyroid cancer, glioblastoma, Kaposi’s sarcoma, Castleman’s disease, uterine
leiomyosarcoma, melanoma etc), hematological cancers (e.g., lymphoma, leukemia
such as acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML) or
multiple myeloma), and skin cancer such as cutaneous T-cell lymphoma (CTCL) and
ous B—cell ma. Example CTCLs e Sezary syndrome and mycosis
des.
In some embodiments, the dosage forms described herein, or in combination
with other JAK inhibitors, such as those ed in US. Ser. No. 11/637,545, which
is incorporated herein by reference in its entirety, can be used to treat inflammation-
associated cancers. In some embodiments, the cancer is ated with inflammatory
bowel disease. In some embodiments, the inflammatory bowel disease is ulcerative
colitis. In some embodiments, the inflammatory bowel disease is Crohn’s disease. In
some embodiments, the inflammation-associated cancer is colitis—associated cancer.
In some embodiments, the inflammation-associated cancer is colon cancer or
colorectal cancer. In some embodiments, the cancer is gastric cancer, gastrointestinal
carcinoid tumor, gastrointestinal stromal tumor (GIST), adenocarcinoma, small
intestine cancer, or rectal cancer.
JAK—associated diseases can further include those characterized by expression
of: JAK2 mutants such as those having at least one mutation in the pseudo-kinase
domain (e.g., JAK2V617F); JAKZ mutants having at least one mutation outside of
the pseudo-kinase domain; JAKl s; JAK3 mutants; erythropoietin receptor
(EPOR) mutants; or deregulated expression of CRLF2.
JAK—associated es can further include myeloproliferative disorders
(MPDs) such as polycythemia vera (PV), essential thrombocythemia (ET),
myelofibrosis with myeloid metaplasia (MMM), primary myelofibrosis (PMF),
chronic myelogenous leukemia (CML), chronic myelomonocytic leukemia (CMML),
hypereosinophilic syndrome (HES), ic mast cell disease (SMCD), and the like.
In some embodiments, the myeloproliferative disorder is myelofibrosis (e.g., primary
myelofibrosis (PMF) or post polycythemia vera/essential thrombocythemia
myelofibrosis (Post-PV/ET MF)). In some ments, the myeloproliferative
er is post— essential thrombocythemia myeloflbrosis (Post—ET). In some
embodiments, the roliferative disorder is post themia vera myelofibrosis
(Post-PV MF).
In some embodiments, dosage forms described herein can be used to treat
pulmonary arterial hypertension.
The present invention further provides a method of treating dermatological
side effects of other pharmaceuticals by administration of the dosage forms of the
invention. For example, numerous ceutical agents result in unwanted allergic
reactions which can manifest as acneiform rash or related dermatitis. Example
pharmaceutical agents that have such undesirable side effects include anti—cancer
drugs such as gefltinib, cetuximab, erlotinib, and the like. The dosage forms of the
invention can be administered systemically in ation with (e.g., aneously
or sequentially) the pharmaceutical agent having the undesirable dermatological side
effect.
r JAK-associated diseases include ation and inflammatory
diseases. Example inflammatory diseases include sarcoidosis, inflammatory diseases
of the eye (e. g., iritis, uveitis, scleritis, conjunctivitis, or related disease),
inflammatory diseases of the respiratory tract (e. g., the upper respiratory tract
including the nose and sinuses such as rhinitis or sinusitis or the lower respiratory
tract including bronchitis, chronic obstructive pulmonary disease, and the like),
inflammatory myopathy such as myocarditis, and other inflammatory es. In
some embodiments, the inflammation disease of the eye is blepharitis.
The dosage forms described herein can further be used to treat ischemia
reperfusion injuries or a disease or condition related to an inflammatory ic
event such as stroke or cardiac arrest. The dosage forms described herein can further
be used to treat xin—driven disease state (e. g., complications after bypass
surgery or chronic endotoxin states contributing to chronic cardiac failure). The
dosage forms described herein can further be used to treat ia, cachexia, or
fatigue such as that resulting from or associated with cancer. The dosage forms
described herein can further be used to treat restenosis, dermitis, or fibrosis.
The dosage forms described herein can further be used to treat conditions associated
with a or astrogliosis such as, for example, diabetic retinopathy, cancer, or
neurodegeneration. See, e.g., Dudley, A.C. et al. Biochem. J. 2005, 390(Pt —36
and Sriram, K. et a]. J. Biol. Chem. 2004, 279(19):19936-47. Epub 2004 Mar 2, both
of which are incorporated herein by reference in their entirety. The JAK inhibitors
bed herein can be used to treat Alzheimer’s disease.
The dosage forms described herein can further be used to treat other
inflammatory diseases such as ic inflammatory response syndrome (SIRS) and
septic shock.
The dosage forms bed herein can further be used to treat gout and
increased prostate size due to, e.g., benign prostatic hypertrophy or benign prostatic
hyperplasia.
Further JAK—associated diseases include bone resorption diseases such as
osteoporosis, osteoarthritis. Bone resorption can also be associated with other
conditions such as al imbalance and/or hormonal therapy, autoimmune disease
(e.g. osseous sarcoidosis), or cancer (e.g. myeloma). The reduction of the bone
resorption due to the the compound of Formula I can be about 10%, about 20%,
about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, or about 90%.
In some embodiments, the dosage forms bed herein can further be used
to treat a dry eye disorder. As used herein, “dry eye er” is intended to
encompass the disease states summarized in a recent official report of the Dry Eye
Workshop (DEWS), which defined dry eye as “a multifactorial e of the tears
and ocular surface that results in symptoms of discomfort, Visual disturbance, and tear
film instability with potential damage to the ocular surface. It is accompanied by
increased osmolarity of the tear film and inflammation of the ocular surface.” Lemp,
“The Definition and fication of Dry Eye Disease: Report of the Definition and
Classification Subcommittee of the International Dry Eye Workshop”, The Ocular
Surface, 5(2), 75—92 April 2007, which is incorporated herein by reference in its
entirety. In some embodiments, the dry eye disorder is selected from aqueous tear—
deficient dry eye (ADDE) or evaporative dry eye disorder, or appropriate
combinations f. In some embodiments, the dry eye disorder is Sjogren
syndrome dry eye (SSDE). In some embodiments, the dry eye disorder is non—
Sjogren syndrome dry eye (NSSDE).
In a further aspect, the t invention provides a method of treating
conjunctivitis, uveitis (including chronic uveitis), chorioditis, retinitis, cyclitis,
sclieritis, episcleritis, or iritis; treating inflammation or pain related to l
transplant, LASIK (laser assisted in situ keratomileusis), photorefractive keratectomy,
or LASEK (laser assisted sub—epithelial keratomileusis); inhibiting loss of visual
acuity related to corneal transplant, LASIK, photorefractive keratectomy, or LASEK;
or inhibiting lant rejection in a patient in need thereof, sing administering
to the patient a dosage form of the invention.
Additionally, the dosage forms of the ion, or in ation with other
JAK inhibitors, such as those reported in US. Ser. No. 11/637,545, which is
incorporated herein by nce in its entirety, can be used to treat respiratory
dysfunction or failure associated with viral infection, such as influenza and SARS.
In some ments, the present invention provides a dosage form as
described in any of the embodiments herein, for use in a method of treating any of the
diseases or disorders described herein. In some embodiments, the present invention
provides the use of a dosage form as described in any of the embodiments herein, for
the preparation of a ment for use in a method of treating any of the diseases or
disorders described herein.
In some embodiments, the present invention provides a dosage form as
described herein, or a pharmaceutically acceptable salt thereof, for use in a method of
modulating JAKl. In some embodiments, the present invention also provides use of a
dosage form as bed herein, or a ceutically acceptable salt f, for the
preparation of a medicament for use in a method of modulating JAKl.
As used herein, the term “individual” is a human. In some embodiments, the
human is an adult subject.
As used herein, the term “treating” or “treatment” refers to one or more of (l)
inhibiting the disease; for example, inhibiting a disease, condition or disorder in an
individual who is experiencing or displaying the pathology or symptomatology of the
disease, condition or disorder (i.e., arresting r development of the pathology
and/or symptomatology); and (2) ameliorating the disease; for example, ameliorating
a disease, condition or disorder in an individual who is encing or displaying the
pathology or symptomatology of the disease, condition or disorder (i.e., reversing the
pathology and/or symptomatology) such as decreasing the severity of disease.
Combination Therapies
One or more additional pharmaceutical agents such as, for example,
chemotherapeutics, anti-inflammatory , steroids, immunosuppressants, as well
as Bcr—Abl, Flt-3, RAF and FAK kinase inhibitors such as, for e, those
described in WC 2006/0563 99, which is incorporated herein by reference in its
entirety, or other agents can be used in combination with the dosage forms described
herein for treatment of JAK—associated diseases, disorders or conditions. The one or
more additional pharmaceutical agents can be stered to a t
simultaneously or sequentially.
Example chemotherapeutics include proteosome inhibitors (e.g., bortezomib),
thalidomide, id, and maging agents such as melphalan, doxorubicin,
cyclophosphamide, vincristine, etoposide, carmustine, and the like.
Example steroids include coriticosteroids such as dexamethasone or
prednisone.
Example l inhibitors include the compounds, and pharmaceutically
acceptable salts thereof, of the genera and species disclosed in US. Pat. No.
,521,184, WO 04/005281, and US. Ser. No. ,491, all ofwhich are
incorporated herein by reference in their entirety.
Example suitable Flt-3 tors include compounds, and their
pharmaceutically acceptable salts, as disclosed in WC 03/037347, WO 03/099771,
and WO 04/046120, all of which are incorporated herein by reference in their entirety.
Example suitable RAF inhibitors include compounds, and their
ceutically acceptable salts, as disclosed in WO 95 and WO 05/028444,
both of which are incorporated herein by reference in their entirety.
Example suitable FAK inhibitors include compounds, and their
pharmaceutically acceptable salts, as disclosed in WC 04/080980, WO 04/056786,
WO 03/024967, WO 01/064655, WO 595, and WO 01/014402, all ofwhich
are orated herein by reference in their entirety.
In some ments, one or more of the dosage forms of the invention can
be used in combination with one or more other kinase inhibitors including imatinib,
particularly for treating patients resistant to imatinib or other kinase inhibitors.
In some embodiments, one or more dosage forms of the invention can be used
in combination with a chemotherapeutic in the treatment of cancer, such as multiple
myeloma, and may improve the treatment response as compared to the response to the
chemotherapeutic agent alone, without exacerbation of its toxic effects. Examples of
additional pharmaceutical agents used in the treatment of le myeloma, for
example, can include, without limitation, melphalan, melphalan plus prednisone
[MP], doxorubicin, dexamethasone, and Velcade (bortezomib). Further additional
agents used in the treatment of multiple myeloma e Bcr-Abl, Flt-3, RAF and
FAK kinase inhibitors. Additive or synergistic effects are ble outcomes of
ing a dosage form of the present invention with an additional agent.
Furthermore, resistance of multiple myeloma cells to agents such as dexamethasone
may be reversible upon treatment with a dosage form of the present invention. The
agents can be combined with the present compounds in a single or continuous dosage
form, or the agents can be administered simultaneously or tially as separate
dosage forms.
In some embodiments, a corticosteroid such as dexamethasone is administered
to a patient in combination with at the dosage form of the invention where the
thasone is administered intermittently as opposed to continuously.
In some further embodiments, combinations of one or more IAK inhibitors of
the invention with other eutic agents can be stered to a t prior to,
during, and/or after a bone marrow transplant or stem cell transplant.
In some ments, the additional therapeutic agent is fluocinolone
acetonide (Retisert®), or rimexolone (AL-2178, Vexol, Alcon).
In some embodiments, the additional therapeutic agent is cyclosporine
(Restasis®).
In some embodiments, the additional therapeutic agent is a osteroid. In
some ments, the corticosteroid is triamcinolone, dexamethasone, fluocinolone,
cortisone, prednisolone, or flumetholone.
In some embodiments, the additional therapeutic agent is ed from
DehydrexTM (Holles Labs), Civamide (Opko), sodium hyaluronate (Vismed,
Lantibio/TRB Chemedia), cyclosporine (ST-603, Sirion Therapeutics), ARG101(T)
sterone, is), AGR1012(P) (Argentis), ecabet sodium (Senju-Ista),
gefarnate (Santen), 15-(s)-hydroxyeicosatetraenoic acid (15(S)-HETE), cevilemine,
doxycycline (ALTY-0501, Alacrity), minocycline, iDestrinTM (NP50301, Nascent
Pharmaceuticals), cyclosporine A (Nova22007, Novagali), oxytetracycline
(Duramycin, MOLI1901, Lantibio), CF101 (2S,3S,4R,5R)—3,4—dihydroxy—5—[6—[(3—
iodophenyl)methylamino]purinyl]—N—methy1-oxolanecarbamyl, Can-Fite
Biopharma), voclosporin (LX212 or LX214, Lux Biosciences), ARG103 (Agentis),
RX-10045 (synthetic resolvin , Resolvyx), DYN15 (Dyanmis Therapeutics),
rivoglitazone (DEOl 1, Daiichi Sanko), TB4 (RegeneRx), OPH-Ol (Ophtalmis
Monaco), PCSlOl (Pericor Science), REV1—31 (Evolutec), Lacritin (Senju),
rebamipide (Otsuka—Novartis), OT-551 (Othera), PAI-2 (University of Pennsylvania
and Temple University), rpine, imus, pimecrolimus (AMS981, Novartis),
loteprednol etabonate, rituximab, diquafosol tetrasodium (INS365, Inspire), KLS—
0611 (Kissei Pharmaceuticals), dehydroepiandrosterone, anakinra, efalizumab,
mycophenolate sodium, etanercept l®), hydroxychloroquine, NGX267
(TorreyPines Therapeutics), a, gemcitabine, oxaliplatin, L-asparaginase, or
thalidomide.
In some embodiments, the additional therapeutic agent is an anti-angiogenic
agent, cholinergic agonist, TRP-l receptor modulator, a calcium channel blocker, a
mucin secretagogue, MUCl ant, a calcineurin inhibitor, a osteroid, a
P2Y2 receptor t, a muscarinic receptor agonist, an mTOR inhibitor, another
JAK inhibitor, Ber-Ab] kinase inhibitor, Flt-3 kinase inhibitor, RAF kinase inhibitor,
and FAK kinase inhibitor such as, for example, those described in WO 56399,
which is incorporated herein by reference in its entirety. In some embodiments, the
additional therapeutic agent is a tetracycline derivative (e.g., minocycline or
doxycline). In some embodiments, the additional therapeutic agent binds to FKBP12.
In some embodiments, the additional therapeutic agent is an alkylating agent
or DNA cross—linking agent; an anti-metabolite/demethylating agent (e.g., 5-
flurouracil, capecitabine or azacitidine); an anti-hormone therapy (e. g., hormone
receptor antagonists, SERMs, or aromotase inhibitor); a mitotic inhibitor (e. g.
Vincristine or paclitaxel); an topoisomerase (I or II) tor (e.g. mitoxantrone and
irinotecan); an apoptotic inducers (e. g. ABT—737); a nucleic acid therapy (e. g.
antisense or RNAi); nuclear receptor ligands (e.g., agonists and/or antagonists: all-
trans retinoic acid or bexarotene); epigenetic targeting agents such as histone
deacetylase inhibitors (e. g. vorinostat), hypomethylating agents (e.g. decitabine);
regulators of protein stability such as Hsp90 inhibitors, ubiquitin and/or ubiquitin like
conjugating or deconjugating molecules; or an EGFR inhibitor (erlotinib).
In some embodiments, the additional therapeutic agent(s) are demulcent eye
drops (also known as “artificial tears”), which include, but are not limited to,
compositions containing polyvinylalcohol, hydroxypropyl methylcellulose, in,
hylene glycol (e.g. PEG400), or carboxymethyl cellulose. Artificial tears can
help in the treatment of dry eye by compensating for reduced ning and
lubricating ty of the tear film. In some embodiments, the additional eutic
agent is a mucolytic drug, such as N-acetyl-cysteine, which can ct with the
mucoproteins and, therefore, to decrease the viscosity of the tear film.
In some embodiments, the additional therapeutic agent includes an antibiotic,
antiviral, antifungal, anesthetic, anti-inflammatory agents including steroidal and non-
steroidal anti-inflammatories, and anti-allergic agents. Examples of suitable
medicaments include aminoglycosides such as in, gentamycin, ycin,
streptomycin, netilmycin, and kanamycin; fluoroquinolones such as ciprofloxacin,
norfloxacin, in, trovafloxacin, lomefloxacin, xacin, and enoxacin;
naphthyridine; sulfonamides; xin; chloramphenicol; neomycin; mycin;
colistimethate; bacitracin; vancomycin; tetracyclines; rifampin and its tives
(“rifampins”); cycloserine; beta-lactams; cephalosporins; amphotericins; fluconazole;
flucytosine; natamycin; miconazole; nazole; corticosteroids; diclofenac;
flurbiprofen; ketorolac; suprofen; cromolyn; lodoxamide; levocabastin; naphazoline;
antazoline; pheniramine; or azalide antibiotic.
It is further appreciated that certain features of the ion, which are, for
y, described in the context of separate embodiments, can also be provided in
combination in a single embodiment (as if the embodiments of the specification are
written as multiply dependent claims).
e 1. Preparation of Sustained Release ations
Sustained release tablets were prepared with the excipients being in the
amounts shown in the table below. Protocol A was used for the SR1 tablets, protocol
B was used for the SR2 tablets, Protocol C was used for the SR3 tablets and the 25
mg SR tablets, and Protocol D was used for the SR4 tablets.
Protocol A:
Step 1. Individually screen the adipic acid salt of the compound of
Formula I, rystalline cellulose, hypromelloses cel K100 LV and
Methocel K4M), and lactose monohydrate.
Step 2. Transfer the ed material from Step 1 to a suitable blender
and mix.
Step 3. Transfer the blend from Step 2 to a suitable granulator and mix.
Step 4. Add purified water while mixing.
Step 5. Transfer the granules from Step 4 into a suitable dryer and dry
until LCD is less than 3%.
Step 6. Screen the granules from Step 5.
Step 7. Mix screened Magnesium Stearate with granules in Step 6 in a
suitable blender.
Step 8. ss the final blend in Step 7 on a suitable rotary tablet
press.
Protocol B:
Step 1. Individually screen the adipic acid salt of the compound of
Formula I, microcrystalline cellulose, hypromellose and pregelatinized starch.
Step 2. Transfer the screened material from Step 1 to a suitable blender
and mix.
Step 3. Transfer the blend from Step 2 to a suitable ator and mix.
Step 4. Add purified water while .
Step 5. Transfer the granules from Step 4 into a suitable dryer and dry
until LCD is less than 3%.
Step 6. Screen the granules from Step 5.
Step 7. Individually screened polyox, butylated hydroxytoluene and
colloidal silicone dioxide.
Step 8. Transfer the es from Step 6 and material from Step 7 into
a suitable r and mix.
Step 9. Add screened Magnesium Stearate to the material in Step 8 and
continue blending.
Step 10. Compress the final blend in Step 9 on a suitable rotary tablet
press.
Protocol C:
Step 1. Individually screen e monohydrate, the adipic acid salt of
the compound of Formula I, microcrystalline cellulose and hypromelloses through a
suitable screen.
Step 2. Transfer the screened material from Step 1 to a suitable blender
and mix.
Step 3. Transfer the blend from Step 2 to a suitable granulator and mix.
Step 4. Add purified water while mixing.
Step 5. Screen wet granules through a suitable screen.
Step 6. Transfer the granules from Step 5 into a suitable dryer and dry
until LCD is less than 3%.
Step 7. Mill the granules from Step 6.
Step 8. Mix screened magnesium stearate with granules in Step 7 in a
suitable blender.
Step 9. ss the final blend in Step 8 on a suitable rotary tablet
press.
Protocol D:
Step 1. dually screen pregelatinized starch, the adipic acid salt of
the compound of Formula I, hypromellose, and a portion of required microcrystalline
cellulose through a suitable screen.
Step 2. Transfer the screened material from Step 1 to a suitable blender
and mix.
Step 3. Transfer the blend from Step 2 to a suitable granulator and mix.
Step 4. Add purified water while mixing.
Step 5. Screen wet granules through a suitable screen.
Step 6. Transfer the granules from Step 5 into a le dryer and dry
until LOD is less than 3%.
Step 7. Mill the granules from Step 6.
Step 8. Screen the remaining portion of microcrystalline cellulose and
half of the sodium bicarbonate.
Step 9. Transfer the milled granules from Step 7 and screened
materials from Step 8 into a suitable blender and mix.
Step 10. Screen the remaining portion of sodium bicarbonate and mix
with blend in Step 9.
Step 11. Screen magnesium te and mix with blend in Step 10.
Step 12. Compress the final blend in Step 11 on a suitable rotary tablet
press.
SR1: Composition of 100 mg ned Release Tablets
Component Function Weight (mg/tablet) ition
(wt%)
Adipic acid salt of the Active 126.422 21.1
compound of Formula I a
MlClOCl'ystalllne Cellulose"m
Hypromellose
Release Control 10.0
(Methocel )
ellose
Release Control 10.0
(Methocel K4M)
Component Function Weight (mg/tablet) Composition
(wt%)
Lactose Monohydrate 290.5 8
Purified Water c Granulating q.s.
Liquid
a Conversion factor for adipate salt to free base is 0.7911
b Added after ation
c Removed during processing
SR2: Composition of 100 mg Sustained Release Tablets
Component Function Weight Composition
blet) (wt%)
Adipic acid salt of the Active
126.4 a 21.1
compound of Formula Ia
Microcrystalline Cellulose 180.0
ellose .
(MethoceleOLw "n
hylene Oxide
Release Control 1800
(Polyox WRS 1105) b
Pregelatinized Starch 101.6
Butylated Hydroxytoluene b 0.012 0.002
Purified Water ° Granulating
‘1' s
Liquid '
a Conversion factor for adipate salt to free base is 0.7911
b Added after granulation
c Removed during processing
SR3 (100 mg): Composition of 100 mg Sustained Release s
Component Function Weight Composition
(mg/tablet) (wt%)
Adipic acid salt of the Active
126.4 a 21.1
compound of Formula I21
Mlcrocrystalllne Flller
108.0 18.0
Cellulose
Hypromellose
Release Control 42 0 7 0
(Methocel KIOOLV) ' '
Hypromellose
Rdeasecmtml
<Methoce1K4M>
Purified Water C Granulating
q‘ s
Liquid '
a Conversion factor for adipate salt to free base is 0.7911
b Added after granulation
c Removed during processing
SR4: Composition of 100 mg ned e Tablets
ent Function Weight (mg/tablet) Composition
(wt%)
AdlplC ac1d salt of the Actlve
126.4 a 21.1
compound of Formula I21
Microcrystalline .
Hyprome11056
Release Control 210.0 35.0
(Methocel KIOOLV)
————
a Conversion factor for adipate salt to free base is 0.7911
b Added after granulation
c Removed during processing
d Partial added before and partial added after granulation
25mg SR: Composition of 25 mg Sustained e Tablets
Component Function Weight Composition
blet) (wt%)
Adlpic ac1d salt of the Active
31.6 a 12.6
compound of Formula Ial
Microcrystamnecaulose 105.0
Hypromellose,
Hypromellose,
Release Control 25.0 10.0
(Methocel K4M)
C Granulating
a Conversion factor for adipate salt to free base is 0.7911
b Added after granulation
c Removed during processing
Example 2. ation of the IR Formulation of the Compound of Formula I
The IR formulation used in the studies in Example 3 was prepared as 50 mg
capsules with the composition shown in the table below according to Protocol E
below.
Protocol E:
Step 1. x the required amount of the adipic acid salt of the compound of
Formula I and an approximately equal amount of silicified microcrystalline cellulose
(SMCC).
Step 2. Pass the mixture in Step 1 through a suitable screen (for example 40
mesh).
Step 3. Screen the remaining SMCC through the same screen used in Step 2.
Step 4. Blend the ed SMCC from Step 3 along with mixture from Step 2
in a suitable blender (for example Turbula blender) for approximately 5 minutes.
Step 5. Fill the blend into capsules to desired fill weight.
INGREDIENT WEIGHT QUANTITY
COMPOSITION PER UNIT
(0%) (mg)
Adipic acid salt of the compound of
.11 6320*
Silicified Microcrystalline Cellulose, NF
64.89
lv SMCC HD 90)
TOTAL 100.00 %
#2 Capsules, Hard Gelatin, White
Opaque
* Adipic acid salt of the nd of Formula I with salt conversion factor of 0.7911
Example 3. Relative Bioavailability Study of Sustained Release Dosage Forms
A total of 72 healthy adult subjects were enrolled in 6 cohorts (12 subjects per
cohort) and randomized to treatment sequences within each cohort according to a
randomization schedule. All treatments were single-dose administrations of the
compound of Formula I. There was a washout period of 7 days between the treatment
periods.
The SR1, SR2, SR3, and SR4 formulations were evaluated in Cohort l, Cohort
2, Cohort 3, and Cohort 4, respectively (see Example 1 for SR1, SR2, SR3, SR4, and
mg SR tablets used in study). The subjects received the IR and SR treatments
according to a 3—way crossover :
Treatment A: 300 mg (6 X 50 mg capsule) IR formulation of the compound of
Formula 1 stered orally after an overnight fast of at least 10 hours.
Treatment B: 300 mg (3 X 100 mg tablets) SR formulation of the compound
of a 1 administered orally after an overnight fast of at least 10 hours.
Treatment C: 300 mg (3 X 100 mg tablets) SR formulation of the compound
of Formula I administered orally after a high-fat meal.
The subjects in Cohort 5 ed the following treatments in a 2—way
crossover :
Treatment A: 300 mg (3 X 100 mg tablets of the compound of Formula I) SR3
administered orally after an overnight fast of at least 10 hours.
Treatment B: 300 mg (3 X 100 mg tablets of the compound of Formula I) SR3
administered orally after a medium—fat meal.
The subjects in Cohort 6 received the following ents in a 3—way
crossover design:
Treatment A: 50 mg (2 X 25 mg tablets of the nd of Formulal (25 mg
SR s from e 1)) administered orally after an overnight fast of at least 10
hours.
Treatment B: 50 mg (2 X 25 mg tablets of the compound of Formula I (25 mg
SR tablets from Example 1)) administered orally after a high-fat meal.
Treatment C: 100 mg (l X 100 mg tablets) SR3 administered orally after an
overnight fast of at least 10 hours.
Blood samples for determination of plasma concentrations of the compound of
Formula I were collected using lavender top (K2EDTA) Vacutainer® tubes at 0, 0.25,
0.5, l, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose.
Plasma samples were assayed by a validated, GLP, LC/MS/MS method with a
linear range of 5.0 to 5000 nM. Table 1 summarizes the accuracy and precision (CV
%) of the assay quality control samples during the analysis of the plasma samples
from this study.
Table 1: Accuracy and ion of the Plasma Assay Quality Control
Samples
-------- Low QC --------- -—----- Middle QC ------- —-------- High QC --
Analyte CV CV CV
Theo Accuracy Theo Accuracy Theo Accuracy
(Unit). % % %
Compound
of 15.0 99.0% 4.6% 250 101% 4.2% 4000 99.5% 2.2%
CV% =percent coefficient of variability; QC = quality control; Theo = theoretical or nominal
concentration.
For the PK analysis, the actual sample collection times were used. For any
sample with missing actual collection time, the scheduled time was used provided that
there was no protocol ion noted for the collection of these samples.
Standard noncompartmental PK methods were used to analyze the data for the
plasma concentration of the compound of Formula using Phoenix WinNonlin version
6.0 (Pharsight Corporation, in View, CA). Thus, Cmax and Tmax were taken
directly from the ed plasma concentration data. The terminal-phase disposition
rate constant (M) was estimated using a log-linear regression of the concentration data
in the terminal disposition phase, and W. was estimated as ln(2)/?tz. AUCO-t was
estimated using the linear oidal rule for sing concentrations and the log-
trapezoidal rule for decreasing concentrations, and the total AUCo.oo was calculated as
AUCO—t + Ct/kz. The oral—dose clearance (CL/F) was estimated as Dose/AUCo-oo, and
the terminal-phase volume of distribution (Vz/F) was estimated as Dose/[AUCo—oo*?tz].
The log—transformed Cmax and AUC values (after dose normalization, where
the doses were different) were compared between the fasted and fed dosing
treatments, and n the SR and IR dosing treatments, using a crossover ANOVA
(fixed factor = treatment, sequence and period, random effect = subject (sequence)).
The adjusted geometric mean ratios of Cmax and AUC between the treatments
(reference = IR or fasted administration of SR) and the corresponding 90% nce
intervals (CIs) were determined. In addition, the correlation between the observed
food effect of a high-fat meal on AUCo.00 and the relative bioavailability of the SR
formulations (with reference to the IR capsule) were explored by a quantile plot using
the data from all subjects who completed Treatment A, B, and C in Cohorts l to 4.
The statistical is was performed using Phoenix WinNonlin version 6.0.
presents plasma concentrations of the compound of Formula I (mean :
SE) for the subjects in Cohorts 1 to 4 following Treatment A (300 mg IR
administration in fasted state), Treatment B (300 mg SR stration in fasted
state), and ent C (300 mg SR administration with a high-fat meal).
compares the effect of a high-fat meal and -fat meal on the mean PK profile
following a single-dose 300 mg (3 X 100mg) administration of the compound of
Formula I SR3 tablets. presents plasma concentrations of the compound of
Formula I (mean :: SE) for the subjects in Cohort 6 following Treatment A (2 X 25 mg
SR tablet administration in fasted state), Treatment B (2 X 25 mg SR tablet with a
high—fat meal), and Treatment C (l X 100 mg SR3 administration in fasted state).
Tables 2A, 2B, 3A and 3B summarize mean PK parameters for subjects in
Cohorts l to 4, the ve ilability (reference = IR capsule) and food effect
(high-fat meal) for the 100 mg strength SRl-SR4 tablets. Table 4A and 4B
summarize mean PK parameters for ts in Cohort 5, and food effect m—fat
meal) for the 100 mg strength SR3 tablet. Table 5A and 5B summarizes mean PK
parameters for subjects in Cohort 6, the dose—normalized relative bioavailability
(reference = 100 mg SR3 tablet), and the food effect (high-fat meal) for the 25 mg SR
tablet.
Table 2A
Cmax Tmax tl/z
Cohort/Treatment 11 CmaX/Cl2h
(HM) (h) (h
Cohort 1
12 2.29 i 1 0
300 mg IR 197 :: 147 2.0 :: 0.27
0.50 (0.50—
(fasted) 159 2.0
2.24 2.0)
12 0 341 4 1 3
300 mg SR1 13.2 :: 7.8 9.2 :: 4.5
0 13 (0.50—
(fasted) 11.6
0.317 3.0)
12 0.610 d:
300 mg SR1 4'0 18.0 :: 6.4 3.2 :: 1.4
0.14
(high-fat meal) (2.0-8.0) 16.8 3.0
0.595
Cohort 2
12 2.05 d: 1.0
300 mg IR 130 i: 72.9 2.1 4: 0.34
0.67 (0.50—
(fasted) 112 2.1
1.92 3.0)
12 0.191 i
300 mg SR2 2'5 11.4 :1: 9.9 11 d: 8.4
0.10
(fasted) (1.040) 8.60 9.23
0.172
12 0.470 i
300 mg SR2 6'0 11.0 :1: 4.0 3.5 3:26
0.16
(high-fat meal) (1.5-6.0) 10.4 3.0
0443
Cohort 3
11 2.35 i 1.0
300 mg IR 136 :: 70.8 2.2 :: 0.53
0.41 (0.50—
(fasted) 120 2.2
2.31 2.0)
11 0.553 4 1.5
300 mg SR3 22.9 :: 13.4 9.8 :: 8.5
0.24 (0.50—
(fasted)
0.502 3.0)
12 1.05 i
300 mg SR3 4‘0 34.92: 15.8 3.3 :: 1.2
0.47
(high-fat meal) .0) 30.8 3.1
0.968
Cohort 4
12 2.94 i 1.0
300 mg IR 170 i 58.6 2.14:0.58
0.98 (0.25-
(fasted) 162 2.1
2.78 1.5)
12 0.321 i
300 mg SR4 2'0 10.3 i 6.0 7.3 i 5.3
0.27
(fasted) (1.5-8.1) 8.92 6.0
0.249
12 0.549 i
300 mg SR4 4'0 12.8 i 14.8 4.9 i 2.6
0.28
fat meal) (2.0-16) 6.06 4.4
0.481
Table 2B
Cohort/Treatment (1:512:13 83/31:; 8417141;
Cohort 1
300 mg IR 4148;: 4.45 1.00 127 27.1
(fasted) 4:33 4.35 124
300 mg SR1 10555: 1.65 0.54 359 106
(fasted) 1:47 1.57 345
300 mg SR1 268:: 2.91 0.65 194 39.9
(high-fat meal) 2:82 2.85 190
Cohort 2
300 mg IR 41435;: 4.47 :1: 1.36 134 :1: 50.1
(fasted) 4:24 4.27 127
300 mg SR2 37 1.17: 0.43 510:: 148
(fasted) 0.95 1.11 488
300 mg SR2 204:; 2.52 0.72 235 83.5
(high-fat meal) 2:38 2.42 224
Cohort 3
300 mg IR 55);); 5.03 :1: 1.34 115 :1: 32.4
(fasted) 4:83 4.87 111
300 mg SR3 20273: 2.39 :1: 0.70 248 i— 82.8
d) 2:17 2.29 236
300 mg SR3 3i5153i 3.59 :1: 1.13 165 :1: 50.2
(high-fat meal) 3:40 3.44 158
Cohort 4
300 mg IR 52213;: 5.25 2.15 117 39.8
(fasted) 4:88 4.90 111
300 mg SR4 2621: 1 70 1.25 456 259
(fasted) 1:31 1.40 387
300 mg SR4 20?: 3.13 1.20 200 80.0
(high-fat meal) 2:78 2.92 186
Table 3A
Cmax Tmax t1/2
Cohort/Treatment Cmax/C12h
(HM) (h) (h)
SR1 fasted vs IR 14.2%
-17.5%)
SR1 fed vs fasted 188%
(152%-232%)
SR2 fasted vs IR 8.9%
(6.7%-11.9%)
SR2 fed vs fasted 258%
(193%-344%)
SR3 fasted vs IR 22.3%
(17.4%-28.6%)
SR3 fed vs fasted 191%
(150%-244%)
SR4 fasted vs IR 9.0%
(6.8%-11.9%)
SR4 fed vs fasted 193%
( 146%—256%)
PK parameter values are mean :: SD and geometric mean except for Tm“, Where median
(90%
confidence interval) is reported.
Table 3B
/Treatment (1:{15/1311) 3:10) 841;]:
Geometric Mean Relative Bioavailability and the 90% nce Intervals
SR1 fasted vs IR 34.1% 36.1%
(31.3%-37.0%) (33.3%—39.2%)
SR1 fed vs fasted 191% 181%
(176%-208%) (167%-196%)
SR2 fasted vs IR 22.4% 26.0%
(18.3%-27.4%) (21.6%-31.3%)
SR2 fed vs fasted 250% 218%
(204%-306%) (181%-262%)
SR3 fasted vs IR 45.4% 47.5%
(39.6%-52.0%) (41.9%-53.9%)
SR3 fed vs fasted 151% 145%
(132%-173%) (128%-164%)
SR4 fasted vs IR 26.9% 28.5%
(21.6%-33.4%) (23.2%-35.l%)
SR4 fed vs fasted 213% 215%
(171%-264%) (172%-268%)
PK parameter values are mean :: SD and geometric mean except for Tm“, where median
(90% confidence interval) is reported.
Table 4A
Cohort/Tl‘e Cmax Tmax t‘/2
’1 Cmax/C12“
atment (uM) (h) (h)
Cohort 5
300 mg SR3 12 0.619i0.41 1.75 22.8 i: 16.7 7.73:5.2
d) 0.523 (0.5040) 17.8 6.2
ffiiifgfifi 12
0.875 i 0.47 2.5 40.6 :1: 22.7 3.6 2.0
0.764 (1.560) 31.2 3.3
meal)
Geometric Mean Relative Bioavailability and the 90% Confidence Intervals
146%
SR3 fed vs fasted
(105%—202%)
Pharmacokinetic ter values are mean i SD and geometric mean except for Tm“, where median
(90% confidence interval) is reported.
Table 4B
Cohort/Tre AUCO-t u CL/F
atment (11M*h) (11M*h) (L/h)
Cohort 5
300 mg SR3 2.46d:1.13 2.58i1.12 251 :: 105
(fasted) 2.23 2.36 230
300 mg SR3
2.98 d: 1.34 3.02 d: 1.35 215d:94.2
(medium-fat
2.72 2.76 196
meal)
Geometric Mean Relative Bioavailability and the 90% Confidence Intervals
SR3 fed vs 0
fasted (102A;146A>)0 0 . 0-
137%)
Pharmacokinetic parameter values are mean :: SD and geometric mean except for Tmax,
where median
(90% confidence interval) is reported.
Table 5A
Cmax Tmax CmaX/C12 tl/z
Cohort/Treatment 11
(nM) (h) h (h)
Cohort 6
2 X 25 mg SR3 12 55.1 i 30.3 1.3 4.0 :: 2.6
(fasted) 48.0 (0.50-4.0) 3.4
2 X 25 mg SR3 12 80.3 d: 27.3 3.0 2.2 i 0.4
(high-fat meal) 76.7 (1.5-6.0) 2.2
1X 100mg SR3 11 174i69.5 1.8 3.0i1.3
(fasted) 161 (050-40) 2.7
ric Mean Relative Bioavailability and the 90% nce
Intervals
2 X 25 mg SR3 fed 160%
vs fasted (129%-199%)
2 x 25 mg SR3 vs 1 x 58.7%}
100 mg SR3 (fasted) (46.9%-73.5%)
NC = not calculated because of significant s of mismatching Tlast within the subjects
between treatments; NR = not reported because significant numbers of C1211 values were BQL.
PK parameter values are mean i SD and geometric mean except for Tm, where median (90%
nce interval) is reported.
i) Statistical comparison
was dose-normalized.
Table 5B
Cohort/Treatme AUC0-t AUCMo CL/F
nt (nM*h) (nM*h) (L/h)
Cohort 6
2 X 25 mg SR3 205 i 103 243 d: 99.9 429 :: 167
(fasted) 183 226 400
2 X 25 mg SR3 333 :: 104 376 :: 94.6 253 :: 57.7
(high-fat meal) 319 366 247
1X 100 mg SR3 671 i230 704i230 280::815
(fasted) 639 673 268
Geometric Mean Relative Bioavailability and the 90% Confidence
Intervals
2 X 25 mg SR3 fed 174% 158%
vs fasted (150%-202%) 182%
2 X 25 mg SR3 vs 1 X 66.1%”
100 mg SR3 (fasted) (57.5%-75.9‘%
NC = not calculated because of significant numbers of ching Tm within the subjects
between treatments; NR = not reported because significant s of C12h values were BQL.
PK parameter values are mean fl: SD and geometric mean except for Tmax, where median (90%
confidence interval) is reported.
0 Statistical comparison
was dose-normalized.
The mean PK profiles following the fasting single-dose administration of 300
mg IR capsules were similar among the subjects in Cohorts l to 4 (.
Compared to the IR formulation, following fasting single-dose administration of the
SR1-SR4 formulations (3 X 100 mg tablets), the observed plasma median Tmax values
were moderately prolonged (by 0.3 to 1.5 hours) with significantly reduced mean Cmax
values (the upper bounds of the 90% CI for the geometric mean Cmax ratios were
< 30%), suggesting decreased absorption rate of the compound of Formula I for the
SR tablets. The apparent mean disposition tI/z observed in the terminal phase was
significantly longer, ranging from 7.3 to 11 hours for SR1-SR4, as compared to about
2 hours for the IR capsule, indicating that the systemic elimination of the compound
of Formula I was likely imited by its absorption, which was ned in the
terminal disposition phase. As a result of lower Cmax and longer disposition tI/z, the
Cmax/ C1211 ratios were significantly lower for the SR tablets compared to the IR
capsule for the same subjects studied. The ric mean Cmax/Cth ratios were
11.6—, 8.6—, 19.3—, and 89—fold, respectively, for SR1, SR2, SR3, and SR4 s, as
compared to 112- to l62-fold for the IR capsules administered in the fasted state.
For administration in the fasted state, the 4 SR s showed d relative
bioavailability compared to the IR capsule dosed in the same subjects. The percent
geometric mean ratios (90% CI) of Cmax were 14.2 % (11.4%—l7.5%), 8.9% (6.7%—
, 22.3% (17.4%—28.6%) and 9.0% (6.8%—11.9%) for SR1, SR2, SR3, and SR4,
respectively. The percent geometric mean ratios (90% CI) of AUCo-oo were 36.1 %
(33.3%—39.2%), 26.0% (21.6%—31.3%), 47.5% (41.9%—53.9%), and 28.5% (23.2%—
.1%) for SR1, SR2, SR3, and SR4, reSpectively. SR3 and SR1 demonstrated the
best and second best relative bioavailability, respectively, among the SR ations
tested.
Dosed in the fasted state, the intersubj ect variability as measured by percent
coefficient of variability (CV%) in plasma re was significantly higher for the
gastroretentive formulation SR4, but comparable among the 3 regular SR s
designed for intestinal release. The intersubj ect CV% for the 100 mg SR1 tablet was
39% and 33% for Cmax and AUCO—oo, respectively. The ubject CV% for the 100
mg SR2 tablet was 50% and 37% for Cmax and AUCO—oo, respectively. The intersubject
CV% for the 100 mg SR3 tablet was 43% and 29% for Cmax and AUC0_oo,
respectively. The intersubject CV% for the 100 mg SR4 tablet was 83% and 73% for
Cmax and AUCo-oo, respectively. Pooling all subjects in s 1—5 (n = 59) who were
administered 300 mg IR in the fasted state, the intersubj ect CV% was 49% and 39%
for Cmax and AUCO—oo, respectively, comparable to the CV% values ed for SR1,
SR2, and SR3.
A positive food effect was observed for all SR ations studied at the
300 mg (3 X 100 mg) dose level. Administered after a high-fat meal, geometric mean
Cmax and o values increased by 88% and 81%, respectively, for SR1; by 158%
and 118%, respectively; for SR2; by 91% and 45%; respectively; for SR3; and by
93% and 115%; respectively; for SR4. The food effect was moderate for a medium—
fat meal as compared to a high—fat meal, as suggested by the data for SR3 in Cohort 5.
For SR3, Cmax and AUCo-oo values increased by 46% and 17%, respectively, when it
was administered following a standardized medium-fat meal. Administration with
food did not significantly change the intersubj ect CV% in compound of al
plasma exposure for SR1, SR2, and SR3, which are SR formulations designed for
intra—intestinal e. For SR4, which is a gastroretentive SR formulation, the
intersubj ect CV% in plasma exposures appeared to be significantly reduced with a
concomitant high-fat meal.
This study also explored the dose-normalized relative bioavailability of the
25 mg SR tablet in reference to the 100 mg SR3 tablet. For the subjects in Cohort 6,
the dose—normalized Cmax and AUCO-oo percent geometric mean ratio for the 2 X 25 mg
SR3 treatment was 59% and 66%, respectively, versus the 1 X 100 mg SR3
administration in the fasted state. However, due to the supralinear dose-exposure
relationship for the compound of Formula I, the relative bioavailability of the 25 mg
SR tablet may be underestimated. For the 2 X 25 mg SR dose, a high-fat meal
increased compound of Formula I Cmax and o by 60% and 58%, respectively.
For the four SR formulations ted, the observed apparent disposition tvz
was comparable, and the Cmax/Cth ratios from a g single—dose administration
(which is used as a proxy for P/T ratio from twice-daily stration) were similar
among SR1, SR2, and SR4 (~10-fold) and moderately higher for SR3 old).
Overall, all 4 SR formulations demonstrated a significantly flatter PK profile
compared the IR capsule, meeting an important objective for sustained release.
Bioavailability of orally administered drug ts may be defined by the rate and
extent of the drug absorption into systemic circulation. A reduction in drug
absorption rate by limiting the drug release rate from drug products is a design
requirement in sustained release formulations. Therefore, for SR formulations, the
extent of the compound of Formula 1 absorption as ed by the plasma AUCO—oo is
used as the primary endpoint to assess the ve bioavailability. Thus, the mean
relative bioavailability is similar between SR2 (26%) and SR4 (29%), which was
ly lower than that of SR1 (36%). The best relative ilability was observed
for SR3 (48%). The results are in line with the in vitro ution profiles obtained
before conducting this study.
There was an nt inverse correlation between the food effect and relative
ilability for the SR formulations. On average, dosed with a high—fat meal, the
food—effect measured by the increase in AUCO—oo was the greatest for SR2 (118%) and
SR4 (115%), which was lower than that for SR1 (81%). The smallest food effect was
observed for SR3 (45%). This correlation was also apparent when the data from all
the subjects were pooled together. A quantile plot using the pooled individual data
(divided into 5 bins with 9 subjects per bin) suggests that the food effect was more
significant (> 2-fold increase in AUC) for the subjects with relative bioavailability
less than 35%, regardless of the formulation. The food effect was moderate (~50% or
less increase in AUC) for the subjects with relative ilability greater than 40%,
less of formulation. SR3 delivered a mean relative bioavailability of 48% and
is likely to be associated with a moderate food effect. In fact, when the SR3 tablet (3
X 100 mg) was dosed with a medium-fat meal (which is a more l daily diet), the
observed increase in geometric mean AUCO—oo was only 17%, suggesting that this
formulation may be administered without regard to medium- or low-fat meals. From
the perspective of avoiding significant food effect, SR3 is superior to the other
formulations.
Example 4. Clinical Results in Phase 2a in patients with active rheumatoid
arthritis (RA)
An initial 28 day part of the study was conducted in order to select doses
moving forward, guiding dose selection for the 3 month second part of the study. Part
2 of the study was randomized, double—blind, o controlled (sponsor ded)
with treatment for 84 days. Sixty subjects to be randomized, using the same
population as in Part 1: single cohort, five parallel treatment , 12 subjects each:
100 mg SR3 tablets BID; 300 mg (3 x 100 mg SR3 tablets) QD; 200 mg (2 x 100 mg
SR3 tablets) BID; 600 mg (6 x 100 mg SR3 tablets) QD; and placebo. Interim data
was submitted to ACR (American College of Rheumatology) 2013 (n=40 subjects
who completed day 84). The ACR scores at 3 months re shown in Table 6. The
ACR scores for the 600 mg QD are unprecedented as compared to other JAK
inhibitors that are approved for treatment of RA. For example, the approved product
for tofacitinib citrate (5 mg BID) showed much lower ACR scores at 3 months: 59%
(ACR20), 31% ), and 15% (ACR70) (Table 5 of XELJANZ® — tofacitinib
citrate tablet — label).
Table 6
Placebo 100 mg 300 mg QD 200 mg 600 mg QD
BID BID
The percent change from baseline for hemoglobin was also studied for each of
the dosing regimens (. As can be seen in the 200 mg BID dose showed
a drop away from the baseline compared to the other doses which tended to stay Close
to the placebo levels. For example, the 600 mg QD dose did not show the same
downward trend as shown for the BID dose. However, as can be seen in Table 6, the
once—daily dosing (600 mg QD) did not compromise efficacy compared with the BID
doses. This tes that the once-daily dosing (such as 600 mg QD) may achieve
maximal efficacy without inducing side-effects such anemia. As shown in and
Table 6, the 600 mg QD dose has robust y with trivial change in hemoglobin
levels.
It is believed that this efficacy/side—effect profile may be due to the QD dose
achieving maximal JAKl signaling (tied to efficacy) with low JAK2 inhibition at the
trough, as JAK2 signaling is tied to hematopoiesis. This hypothesis is supported by
the PK derived JAKl (IL-6) and JAK2 (TPO) inhibition data for the compound of
Formula at various doses (Table 7). In particular, the 600 mg QD dose showed
similar average IL-6 inhibition to the 200 mg BID and 400 mg BID doses (61%
versus 64% and 69%), but lower trough TPO inhibition in comparison to the 200 mg
BID and 400 mg BID doses (4% versus 13% and 16%). The trough IL-6 inhibition
for the 600 mg QD dose is also lower than the trough IL-6 inhibition for the 200 mg
BID and 400 mg BID doses, which suggests that there may be a reduction in infection
from the QD dose.
Table 7
Dose regimen Average IL-6 Trough IL-6 Average TPO Trough TPO
inhibition tion inhibition inhibition
100 mg QD 30% 7% 7% <1%
200 mg QD 39% 11% 11% <1%
300 mg QD
600 mg QD
100 mg BID
200 mg BID
400 mg BID
e 5. Clinical Results in ts with Plaque Psoriasis
A double—blind (sponsor unblinded), randomized, o controlled study
was conducted in approximately 48 ts treated for 28 days. Eligibility
requirements included: active plaque psoriasis for at least 6 months at ing;
body surface area (BSA) of plaque psoriasis of 2 5%; psoriasis area and severity
index (PASI) score of 2 5; static physician’s global assessment (sPGA) score of Z 3;
inadequate response to topical therapies; innovative design allowing rapid progress
between doses, with conservative safety assessment. Four staggered dose groups of
12 ts each (9 active and 3 PBO) progressing from 100 mg QD to 200 mg QD to
200 mg BID to 600 mg QD. Once the 4th subject (block of 3 active 1 PBO)
completed 28 days administration without a Grade 3 or higher AE, the next group of
12 subjects initiated treatment with the next highest dose; while the first 4 subjects in
this group are treated for 28 days, the 1st group is filled
60 subjects with moderate to severe psoriasis were randomized. There were five
treatment groups: placebo, 100 mg QD, 200 mg QD, 200 mg BID and 600 mg QD. A
sequential method of recruitment was used, increasing from the lowest dose to the
highest, each after the completion of 28 days for the first four subjects in the previous
dose. The results at 28 days are show in Table 8 (PASI 50 is Psoriasis Area and
Severity Index). These PASI 50 score of 81.8% for the 600 mg QD dose are
unprecedented as compared to other JAK inhibitors that are in development for
treatment of psoriasis. For example, 5 mg tofacitinib (also known as tasocitinib)
showed lower PASI 50 score of 65.3% at 12 weeks (published on
http://press.pfizer.com on 10/7/2010). The 5 mg tofacitinib dose is the approved
dosage level for RA for safety reasons in the US.
Table 8
Placebo 100 mg 200 mg QD 200 mg 600 mg QD
BID BID
Mean % —12.5% —35.2% —42.4%
change
sPGA
% sPGA 0 33.3% 45.5%
(clear or
% PASI 50 8.3% 22.2% 66.7% 44.4% 81.8%
Example 6. abel Phase 11 Study in Patients with Myelofibrosis
In this study, ts with age 218 years, a sis of primary myelofibrosis
(PMF) or post—polycythemia vera MF or post-essential thrombocythemia MF
(JAK2V617F positive or ve on ), platelet counts 2 50 X 109/L,
hemoglobin levels 2 8.0 g/dL (transfusions permitted to achieve these levels),
intermediate-l or higher per DIPSS criteria, and palpable spleen or prior splenectomy
were enrolled. Three different dose cohorts were assessed: (1) 100 mg SR3 tablets
BID) (2) 200 mg (2 x 100 mg SR3 tablets) BID; and (3) 600 mg (6 X 100 mg SR3
tablets) QD. a)-(b) show interim results with t to proportion of subjects
with 2 50% reduction in total symptom score (TSS) in each dose group per the
d Myelofibrosis Symptom Assessment Form (MFSAF) V3.0 electronic diary
at week 12 compared with baseline (The d MFSAF V3.0 comprises 19
questions assessing MF—related symptoms on a scale of 0 (absent) to 10 (worst
imaginable». a) depicts the percentage of patients having a 2 50% reduction
in TSS at week 12 by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD)
(patients who discontinued prior to the week 12 visit were considered nonresponders).
b) depicts the percent change in TSS from baseline at week 12 by dose cohort
(100 mg BID, 200 mg BID, and 600 mg QD) (only patients with baseline and week
12 data were included). a) depicts mean obin levels (g/dL) over time
by dose cohort (100 mg BID, 200 mg BID, and 600 mg QD) (interim results of study
for all patients). b) s mean hemoglobin levels (g/dL) over time by dose
cohort (100 mg BID, 200 mg BID, and 600 mg QD) at 48 weeks. c) depicts
mean hemoglobin levels (g/dL) over time by dose cohort at 48 weeks as an average
for three dose cohorts as compared to individuals dosed with o or tinib
(ruxolitinib was dosed according to the label for Jakafi®). The data show an increase
in obin levels for the 600 mg QD dose. Finally, Table 9 below show interim
hematology laboratory results (new and worsening) for each dose cohort. Table 9a
shows the hematology laboratory results (new and worsening) for each dose cohort
after long exposure.
Table 9
Days of Exposure, 102.5 169.0 16.0
median (range) (23.0, 376.0) (22.0, 339.0) (1.0, 196.0)
Anemia, Grade 3 3/9 (33.3) 12/42 (28.6) 2/29 (6.9)
Thrombocytopenia
Grade 3 4/9 (44.4) 12/44 (27.3) 1/29 (3.4)
Grade 4 0/9 (0) 2/45 (4.4) 0/29 (0)
Table 93
Event n/N % 100 mg BID 200 mg BID 600 mg QD
Days of Exposure, 102.0 254.0 192.0
median (range) (23,519) (28,343)
Anemia, Grade 3 3/10 (30.0) 19/45 (42.2) 8/32 (25.0)
Thrombocytopenia
Grade 3 4/10 (40.0) 13/45 (28.9) 4/32 (12.5)
Grade 4 0/10 (0.0) 3/45 (6.7) 1/32 (3.1)
Example A: In vitro JAK Kinase Assay
The compound of Formula 1 herein was tested for inhibitory ty of JAK
targets according to the following in vitro assay described in Park et al, Analytical
Biochemistry 1999, 269, 94—104. The catalytic domains of human JAK] (a.a. 837-
1142) and JAK2 (a.a. 828-1132) with an N—terminal His tag were expressed using
baculovirus in insect cells and purified. The catalytic activity of JAKl and JAK2 was
assayed by measuring the phosphorylation of a biotinylated peptide. The
phosphorylated e was detected by homogenous time resolved fluorescence
(HTRF). ICsos of compounds were ed for each kinase in the 40 microL
reactions that contain the enzyme, ATP and 500 nM peptide in 50 mM Tris (pH 7.8)
buffer with 100 mM NaCl, 5 mM DTT, and 0.1 mg/mL (0.01%) BSA. For the 1 mM
leo measurements, ATP concentration in the reactions was 1 mM. Reactions were
carried out at room temperature for 1 hr and then stopped with 20 uL 45 mM EDTA,
300 nM SA-APC, 6 nM Eu-Py20 in assay buffer (Perkin Elmer, Boston, MA).
Binding to the um d antibody took place for 40 minutes and HTRF signal
was measured on a Fusion plate reader (Perkin Elmer, , MA). The compound
of a I and the adipic acid salt had an leo at JAKl of S 5 nM (measured at 1
mM ATP) with a JAK2/JAK1 ratio of > 10 (measured at 1 mM ATP).
Example B: Cellular Assays
Cancer cell lines dependent on cytokines and hence JAK/STAT signal
transduction, for growth, can be plated at 6000 cells per well (96 well plate format) in
RPMI 1640, 10% FBS, and l nG/mL of appropriate cytokine. Compounds can be
added to the cells in DMSO/media (final tration 0.2% DMSO) and incubated
for 72 hours at 37 OC, 5% C02. The effect of compound on cell viability is assessed
using the CellTiter-Glo scent Cell Viability Assay ga) ed by
TopCount n Elmer, Boston, MA) quantitation. Potential off-target effects of
compounds are measured in parallel using a non-JAK driven cell line with the same
assay readout. All experiments are typically performed in duplicate.
The above cell lines can also be used to examine the effects of compounds on
phosphorylation of JAK kinases or potential downstream substrates such as STAT
proteins, Akt, Shp2, or Erk. These experiments can be performed following an
overnight cytokine starvation, followed by a brief preincubation with compound (2
hours or less) and ne stimulation of approximately 1 hour or less. Proteins are
then ted from cells and analyzed by ques familiar to those schooled in the
art ing n blotting or ELISAs using antibodies that can differentiate
between orylated and total protein. These experiments can utilize normal or
cancer cells to investigate the activity of compounds on tumor cell survival biology or
on mediators of inflammatory disease. For example, with regards to the latter,
cytokines such as IL-6, IL-12, IL-23, or IFN can be used to stimulate JAK activation
resulting in orylation of STAT protein(s) and potentially in transcriptional
profiles (assessed by array or qPCR technology) or production and/or secretion of
proteins, such as lL-l7. The ability of compounds to inhibit these cytokine mediated
effects can be measured using techniques common to those schooled in the art.
Compounds herein can also be tested in cellular models designed to evaluate
their potency and activity against mutant JAKs, for example, the JAK2V617F
on found in myeloid proliferative disorders. These experiments often utilize
cytokine dependent cells of hematological lineage (e. g. BaF/3) into which the wild-
type or mutant JAK kinases are ectopically expressed (James, C., et al. Nature
434:1144—1148; Staerk, J., et a3. JBC 280:41893—41899). Endpoints include the
effects of compounds on cell survival, proliferation, and orylated JAK, STAT,
Akt, or Erk proteins.
n compounds herein can be evaluated for their activity inhibiting T-cell
proliferation. Such as assay can be considered a second cytokine (226. JAK) driven
proliferation assay and also a simplistic assay of immune suppression or inhibition of
immune activation. The following is a brief outline of how such experiments can be
performed. Peripheral blood mononuclear cells (PBMCs) are prepared from human
whole blood samples using Ficoll Hypaque separation method and T-cells (fraction
2000) can be obtained from PBMCs by elutriation. y isolated human T-cells can
be maintained in culture medium (RPMI 1640 supplemented with10% fetal bovine
serum, 100 U/ml penicillin, 100 ug/ml streptomycin) at a density of 2 x 106 cells/ml at
37 °C for up to 2 days. For IL—2 stimulated cell proliferation analysis, T—cells are first
treated with Phytohemagglutinin (PHA) at a final concentration of 10 ug/mL for 72h.
After g once with PBS, 6000 cells/well are plated in 96—well plates and treated
with compounds at different trations in the culture medium in the presence of
100 U/mL human IL-2 ec-Tany TechnoGene; Rehovot, Israel). The plates are
incubated at 37 °C for 72h and the proliferation index is assessed using CellTiter—Glo
Luminescent ts following the manufactory ted protocol (Promega;
Madison, WI).
Example C: In vivo anti-tumor efficacy
Compounds herein can be ted in human tumor xenograft models in
immune compromised mice. For example, a tumorigenic variant of the INA-6
plasmacytoma cell line can be used to inoculate SCID mice subcutaneously (Burger,
R., et a]. Hematol J. 2:42—53, 2001). Tumor bearing animals can then be randomized
into drug or vehicle treatment groups and different doses of compounds can be
administered by any number of the usual routes including oral, i.p., or continuous
infusion using implantable pumps. Tumor growth is ed over time using
rs. Further, tumor samples can be harvested at any time after the initiation of
treatment for analysis as described above (Example B) to evaluate compound effects
on JAK activity and downstream ing pathways. In addition, selectivity of the
compound(s) can be assessed using xenograft tumor models that are driven by other
know kinases (e.g. l) such as the K562 tumor model.
Example D: Murine Skin Contact Delayed Hypersensitivity se Test
Compounds herein can also be tested for their efficacies (of ting JAK
targets) in the T—cell driven murine delayed hypersensitivity test model. The murine
skin contact d—type hypersensitivity (DTH) response is considered to be a valid
model of al contact dermatitis, and other T-lymphocyte mediated immune
disorders of the skin, such as psoriasis (Immunol Today. 1998 Jan;l9(l):37—44).
Murine DTH shares multiple characteristics with psoriasis, including the immune
infiltrate, the accompanying increase in inflammatory cytokines, and keratinocyte
hyperproliferation. Furthermore, many classes of agents that are efficacious in
treating psoriasis in the clinic are also effective inhibitors of the DTH response in
mice (Agents Actions. 1993 Jan;38(l—2): 1 16-21).
On Day 0 and l, Balb/c mice are sensitized with a topical application, to their
shaved abdomen with the antigen 2,4,dinitro-fluorobenzene (DNFB). On day 5, ears
are measured for thickness using an engineer’s micrometer. This measurement is
recorded and used as a baseline. Both of the animals’ ears are then challenged by a
topical application of DNFB in a total of 20 uL (10 uL on the internal pinna and 10
uL on the al pinna) at a concentration of 0.2%. Twenty—four to seventy—two
hours after the nge, ears are measured again. ent with the test
compounds is given throughout the sensitization and challenge phases (day -l to day
7) or prior to and throughout the nge phase (usually afternoon of day 4 to day
7). Treatment of the test compounds (in different concentration) is administered
either systemically or topically (topical application of the treatment to the ears).
Efficacies of the test compounds are indicated by a reduction in ear swelling
comparing to the situation without the treatment. nds causing a reduction of
% or more were considered efficacious. In some experiments, the mice are
challenged but not sensitized (negative control).
The tive effect (inhibiting tion of the JAK-STAT pathways) of the
test compounds can be confirmed by immunohistochemical analysis. Activation of
the JAK-STAT pathway(s) results in the formation and translocation of functional
transcription factors. Further, the influx of immune cells and the increased
proliferation of keratinocytes should also e unique expression profile changes
in the ear that can be investigated and quantified. Formalin fixed and n
embedded ear sections (harvested after the challenge phase in the DTH model) are
subjected to immunohistochemical is using an antibody that specifically
interacts with orylated STAT3 (clone 58El2, Cell Signaling Technologies).
The mouse ears are treated with test compounds, e, or dexamethasone (a
clinically efficacious treatment for psoriasis), or without any treatment, in the DTH
model for comparisons. Test compounds and the dexamethasone can produce similar
riptional changes both qualitatively and quantitatively, and both the test
compounds and dexamethasone can reduce the number of infiltrating cells. Both
ically and topical stration of the test compounds can produce inhibitive
effects, i.e., reduction in the number of infiltrating cells and inhibition of the
transcriptional changes.
Example E: In vivo anti-inflammatory activity
Compounds herein can be evaluated in rodent or dent models designed
to replicate a single or complex inflammation response. For instance, rodent models
of arthritis can be used to evaluate the therapeutic potential of compounds dosed
preventatively or eutically. These models include but are not limited to mouse
or rat collagen—induced arthritis, rat adjuvant-induced arthritis, and collagen antibody-
induced arthritis. Autoimmune diseases including, but not limited to, multiple
sclerosis, type I-diabetes mellitus, uveoretinitis, thyroditis, myasthenia gravis,
immunoglobulin nephropathies, myocarditis, airway sensitization (asthma), lupus, or
s may also be used to evaluate the therapeutic potential of compounds herein.
These models are well established in the research community and are familiar to those
schooled in the art (Current Protocols in Immunology, Vol 3., Coligan, J.E. et a1,
Wiley Press; Methods in Molecular Biology: Vol. 225, Inflammation Protocols,
Winyard, PG. and Willoughby, D.A., Humana Press, 2003.).
Example F: Animal Models for the Treatment of Dry Eye, s, and
Conjunctivitis
Agents may be evaluated in one or more preclinical models of dry eye known
to those schooled in the art including, but not limited to, the rabbit concanavalin A
(ConA) lacrimal gland model, the scopolamine mouse model (subcutaneous or
transdermal), the Botulinumn mouse lacrimal gland model, or any of a number of
neous rodent mmune models that result in ocular gland dysfunction (e.g.
NOD-SCID, MRL/lpr, or NZB/NZW) (Barabino et al., Experimental Eye ch
2004, 79, 1 and Schrader et al., Developmental Opthalmology, Karger 2008,
41, 298-312, each of which is incorporated herein by reference in its entirety).
Endpoints in these models may include histopathology of the ocular glands and eye
(cornea, etc.) and possibly the classic Schirmer test or modified versions thereof
(Barabino et al.) which measure tear production. Activity may be assessed by dosing
via multiple routes of administration (eg. systemic or topical) which may begin prior
to or after measurable disease exists.
Agents may be evaluated in one or more preclinical models of uveitis known
to those schooled in the art. These include, but are not limited to, models of
mental mune uveitis (EAU) and endotoxin induced uveitis (EIU). EAU
experiements may be performed in the , rat, or mouse and may involve passive
or activate immunization. For instance, any of a number or retinal antigens may be
used to sensitize animals to a relevant immunogen after which animals may be
challenged ocuarly with the same antigen. The EIU model is more acute and involves
local or systemic administration of lysaccaride at sublethal doses. Endpoints
for both the EIU and EAU models may include fundoscopic exam, histopathology
amongst . These models are reviewed by Smith et a1. (Immunology and Cell
Biology 1998, 76, 497—512, which is incorporated herein by reference in its entirety).
Activity is assessed by dosing via multiple routes of administration (e. g. systemic or
topical) which may begin prior to or after measurable disease . Some models
listed above may also develop scleritis/episcleritis, chorioditis, cyclitis, or iritis and
are therefore useful in investigating the potential activity of compounds for the
therapeutic treatment of these diseases.
Agents may also be evaluated in one or more preclinical models of
conjunctivitis known those schooled in the art. These include, but are not limited to,
rodent models utilizing guinea—pig, rat, or mouse. The guinea-pig models include
those utilizing active or passive immunization and/or immune challenge ols
with antigens such as min or ragweed (reviewed in Groneberg, D.A., et al.,
Allergy 2003, 58, 1101—1113, which is incorporated herein by reference in its
entirety). Rat and mouse models are r in general design to those in the guinea-
pig (also reviewed by Groneberg). Activity may be assessed by dosing via multiple
routes of administration (e. g. systemic or topical) which may begin prior to or after
measurable disease exists. Endpoints for such studies may include, for e,
histological, immunological, biochemical, or molecular analysis of ocular tissues such
as the conjunctiva.
Example G: In vivo protection of bone
Compounds may be evaluated in various preclinical models of osteopenia,
osteoporosis, or bone resorption known to those schooled in the art. For e,
ovariectomized rodents may be used to evaluate the ability of nds to affect
signs and markers of bone remodeling and/or density (W.S.S. Jee and W. Yao, J
Musculoskel. Nueron. Interact, 2001, 1(3), 193—207, which is orated herein by
reference in its entirety). Alternatively, bone density and architecture may be
evaluated in control or nd treated rodents in models of therapy (e.g.
glucocorticoid) induced osteopenia (Yao, et a1. Arthritis and Rheumatism, 2008,
58(6), 3485-3497; and id. , 1674-1686, both of which are incorporated herein
by reference in its entirety). In addition, the effects of compounds on bone resorption
and density may be evaluable in the rodent models of arthritis discussed above
(Example E). Endpoints for all these models may vary but often e ogical
and radiological assessments as well as immunohisotology and appropriate
biochemical markers of bone remodeling.
Claims (9)
1. A ned release composition, comprising: (i) {1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H- pyrrolo[2,3-d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile, or a pharmaceutically acceptable salt thereof; (ii) a first hypromellose characterized by having an apparent viscosity at a concentration of 2% in water of 80 cP to 120 cP; (iii) a second hypromellose, characterized by having an apparent viscosity at a concentration of 2% in water of 3000 cP to 5600 cP, wherein the composition comprises 8% to 20% of the first and second hypromelloses; (iv) 16% to 22% by weight of microcrystalline cellulose; and (v) 45% to 55% by weight of lactose monohydrate.
2. The sustained release composition ing to claim 1, wherein the composition comprises 10% to 15% by weight of the one or more hypromelloses.
3. The sustained release ition according to claim 1 or claim 2, wherein the composition comprises 0.3% to 0.7% by weight of magnesium stearate.
4. A sustained release composition, sing: (i) 100 mg on a free base basis of {1-{1-[3-fluoro (trifluoromethyl)isonicotinoyl]piperidinyl}[4-(7H-pyrrolo[2,3- d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile, or a pharmaceutically acceptable salt thereof; (ii) a first hypromellose characterized by having an apparent ity at a concentration of 2% in water of 80 cP to 120 cP; (iii) a second hypromellose, characterized by having an apparent viscosity at a concentration of 2% in water of 3000 cP to 5600 cP, wherein the composition comprises 10% to 15% of the first and second elloses; (iv) 16% to 22% by weight of microcrystalline cellulose; and (v) 45% to 55% by weight of lactose monohydrate.
5. The sustained release composition ing to any one of claims 1 to 4, n oral administration of one or more of the sustained release compositions to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (C12h) of {1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidinyl}[4- (7H-pyrrolo[2,3-d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile of 9 to 40.
6. The sustained release composition according to any one of claims 1 to 4, wherein oral administration of one or more of the sustained release compositions to a fasted individual provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (C12h) of {1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidinyl}[4- (7H-pyrrolo[2,3-d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile of 15 to 30.
7. The sustained release ition ing to any one of claims 1 to 6, wherein oral administration of one or more of the sustained release compositions to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (C12h) of {1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin yl}[4-(7H-pyrrolo[2,3-d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile of 10 to 70.
8. The sustained release composition according to any one of claims 1 to 6, wherein oral administration of one or more of the sustained e compositions to an individual after a high-fat meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (C12h) of {1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin yl}[4-(7H-pyrrolo[2,3-d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile of 15 to 50.
9. The sustained release composition according to any one of claims 1 to 6, wherein oral administration of one or more of the sustained release compositions to an individual after a at meal provides a ratio of mean peak plasma concentration (Cmax) to mean 12-hour plasma concentration (C12h) of {1-{1-[3-fluoro(trifluoromethyl)isonicotinoyl]piperidin yl}[4-(7H-pyrrolo[2,3-d]pyrimidinyl)-1H-pyrazolyl]azetidinyl}acetonitrile of 25 to 45.
Applications Claiming Priority (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201361863325P | 2013-08-07 | 2013-08-07 | |
US61/863,325 | 2013-08-07 | ||
US201361913066P | 2013-12-06 | 2013-12-06 | |
US61/913,066 | 2013-12-06 | ||
NZ717230A NZ717230B2 (en) | 2013-08-07 | 2014-08-06 | Sustained release dosage forms for a jak1 inhibitor |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ756084A NZ756084A (en) | 2021-04-30 |
NZ756084B2 true NZ756084B2 (en) | 2021-08-03 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2021202916B2 (en) | Sustained release dosage forms for a jak1 inhibitor | |
NZ756084B2 (en) | Sustained release dosage forms for a jak1 inhibitor | |
NZ756083B2 (en) | Sustained release dosage forms for a jak1 inhibitor | |
NZ756083A (en) | Sustained release dosage forms for a jak1 inhibitor | |
NZ717230B2 (en) | Sustained release dosage forms for a jak1 inhibitor | |
BR112016002571B1 (en) | SUSTAINED RELEASE TABLETS WITH A JAK1 INHIBITOR | |
EA043723B1 (en) | JAK1 INHIBITOR TIME-RELEASE TABLET |