NZ621215B2 - Treatment of multiple sclerosis with combination of laquinimod and interferon-beta - Google Patents
Treatment of multiple sclerosis with combination of laquinimod and interferon-beta Download PDFInfo
- Publication number
- NZ621215B2 NZ621215B2 NZ621215A NZ62121512A NZ621215B2 NZ 621215 B2 NZ621215 B2 NZ 621215B2 NZ 621215 A NZ621215 A NZ 621215A NZ 62121512 A NZ62121512 A NZ 62121512A NZ 621215 B2 NZ621215 B2 NZ 621215B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- laquinimod
- interferon
- amount
- multiple sclerosis
- another embodiment
- Prior art date
Links
- 229960004577 laquinimod Drugs 0.000 title claims abstract description 259
- GKWPCEFFIHSJOE-UHFFFAOYSA-N Laquinimod Chemical compound OC=1C2=C(Cl)C=CC=C2N(C)C(=O)C=1C(=O)N(CC)C1=CC=CC=C1 GKWPCEFFIHSJOE-UHFFFAOYSA-N 0.000 title claims abstract description 257
- 201000006417 multiple sclerosis Diseases 0.000 title claims abstract description 132
- 108090000467 Interferon beta Proteins 0.000 title description 18
- 102000003996 Interferon beta Human genes 0.000 title description 18
- 229960001388 Interferon-beta Drugs 0.000 title description 18
- 239000000203 mixture Substances 0.000 claims abstract description 97
- 238000002560 therapeutic procedure Methods 0.000 claims abstract description 73
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 51
- 206010071068 Clinically isolated syndrome Diseases 0.000 claims abstract description 36
- 239000003814 drug Substances 0.000 claims description 76
- 241000282414 Homo sapiens Species 0.000 claims description 69
- 229940079593 drugs Drugs 0.000 claims description 47
- 201000010099 disease Diseases 0.000 claims description 41
- 239000003795 chemical substances by application Substances 0.000 claims description 40
- 230000000694 effects Effects 0.000 claims description 40
- 210000004556 Brain Anatomy 0.000 claims description 34
- 230000002354 daily Effects 0.000 claims description 33
- 102000014150 Interferons Human genes 0.000 claims description 28
- 108010050904 Interferons Proteins 0.000 claims description 28
- 230000001603 reducing Effects 0.000 claims description 26
- 229940079322 interferon Drugs 0.000 claims description 25
- 206010061818 Disease progression Diseases 0.000 claims description 22
- 230000036541 health Effects 0.000 claims description 20
- 230000001965 increased Effects 0.000 claims description 16
- 238000009825 accumulation Methods 0.000 claims description 13
- 239000002775 capsule Substances 0.000 claims description 13
- 239000007787 solid Substances 0.000 claims description 13
- 230000000737 periodic Effects 0.000 claims description 12
- FBPFZTCFMRRESA-KAZBKCHUSA-N D-Mannitol Natural products OC[C@@H](O)[C@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KAZBKCHUSA-N 0.000 claims description 11
- 229960001375 Lactose Drugs 0.000 claims description 11
- 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 claims description 11
- FBPFZTCFMRRESA-KVTDHHQDSA-N Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 claims description 11
- 239000000945 filler Substances 0.000 claims description 11
- 239000008101 lactose Substances 0.000 claims description 11
- 239000000594 mannitol Substances 0.000 claims description 11
- 235000010355 mannitol Nutrition 0.000 claims description 11
- 238000004519 manufacturing process Methods 0.000 claims description 10
- 239000003937 drug carrier Substances 0.000 claims description 9
- 230000003862 health status Effects 0.000 claims description 9
- 230000003647 oxidation Effects 0.000 claims description 9
- 238000007254 oxidation reaction Methods 0.000 claims description 9
- 239000003638 reducing agent Substances 0.000 claims description 9
- 239000000969 carrier Substances 0.000 claims description 8
- 230000001771 impaired Effects 0.000 claims description 8
- 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 claims description 8
- 230000001537 neural Effects 0.000 claims description 8
- 150000002500 ions Chemical class 0.000 claims description 7
- 239000000314 lubricant Substances 0.000 claims description 7
- 239000001301 oxygen Substances 0.000 claims description 7
- MYMOFIZGZYHOMD-UHFFFAOYSA-N oxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 claims description 7
- 229910052760 oxygen Inorganic materials 0.000 claims description 7
- 239000002245 particle Substances 0.000 claims description 7
- 206010057668 Cognitive disease Diseases 0.000 claims description 6
- 210000003205 Muscles Anatomy 0.000 claims description 6
- 229940035295 Ting Drugs 0.000 claims description 6
- 239000011248 coating agent Substances 0.000 claims description 6
- 238000000576 coating method Methods 0.000 claims description 6
- 239000003246 corticosteroid Substances 0.000 claims description 6
- 230000003247 decreasing Effects 0.000 claims description 6
- 230000001939 inductive effect Effects 0.000 claims description 6
- XEEYBQQBJWHFJM-UHFFFAOYSA-N iron Chemical group [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 claims description 6
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 claims description 6
- 229940021182 non-steroidal anti-inflammatory drugs Drugs 0.000 claims description 6
- 229940064701 Corticosteroid nasal preparations for topical use Drugs 0.000 claims description 5
- 229960001334 Corticosteroids Drugs 0.000 claims description 5
- 210000003141 Lower Extremity Anatomy 0.000 claims description 5
- 108090001123 antibodies Proteins 0.000 claims description 5
- 102000004965 antibodies Human genes 0.000 claims description 5
- 239000007884 disintegrant Substances 0.000 claims description 5
- 230000005713 exacerbation Effects 0.000 claims description 5
- 239000003018 immunosuppressive agent Substances 0.000 claims description 5
- 230000001976 improved Effects 0.000 claims description 5
- 230000003340 mental Effects 0.000 claims description 5
- 229940045902 sodium stearyl fumarate Drugs 0.000 claims description 5
- STFSJTPVIIDAQX-LTRPLHCISA-M sodium;(E)-4-octadecoxy-4-oxobut-2-enoate Chemical group [Na+].CCCCCCCCCCCCCCCCCCOC(=O)\C=C\C([O-])=O STFSJTPVIIDAQX-LTRPLHCISA-M 0.000 claims description 5
- 229940083878 topical for treatment of hemorrhoids and anal fissures Corticosteroids Drugs 0.000 claims description 5
- 230000003442 weekly Effects 0.000 claims description 5
- 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 description 4
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 claims description 4
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 claims description 4
- 108010005716 Interferon beta-1a Proteins 0.000 claims description 4
- 229960004461 Interferon beta-1a Drugs 0.000 claims description 4
- 229960001021 Lactose Monohydrate Drugs 0.000 claims description 4
- 239000006096 absorbing agent Substances 0.000 claims description 4
- 239000002274 desiccant Substances 0.000 claims description 4
- 229960001855 mannitol Drugs 0.000 claims description 4
- 229920000642 polymer Polymers 0.000 claims description 4
- 239000008109 sodium starch glycolate Substances 0.000 claims description 4
- 229940079832 sodium starch glycolate Drugs 0.000 claims description 4
- 229920003109 sodium starch glycolate Polymers 0.000 claims description 4
- 239000000600 sorbitol Substances 0.000 claims description 4
- 235000010356 sorbitol Nutrition 0.000 claims description 4
- OQANPHBRHBJGNZ-FYJGNVAPSA-N (3E)-6-oxo-3-[[4-(pyridin-2-ylsulfamoyl)phenyl]hydrazinylidene]cyclohexa-1,4-diene-1-carboxylic acid Chemical compound C1=CC(=O)C(C(=O)O)=C\C1=N\NC1=CC=C(S(=O)(=O)NC=2N=CC=CC=2)C=C1 OQANPHBRHBJGNZ-FYJGNVAPSA-N 0.000 claims description 3
- XXSMGPRMXLTPCZ-UHFFFAOYSA-N Hydroxychloroquine Chemical compound ClC1=CC=C2C(NC(C)CCCN(CCO)CC)=CC=NC2=C1 XXSMGPRMXLTPCZ-UHFFFAOYSA-N 0.000 claims description 3
- 229960004171 Hydroxychloroquine Drugs 0.000 claims description 3
- SERLAGPUMNYUCK-DCUALPFSSA-N Isomalt Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO[C@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O SERLAGPUMNYUCK-DCUALPFSSA-N 0.000 claims description 3
- MBBZMMPHUWSWHV-BDVNFPICSA-N Meglumine Chemical group CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 claims description 3
- 229960003194 Meglumine Drugs 0.000 claims description 3
- 229960001940 Sulfasalazine Drugs 0.000 claims description 3
- 230000001472 cytotoxic Effects 0.000 claims description 3
- 231100000433 cytotoxic Toxicity 0.000 claims description 3
- 239000003623 enhancer Substances 0.000 claims description 3
- 230000002708 enhancing Effects 0.000 claims description 3
- 150000002344 gold compounds Chemical class 0.000 claims description 3
- 239000007927 intramuscular injection Substances 0.000 claims description 3
- 238000010255 intramuscular injection Methods 0.000 claims description 3
- 229910052742 iron Inorganic materials 0.000 claims description 3
- 239000000905 isomalt Substances 0.000 claims description 3
- 235000010439 isomalt Nutrition 0.000 claims description 3
- GUBGYTABKSRVRQ-QKKXKWKRSA-N lactose group Chemical group OC1[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@H](O)[C@@H](O)[C@@H](O)[C@H](O2)CO)[C@H](O1)CO GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 claims description 3
- 239000007788 liquid Substances 0.000 claims description 3
- 235000019359 magnesium stearate Nutrition 0.000 claims description 3
- 238000004806 packaging method and process Methods 0.000 claims description 3
- 239000000049 pigment Substances 0.000 claims description 3
- 229920001098 polystyrene-block-poly(ethylene/propylene) Polymers 0.000 claims description 3
- 150000003873 salicylate salts Chemical class 0.000 claims description 3
- 229960002920 sorbitol Drugs 0.000 claims description 3
- 239000007921 spray Substances 0.000 claims description 3
- 206010022114 Injury Diseases 0.000 claims description 2
- 230000004064 dysfunction Effects 0.000 claims description 2
- 229920001903 high density polyethylene Polymers 0.000 claims description 2
- 239000004700 high-density polyethylene Substances 0.000 claims description 2
- 230000035699 permeability Effects 0.000 claims description 2
- 239000000741 silica gel Substances 0.000 claims description 2
- 229910002027 silica gel Inorganic materials 0.000 claims description 2
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N silicium dioxide Chemical group O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 claims description 2
- 206010059512 Apoptosis Diseases 0.000 claims 1
- 230000006907 apoptotic process Effects 0.000 claims 1
- 230000000007 visual effect Effects 0.000 claims 1
- 238000002360 preparation method Methods 0.000 abstract description 20
- 230000000996 additive Effects 0.000 description 57
- 230000003902 lesions Effects 0.000 description 42
- FHEAIOHRHQGZPC-KIWGSFCNSA-N acetic acid;(2S)-2-amino-3-(4-hydroxyphenyl)propanoic acid;(2S)-2-aminopentanedioic acid;(2S)-2-aminopropanoic acid;(2S)-2,6-diaminohexanoic acid Chemical compound CC(O)=O.C[C@H](N)C(O)=O.NCCCC[C@H](N)C(O)=O.OC(=O)[C@@H](N)CCC(O)=O.OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 FHEAIOHRHQGZPC-KIWGSFCNSA-N 0.000 description 28
- 108010072051 Glatiramer Acetate Proteins 0.000 description 24
- 229960003776 glatiramer acetate Drugs 0.000 description 23
- 238000002595 magnetic resonance imaging Methods 0.000 description 18
- 230000002401 inhibitory effect Effects 0.000 description 15
- 230000000977 initiatory Effects 0.000 description 15
- 238000002648 combination therapy Methods 0.000 description 14
- 230000037230 mobility Effects 0.000 description 14
- 208000007400 Relapsing-Remitting Multiple Sclerosis Diseases 0.000 description 13
- 230000001186 cumulative Effects 0.000 description 12
- 230000000926 neurological Effects 0.000 description 10
- 210000003169 Central Nervous System Anatomy 0.000 description 9
- 239000011780 sodium chloride Substances 0.000 description 9
- 235000002639 sodium chloride Nutrition 0.000 description 9
- 230000001225 therapeutic Effects 0.000 description 9
- 230000003993 interaction Effects 0.000 description 8
- 230000005415 magnetization Effects 0.000 description 8
- 238000000034 method Methods 0.000 description 8
- 229940068196 placebo Drugs 0.000 description 8
- 239000000902 placebo Substances 0.000 description 8
- 229940003504 Avonex Drugs 0.000 description 7
- 229940077362 Extavia Drugs 0.000 description 7
- 102100012588 MOG Human genes 0.000 description 7
- 229940038850 Rebif Drugs 0.000 description 7
- 150000001875 compounds Chemical class 0.000 description 7
- 239000003981 vehicle Substances 0.000 description 7
- -1 e drate Substances 0.000 description 6
- 206010003694 Atrophy Diseases 0.000 description 6
- 241000229754 Iva xanthiifolia Species 0.000 description 6
- 241001465754 Metazoa Species 0.000 description 6
- 231100000494 adverse effect Toxicity 0.000 description 6
- 238000004364 calculation method Methods 0.000 description 6
- 238000006243 chemical reaction Methods 0.000 description 6
- 230000001684 chronic Effects 0.000 description 6
- 238000003745 diagnosis Methods 0.000 description 6
- 239000002552 dosage form Substances 0.000 description 6
- 150000003839 salts Chemical class 0.000 description 6
- 238000007920 subcutaneous administration Methods 0.000 description 6
- 230000001629 suppression Effects 0.000 description 6
- 210000001519 tissues Anatomy 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- 229940021459 Betaseron Drugs 0.000 description 5
- 229940038717 Copaxone Drugs 0.000 description 5
- 210000002683 Foot Anatomy 0.000 description 5
- 210000004027 cells Anatomy 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 235000013305 food Nutrition 0.000 description 5
- 231100000486 side effect Toxicity 0.000 description 5
- 239000000126 substance Substances 0.000 description 5
- 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 4
- 208000000999 Encephalomyelitis, Autoimmune, Experimental Diseases 0.000 description 4
- KKGQTZUTZRNORY-UHFFFAOYSA-N Fingolimod Chemical compound CCCCCCCCC1=CC=C(CCC(N)(CO)CO)C=C1 KKGQTZUTZRNORY-UHFFFAOYSA-N 0.000 description 4
- AIONOLUJZLIMTK-AWEZNQCLSA-N Hesperetin Chemical compound C1=C(O)C(OC)=CC=C1[C@H]1OC2=CC(O)=CC(O)=C2C(=O)C1 AIONOLUJZLIMTK-AWEZNQCLSA-N 0.000 description 4
- 210000002966 Serum Anatomy 0.000 description 4
- 229940032147 Starch Drugs 0.000 description 4
- 229920002472 Starch Polymers 0.000 description 4
- 230000001363 autoimmune Effects 0.000 description 4
- 239000000839 emulsion Substances 0.000 description 4
- 229960000556 fingolimod Drugs 0.000 description 4
- 230000002757 inflammatory Effects 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 239000008107 starch Substances 0.000 description 4
- 235000019698 starch Nutrition 0.000 description 4
- 230000002459 sustained Effects 0.000 description 4
- 201000010874 syndrome Diseases 0.000 description 4
- 239000001100 (2S)-5,7-dihydroxy-2-(3-hydroxy-4-methoxyphenyl)chroman-4-one Substances 0.000 description 3
- 229920001817 Agar Polymers 0.000 description 3
- 210000004369 Blood Anatomy 0.000 description 3
- 229940077737 Brain-Derived Neurotrophic Factor Drugs 0.000 description 3
- 102000004219 Brain-Derived Neurotrophic Factor Human genes 0.000 description 3
- 108090000715 Brain-Derived Neurotrophic Factor Proteins 0.000 description 3
- 239000004255 Butylated hydroxyanisole Substances 0.000 description 3
- CZBZUDVBLSSABA-UHFFFAOYSA-N Butylated hydroxyanisole Chemical compound COC1=CC=C(O)C(C(C)(C)C)=C1.COC1=CC=C(O)C=C1C(C)(C)C CZBZUDVBLSSABA-UHFFFAOYSA-N 0.000 description 3
- 239000004322 Butylated hydroxytoluene Substances 0.000 description 3
- 206010012305 Demyelination Diseases 0.000 description 3
- 108010010803 Gelatin Proteins 0.000 description 3
- 229940047124 Interferons Drugs 0.000 description 3
- 108010035649 Natalizumab Proteins 0.000 description 3
- 208000002193 Pain Diseases 0.000 description 3
- 206010033775 Paraesthesia Diseases 0.000 description 3
- GEHJYWRUCIMESM-UHFFFAOYSA-L Sodium sulfite Chemical compound [Na+].[Na+].[O-]S([O-])=O GEHJYWRUCIMESM-UHFFFAOYSA-L 0.000 description 3
- 210000003932 Urinary Bladder Anatomy 0.000 description 3
- 210000002700 Urine Anatomy 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 239000002253 acid Substances 0.000 description 3
- 239000008272 agar Substances 0.000 description 3
- 235000010419 agar Nutrition 0.000 description 3
- 230000003113 alkalizing Effects 0.000 description 3
- 230000003078 antioxidant Effects 0.000 description 3
- 239000003963 antioxidant agent Substances 0.000 description 3
- 235000006708 antioxidants Nutrition 0.000 description 3
- 230000003376 axonal Effects 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 235000019282 butylated hydroxyanisole Nutrition 0.000 description 3
- 235000010354 butylated hydroxytoluene Nutrition 0.000 description 3
- 230000001054 cortical Effects 0.000 description 3
- 238000003379 elimination reaction Methods 0.000 description 3
- 229920000159 gelatin Polymers 0.000 description 3
- 239000008273 gelatin Substances 0.000 description 3
- 235000019322 gelatine Nutrition 0.000 description 3
- 235000011852 gelatine desserts Nutrition 0.000 description 3
- 230000002068 genetic Effects 0.000 description 3
- 238000003384 imaging method Methods 0.000 description 3
- 238000009533 lab test Methods 0.000 description 3
- 229960005027 natalizumab Drugs 0.000 description 3
- 230000036407 pain Effects 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- JWHPPWBIIQMBQC-UHFFFAOYSA-M sodium;5-chloro-3-[ethyl(phenyl)carbamoyl]-1-methyl-2-oxoquinolin-4-olate Chemical compound [Na+].[O-]C=1C2=C(Cl)C=CC=C2N(C)C(=O)C=1C(=O)N(CC)C1=CC=CC=C1 JWHPPWBIIQMBQC-UHFFFAOYSA-M 0.000 description 3
- 231100000730 tolerability Toxicity 0.000 description 3
- 239000003053 toxin Substances 0.000 description 3
- GVJHHUAWPYXKBD-IEOSBIPESA-N α-tocopherol Chemical compound OC1=C(C)C(C)=C2O[C@@](CCC[C@H](C)CCC[C@H](C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-IEOSBIPESA-N 0.000 description 3
- WMBWREPUVVBILR-WIYYLYMNSA-N (-)-Epigallocatechin-3-o-gallate Chemical compound O([C@@H]1CC2=C(O)C=C(C=C2O[C@@H]1C=1C=C(O)C(O)=C(O)C=1)O)C(=O)C1=CC(O)=C(O)C(O)=C1 WMBWREPUVVBILR-WIYYLYMNSA-N 0.000 description 2
- VHJLVAABSRFDPM-UHFFFAOYSA-N 1,4-dimercaptobutane-2,3-diol Chemical compound SCC(O)C(O)CS VHJLVAABSRFDPM-UHFFFAOYSA-N 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N 2-mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- YXOLAZRVSSWPPT-UHFFFAOYSA-N 3,5,7,2',4'-Pentahydroxyflavonol Chemical compound OC1=CC(O)=CC=C1C1=C(O)C(=O)C2=C(O)C=C(O)C=C2O1 YXOLAZRVSSWPPT-UHFFFAOYSA-N 0.000 description 2
- 108010082126 Alanine Transaminase Proteins 0.000 description 2
- 239000004382 Amylase Substances 0.000 description 2
- 102000013142 Amylases Human genes 0.000 description 2
- 108010065511 Amylases Proteins 0.000 description 2
- KZNIFHPLKGYRTM-UHFFFAOYSA-N Apigenin Chemical compound C1=CC(O)=CC=C1C1=CC(=O)C2=C(O)C=C(O)C=C2O1 KZNIFHPLKGYRTM-UHFFFAOYSA-N 0.000 description 2
- 206010003246 Arthritis Diseases 0.000 description 2
- 206010003591 Ataxia Diseases 0.000 description 2
- 206010004146 Basal cell carcinoma Diseases 0.000 description 2
- WUADCCWRTIWANL-UHFFFAOYSA-N Biochanin A Chemical compound C1=CC(OC)=CC=C1C1=COC2=CC(O)=CC(O)=C2C1=O WUADCCWRTIWANL-UHFFFAOYSA-N 0.000 description 2
- 208000001183 Brain Injury Diseases 0.000 description 2
- 210000000133 Brain Stem Anatomy 0.000 description 2
- 229940043253 Butylated Hydroxyanisole Drugs 0.000 description 2
- 229940095259 Butylated Hydroxytoluene Drugs 0.000 description 2
- 102100004057 CYP3A4 Human genes 0.000 description 2
- 101710007540 CYP3A4 Proteins 0.000 description 2
- OSGAYBCDTDRGGQ-UHFFFAOYSA-L Calcium sulfate Chemical compound [Ca+2].[O-]S([O-])(=O)=O OSGAYBCDTDRGGQ-UHFFFAOYSA-L 0.000 description 2
- RTIXKCRFFJGDFG-UHFFFAOYSA-N Chrysin Chemical compound C=1C(O)=CC(O)=C(C(C=2)=O)C=1OC=2C1=CC=CC=C1 RTIXKCRFFJGDFG-UHFFFAOYSA-N 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N D-Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 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 2
- 238000008789 Direct Bilirubin Methods 0.000 description 2
- 206010067671 Disease complication Diseases 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 210000003414 Extremities Anatomy 0.000 description 2
- 102100010966 GPT Human genes 0.000 description 2
- 229940014689 Gilenya Drugs 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 206010062016 Immunosuppression Diseases 0.000 description 2
- 206010061218 Inflammation Diseases 0.000 description 2
- 108010005714 Interferon beta-1b Proteins 0.000 description 2
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 2
- 241001490312 Lithops pseudotruncatella Species 0.000 description 2
- 206010025135 Lupus erythematosus Diseases 0.000 description 2
- IQPNAANSBPBGFQ-UHFFFAOYSA-N Luteolin Chemical compound C=1C(O)=CC(O)=C(C(C=2)=O)C=1OC=2C1=CC=C(O)C(O)=C1 IQPNAANSBPBGFQ-UHFFFAOYSA-N 0.000 description 2
- 229920002521 Macromolecule Polymers 0.000 description 2
- LXCFILQKKLGQFO-UHFFFAOYSA-N Methylparaben Chemical compound COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 2
- KKZJGLLVHKMTCM-UHFFFAOYSA-N Mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 2
- 229960001156 Mitoxantrone Drugs 0.000 description 2
- FTVWIRXFELQLPI-ZDUSSCGKSA-N Naringenin Chemical compound C1=CC(O)=CC=C1[C@H]1OC2=CC(O)=CC(O)=C2C(=O)C1 FTVWIRXFELQLPI-ZDUSSCGKSA-N 0.000 description 2
- 208000009025 Nervous System Disease Diseases 0.000 description 2
- 210000004940 Nucleus Anatomy 0.000 description 2
- 208000003435 Optic Neuritis Diseases 0.000 description 2
- 108010081690 Pertussis Toxin Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 229940083542 Sodium Drugs 0.000 description 2
- PPASLZSBLFJQEF-RKJRWTFHSA-M Sodium erythorbate Chemical compound [Na+].OC[C@@H](O)[C@H]1OC(=O)C(O)=C1[O-] PPASLZSBLFJQEF-RKJRWTFHSA-M 0.000 description 2
- 229940091252 Sodium supplements Drugs 0.000 description 2
- QIQXTHQIDYTFRH-UHFFFAOYSA-N Stearic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 2
- CZMRCDWAGMRECN-GDQSFJPYSA-N Sucrose Natural products O([C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@H](CO)O1)[C@@]1(CO)[C@H](O)[C@@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-GDQSFJPYSA-N 0.000 description 2
- 210000001744 T-Lymphocytes Anatomy 0.000 description 2
- CXQWRCVTCMQVQX-LSDHHAIUSA-N Taxifolin Chemical compound C1([C@@H]2[C@H](C(C3=C(O)C=C(O)C=C3O2)=O)O)=CC=C(O)C(O)=C1 CXQWRCVTCMQVQX-LSDHHAIUSA-N 0.000 description 2
- 231100000765 Toxin Toxicity 0.000 description 2
- 206010044565 Tremor Diseases 0.000 description 2
- VILCJCGEZXAXTO-UHFFFAOYSA-N Triethylenetetramine Chemical compound NCCNCCNCCN VILCJCGEZXAXTO-UHFFFAOYSA-N 0.000 description 2
- KWIUHFFTVRNATP-UHFFFAOYSA-N Trimethylglycine Chemical compound C[N+](C)(C)CC([O-])=O KWIUHFFTVRNATP-UHFFFAOYSA-N 0.000 description 2
- 229940079023 Tysabri Drugs 0.000 description 2
- 208000001756 Virus Disease Diseases 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 230000001154 acute Effects 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 230000000172 allergic Effects 0.000 description 2
- 235000019418 amylase Nutrition 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 230000003110 anti-inflammatory Effects 0.000 description 2
- 230000030741 antigen processing and presentation Effects 0.000 description 2
- 201000008937 atopic dermatitis Diseases 0.000 description 2
- 239000011230 binding agent Substances 0.000 description 2
- 201000004569 blindness Diseases 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 238000010322 bone marrow transplantation Methods 0.000 description 2
- 231100000874 brain damage Toxicity 0.000 description 2
- 239000002738 chelating agent Substances 0.000 description 2
- 210000000038 chest Anatomy 0.000 description 2
- 230000001149 cognitive Effects 0.000 description 2
- 230000000052 comparative effect Effects 0.000 description 2
- 239000002131 composite material Substances 0.000 description 2
- 229920001577 copolymer Polymers 0.000 description 2
- 230000000875 corresponding Effects 0.000 description 2
- 230000016396 cytokine production Effects 0.000 description 2
- 239000002254 cytotoxic agent Substances 0.000 description 2
- 231100000599 cytotoxic agent Toxicity 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- 230000001712 encephalitogenic Effects 0.000 description 2
- 201000002491 encephalomyelitis Diseases 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 229960001587 hesperetin Drugs 0.000 description 2
- 235000010209 hesperetin Nutrition 0.000 description 2
- 230000001506 immunosuppresive Effects 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 229960003161 interferon beta-1b Drugs 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 229930013032 isoflavonoids Natural products 0.000 description 2
- 235000012891 isoflavonoids Nutrition 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- 229920000609 methyl cellulose Polymers 0.000 description 2
- 239000001923 methylcellulose Substances 0.000 description 2
- 235000010981 methylcellulose Nutrition 0.000 description 2
- ZAHQPTJLOCWVPG-UHFFFAOYSA-N mitoxantrone dihydrochloride Chemical compound Cl.Cl.O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO ZAHQPTJLOCWVPG-UHFFFAOYSA-N 0.000 description 2
- 230000000051 modifying Effects 0.000 description 2
- 230000004112 neuroprotection Effects 0.000 description 2
- 239000000546 pharmaceutic aid Substances 0.000 description 2
- 230000000275 pharmacokinetic Effects 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 230000035935 pregnancy Effects 0.000 description 2
- 230000000750 progressive Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 230000001568 sexual Effects 0.000 description 2
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 235000015424 sodium Nutrition 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 235000010265 sodium sulphite Nutrition 0.000 description 2
- 238000004611 spectroscopical analysis Methods 0.000 description 2
- 239000007929 subcutaneous injection Substances 0.000 description 2
- 238000010254 subcutaneous injection Methods 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 108020003112 toxins Proteins 0.000 description 2
- 238000002562 urinalysis Methods 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N β-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- PFTAWBLQPZVEMU-UKRRQHHQSA-N (-)-epicatechin Chemical compound C1([C@H]2OC3=CC(O)=CC(O)=C3C[C@H]2O)=CC=C(O)C(O)=C1 PFTAWBLQPZVEMU-UKRRQHHQSA-N 0.000 description 1
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2R,3R,4S,5R,6S)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2S,3R,4S,5R,6R)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2R,3R,4S,5R,6R)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 1
- AOFUBOWZWQFQJU-SNOJBQEQSA-N (2R,3S,4S,5R)-2,5-bis(hydroxymethyl)oxolane-2,3,4-triol;(2S,3R,4S,5S,6R)-6-(hydroxymethyl)oxane-2,3,4,5-tetrol Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O.OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@@H]1O AOFUBOWZWQFQJU-SNOJBQEQSA-N 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N 1,2-ethanediamine Chemical compound NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- JUSMHIGDXPKSID-DVKNGEFBSA-N 1-thio-β-D-glucopyranose Chemical compound OC[C@H]1O[C@@H](S)[C@H](O)[C@@H](O)[C@@H]1O JUSMHIGDXPKSID-DVKNGEFBSA-N 0.000 description 1
- HVQAJTFOCKOKIN-UHFFFAOYSA-N 3-Hydroxyflavone Chemical compound O1C2=CC=CC=C2C(=O)C(O)=C1C1=CC=CC=C1 HVQAJTFOCKOKIN-UHFFFAOYSA-N 0.000 description 1
- PJUIMOJAAPLTRJ-UHFFFAOYSA-N 3-Mercaptopropane-1,2-diol Chemical compound OCC(O)CS PJUIMOJAAPLTRJ-UHFFFAOYSA-N 0.000 description 1
- NUKYPUAOHBNCPY-UHFFFAOYSA-N 4-Aminopyridine Chemical compound NC1=CC=NC=C1 NUKYPUAOHBNCPY-UHFFFAOYSA-N 0.000 description 1
- GZSOSUNBTXMUFQ-NJGQXECBSA-N 5,7,3'-Trihydroxy-4'-methoxyflavone 7-O-rutinoside Natural products O(C[C@@H]1[C@@H](O)[C@H](O)[C@@H](O)[C@H](Oc2cc(O)c3C(=O)C=C(c4cc(O)c(OC)cc4)Oc3c2)O1)[C@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@H](C)O1 GZSOSUNBTXMUFQ-NJGQXECBSA-N 0.000 description 1
- HRZFUMHJMZEROT-UHFFFAOYSA-L 7681-57-4 Chemical compound [Na+].[Na+].[O-]S(=O)S([O-])(=O)=O HRZFUMHJMZEROT-UHFFFAOYSA-L 0.000 description 1
- 229940023476 Agar Drugs 0.000 description 1
- 206010001584 Alcohol abuse Diseases 0.000 description 1
- 229940117893 Apigenin Drugs 0.000 description 1
- 102000000546 Apoferritins Human genes 0.000 description 1
- 108010002084 Apoferritins Proteins 0.000 description 1
- 206010003210 Arteriosclerosis Diseases 0.000 description 1
- 241000209134 Arundinaria Species 0.000 description 1
- 241000416162 Astragalus gummifer Species 0.000 description 1
- 206010003816 Autoimmune disease Diseases 0.000 description 1
- 210000003050 Axons Anatomy 0.000 description 1
- 102100004569 BFSP1 Human genes 0.000 description 1
- 101700023229 BFSP1 Proteins 0.000 description 1
- 229940092782 Bentonite Drugs 0.000 description 1
- NNUSCVFWUPARGJ-UHFFFAOYSA-N Bilirubin Chemical compound N1C(=O)C(C)=C(C=C)\C1=C\C1=NC(CC=2[C](C(C)=C(\C=C/3C(=C(C=C)C(=O)N\3)C)N=2)CCC(O)=O)=C(CCC(O)=O)[C]1C NNUSCVFWUPARGJ-UHFFFAOYSA-N 0.000 description 1
- 206010006784 Burning sensation Diseases 0.000 description 1
- 210000001266 CD8-Positive T-Lymphocytes Anatomy 0.000 description 1
- 241000220450 Cajanus cajan Species 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 206010007558 Cardiac failure chronic Diseases 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 description 1
- PTOAARAWEBMLNO-KVQBGUIXSA-N Cladribine Chemical compound C1=NC=2C(N)=NC(Cl)=NC=2N1[C@H]1C[C@H](O)[C@@H](CO)O1 PTOAARAWEBMLNO-KVQBGUIXSA-N 0.000 description 1
- 206010009696 Clumsiness Diseases 0.000 description 1
- 206010010774 Constipation Diseases 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 241000276573 Cottidae Species 0.000 description 1
- 229940109239 Creatinine Drugs 0.000 description 1
- 206010011401 Crohn's disease Diseases 0.000 description 1
- 229960001681 Croscarmellose Sodium Drugs 0.000 description 1
- 229920002785 Croscarmellose sodium Polymers 0.000 description 1
- 208000000264 Deglutition Disorders Diseases 0.000 description 1
- 208000004275 Demyelinating Disease Diseases 0.000 description 1
- 229940099217 Desferal Drugs 0.000 description 1
- MKXZASYAUGDDCJ-SZMVWBNQSA-N Dextromethorphan Chemical compound C1CCC[C@H]2[C@@]3([H])N(C)CC[C@]21C1=CC(OC)=CC=C1C3 MKXZASYAUGDDCJ-SZMVWBNQSA-N 0.000 description 1
- 206010012601 Diabetes mellitus Diseases 0.000 description 1
- NEFBYIFKOOEVPA-UHFFFAOYSA-K Dicalcium phosphate Chemical compound [Ca+2].[Ca+2].[O-]P([O-])([O-])=O NEFBYIFKOOEVPA-UHFFFAOYSA-K 0.000 description 1
- 235000019739 Dicalciumphosphate Nutrition 0.000 description 1
- 208000003164 Diplopia Diseases 0.000 description 1
- 241000668709 Dipterocarpus costatus Species 0.000 description 1
- 206010013654 Drug abuse Diseases 0.000 description 1
- 206010013700 Drug hypersensitivity Diseases 0.000 description 1
- 206010013710 Drug interaction Diseases 0.000 description 1
- 206010013950 Dysphagia Diseases 0.000 description 1
- 208000010228 Erectile Dysfunction Diseases 0.000 description 1
- NUVBSKCKDOMJSU-UHFFFAOYSA-N Ethylparaben Chemical compound CCOC(=O)C1=CC=C(O)C=C1 NUVBSKCKDOMJSU-UHFFFAOYSA-N 0.000 description 1
- 206010016059 Facial pain Diseases 0.000 description 1
- 206010016256 Fatigue Diseases 0.000 description 1
- 239000004606 Fillers/Extenders Substances 0.000 description 1
- UIWYJDYFSGRHKR-UHFFFAOYSA-N Gadolinium Chemical compound [Gd] UIWYJDYFSGRHKR-UHFFFAOYSA-N 0.000 description 1
- 229910052688 Gadolinium Inorganic materials 0.000 description 1
- 229940045109 Genistein Drugs 0.000 description 1
- ZCOLJUOHXJRHDI-CMWLGVBASA-N Genistin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1OC1=CC(O)=C2C(=O)C(C=3C=CC(O)=CC=3)=COC2=C1 ZCOLJUOHXJRHDI-CMWLGVBASA-N 0.000 description 1
- RWSXRVCMGQZWBV-WDSKDSINSA-N Glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 1
- 229960003180 Glutathione Drugs 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- 206010018651 Graft versus host disease Diseases 0.000 description 1
- 208000009329 Graft vs Host Disease Diseases 0.000 description 1
- 108010023302 HDL Cholesterol Proteins 0.000 description 1
- 229940025878 Hesperidin Drugs 0.000 description 1
- QUQPHWDTPGMPEX-YEDPJISVSA-N Hesperidin Natural products O(C[C@H]1[C@@H](O)[C@H](O)[C@@H](O)[C@H](Oc2cc(O)c3C(=O)C[C@@H](c4cc(O)c(OC)cc4)Oc3c2)O1)[C@H]1[C@@H](O)[C@H](O)[C@@H](O)[C@H](C)O1 QUQPHWDTPGMPEX-YEDPJISVSA-N 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 229940088597 Hormone Drugs 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- 241000257303 Hymenoptera Species 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 101700011451 IFNB1 Proteins 0.000 description 1
- 102100015720 IFNB1 Human genes 0.000 description 1
- 229960003444 IMMUNOSUPPRESSANTS Drugs 0.000 description 1
- 210000000987 Immune System Anatomy 0.000 description 1
- 102000018358 Immunoglobulins Human genes 0.000 description 1
- 108060003951 Immunoglobulins Proteins 0.000 description 1
- 206010021639 Incontinence Diseases 0.000 description 1
- 210000004969 Inflammatory Cells Anatomy 0.000 description 1
- 206010021972 Inflammatory bowel disease Diseases 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- 108010028554 LDL Cholesterol Proteins 0.000 description 1
- 239000004367 Lipase Substances 0.000 description 1
- 229940040461 Lipase Drugs 0.000 description 1
- 210000004185 Liver Anatomy 0.000 description 1
- 208000009856 Lung Disease Diseases 0.000 description 1
- 208000005777 Lupus Nephritis Diseases 0.000 description 1
- 210000004698 Lymphocytes Anatomy 0.000 description 1
- 208000008466 Metabolic Disease Diseases 0.000 description 1
- 230000036740 Metabolism Effects 0.000 description 1
- 229960002900 Methylcellulose Drugs 0.000 description 1
- VHRSUDSXCMQTMA-PJHHCJLFSA-N Methylprednisolone Chemical compound C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)CO)CC[C@H]21 VHRSUDSXCMQTMA-PJHHCJLFSA-N 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 210000000274 Microglia Anatomy 0.000 description 1
- 206010027926 Monoplegia Diseases 0.000 description 1
- 208000008238 Muscle Spasticity Diseases 0.000 description 1
- 206010028323 Muscle pain Diseases 0.000 description 1
- 206010028334 Muscle spasms Diseases 0.000 description 1
- 206010052904 Musculoskeletal stiffness Diseases 0.000 description 1
- 208000000112 Myalgia Diseases 0.000 description 1
- 210000003007 Myelin Sheath Anatomy 0.000 description 1
- 208000003926 Myelitis Diseases 0.000 description 1
- DCYOADKBABEMIQ-FLCVNNLFSA-N Myricitrin Natural products O([C@H]1[C@@H](O)[C@H](O)[C@@H](O)[C@H](C)O1)C1=C(c2cc(O)c(O)c(O)c2)Oc2c(c(O)cc(O)c2)C1=O DCYOADKBABEMIQ-FLCVNNLFSA-N 0.000 description 1
- CVRXLMUYFMERMJ-UHFFFAOYSA-N N,N,N',N'-tetrakis(pyridin-2-ylmethyl)ethane-1,2-diamine Chemical compound C=1C=CC=NC=1CN(CC=1N=CC=CC=1)CCN(CC=1N=CC=CC=1)CC1=CC=CC=N1 CVRXLMUYFMERMJ-UHFFFAOYSA-N 0.000 description 1
- FFDGPVCHZBVARC-UHFFFAOYSA-N N,N-dimethylglycine zwitterion Chemical compound CN(C)CC(O)=O FFDGPVCHZBVARC-UHFFFAOYSA-N 0.000 description 1
- WHNWPMSKXPGLAX-UHFFFAOYSA-N N-Vinylpyrrolidone Chemical compound C=CN1CCCC1=O WHNWPMSKXPGLAX-UHFFFAOYSA-N 0.000 description 1
- 102000003945 NF-kappa B Human genes 0.000 description 1
- 108010057466 NF-kappa B Proteins 0.000 description 1
- IYRGXJIJGHOCFS-UHFFFAOYSA-N Neocuproine Chemical compound C1=C(C)N=C2C3=NC(C)=CC=C3C=CC2=C1 IYRGXJIJGHOCFS-UHFFFAOYSA-N 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 241001182492 Nes Species 0.000 description 1
- 208000000434 Neuritis, Autoimmune, Experimental Diseases 0.000 description 1
- GUBGYTABKSRVRQ-DCSYEGIMSA-N OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O Chemical compound OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-DCSYEGIMSA-N 0.000 description 1
- 230000035536 Oral bioavailability Effects 0.000 description 1
- 206010033799 Paralysis Diseases 0.000 description 1
- VVNCNSJFMMFHPL-VKHMYHEASA-N Penicillamine Chemical compound CC(C)(S)[C@@H](N)C(O)=O VVNCNSJFMMFHPL-VKHMYHEASA-N 0.000 description 1
- 210000000578 Peripheral Nerves Anatomy 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 241001272996 Polyphylla fullo Species 0.000 description 1
- RWPGFSMJFRPDDP-UHFFFAOYSA-L Potassium metabisulfite Chemical compound [K+].[K+].[O-]S(=O)S([O-])(=O)=O RWPGFSMJFRPDDP-UHFFFAOYSA-L 0.000 description 1
- BHZRJJOHZFYXTO-UHFFFAOYSA-L Potassium sulfite Chemical compound [K+].[K+].[O-]S([O-])=O BHZRJJOHZFYXTO-UHFFFAOYSA-L 0.000 description 1
- 229940069328 Povidone Drugs 0.000 description 1
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N Prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 1
- 206010063401 Primary progressive multiple sclerosis Diseases 0.000 description 1
- 206010053395 Progressive multiple sclerosis Diseases 0.000 description 1
- 229940075579 Propyl Gallate Drugs 0.000 description 1
- 206010037175 Psychiatric disease Diseases 0.000 description 1
- 229960001285 Quercetin Drugs 0.000 description 1
- REFJWTPEDVJJIY-UHFFFAOYSA-N Quercetin Chemical compound C=1C(O)=CC(O)=C(C(C=2O)=O)C=1OC=2C1=CC=C(O)C(O)=C1 REFJWTPEDVJJIY-UHFFFAOYSA-N 0.000 description 1
- 206010038683 Respiratory disease Diseases 0.000 description 1
- 206010039073 Rheumatoid arthritis Diseases 0.000 description 1
- 241000220010 Rhode Species 0.000 description 1
- IKGXIBQEEMLURG-BKUODXTLSA-N Rutin Chemical compound O[C@H]1[C@H](O)[C@@H](O)[C@H](C)O[C@@H]1OC[C@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](OC=2C(C3=C(O)C=C(O)C=C3OC=2C=2C=C(O)C(O)=CC=2)=O)O1 IKGXIBQEEMLURG-BKUODXTLSA-N 0.000 description 1
- 229940081967 Rutin Drugs 0.000 description 1
- 229960005055 SODIUM ASCORBATE Drugs 0.000 description 1
- 206010040030 Sensory loss Diseases 0.000 description 1
- 102000018674 Sodium Channels Human genes 0.000 description 1
- 108010052164 Sodium Channels Proteins 0.000 description 1
- WXMKPNITSTVMEF-UHFFFAOYSA-M Sodium benzoate Chemical compound [Na+].[O-]C(=O)C1=CC=CC=C1 WXMKPNITSTVMEF-UHFFFAOYSA-M 0.000 description 1
- JVBXVOWTABLYPX-UHFFFAOYSA-L Sodium dithionite Chemical compound [Na+].[Na+].[O-]S(=O)S([O-])=O JVBXVOWTABLYPX-UHFFFAOYSA-L 0.000 description 1
- BCKXLBQYZLBQEK-KVVVOXFISA-M Sodium oleate Chemical compound [Na+].CCCCCCCC\C=C/CCCCCCCC([O-])=O BCKXLBQYZLBQEK-KVVVOXFISA-M 0.000 description 1
- RYYKJJJTJZKILX-UHFFFAOYSA-M Sodium stearate Chemical compound [Na+].CCCCCCCCCCCCCCCCCC([O-])=O RYYKJJJTJZKILX-UHFFFAOYSA-M 0.000 description 1
- 240000006394 Sorghum bicolor Species 0.000 description 1
- 235000011684 Sorghum saccharatum Nutrition 0.000 description 1
- 208000005392 Spasm Diseases 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 208000006011 Stroke Diseases 0.000 description 1
- PZBFGYYEXUXCOF-UHFFFAOYSA-N TCEP Chemical compound OC(=O)CCP(CCC(O)=O)CCC(O)=O PZBFGYYEXUXCOF-UHFFFAOYSA-N 0.000 description 1
- 235000009430 Thespesia populnea Nutrition 0.000 description 1
- YODZTKMDCQEPHD-UHFFFAOYSA-N Thiodiglycol Chemical compound OCCSCCO YODZTKMDCQEPHD-UHFFFAOYSA-N 0.000 description 1
- 229950006389 Thiodiglycol Drugs 0.000 description 1
- 229960001295 Tocopherol Drugs 0.000 description 1
- 238000008050 Total Bilirubin Reagent Methods 0.000 description 1
- 229940116362 Tragacanth Drugs 0.000 description 1
- 229920001615 Tragacanth Polymers 0.000 description 1
- 229960001124 Trientine Drugs 0.000 description 1
- 206010044652 Trigeminal neuralgia Diseases 0.000 description 1
- HWKQNAWCHQMZHK-UHFFFAOYSA-N Trolnitrate Chemical compound [O-][N+](=O)OCCN(CCO[N+]([O-])=O)CCO[N+]([O-])=O HWKQNAWCHQMZHK-UHFFFAOYSA-N 0.000 description 1
- 206010052769 Vertigos Diseases 0.000 description 1
- 206010047461 Viral infection Diseases 0.000 description 1
- 206010047513 Vision blurred Diseases 0.000 description 1
- 206010047571 Visual impairment Diseases 0.000 description 1
- 241000289690 Xenarthra Species 0.000 description 1
- 239000008186 active pharmaceutical agent Substances 0.000 description 1
- 238000007792 addition Methods 0.000 description 1
- 230000002730 additional Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000000240 adjuvant Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 201000003082 alcohol use disease Diseases 0.000 description 1
- 201000005794 allergic hypersensitivity disease Diseases 0.000 description 1
- 229940087168 alpha Tocopherol Drugs 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminum Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007172 antigens Proteins 0.000 description 1
- 102000038129 antigens Human genes 0.000 description 1
- 235000008714 apigenin Nutrition 0.000 description 1
- 230000037007 arousal Effects 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 125000003289 ascorbyl group Chemical group [H]O[C@@]([H])(C([H])([H])O*)[C@@]1([H])OC(=O)C(O*)=C1O* 0.000 description 1
- 201000001320 atherosclerosis Diseases 0.000 description 1
- 230000002238 attenuated Effects 0.000 description 1
- 201000009596 autoimmune hypersensitivity disease Diseases 0.000 description 1
- ZETCGWYACBNPIH-UHFFFAOYSA-N azane;sulfurous acid Chemical compound N.OS(O)=O ZETCGWYACBNPIH-UHFFFAOYSA-N 0.000 description 1
- 239000000440 bentonite Substances 0.000 description 1
- 229910000278 bentonite Inorganic materials 0.000 description 1
- 235000012216 bentonite Nutrition 0.000 description 1
- 229960003237 betaine Drugs 0.000 description 1
- 229960000074 biopharmaceuticals Drugs 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 238000004820 blood count Methods 0.000 description 1
- 210000003008 brain-resident macrophage Anatomy 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 235000011132 calcium sulphate Nutrition 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
- 230000015556 catabolic process Effects 0.000 description 1
- 150000001765 catechin Chemical class 0.000 description 1
- 229930016253 catechin Natural products 0.000 description 1
- 235000005487 catechin Nutrition 0.000 description 1
- 230000001364 causal effect Effects 0.000 description 1
- 238000002659 cell therapy Methods 0.000 description 1
- 230000002490 cerebral Effects 0.000 description 1
- 229940121538 choriogonadotropin beta Drugs 0.000 description 1
- 229940043370 chrysin Drugs 0.000 description 1
- 235000015838 chrysin Nutrition 0.000 description 1
- 229960002436 cladribine Drugs 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 230000000295 complement Effects 0.000 description 1
- 230000002254 contraceptive Effects 0.000 description 1
- 239000003433 contraceptive agent Substances 0.000 description 1
- 239000002872 contrast media Substances 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 230000003131 corticotroph Effects 0.000 description 1
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 1
- 235000010947 crosslinked sodium carboxy methyl cellulose Nutrition 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 239000007857 degradation product Substances 0.000 description 1
- 230000001419 dependent Effects 0.000 description 1
- IDDIJAWJANBQLJ-UHFFFAOYSA-N desferrioxamine B mesylate Chemical compound [H+].CS([O-])(=O)=O.CC(=O)N(O)CCCCCNC(=O)CCC(=O)N(O)CCCCCNC(=O)CCC(=O)N(O)CCCCCN IDDIJAWJANBQLJ-UHFFFAOYSA-N 0.000 description 1
- 229940038472 dicalcium phosphate Drugs 0.000 description 1
- 229910000390 dicalcium phosphate Inorganic materials 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 230000003292 diminished Effects 0.000 description 1
- GZSOSUNBTXMUFQ-YFAPSIMESA-N diosmin Chemical compound C1=C(O)C(OC)=CC=C1C(OC1=C2)=CC(=O)C1=C(O)C=C2O[C@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO[C@H]2[C@@H]([C@H](O)[C@@H](O)[C@H](C)O2)O)O1 GZSOSUNBTXMUFQ-YFAPSIMESA-N 0.000 description 1
- 229960004352 diosmin Drugs 0.000 description 1
- 201000009910 diseases by infectious agent Diseases 0.000 description 1
- QXNVGIXVLWOKEQ-UHFFFAOYSA-N disodium Chemical compound [Na][Na] QXNVGIXVLWOKEQ-UHFFFAOYSA-N 0.000 description 1
- 231100000673 dose–response relationship 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
- 239000003792 electrolyte Substances 0.000 description 1
- 238000002451 electron ionisation mass spectrometry Methods 0.000 description 1
- 125000002587 enol group Chemical group 0.000 description 1
- 229930003335 epicatechin Natural products 0.000 description 1
- 235000012734 epicatechin Nutrition 0.000 description 1
- 229940030275 epigallocatechin gallate Drugs 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 229960001617 ethyl hydroxybenzoate Drugs 0.000 description 1
- 235000010228 ethyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000004403 ethyl p-hydroxybenzoate Substances 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 229960004979 fampridine Drugs 0.000 description 1
- 235000020937 fasting conditions Nutrition 0.000 description 1
- 230000002349 favourable Effects 0.000 description 1
- SRMBQCVUAVULDJ-UHFFFAOYSA-N ferrioxamine B Chemical compound [Fe+3].CC(=O)N([O-])CCCCCNC(=O)CCC(=O)N([O-])CCCCCNC(=O)CCC(=O)N([O-])CCCCCN SRMBQCVUAVULDJ-UHFFFAOYSA-N 0.000 description 1
- 238000009093 first-line therapy Methods 0.000 description 1
- VHBFFQKBGNRLFZ-UHFFFAOYSA-N flavone Chemical compound O1C2=CC=CC=C2C(=O)C=C1C1=CC=CC=C1 VHBFFQKBGNRLFZ-UHFFFAOYSA-N 0.000 description 1
- 235000011949 flavones Nutrition 0.000 description 1
- 229930003944 flavones Natural products 0.000 description 1
- 150000002215 flavonoids Chemical class 0.000 description 1
- 229930003935 flavonoids Natural products 0.000 description 1
- 235000017173 flavonoids Nutrition 0.000 description 1
- 229930008690 flavonol Natural products 0.000 description 1
- 235000011957 flavonols Nutrition 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 238000005755 formation reaction Methods 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 230000005021 gait Effects 0.000 description 1
- 230000002496 gastric Effects 0.000 description 1
- 230000014509 gene expression Effects 0.000 description 1
- TZBJGXHYKVUXJN-UHFFFAOYSA-N genistein Natural products C1=CC(O)=CC=C1C1=COC2=CC(O)=CC(O)=C2C1=O TZBJGXHYKVUXJN-UHFFFAOYSA-N 0.000 description 1
- 235000006539 genistein Nutrition 0.000 description 1
- 230000001434 glomerular Effects 0.000 description 1
- 230000024924 glomerular filtration Effects 0.000 description 1
- 235000001727 glucose Nutrition 0.000 description 1
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 1
- 229910052737 gold Inorganic materials 0.000 description 1
- 239000010931 gold Substances 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- QUQPHWDTPGMPEX-QJBIFVCTSA-N hesperidin Chemical compound C1=C(O)C(OC)=CC=C1[C@H]1OC2=CC(O[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@@H](CO[C@H]4[C@@H]([C@H](O)[C@@H](O)[C@H](C)O4)O)O3)O)=CC(O)=C2C(=O)C1 QUQPHWDTPGMPEX-QJBIFVCTSA-N 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- UFHFLCQGNIYNRP-UHFFFAOYSA-N hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 230000009610 hypersensitivity Effects 0.000 description 1
- 230000002519 immonomodulatory Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 239000002955 immunomodulating agent Substances 0.000 description 1
- 230000002584 immunomodulator Effects 0.000 description 1
- 229940121354 immunomodulators Drugs 0.000 description 1
- 230000001861 immunosuppresant Effects 0.000 description 1
- 201000001881 impotence Diseases 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000004968 inflammatory condition Effects 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 239000003838 injectable contraceptive agent Substances 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 238000009114 investigational therapy Methods 0.000 description 1
- 230000002427 irreversible Effects 0.000 description 1
- 150000003817 isoflavonoid derivatives Chemical class 0.000 description 1
- 150000003816 isoflavonoids Chemical class 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 102000004882 lipase Human genes 0.000 description 1
- 235000019421 lipase Nutrition 0.000 description 1
- 108090001060 lipase Proteins 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 201000009673 liver disease Diseases 0.000 description 1
- 238000011068 load Methods 0.000 description 1
- 238000010234 longitudinal analysis Methods 0.000 description 1
- 231100000896 loss of balance Toxicity 0.000 description 1
- 235000009498 luteolin Nutrition 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
- 230000036210 malignancy Effects 0.000 description 1
- PWHULOQIROXLJO-UHFFFAOYSA-N manganese Chemical compound [Mn] PWHULOQIROXLJO-UHFFFAOYSA-N 0.000 description 1
- 229910052748 manganese Inorganic materials 0.000 description 1
- 239000011572 manganese Substances 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000001404 mediated Effects 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 230000004630 mental health Effects 0.000 description 1
- 230000002503 metabolic Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000035786 metabolism Effects 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- AFVFQIVMOAPDHO-UHFFFAOYSA-M methanesulfonate Chemical class CS([O-])(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-M 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 229960002216 methylparaben Drugs 0.000 description 1
- 229960004584 methylprednisolone Drugs 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 235000007708 morin Nutrition 0.000 description 1
- DCYOADKBABEMIQ-OWMUPTOHSA-N myricitrin Chemical compound O[C@@H]1[C@H](O)[C@@H](O)[C@H](C)O[C@H]1OC1=C(C=2C=C(O)C(O)=C(O)C=2)OC2=CC(O)=CC(O)=C2C1=O DCYOADKBABEMIQ-OWMUPTOHSA-N 0.000 description 1
- 235000007625 naringenin Nutrition 0.000 description 1
- 229940117954 naringenin Drugs 0.000 description 1
- 230000007459 negative regulation of leukocyte migration Effects 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 230000000324 neuroprotective Effects 0.000 description 1
- 230000001264 neutralization Effects 0.000 description 1
- 239000002547 new drug Substances 0.000 description 1
- 230000003000 nontoxic Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000036220 oral bioavailability Effects 0.000 description 1
- 239000003539 oral contraceptive agent Substances 0.000 description 1
- 239000006186 oral dosage form Substances 0.000 description 1
- 230000001314 paroxysmal Effects 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000001717 pathogenic Effects 0.000 description 1
- 244000052769 pathogens Species 0.000 description 1
- 230000001575 pathological Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229960001639 penicillamine Drugs 0.000 description 1
- 238000011056 performance test Methods 0.000 description 1
- 230000002688 persistence Effects 0.000 description 1
- 239000008024 pharmaceutical diluent Substances 0.000 description 1
- 230000002974 pharmacogenomic Effects 0.000 description 1
- 230000036231 pharmacokinetics Effects 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 238000002616 plasmapheresis Methods 0.000 description 1
- 229920000314 poly p-methyl styrene Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 229940043349 potassium metabisulfite Drugs 0.000 description 1
- 235000010263 potassium metabisulphite Nutrition 0.000 description 1
- 235000019252 potassium sulphite Nutrition 0.000 description 1
- 229960004618 prednisone Drugs 0.000 description 1
- 230000000770 pro-inflamatory Effects 0.000 description 1
- ZTHYODDOHIVTJV-UHFFFAOYSA-N propyl 3,4,5-trihydroxybenzoate Chemical compound CCCOC(=O)C1=CC(O)=C(O)C(O)=C1 ZTHYODDOHIVTJV-UHFFFAOYSA-N 0.000 description 1
- 235000010388 propyl gallate Nutrition 0.000 description 1
- 239000000473 propyl gallate Substances 0.000 description 1
- 230000001681 protective Effects 0.000 description 1
- 201000004681 psoriasis Diseases 0.000 description 1
- 235000005875 quercetin Nutrition 0.000 description 1
- 229930002344 quercetin Natural products 0.000 description 1
- OXGUCUVFOIWWQJ-HQBVPOQASA-M quercitrin-7-olate Chemical compound O[C@@H]1[C@H](O)[C@@H](O)[C@H](C)O[C@H]1OC1=C(C=2C=C(O)C(O)=CC=2)OC2=CC([O-])=CC(O)=C2C1=O OXGUCUVFOIWWQJ-HQBVPOQASA-M 0.000 description 1
- 230000004461 rapid eye movement Effects 0.000 description 1
- 230000002829 reduced Effects 0.000 description 1
- 230000000268 renotropic Effects 0.000 description 1
- 230000033764 rhythmic process Effects 0.000 description 1
- 229930002876 rutin Natural products 0.000 description 1
- 235000005493 rutin Nutrition 0.000 description 1
- 229960004555 rutoside Drugs 0.000 description 1
- 238000004826 seaming Methods 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 230000001953 sensory Effects 0.000 description 1
- 230000006403 short-term memory Effects 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 235000010378 sodium ascorbate Nutrition 0.000 description 1
- 235000010234 sodium benzoate Nutrition 0.000 description 1
- 239000004299 sodium benzoate Substances 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- 239000004289 sodium hydrogen sulphite Substances 0.000 description 1
- 229940001584 sodium metabisulfite Drugs 0.000 description 1
- 235000010262 sodium metabisulphite Nutrition 0.000 description 1
- 159000000000 sodium salts Chemical group 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 239000000934 spermatocidal agent Substances 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 238000002660 stem cell treatment Methods 0.000 description 1
- 230000001954 sterilising Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 150000003431 steroids Chemical class 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 230000000152 swallowing Effects 0.000 description 1
- 238000002636 symptomatic treatment Methods 0.000 description 1
- 230000002195 synergetic Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- FAGUFWYHJQFNRV-UHFFFAOYSA-N tetraethylenepentamine Chemical compound NCCNCCNCCNCCN FAGUFWYHJQFNRV-UHFFFAOYSA-N 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 229940035024 thioglycerol Drugs 0.000 description 1
- 229960000984 tocofersolan Drugs 0.000 description 1
- 235000010384 tocopherol Nutrition 0.000 description 1
- 239000011732 tocopherol Substances 0.000 description 1
- 229930003799 tocopherols Natural products 0.000 description 1
- 230000036409 touch and pain Effects 0.000 description 1
- 239000000196 tragacanth Substances 0.000 description 1
- 235000010487 tragacanth Nutrition 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- 230000001960 triggered Effects 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 108010038909 turmerin Proteins 0.000 description 1
- 201000002282 venous insufficiency Diseases 0.000 description 1
- 231100000889 vertigo Toxicity 0.000 description 1
- 230000017613 viral reproduction Effects 0.000 description 1
- 230000004382 visual function Effects 0.000 description 1
- 230000004393 visual impairment Effects 0.000 description 1
- 239000001993 wax Substances 0.000 description 1
- 230000036642 wellbeing Effects 0.000 description 1
- 239000000230 xanthan gum Substances 0.000 description 1
- 229920001285 xanthan gum Polymers 0.000 description 1
- 235000010493 xanthan gum Nutrition 0.000 description 1
- 229940082509 xanthan gum Drugs 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- 239000002076 α-tocopherol Substances 0.000 description 1
- 235000004835 α-tocopherol Nutrition 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61J—CONTAINERS SPECIALLY ADAPTED FOR MEDICAL OR PHARMACEUTICAL PURPOSES; DEVICES OR METHODS SPECIALLY ADAPTED FOR BRINGING PHARMACEUTICAL PRODUCTS INTO PARTICULAR PHYSICAL OR ADMINISTERING FORMS; DEVICES FOR ADMINISTERING FOOD OR MEDICINES ORALLY; BABY COMFORTERS; DEVICES FOR RECEIVING SPITTLE
- A61J1/00—Containers specially adapted for medical or pharmaceutical purposes
- A61J1/03—Containers specially adapted for medical or pharmaceutical purposes for pills or tablets
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61J—CONTAINERS SPECIALLY ADAPTED FOR MEDICAL OR PHARMACEUTICAL PURPOSES; DEVICES OR METHODS SPECIALLY ADAPTED FOR BRINGING PHARMACEUTICAL PRODUCTS INTO PARTICULAR PHYSICAL OR ADMINISTERING FORMS; DEVICES FOR ADMINISTERING FOOD OR MEDICINES ORALLY; BABY COMFORTERS; DEVICES FOR RECEIVING SPITTLE
- A61J1/00—Containers specially adapted for medical or pharmaceutical purposes
- A61J1/14—Details; Accessories therefor
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2300/00—Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/47—Quinolines; Isoquinolines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/47—Quinolines; Isoquinolines
- A61K31/4704—2-Quinolinones, e.g. carbostyril
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/21—Interferons [IFN]
- A61K38/215—IFN-beta
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/02—Inorganic compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/16—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
- A61K47/18—Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/26—Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/02—Ophthalmic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
Abstract
This disclosure provides a method of treating a patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome comprising administering to the patient laquinimod as an add-on therapy to or in combination with interferon-?. This invention also provides a package and a pharmaceutical composition comprising laquinimod and interferon-? for treating a patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome. This disclosure also provides laquinimod for use as an add-on therapy or in combination with interferon-? in treating a patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome. This disclosure further provides use of laquinimod and interferon-? in the preparation of a combination for treating a patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome. utical composition comprising laquinimod and interferon-? for treating a patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome. This disclosure also provides laquinimod for use as an add-on therapy or in combination with interferon-? in treating a patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome. This disclosure further provides use of laquinimod and interferon-? in the preparation of a combination for treating a patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome.
Description
W0 2013I016686 PCT/U52012/048689
TREATMENT OF MULTIPLE SCLEROSIS WITH COMBINATION OF LA!QUINIMOD
AND INTERFERON-BETA
This application claims benefit of US. Provisional Application No. 61/512,817, filed July 28, 2011,
the entire content of which is hereby incorporated by reference .
Throughout this ation, various publications are referred to by first author and year of
publication. Full citations for these publications are presented in a References section immediately
before the claims. Disclosures of the documents and publications cited are hereby incorporated by
reference in their entireties into this application in order to more fully describe the state of the art as
of the date ofthe invention described herein.
ound
Multiple sis (MS) is a neurological disease affecting more than 1 million people worldwide. It
is the most common cause of neurological disability in young and middle—aged adults and has a major
physical, logical, social and financial impact on subjects and their families, friends and bodies
responsible for health care (EMEA Guideline, 2006).
It is generally assumed that MS is mediated by some kind of autoimmune process possibly triggered
by infection and superimposed upon a genetic predisposition. It is a chronic inflammatory condition
that damages the myelin of the Central Nervous System (CNS). The pathogenesis of MS is
characterized by the infiltration of active T—cells from the circulation directed against myelin
antigens into the CNS (Bjartmar, 2002). In addition to the inflammatory phase in MS, axonal loss
occurs early in the course of the disease and can be extensive over time, leading to the subsequent
pment of progressive, permanent, ogic ment and, frequently, severe disability
(Neuhaus, 2003). ms associated with the disease include fatigue, spasticity, ataxia, weakness,
bladder and bowel bances, sexual ction, pain, tremor, paroxysmal manifestations, visual
impairment, psychological problems and cognitive dysfiinction (EMEA Guideline, 2006).MS e
activity can be monitored by cranial scans, including magnetic resonance imaging (MRI) of the brain,
accumulation of disability, as well as rate and severity of relapses. The diagnosis of clinically definite
MS as determined by the Poser criteria (Poser, 1983) requires at least two neurological events
suggesting demyelination in the CNS separated in time and in location. A clinically isolated syndrome
(CIS) is a single monosymptomatic attack suggestive of MS, such as optic neuritis, brain stem
symptoms, and l myelitis. Patients with CIS that experience a second al attack are
generally considered to have ally definite multiple sclerosis (CDMS). Over 80 percent of
ts with a CIS and MR1 lesions go on to develop MS, while approximately 20 percent have a
self—limited process (Brex, 2002; Frohman, 2003).
W0 20131016686 PCT/U82012/048689
Various MS disease stages and/or types are described in Multiple Sclerosis Therapeutics (Duntiz,
1999). Among them, relapsing-remitting multiple sclerosis (RRMS) is the most common form at the
time of initial diagnosis. Many ts with RRMS have an initial relapsing—remitting course for 5-
years, which then advances into the secondary ssive MS (SPMS) disease course. Relapses
result from inflammation and demyelination, s restoration of nerve conduction and remission
is accompanied by resolution of inflammation, redistribution of sodium channels on demyelinated
axons and remyelination (Neuhaus, 2003; Noseworthy, 2000).
In April 2001, an international panel in association with the National MS Society of America
recommended diagnostic ia for multiple sis. These criteria became known as the
McDonald ia. The McDonald Criteria make use of MRI techniques and are intended to e
the Poser Criteria and the older Schumacher Criteria (McDonald, 2001). The McDonald ia was
d in March 2005 by an international panel (Polman, 2005) and updated again in 2010 (Polman,
2011).
Intervention with disease-modifying therapy at relapsing stages of MS is suggested to reduce and/or
prevent accumulating neurodegeneration (Hohlfeld, 2000; De Stefano, 1999). There are currently a
number of disease—modifying medications ed for use in relapsing MS (RMS), which includes
RRMS and SPMS (The Disease Modifying Drug Brochure, 2006). These e interferon beta l-a
(Avonex® and Rebif®), interferon beta l-b (Betaseron®), glatiramer acetate (Copaxone®),
ntrone (Novantrone®), natalizumab (Tysabri®) and fingolimod (Gilenya®). Most of them are
ed to act as immunomodulators. Mitoxantrone and natalizumab are believed to act as
immunesuppressants. However, the mechanisms of action of each have been only partly elucidated.
Immunosuppressants or cytotoxic agents are used in some subjects after failure of conventional
therapies. However, the relationship between s of the immune response induced by these
agents and the clinical efficacy in MS is far from settled (EMEA Guideline, 2006).
Other therapeutic approaches include symptomatic treatment which refers to all therapies applied to
improve the symptoms caused by the disease (EMEA Guideline, 2006) and ent of acute
relapses with corticosteroids. While ds do not affect the course of MS over time, they can
reduce the duration and severity of attacks in some subjects.
Laquinimod
Laquinimod is a novel synthetic compound with high oral bioavailability which has been suggested as
an oral formulation for the treatment of Multiple Sclerosis (MS) (Pohnan, 2005; Sandberg-Wollheim,
2005). Laquinimod and its sodium salt form are described, for example, in US. Patent No. 6,077,851.
PCT/U82012/048689
The mechanism of action of imod is not fiilly understood. Animal studies show it causes a Thl
(T helper 1 cell, produces pro-inflammatory cytokines) to Th2 (T helper 2 cell, produces anti-
inflammatory nes) shift with an anti-inflammatory profile (Yang, 2004; Brfick, 2011). Another
study demonstrated (mainly via the NFkB pathway) that laquinimod induced suppression of genes
related to antigen presentation and corresponding inflammatory ys (Gurevich, 2010). Other
suggested potential mechanisms of action include inhibition of leukocyte migration into the CNS,
se of axonal integrity, modulation of cytokine production, and increase in levels of brain—
derived neurotrophic factor (BDNF) (Runstrom, 2006; Briick, 2011).
Laquinimod showed a favorable safety and tolerability profile in two phase III trials (Results of Phase
III BRAVO Trial Reinforce Unique Profile of Laquinimod for Multiple sis Treatment; Teva
Pharma, Active Biotech Post Positive Laquinimod Phase 3 ALLEGRO s).
Interferon beta (IFN—lfl)
Interferons (IFNs) are cytokines produced and released by host cells in response to the presence of
pathogens and allow ication between cells to trigger the protective defenses of the immune
system. lFN—B has been used over that past 15 years as treatment for RRMS. IFNs’ complex
mechanisms of action are not yet completely elucidated. Commercially available IFN-B include
Avonex®, Betaseron®, Extavia® and Rebif®.
Add-On/Combination Therapy
The effects of add-0n or combination y using laquinimod and interferon—B on MS patients have
not been reported.
The administration of two drugs to treat a given condition, such as multiple sclerosis, raises a number
of potential problems. In vivo interactions between two drugs are complex. The effects of any single
drug are related to its absorption, distribution, and elimination. When two drugs are introduced into
the body, each drug can affect the absorption, distribution, and elimination of the other and hence,
alter the effects of the other. For ce, one drug may t, te or induce the production of
enzymes involved in a lic route of elimination of the other drug (Guidance for Industry, 1999).
In one example, combined administration of GA and interferon (IFN) has been experimentally shown
to abrogate the clinical effectiveness of either therapy (Brod 2000). In another experiment, it was
reported that the addition of prednisone in ation therapy with IFN—B antagonized its up-
regulator . Thus, when two drugs are administered to treat the same condition, it is
ictable whether each will complement, have no effect on, or interfere with, the therapeutic
activity of the other in a human subject.
PCT/U82012/048689
Not only may the interaction between two drugs affect the intended therapeutic activity of each drug,
but the ction may se the levels of toxic metabolites (Guidance for Industry, 1999). The
interaction may also heighten or lessen the side effects of each drug. Hence, upon administration of
two drugs to treat a disease, it is unpredictable what change will occur in the negative side profile of
each drug. In one example, the combination of natalizumab and interferon B-la was observed to
increase the risk of unanticipated side effects (Vollmer, 2008; Rudick 2006; Kleinschmidt—DeMasters,
2005; Langer—Gould 2005).
Additionally, it is difficult to accurately predict when the effects of the interaction between the two
drugs will become manifest. For example, metabolic interactions n drugs may become
apparent upon the initial administration of the second drug, after the two have reached a steady—state
tration or upon discontinuation of one of the drugs (Guidance for Industry, 1999).
ore, the state of the art at the time of filing is that the effects of an add-on or ation
therapy of two drugs, in particular laquinimod and IFN—B, cannot be predicted until the results of a
formal combination study are available.
W0 2013(016686 PCT/U82012/048689
Brief Description of the Drawings
Figure 1: Figure 1 is a graphical representation of the activity of interferon-B stered
daily, subcutaneous (5.0). alone or in combination with laquinimod in chronic EAE in
C57 BI mice. The graph shows the mean clinical score for the EAE rodents in each
group (on the y—axis) against the days (on the X—axis).
y of the Invention
In a first aspect, the present invention provides the use of 0.6mg laquinimod in the manufacture of a
medicament for the ent of a human patient afflicted with multiple sclerosis or presenting a
clinically isolated syndrome, wherein the medicament is prepared as an add-on therapy to or in
combination with an amount of interferon—B, wherein the amounts when taken together is more
effective to treat the human patient than when each agent at the same amount is administered alone.
In another aspect, the present invention provides the use of 0.6mg laquinimod and an amount of
interferon-l3 in the manufacture of a medicament for the treatment of a human patient afflicted with
le sclerosis or presenting a ally isolated syndrome, wherein the amounts when taken
together is more effective to treat the human patient than when each agent at the same amount is
administered alone.
In another aspect the present invention provides the use of an amount of laquinimod in the
manufacture of a medicament for the treatment of a human patient ed with multiple sclerosis or
presenting a clinically isolated syndrome, wherein the medicament is prepared as an add-on therapy
to or in combination with an amount of interferon-B (IFN—B), n the amounts when taken
together are more effective to treat the human patient than when each agent at the same amount is
administered alone.
In a further aspect, the present invention es the use of an amount of laquinimod and an amount
of interferon-B in the manufacture of a medicament for the treatment of a human patient afflicted with
le sclerosis or presenting a clinically isolated syndrome, wherein the s when taken
together are more effective to treat the human t than when each agent at the same amount is
administered alone.
In a still further aspect, the t invention provides a package comprising: a) a first pharmaceutical
composition comprising an amount of laquinimod and a pharmaceutically acceptable carrier; b) a
second pharmaceutical ition comprising an amount of interferon-6 and a pharmaceutically
acceptable carrier; and instructions for use of the first and second pharmaceutical compositions
er to treat a human patient afflicted with multiple sclerosis or presenting a clinically isolated
me, wherein the amount of laquinimod and the amount of interferon-6 when taken together are
more effective to treat the human patient than each agent at the same amount is administered alone.
This invention provides a method of treating a human patient afflicted with multiple sclerosis or
presenting a clinically isolated syndrome comprising orally administering to the patient a daily dose of
0.6mg laquinimod, and periodically administering to the patient a pharmaceutically effective amount of
interferon-B, wherein the amounts when taken together is more effective to treat the human patient than
when each agent is administered alone.
This invention provides a method of treating a human t afflicted with multiple sclerosis or
presenting a clinically isolated syndrome sing periodically administering to the patient an
amount of laquinimod and an amount of interferon-B, wherein the amounts when taken er are
effective to treat the human patient.
This invention also provides a package comprising a) a first pharmaceutical composition comprising
an amount of laquinimod and a pharmaceutically acceptable carrier; b) a second pharmaceutical
ition comprising an amount of interferon—B and a pharmaceutically acceptable carrier; and c)
instructions for use of the first and second ceutical itions together to treat a human
patient afflicted with multiple sclerosis or ting a clinically isolated syndrome.
This invention also provides laquinimod for use as an add-on therapy or in combination with
interferon-fl in treating a human patient afflicted with multiple sclerosis or presenting a clinically
isolated syndrome.
This invention also provides a pharmaceutical composition comprising an amount of laquinimod and
an amount of interferon-B for use in treating a human patient afflicted with le sclerosis or
presenting a clinically isolated syndrome, wherein the laquinimod and the interferon-6 are
administered simultaneously or contemporaneously.
This invention also provides a pharmaceutical composition comprising an amount of laquinimod and
an amount of interferon-B.
This invention also provides use of an amount of laquinimod and an amount of interferon-B in the
preparation of a combination for ng a human patient afflicted with multiple sclerosis or
ting a clinically isolated syndrome wherein the laquinimod and the eron-B are
administered simultaneously or poraneously.
This ion also es pharmaceutical ition comprising an amount of laquinimod for
use in treating a subject afflicted with multiple sclerosis or presenting a clinically isolated syndrome
as an add-on therapy or in combination with interferon-ti by periodically administering the
pharmaceutical composition and the interferon-B to the subject.
This invention r provides pharmaceutical composition comprising an amount of interferon-B for
use treating a subject afflicted with multiple sclerosis or presenting a clinically isolated syndrome as
an add-on therapy or in combination with laquinimod by periodically administering the
pharmaceutical ition and the laquinimod to the subject.
WO 16686 PCT/U82012/048689
Detailed Description of the Invention
This invention provides a method of treating a human patient afflicted with le sclerosis or
presenting a clinically isolated syndrome comprising orally administering to the patient a daily dose of
0.6mg imod, and periodically administering to the patient a pharmaceutically effective amount of
interferon—B, wherein the amounts when taken together is more effective to treat the human t than
when each agent is administered alone.
In one embodiment, the multiple sclerosis is relapsing multiple sis. In another embodiment, the
ing le sclerosis is relapsing-remitting multiple sclerosis.
In one ment, the amount of imod and the amount of interferon-[3 when taken together is
effective to reduce a symptom of multiple sclerosis in the human t. In another ment, the
symptom is a MRI—monitored multiple sclerosis disease activity, relapse rate, accumulation of physical
disability, frequency of relapses, frequency of al exacerbation, brain atrophy, risk for confirmed
progression, or time to confirmed disease progression.
In one embodiment, the accumulation of physical disability is assessed by the time to confirmed disease
ssion as measured by Kurtzke Expanded Disability Status Scale (EDSS) score. In another
embodiment, the patient had an EDSS score of 0—5 prior to administration of laquinimod. In another
embodiment, the patient had an EDSS score of 1-5 .5 prior to administration of imod. In another
embodiment, the patient had an EDSS score of 0-5.5 prior to stration of laquinimod. In another
embodiment, the patient had an EDSS score of 5.5 or greater prior to administration of laquinimod. In
another embodiment, confirmed disease progression is a I point increase of the EDSS score. In yet
another embodiment, confirmed disease progression is a 0.5 point increase of the EDSS score.
In one embodiment, time to confirmed disease progression is increased by 10-100%. In another
embodiment, time to confirmed disease progression is increased by 20—80%. In another embodiment,
time to confirmed disease progression is increased by 20-60%. In another embodiment, time to
confirmed disease progression is increased by 30—50%. In yet another embodiment, time to confirmed
disease progression is increased by at least 50%.
In one embodiment, laquinimod is laquinimod sodium. In another embodiment, the interferon-[3 is
administered via subcutaneous injection or intramuscular injection.
In one ment, the interferon-l3 is interferon beta-la. In another embodiment, the interferon-[3 is
interferon beta~lb.
In one embodiment, the interferon—B is administered intramuscularly. In another embodiment, the
interferon—B is stered subcutaneously. In another embodiment, the interferon-B is administered
PCT/U82012/048689
1-5 times a month. In another embodiment, the interferon-B is stered 1-3 times a month. In
another embodiment, the interferon-l3 is administered 1-5 times a week. In another embodiment, the
interferon—B is administered 1—3 times a week. In r embodiment, the interferon—B is
administered 1-5 times a day. In another embodiment, the interferon-B is administered 1-3 times a
day. In another embodiment, the interferon-B is administered every other day. In yet another
embodiment, the eron-B is stered daily.
In one embodiment, the amount interferon—B stered is about 10-300 meg. In another
embodiment, the amount interferon-B administered is about 30-250 meg. In another embodiment, the
amount interferon—B administered is about 30-440 meg. In another embodiment, the amount
interferon—B administered is about 22-44 meg. In another embodiment, the amount interferon—B
administered is about 30 meg. In another embodiment, the amount eron-[3 administered is about
250 meg.
In one embodiment, the interferon-B is interferon beta-1a and is administered intramuscularly at 30
mcg, once weekly. In another embodiment, the interferon-[3 is interferon beta-lb and is administered
subcutaneously at 0.25 mg, every other day. In another embodiment, the interferon-B is interferon
b and is stered subcutaneously at 0.25 mg, every other day. In yet another embodiment,
the eron-[3 is interferon beta-1a and is administered subcutaneously at 22-44 meg, three times a
week.
In one embodiment, the administration of laquinimod substantially precedes the administration of
interferon—B. In another embodiment, the administration of interferon—B substantially precedes the
administration of laquinimod.
In an embodiment, the human patient is receiving interferon-[3 therapy prior to initiating laquinimod
therapy. In another embodiment, the human t is receiving interferon-B therapy for at least 24
weeks prior to initiating laquinimod therapy. In another ment, the human patient is receiving
interferon—B therapy for about 24 weeks prior to initiating imod therapy. In r embodiment,
the human patient is receiving interferon-B therapy for at least 28 weeks prior to initiating laquinimod
therapy. In another embodiment, the human patient is ing interferon—[3 therapy for about 28 weeks
prior to initiating laquinimod therapy. In another embodiment, the human t is receiving
eron—B therapy for at least 48 weeks prior to initiating laquinimod therapy. In another
embodiment, the human patient is receiving interferon-B therapy for about 48 weeks prior to initiating
laquinimod therapy. In another embodiment, the human patient is receiving interferon-B therapy for at
least 52 weeks prior to initiating laquinimod therapy. In yet another ment, the human patient is
receiving interferon-B therapy for about 52 weeks prior to initiating laquinimod therapy.
PCT/U82012/048689
In one embodiment, the laquinimod is administered in the morning. In another embodiment, the
laquinimod is administered at night. In one embodiment, the laquinimod is with food. In another
embodiment, the laquinimod is administered without food.
In one embodiment, the eron-B is administered in the morning. In another embodiment, the
interferon—B is administered at night. In one embodiment, the interferon—B is administered with food. In
another embodiment, the interferon—B is administered without food.
In one embodiment, the laquinimod is stered simultaneously with the interferon-B. In another
embodiment, the imod is administered contemporaneously with the interferon—B. In another
embodiment, the laquinimod is administered immediately before or immediately after the interferon~{3.
In another embodiment, the laquinimod is administered within 1 hour before or after the interferon— B. In
another embodiment, the laquinimod is administered within 3 hour before or after the interferon~B. In
another embodiment, the laquinimod is stered within 6 hour before or after the eron—[3. In
another embodiment, the laquinimod is stered within 12 hour before or after the interferon-B. In
another embodiment, the laquinimod is administered within 24 hour before or after the interferon—B.
In one embodiment, the method further comprises administration of nonsteroidal anti-inflammatory
drugs (NSAIDS), salicylates, slow-acting drugs, gold compounds, hydroxychloroquine, sulfasalazine,
combinations of slow-acting drugs, corticosteroids, cytotoxic drugs, immunosuppressive drugs and/or
antibodies.
In an ment, the periodic administration of laquinimod and interferon-B ues for more than
30 days. In another embodiment, the ic stration of laquinimod and interferon—B continues
for more than 42 days. In yet another embodiment, the periodic administration of imod and
interferon-B continues for 6 months or more.
In one embodiment, the administration of laquinimod and interferon-[3 inhibits a symptom of relapsing
multiple sclerosis by at least 20%. In another embodiment, the administration of laquinimod and
interferon-B ts a symptom of relapsing multiple sclerosis by at least 30%. In another embodiment,
the administration of laquinimod and interferon-B inhibits a symptom of relapsing multiple sclerosis by
at least 40%. In another embodiment, the administration of laquinimod and eron-B ts a
symptom of relapsing multiple sclerosis by at least 50%. In another embodiment, the administration of
laquinimod and interferon-B inhibits a symptom of ing le sclerosis by more than 100%. In
another ment, the administration of laquinimod and interferon-B ts a symptom of relapsing
multiple sclerosis by more than 300%. In yet another embodiment, the administration of laquinimod
and interferon—B inhibits a symptom of relapsing multiple sclerosis by more than 1000%.
W0 16686 2012/048689
In one embodiment, each of the amount of laquinimod when taken alone, and the amount of interferon-
B when taken alone is effective to treat the human patient. In another embodiment, either the amount
of laquinimod when taken alone, the amount of interferon-B when taken alone, or each such amount
when taken alone is not effective to treat the human patient.
This invention also provides a method of treating a human patient afflicted with multiple sclerosis or
presenting a clinically isolated syndrome comprising periodically stering to the patient an
amount of laquinimod and an amount of interferon—B (IFN-B), wherein the amounts when taken together
are effective to treat the human patient. In one embodiment, the amount of laquinimod and the amount
of IFN—[i when taken together is more effective to treat the human patient than when each agent is
administered alone.
In one embodiment, the multiple sclerosis is relapsing multiple sclerosis. In another embodiment, the
relapsing multiple sclerosis is relapsing-remitting multiple sclerosis.
In one embodiment, the amount of laquinimod and the amount of interferon-B when taken together is
effective to reduce a symptom of multiple sclerosis in the human patient. In another embodiment, the
symptom is a MRI—monitored multiple sclerosis disease activity, relapse rate, accumulation of physical
disability, frequency of relapses, decreased time to confirmed disease progression, decreased time to
confirmed relapse, frequency of clinical exacerbation, brain atrophy, neuronal dysfunction, neuronal
injury, neuronal degeneration, neuronal sis, risk for confirmed progression, deterioration of
visual function, e, impaired mobility, cognitive impairment, reduction of brain volume,
2O abnormalities observed in whole Brain MTR histogram, deterioration in general health status,
functional status, quality of life, and/or symptom severity on work.
In one embodiment, the amount of imod and the amount of interferon—B when taken together is
effective to se or inhibit reduction of brain volume. In another embodiment, brain volume is
ed by percent brain volume change (PBVC).
In one ment the amount of laquinimod and the amount of eron—[3 when taken together is
effective to increase time to confirmed disease progression. In another embodiment, time to confirmed
disease progression is increased by 20—60%. In yet another embodiment, time to confirmed disease
progression is increased by at least 50%.
In one ment, the amount of laquinimod and the amount of interferon-B when taken together is
effective to decrease abnormalities observed in whole Brain MTR ram.
In one embodiment, the accumulation of physical lity is measured by Kurtzke Expanded Disability
Status Scale (EDSS) score. In another embodiment, the accumulation of physical disability is assessed
by the time to confirmed disease progression as ed by Kurtzke Expanded Disability Status Scale
(EDSS) score. In another embodiment, the patient had an EDSS score of 0—5.5 prior to administration
of laquinimod. In another embodiment, the patient had an EDSS score of 1.5—4.5 prior to
administration of laquinimod. In another embodiment, the patient had an EDSS score of 5.5 or r
prior to administration of laquinimod. In another embodiment, confirmed disease ssion is a 1
point increase of the EDSS score. In yet r embodiment, ed disease progression is a 0.5
point increase of the EDSS score.
In one ment, impaired mobility is assessed by the Timed—25 Foot Walk test. In r
embodiment, impaired mobility is assessed by the 12-Item Multiple Sclerosis Walking Scale (MSWS-
12) self—report questionnaire. In another embodiment, ed mobility is assessed by the Ambulation
Index (AI). In another embodiment, impaired mobility is assessed by the Six~Minute Walk (6MW)
Test. In yet another embodiment, impaired mobility is assessed by the Lower Extremity Manual
Muscle Test (LEMMT) Test.
In one embodiment, the amount of laquinimod and the amount of interferon-B when taken together is
effective to reduce ive impairment. In another embodiment, cognitive impairment is assessed by
the Symbol Digit Modalities Test (SDMT) score.
In one embodiment, general health status is assessed by the EuroQoL (EQSD) questionnaire, Subject
Global Impression (SGI) or Clinician Global Impression of Change (CGIC). In another embodiment,
functional status is measured by the patient’s Short—Form l Health survey (SF—36) Subject
Reported Questionnaire score. In another ment,quality of life is assessed by SF—36, EQSD,
Subject Global Impression (SGI) or Clinician Global Impression of Change (CGIC). In another
embodiment, the patient’s SF—36 mental component y score (MSC) is improved. In another
embodiment, the patient’s SF-36 physical component summary sore (PSC) is improved.
In one embodiment, fatigue is assessed by the EQSD, the patient’s Modified e Impact Scale
(MFIS) score or the French valid versions of the e Impact Scale (EMIF-SEP) score. In another
embodiment,symptom ty on work is ed by the work productivity and activities
impairment General Health (WPAI-GH) questionnaire.
In one embodiment, laquinimod is laquinimod sodium. In another embodiment, laquinimod is
administered via oral administration. In another embodiment, laquinimod is administered daily. In
another embodiment, laquinimod is administered more often than once daily. In another embodiment,
laquinimod is administered less often than once daily.
In one embodiment, the amount laquinimod administered is less than 0.6 . In another
embodiment, the amount laquinimod administered is 0.1-40.0 mg/day. In another embodiment, the
W0 20131’016686
amount laquinimod administered is 0.1—2.5 . In another embodiment, the amount imod
administered is 0.25-2.0 . In another embodiment, the amount imod administered is 0.5-1.2
mg/day. In another embodiment, the amount laquinimod administered is 0.25 mg/day. In another
embodiment, the amount laquinimod administered is 0.3 mg/day. In another embodiment, the amount
laquinimod administered is 0.5 y. In another embodiment, the amount imod administered is
0.6 mg/day. In another embodiment, the amount laquinimod administered is 1.0 mg/day. In another
embodiment, the amount laquinimod administered is 1.2 mg/day. In r embodiment, n the amount
laquinimod administered is 1.5 mg/day. In another embodiment, the amount laquinimod administered is
2.0 mg/day.
In one embodiment, a loading dose of an amount different form the intended dose is administered for a
period of time at the start of the periodic stration. In another ment, the g dose is
double the amount of the ed dose. In yet another embodiment, the g dose administered for
two days at the start of the periodic administration.
In one embodiment, the eron-B is administered via subcutaneous injection or intramuscular
injection. In another embodiment, the interferon-[3 is interferon beta-Ia and is administered
intramuscularly at 30 mcg, once weekly. In another embodiment, the interferon—B is interferon beta-lb
and is administered subcutaneously at 0.25 mg, every other day. In another embodiment, the interferon-
I3 is interferon beta-1b and is administered subcutaneously at 0.25 mg, every other day. In another
embodiment, the eron—B is interferon beta-1a and is administered subcutaneously at 22-44 mcg,
2O three times a week.
In one embodiment, the administration of laquinimod substantially precedes the administration of
interferon—B. In another embodiment, the administration of interferon—B ntially precedes the
administration of Iaquinirnod. In another embodiment, the human patient is receiving interferon-B
therapy prior to initiating imod therapy. In another embodiment, the human patient is receiving
interferon—B therapy for at least 24 weeks prior to initiating laquinimod therapy. In another embodiment,
the human patient is receiving interferon-B therapy for at least 28 weeks prior to initiating laquinimod
therapy. In another embodiment, the human patient is receiving interferon-B therapy for at least 48
weeks prior to ting laquinimod therapy. In another embodiment, the human patient is receiving
interferon-B therapy for at least 52 weeks prior to initiating laquinimod therapy.
In an ment, the method further comprises administration of nonsteroidal anti-inflammatory drugs
(NSAIDs), salicylates, slow-acting drugs, gold compounds, hydroxychloroquine, sulfasalazine,
combinations of slow-acting drugs, corticosteroids, cytotoxic drugs, immunosuppressive drugs and/or
antibodies.
PCT/U82012/048689
In one embodiment, the periodic administration of laquinimod and interferon—[3 continues for at least 3
days. In r ment, the ic administration of laquinimod and interferon—B ues for
more than 30 days. In another embodiment, the periodic administration of imod and interferon—B
continues for more than 42 days. In another embodiment, the periodic administration of laquinimod and
interferon-B continues for 8 weeks or more. In another ment, the periodic administration of
laquinimod and interferon-B continues for at least 12 weeks. In another embodiment, the periodic
administration of laquinimod and interferon—B continues for at least 24 weeks. In another embodiment,
the periodic administration of laquinimod and interferon-B continues for more than 24 weeks. In another
embodiment, the periodic administration of laquinimod and interferon—{3 continues for 6 months or more.
In an embodiment, the administration of laquinimod and interferon-B inhibits a symptom of relapsing
multiple sclerosis by at least 20%. In another embodiment, the stration of laquinimod and
interferon—B inhibits a symptom of ing le sclerosis by at least 30%. In r embodiment,
the administration of laquinimod and interferon-B ts a symptom of relapsing multiple sclerosis by at
least 50%. In another embodiment, the administration of laquinimod and interferon—B inhibits a m
of relapsing multiple. sclerosis by at least 70%. In another embodiment, the administration of laquinimod
and interferon—B inhibits a symptom of relapsing multiple sis by more than 100%. In another
embodiment, the administration of laquinimod and interferon-[i inhibits a m of relapsing multiple
sclerosis by more than 300%. In yet another embodiment, the administration of laquinimod and
interferon-[3 ts a symptom ofrelapsing multiple sclerosis by more than 1000%.
2O In one embodiment, each of the amount of laquinimod when taken alone, and the amount of interferon-
{3 when taken alone is effective to treat the human patient. In another embodiment, either the amount
of laquinimod when taken alone, the amount of interferon-B when taken alone, or each such amount
when taken alone is not effective to treat the human patient.
This invention also provides a package comprising a) a first pharmaceutical composition comprising
an amount of laquinimod and a pharmaceutically acceptable carrier; b) a second pharmaceutical
composition comprising an amount of interferon—[3 and a pharmaceutically acceptable carrier; and c)
instructions for use of the first and second ceutical compositions together to treat a human
patient afflicted with multiple sclerosis or presenting a ally isolated syndrome.
In one embodiment, the first pharmaceutical composition is in liquid form. In another embodiment,
the first pharmaceutical composition is in solid form. In another embodiment, the first pharmaceutical
composition is in capsule form. In r embodiment, the first pharmaceutical ition is in
tablet form. In another embodiment, the tablets are coated with a coating which inhibits oxygen from
contacting the core. In another embodiment, the coating comprises a cellulosic polymer, a detackifier,
a gloss enhancer, and pigment.
W0 2013f016686 PCTfU82012/048689
In one embodiment, the first ceutical composition further comprises mannitol. In another
embodiment, the first pharmaceutical composition further comprises an alkalinizing agent. In another
embodiment, the alkalinizing agent is ine. In another embodiment, the first pharmaceutical
composition further comprises an oxidation reducing agent.
In one embodiment, the first pharmaceutical composition is stable and free of an nizing agent or
an oxidation reducing agent. In another embodiment, the first pharmaceutical composition is free of
an alkalinizing agent and free of an oxidation reducing agent.
In one embodiment, the first ceutical composition is stable and free of disintegrant. In another
embodiment, the first pharmaceutical composition further comprises a ant. In another
embodiment, the lubricant is present in the composition as solid particles. In another embodiment, the
lubricant is sodium stearyl fumarate or magnesium stearate.
In one embodiment the first ceutical composition r comprises a filler. In another
embodiment, the filler is t in the composition as solid particles. In another embodiment, the
filler is lactose, lactose monohydrate, starch, isomalt, mannitol, sodium starch glycolate, sorbitol,
lactose spray dried, lactose anhydrouse, or a ation thereof. In another embodiment, the filler is
mannitol or lactose monohydrate.
In one embodiment, the package filrther ses a desiccant. In another embodiment, the desiccant
is silica gel.
In an embodiment, the first pharmaceutical composition is stable has a moisture content of no more
than 4%. In another embodiment, laquinimod is present in the composition as solid particles.
In an embodiment, the package is a sealed packaging having a moisture bility of not more than
mg/day per liter. In r embodiment, the sealed package is a blister pack in which the
maximum moisture permeability is no more than 0.005 mg/day. In another embodiment, the sealed
package is a bottle. In another embodiment, the bottle is closed with a heat induction liner. In another
embodiment, the sealed e comprises an HDPE bottle. In another embodiment, the sealed
package comprises an oxygen absorbing agent. In another embodiment, the oxygen absorbing agent is
iron.
In an embodiment, the amount of laquinimod in the first composition is less than 0.6 mg. In another
embodiment,the amount of laquinimod in the first ition is 01-400 mg. In another
embodiment,the amount of laquinimod in the first composition is 0.1-2.5 mg. In another embodiment,
the amount of laquinimod in the first composition is 025-20 mg. In another embodiment, the amount
of laquinimod in the first composition is 0.5 -1 .2 mg. In another ment,the amount of laquinimod
PCT/U$2012/048689
in the first composition is 0.25 mg. In another embodiment, the amount of laquinimod in the first
composition is 0.3 mg. In another embodiment, the amount of laquinimod in the first composition is 0.5
mg. In another ment, the amount of imod in the first composition is 0.6 mg. In another
embodiment,the amount of laquinimod in the first composition is 1.0 mg. In another embodiment, the
amount of laquinimod in the first composition is 1.2 mg. In another embodiment, the amount of
laquinimod in the first composition is 1.5 mg. In yet another embodiment, the amount of laquinimod in
the first composition is 2.0 mg.
This invention also es laquinimod for use as an add—on therapy or in combination with
interferon—[3 in treating a human patient afflicted with multiple sclerosis or ting a clinically
isolated syndrome.
This invention also provides a pharmaceutical ition comprising an amount of laquinimod and
an amount of interferon-B for use in treating a human patient afflicted with multiple sclerosis or
presenting a clinically isolated me, n the laquinimod and the interferon-B are
administered simultaneously or poraneously.
This invention also provides a pharmaceutical composition comprising an amount of laquinimod and
an amount of interferon—B.
In one ment, the pharmaceutical composition is in liquid form. In r embodiment, the
pharmaceutical composition is in solid form. In another embodiment, the pharmaceutical composition
is in capsule form. In another embodiment, the pharmaceutical composition is in tablet form. In
another embodiment, the tablets are coated with a coating which inhibits oxygen from contacting the
core. . In another embodiment, the coating ses a cellulosic polymer, a detackifier, a gloss
enhancer, and pigment.
In one embodiment, the pharmaceutical composition further comprises mannitol. In another
embodiment, the pharmaceutical composition further comprises an alkalinizing agent. In another
embodiment, the alkalinizing agent is meglumine. In yet another embodiment, the pharmaceutical
composition further comprises an oxidation ng agent.
In one embodiment, the pharmaceutical composition is free of an nizing agent or an oxidation
reducing agent. In another embodiment, the pharmaceutical composition is free of an alkalinizing
agent and free of an oxidation reducing agent.
In one embodiment, the pharmaceutical composition is stable and free of disintegrant. In another
embodiment, the pharmaceutical composition further comprises a ant. In r embodiment,
PCT/U82012/048689
the lubricant is present in the composition as solid particles. In r ment, the lubricant is
sodium stearyl fumarate or magnesium stearate.
In an embodiment, the pharmaceutical composition further sesa filler. In another embodiment,
the filler is present in the composition as solid les. In another embodiment, the filler is lactose,
e drate, starch, isomalt, mannitol, sodium starch glycolate, sorbitol, lactose spray dried,
lactose anhydrouse, or a combination thereof. In another embodiment, the filler is mannitol or lactose
monohydrate.
In an embodiment, the amount of laquinimod in the composition is less than 0.6 mg. In another
embodiment, the amount of laquinimod in the composition is 01-400 mg. In another embodiment, the
amount of laquinimod in the composition is 0.1-2.5 mg. In another ment, the amount of
laquinimod in the composition is .0 mg. In another embodiment, the amount of laquinimod in the
ition is 0.5—1.2 mg. In another embodiment, the amount of laquinimod in the composition is
0.25 mg. In another embodiment, the amount of laquinimod in the composition is 0.3 mg. In another
embodiment, the amount of laquinimod in the composition is 0.5 mg. In another embodiment, the
amount of imod in the composition is 0.6 mg. In another embodiment, the amount of laquinimod
in the composition is 1.0 mg. In another embodiment, the amount of laquinimod in the composition is
1.2 mg. In another embodiment, the amount of laquinimod in the composition is 1.5 mg. In yet another
embodiment, the amount of laquinimod in the composition is 2.0 mg.
This invention further provides use of an amount of laquinimod and an amount of interferon-B in the
preparation of a combination for treating a human patient afflicted with multiple sclerosis or
presenting a clinically isolated syndrome wherein the laquinimod and the interferon-B are
administered simultaneously or contemporaneously.
This invention also provides pharmaceutical composition comprising an amount of laquinimod for
use in treating a subject afflicted with multiple sclerosis or presenting a ally isolated syndrome
as an add—on y or in combination with interferon—B by ically administering the
pharmaceutical composition and the interferon—B to the subject.
This invention further provides pharmaceutical composition sing an amount of interferon-B for
use treating a subject afflicted with multiple sclerosis or presenting a clinically isolated syndrome as
an add-on therapy or in combination with laquinimod by periodically administering the
pharmaceutical composition and the laquinimod to the subject.
For the foregoing embodiments, each embodiment disclosed herein is plated as being
applicable to each of the other disclosed embodiments. In addition, the elements recited in the
W0 2013f016686 PCT/U82012/048689
packaging and pharmaceutical composition embodiments can be used in the method ments
described .
Laguinimod
Laquinimod mixtures, compositions, and the process for the manufacture thereof are described in,
e.g., U.S. Patent No. 6,077,851, U.S. Patent no. 7,884,208, U.S. Patent No. 7,989,473, U.S. Patent
No. 8,178,127, U.S. Application Publication No. 2010-0055072, U.S. Application Publication No.
2012—0010238, and U.S. Application Publication No. 2012-0010239, each of which is hereby
incorporated by reference in its entireties into this application.
Use of laquinimod for treatment of various conditions, and the corresponding dosages and regimens,
are described in U.S. Patent No. 851 (multiple sclerosis, n—dependent diabetes mellitus,
systemic lupus matosus, rheumatoid arthritis, inflammatory bowel disease, psoriasis,
inflammatory respiratory disorder, atherosclerosis, stroke, and Alzhemier’s disease), U.S. Application
Publication No. 2011-0027219 (Crohn’s disease), U.S. Application Publication No. 2010-0322900
(Relapsing-remitting multiple sclerosis), U.S. Application Publication No. 2011—0034508 (brain-
derived neurotrophic factor —related diseases), U.S. Application ation No. 2011-
0218179 (active lupus nephritis), U.S. Application Publication No. 2011-0218203 atoid
arthritis), U.S. Application Publication No. 2011-0217295 (active lupus arthritis), and U.S.
Application Publication No. 2012—0142730 (reducing fatigue, improving quality of life, and
providing neuroprotection in MS patients), each of which is hereby incorporated by reference in its
2O entireties into this application.
Commercially Available Interferon Beta (IFN-B)
Commercially available IFN—B include ®, Betaseron®, Extavia® and . The
recommended Avonex® dose for treating MS is 30 mcg injected into a muscle once weekly. The
recommended ron® dose for treating MS is 0.25 mg injected (subcutaneously) every other day.
The recommended Extavia® dose for treating MS is 0.25 mg, injected subcutaneously every other
day. The recommended dose of Rebif® for treating MS is 22 mcg or 44 meg, injected subcutaneously
three times a week.
A pharrnaceutically acceptable salt of laquinimod as used in this application includes m, sodium,
potassium, magnesium, calcium, manganese, , zinc, aluminum and iron. Salt formulations of
laquinimod and the process for preparing the same are described, e. g., in U.S. Patent No. 7,589,208
and PCT ational Application Publication No. , which are hereby incorporated
by reference into this application.
Laquinimod can be administered in admixture with suitable pharmaceutical diluents, extenders,
excipients, or carriers (collectively referred to herein as a pharmaceutically able carrier)
suitably ed with respect to the intended form of stration and as consistent with
conventional pharmaceutical practices. The unit will be in a form suitable for oral administration.
Laquinimod can be administered alone but is generally mixed with a aceutically acceptable
carrier, and co—administered in the form of a tablet or capsule, liposome, or as an agglomerated
powder. Examples of suitable solid carriers include lactose, sucrose, gelatin and agar. e or
tablets can be formulated and made easy to swallow or chew; other solid forms include granules, and
bulk powders.
Tablets may contain suitable binders, lubricants, disintegrating agents (disintegrants), coloring ,
flavoring agents, flow—inducing agents, and g agents. For instance, for oral administration in the
dosage unit form of a tablet or capsule, the active drug component can be combined with an oral, non-
toxic, pharmaceutically acceptable, inert carrier such as lactose, gelatin, agar, , sucrose, glucose,
methyl cellulose, dicalcium phosphate, calcium sulfate, mannitol, sorbitol, microcrystalline cellulose
and the like. Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose,
corn starch, natural and tic gums such as , tragacanth, or sodium te, povidone,
carboxymethylcellulose, polyethylene glycol, waxes, and the like. ants used in these dosage
forms include sodium oleate, sodium stearate, sodium benzoate, sodium acetate, sodium chloride,
stearic acid, sodium stearyl fumarate, talc and the like. Disintegrators (disintegrants) include, without
limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, croscarmellose sodium, sodium
starch glycolate and the like.
Specific es of the techniques, pharmaceutically acceptable carriers and excipients that may be
used to formulate oral dosage forms of the present invention are described, e.g., in US. Patent No.
7,589,208, PCT International Application Publication Nos. , , and
2007/146248.
General techniques and compositions for making dosage forms useful in the present invention are
described in the following references: Modern Pharmaceutics, rs 9 and 10 (Banker & Rhodes,
Editors, 1979); Pharmaceutical Dosage Forms: Tablets (Lieberman et al., 1981); Ansel, Introduction
to Pharmaceutical Dosage Forms 2nd Edition (1976); Remington's Pharmaceutical Sciences, 17th ed.
3O (Mack Publishing Company, Easton, Pa., 1985); Advances in Pharmaceutical Sciences (David
Ganderton, Trevor Jones, Eds, 1992); Advances in ceutical Sciences Vol 7. (David
Ganderton, Trevor Jones, James McGinity, Eds, 1995); Aqueous ric Coatings for
ceutical Dosage Forms (Drugs and the Pharmaceutical es, Series 36 (James McGinity,
Ed, 1989); Pharmaceutical Particulate Carriers: eutic Applications: Drugs and the
Pharmaceutical Sciences, Vol 61 (Alain Rolland, Ed, 1993); Drug Delivery to the Gastrointestinal
PCT/U82012/048689
Tract (Ellis Horwood Books in the Biological Sciences. Series in Pharmaceutical Technology; J. G.
Hardy, S. S. Davis, Clive G. Wilson, Eds).; Modern Pharmaceutics Drugs and the Pharmaceutical
Sciences, Vol. 40 (Gilbert S. Banker, Christopher T. Rhodes, Eds). These nces in their entireties
are hereby incorporated by nce into this application.
Disclosed is a method for ng a human patient afflicted with relapsing multiple sclerosis using
laquinimod with interferon-[3 which provides a more efficacious treatment than each agent alone. The
use of laquinimod for relapsing multiple sclerosis had been previously suggested in, e. g., US. Patent
No. 6,077,851. However, the inventors have surprisingly found that the combination of imod
and interferon-beta (IFN—B) is particularly effective for the treatment of relapsing multiple sclerosis as
compared to each agent alone.
Terms
As used herein, and unless stated otherwise, each of the following terms shall have the definition set
forth below.
As used herein, “laquinimod” means laquinimod acid or a ceutically acceptable salt thereof.
As used , an “amount” or “close” of imod as measured in milligrams refers to the
milligrams of laquinimod acid present in a ation, regardless of the form of the preparation. A
“dose of 0.6 mg laquinimod” means the amount of laquinimod acid in a preparation is 0.6 mg,
regardless of the form of the ation. Thus, when in the form of a salt, cg. a laquinimod sodium
salt, the weight of the salt form necessary to provide a dose of 0.6 mg laquinimod would be greater
than 0.6 mg (e.g., 0.64 mg) due to the presence of the additional salt ion.
As used herein, “about” in the context of a numerical value or range means i10% of the numerical
value or range recited or claimed.
As used herein, a composition that is “free” of a chemical entity means that the composition contains,
if at all, an amount of the chemical entity which cannot be avoided although the chemical entity is not
part of the ation and was not affirmatively added during any part of the manufacturing process.
For example, a composition which is “free” of an alkalizing agent means that the alkalizing agent, if
present at all, is a ty component of the composition by weight. Preferably, when a composition
is “free” of a ent, the composition comprises less than 0.1 wt%, 0.05 wt%, 0.02 wt%, or 0.01
wt% of the component.
As used herein, “alkalizing agent” is used interchangeably with the term “alkaline-reacting
component” or “alkaline agent” and refers to any pharmaceutically acceptable excipient which
neutralizes protons in, and raises the pH of, the pharmaceutical composition in which it is used.
PCT/U52012/048689
As used herein, “oxidation reducing agent” refers to a group of als which includes an
“antioxidant”, a “reduction agent” and a “chelating agent”.
As used herein, “antioxidant” refers to a compound selected from the group consisting of erol,
methionine, glutathione, tocotn'enol, dimethyl glycine, betaine, butylated hydroxyanisole, butylated
hydroxytoluene, turmerin, n E, ascorbyl ate, tocopherol, deteroxime mesylate, methyl
paraben, ethyl paraben, butylated hydroxyanisole, butylated hydroxytoluene, propyl gallate, sodium or
potassium metabisulfite, sodium or potassium sulfite, alpha tocopherol or derivatives thereof, sodium
ascorbate, disodium edentate, BHA (butylated yanisole), a pharmaceutically acceptable salt or
ester of the mentioned compounds, and mixtures thereof.
The term “antioxidant” as used herein also refers to Flavonoids such as those selected from the group of
quercetin, morin, naringenin and hesperetin, taxifolin, n, quercitrin, myricitrin, ein, apigenin
and biochanin A, flavone, ridol, isoflavonoids such as the soy isoflavonoid, genistein, catechins
such as the tea catechin epigallocatechin gallate, flavonol, epicatechin, hesperetin, chrysin, diosmin,
hesperidin, luteolin, and rutin.
As used herein, “reduction agent” refers to a compound selected from the group consisting of thiol—
containing compound, thioglycerol, mercaptoethanol, thioglycol, thiodiglycol, ne, thioglucose,
dithiothreitol (DTT), dithio-bis-maleimidoethane (DTME), 2,6-di-tert-butylmethylphenol (BHT),
sodium dithionite, sodium bisulphite, idine sodium sulphite, and ammonium bisulphite."
As used herein, “chelating agent” refers to a compound ed from the group consisting of
penicillamine, trientine, N,N'—diethyldithiocarbamate (DDC), 2,3,2’—tetraamine (2,3,2'-tet), neocuproine,
,N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN), l 1 0-phenanthroline
, (PHE),
tetraethylenepentamine, triethylenetetraamine and tris(2—carboxyethyl) phosphine , ferrioxamine,
CP94, EDTA, deferoxainine B (DFO) as the methanesulfonate salt (also known as desferrioxanilne B
mesylate (DFOM)), desferal from Novartis (previously Ciba-Giegy), and apoferritin.
As used herein, a pharmaceutical composition is “stable” when the composition preserves the physical
stability/integrity andfor chemical stability/integrity of the active pharmaceutical ingredient during
storage. Furthermore, “stable pharmaceutical composition” is characterized by its level of degradation
products not exceeding 5% at 40°C/75%RH after 6 months or 3% at 55°C/75% RH after two weeks,
compared to their level in time zero.
As used herein, “combination” means an lage of reagents for use in therapy either by
aneous or contemporaneous administration. Simultaneous administration refers to
administration of an admixture (Whether a true mixture, a suspension, an emulsion or other physical
combination) of the laquinimod and the lFN-B. In this case, the ation may be the admixture or
PCT/U82012/048689
separate containers of the laquinimod and the IFN—B that are combined just prior to administration.
Contemporaneous stration refers to the separate administration of the imod and the IFN—
B at the same time, or at times sufficiently close together that a synergistic activity relative to the
activity of either the laquinimod or the IFN-B alone is observed.
As used herein, “add—on” or “add—on therapy” means an assemblage of reagents for use in therapy,
wherein the subject receiving the therapy begins a first treatment n of one or more reagents
prior to beginning a second treatment regimen of one or more different reagents in addition to the first
treatment regimen, so that not all of the reagents used in the therapy are started at the same time. For
example, adding laquinimod therapy to a patient already receiving lFN—B therapy.
As used herein, “effective” when referring to an amount of laquinimod and/or interferon—beta (lFN-B)
refers to the quantity of laquinimod and/or interferon—beta (lFN—B) that is sufficient to yield a desired
therapeutic response t undue adverse side s (such as toxicity, irritation, or allergic
response) commensurate with a reasonable benefit/risk ratio when used in the manner of this
ion.
“Administering to the subject” or “administering to the (human) patient” means the giving of,
dispensing of, or application of medicines, drugs, or remedies to a subject/patient to relieve, cure, or
reduce the symptoms associated with a condition, e.g., a pathological condition.
“Treating” as used herein asses, e.g., inducing inhibition, regression, or stasis of a disease or
disorder, e.g., RMS, or ing, suppressing, inhibiting, reducing the severity of, eliminating or
substantially eliminating, or ameliorating a symptom of the disease or disorder. “Treating” as applied
to patients presenting CIS can mean delaying the onset of clinically definite multiple sclerosis
(CDMS), delaying the progression to CDMS, reducing the risk of conversion to CDMS, or reducing
the frequency of relapse in a patient who experienced a first clinical episode consistent with multiple
sclerosis and who has a high risk of developing CDMS.
“Inhibition” of disease progression or disease complication in a subject means ting or reducing
the e ssion and/or disease complication in the subject.
A “symptom” associated with RMS includes any clinical or laboratory manifestation associated with
RMS and is not limited to what the t can feel or observe.
As used herein, “a subject afflicted with ing multiple sclerosis” means a subject who has been
ally diagnosed to have relapsing multiple sclerosis (RMS) which includes ing-remitting
multiple sclerosis (RRMS) and ary Progressive multiple sclerosis (SPMS).
As used herein, a t at “baseline” is as subject prior to stration of laquinimod.
2012/048689
A “patient at risk of developing MS” (i.e. clinically definite MS) as used herein is a patient presenting
any of the known risk factors for MS. The known risk factors for MS include any one of a clinically
isolated syndrome (CIS), a single attack suggestive of MS without a lesion, the presence of a lesion
(in any of the CNS, PNS, or myelin sheath) without a clinical attack, nmental factors
(geographical location, climate, diet, toxins, sunlight), genetics (variation of genes encoding HLA-
DRBl, lpha and lLZR-alpha), and immunological ents (viral infection such as by
Epstein-Barr virus, high avidity CD4+ T cells, CD8+ T cells, anti-NF-L, anti-CSF 114(Glc)).
“Clinically ed syndrome (CIS)” as used herein refers to l) a single clinical attack (used
interchangeably herein with “first clinical event” and “first inating event”) suggestive of MS,
which, for example, presents as an episode of optic neuritis, blurring of vision, diplopia, ntary
rapid eye movement, blindness, loss of balance, tremors, ataxia, vertigo, clumsiness of a limb, lack of
co—ordination, weakness of one or more extremity, altered muscle tone, muscle stiffness, spasms,
tingling, paraesthesia, burning sensations, muscle pains, facial pain, trigeminal neuralgia, stabbing
sharp pains, burning tingling pain, slowing of , slurring of words, changes in rhythm of speech,
dysphagia, fatigue, bladder problems (including urgency, frequency, incomplete emptying and
incontinence), bowel problems (including constipation and loss of bowel l), impotence,
diminished sexual arousal, loss of sensation, ivity to heat, loss of short term memory, loss of
concentration, or loss of judgment or reasoning, and 2) at least one lesion suggestive of MS. In a
specific example, CIS diagnosis would be based on a single clinical attack and at least 2 lesions
suggestive ofMS measuring 6 mm or more in er.
“Relapse Rate” is the number of ed es per unit time. lized relapse rate” is the
mean value of the number of confirmed relapses of each patient multiplied by 365 and divided by the
number of days that patient is on the study drug.
“Expanded lity Status Scale” or “EDSS” is a rating system that is ntly used for
classifying and standardizing the condition of people with multiple sclerosis. The score ranges from
0.0 representing a normal neurological exam to 10.0 representing death due to MS. The score is based
upon neurological testing and examination of functional systems (FS), which are areas of the central
nervous system which l bodily functions. The functional systems are: Pyramidal (ability to
walk), Cerebellar (coordination), Brain stem (speech and swallowing), Sensory (touch and pain),
Bowel and bladder functions, , Mental, and Other (includes any other neurological findings due
to MS) (Kurtzke IF, 1983).
A “confirmed progression” ofEDS S, or “confirmed disease progression” as measured by EDSS score is
defined as a 1 point increase from baseline EDSS sustained for at least 3 months. In addition,
confirmation of progression cannot be made during a relapse.
WO 2013016686
“Adverse event” or “AE” means any rd medical occurrence in a clinical trial subject
administered a medicinal product and which does not have a causal relationship with the ent.
An adverse event can therefore be any unfavorable and unintended sign ing an abnormal
laboratory finding, symptom, or diseases temporally associated with the use of an investigational
medicinal product, whether or not considered related to the investigational medicinal product.
“Gd—enhancing lesion” refers to lesions that result from a breakdown of the brain barrier, which
appear in st studies using gandolinium contrast agents. Gandolinium enhancement provides
information as to the age of a , as Gd-enhancing lesions typically occur within a six week
period of lesion formation.
“Magnetization Transfer Imaging” or “MTI” is based on the magnetization interaction (through
dipolar and/or chemical exchange) between bulk water protons and macromolecular protons.
By applying an off resonance radio frequency pulse to the macromolecular protons, the
saturation of these protons is then transferred to the bulk water protons. The result is a se
in signal (the net magnetization of visible protons is reduced), depending on the magnitude of
MT between tissue macromolecules and bulk water. “MT” or “Magnetization Transfer” refers to
the transfer of longitudinal magnetization from the hydrogen nuclei of water that have restricted
motion to the en nuclei of water that moves with many degrees of m. With MTI, the
ce or absence of macromolecules (e.g. in membranes or brain tissue) can be seen (Mehta,
1996; Grossman, 1994).
“Magnetization Resonance Spectroscopy” or “MRS” is a specialized technique associated with
ic resonance imaging (MRI). MRS is used to measure the levels of different metabolites in
body tissues. The MR signal produces a spectrum of resonances that correspond to different
molecular arrangements of the isotope being “excited”. This signature is used to diagnose certain
metabolic disorders, especially those affecting the brain, (Rosen, 2007) as well as to provide
information on tumor metabolism (Golder, 2007).
As used herein “mobility” refers to any ability relating to walking, walking speed, gait, strength of leg
muscles, leg fianction and the ability to move with or without assistance. Mobility can be evaluated by
one or more of several tests including but not limited to tion Index, Time 25 foot walk, Six-
Minute Walk (6MW), Lower Extremity Manual Muscle Test ) and EDSS. Mobility can also
be reported by the subject, for e by questionnaires, including but not limited to 12—Item
Multiple Sclerosis Walking Scale (MSWS-12). Impaired Mobility refers to any ment, difficulty
or disability relating to mobility.
“Tl-weighted MRI image” refers to an MR—image that emphasizes T1 contrast by which lesions may
be ized. Abnormal areas in a Tl—weighted MRI image are “hypointense” and appear as dark
spots. These spots are generally older lesions.
“T2-weighted MR] image” refers to an MR-image that emphasizes T2 st by which lesions may
be ized. T2 lesions represent new inflammatory activity.
The “Six-Minute Walk (6MW) Test” is a commonly used test developed to assess exercise capacity
in patients with COPD (Guyatt, 1985). It has been used also to measure mobility in multiple sclerosis
patients (Clinical Trials Website).
The “Timed~25 Foot Walk” or “T25—FW” is a quantitative mobility and leg function performance test
based on a timed 25—walk. The patient is directed to one end of a clearly marked 25—foot course and is
instructed to walk 25 feet as quickly as possible, but safely. The time is calculated from the initiation
of the ction to start and ends when the patient has d the 25—foot mark. The task is
immediately administered again by having the patient walk back the same distance. Patients may use
assistive devices when doing this task. The score for the T25-FW is the e of the two completed
trials. This score can be used individually or used as part of the MSFC composite score (National MS
Society Website).
One of the central symptoms of multiple sclerosis is fatigue. Fatigue can be measured by several tests
including but not limited to decrease of French valid versions of the e Impact Scale (EMIF-SEP)
score, and European Quality of Life (EuroQoL) Questionnaire (EQSD). Other tests, including but not
limited to Clinician Global sion of Change (CGIC) and Subject Global sion (SGI), as
well as EQ-SD, can be used to evaluate the general health status and quality of life ofMS patients.
“Ambulation Index” or “Al” is a rating scale developed by Hauser et al. to assess mobility by
evaluating the time and degree of assistance required to walk 25 feet. Scores range from O
(asymptomatic and fully active) to 10 (bedridden). The patient is asked to walk a marked 25-foot
course as quickly and safely as possible. The examiner records the time and type of ance (e.g.,
cane, walker, crutches) needed. (Hauser, 1983)
“EQ—SD” is a standardized questionnaire instrument for use as a measure of health outcome
applicable to a range of health conditions and treatments. It provides a simple ptive profile and
a single index value for health status that can be used in the al and economic evaluation of
health care as well as tion health surveys. EQ—SD was developed by the “EuroQoL” Group
which comprises a network of ational, multilingual, multidisciplinary researchers, originally
from seven centers in England, Finland, the Netherlands, Norway and Sweden. The EQ-SD
questionnaire is in the public domain and can be ed from EuroQoL.
W0 20131016686 PCT/U52012/048689
“SF-36” is a multi-purpose, short-form health survey with 36 ons which yields an 8-scale
profile of onal health and well—being scores as well as psychometrically-based physical and
mental health summary es and a preference—based health utility index. It is a generic measure,
as opposed to one that targets a specific age, disease, or treatment group. The survey is developed by
and can be obtained from QualityMetric, Inc. of Providence, RI.
A “pharmaceutically acceptable carrier” refers to a carrier or excipient that is suitable for use with
humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic
response) commensurate with a reasonable /risk ratio. It can be a pharrnaceutically acceptable
t, suspending agent or vehicle, for delivering the instant compounds to the subject.
It is understood that where a parameter range is provided, all integers within that range, and tenths
thereof, are also ed by the invention. For e, “0.1—2.5mg/day” includes 0.1 mg/day, 0.2
mg/day, 0.3 mg/day, etc. up to 2.5 mg/day.
This invention will be better understood by reference to the Experimental Details which follow, but
those skilled in the art will readily appreciate that the specific experiments detailed are only
illustrative of the invention as described more fully in the claims which follow thereafter.
Experimental s
EXAMPLE 1: Assessment Of Add-on Effect Of Laguinimod In Mice Treated With Glatiramer
Acetate gGA) Or Interferon-beta (IFN—fiz
Mice were treated with a sub—optimal dose of imod (lOmg/kg) alone or add on glatiramer
acetate (12.5mg/kg) or IFN-B 00 IU/mouse). In both cases, the combined treatment ed in
improved efficacy when compared to each treatment alone.
EXAMPLE 2: Activity of interferon~|§ administered daily, subcutaneous (sc). alone or in
combination with laguinimod in chronic EAE in C57 BI mice
Experimental autoimmune alomyelitis (EAE) is an animal model y used with rodents) of
the human CNS demyelinating diseases, including MS. MOG induced EAE in the C57Bl strain of
mice was selected, as it is an established EAE model to test the efficacy of ate molecule for MS
treatement.
In this experiment interferon-[3 is administered daily, subcutaneous (so). alone or in combination with
laquinimod to chronic MOG induced EAE in C57 BI mice. Both were administered from the
beginning ofthe study in the MOG induced EAE in C57Bl mice.
General Design
Disease was induced in all mice by the ion of the encephalitogenic emulsion (MOG/CPA) and
intraperitoneal injection of Pertussis toxin on the first day and 48 hours later. IFN-B at dose levels of
50,000 and 5000,000 IU/mouse was administered by the subcutaneous route, once daily (QD).
Laquinimod at dose levels of 10 and 25 mg/mouse was administered by the oral route, once daily
(QD). Both IFN-B and laquinimod were administered prophylactic from disease induction — Day 1
until termination of the study. Two additional groups of IFN—B at dose level of 500,000 were treated
either prophylactic (Day 1—7) or from onset (Day 8—18) to study activity of IFN—B in prophylactic and
therapeutic .
Materials
eron beta—la ) (Rebif®, 44ug/0.5m1/syringe, equivalent to 1.2 x 107 units
(IU)/0.5ml/syringe), Laquinimod, PBS (Sigma), Pertussiis toxin (Sigma), MOG 35-55 (Mnf
Novatide), te Freund’s Adjuvant (CFA) (Signma), Saline (Mnf—DEMO SA).
Healthy, nulliparous, non-pregnant female mice of the C57BL/6 strain were used in the study. The
animals weighed 18—22 grams and were approximately 8 weeks old on receipt. The body weights of
the animals were recorded on the day of delivery. Overtly healthy animals were assigned to study
groups arbitrarily before treatment commenced.
Procedures
EAE was induced by ing the ecephalitogenic mixture (emulsion) consisting of MOG
(150.0ug/mouse) and CFA ning M. tuberculosis (1mg MG/mlCFA). A volume of 0.2 ml of
encephalitogenic emulsion was injected aneously into the flanks of each mouse (Dose = 0.15
mg MOG and 0.2 mg MT/mouse) Pertussis toxin in 0.2 m1 dosage volume is injected
intraperitonieally on the day of ion and 48 hours later (total amount is 0.2 ug/mouse; 100.0
ng/O.2ml/mouse).
The mice were allocated to the following treatment groups of 13 mice each.
Table 1: Experimental design
Group f Treatment dose/day Administration Regimen
Groups Route
r (treatment
‘initiation
L-AQUINIMQD 10 rug/kg 7:
-LAQUTNIMOD 25mg/kg Oral, QD
: LAQUTNIMOD from Day 1
-IFN-B ‘
500000
SQQD to 30 daily & IFN—B from
, , , ,, ,, . Day 1 to 18 daily
LAQUINIMOD ‘
+ Oral (QD) +
IFNB sc (QD)
LAQUINIMOD + Oral (QD) + :
IFN[3 sc (QD) :
,,, ~
, SC,QD
The mice were administered with the various concentrations of IFN—B (2.5 X106 and 2.5 X 105 IU/ml)
at volume dose level of 200pl/mouse by subcutaneous route equivalent to 50,000 and 500,000
IU/mouse respectively.
The laquinimod ation was administered from Day 1, once daily (QD). Four hours interval was
maintained between administration of laquinimod and lFN—B.
Experimental Observations
All animals were ed once daily to detect if any are moribund. Mice were also d once
weekly. Further, the mice were observed daily from the 8th day post—EAE induction and EAE clinical
signs were scored. The scores were recorded on observation cards ing to the grades described
in Table 2 below.
PCT/U82012/048689
Table 2: Evaluation of the EAE clinical signs
___-_
3 Hind leg wobbly walk - when the mouse walks the hind legs are unsteady
Hind leg paralysis The mouse drags its hind legs but is able to move around using its
fore les
The mouse can't move around, it looks thinner and emaciated.
Full paralysis
All mice with score 1 and above were considered sick. When the first clinical sign appears all mice
were given food soaked in water, which was spread on different places on the bedding of the cages.
For calculation es, the score of s that were sacrificed or died (6) was carried forward.
Interpretation ofResults
Calculation of the incidence of disease gDisease ratio)
. The number of sick animals in each group were summed.
o The incidence of disease was calculated as
N . k . .
. t t d
l 0 INCIDENCE of DISEASE =[W ]
No. of sick mice in control group
. The percent inhibition according to incidence was calculated as
“mber of “Ck ””06 m d group ]
INHIBITION (%)of INCIDENCE = £1——————————————-———-—-—-————N x100
Number of Sick mice in control group
W0 2013l016686 PCT/U52012/048689
ation of the mortalig/moribundig rate (mortalig ratio)
. The number of dead or nd s in each group were summed.
. The mortality of disease was calculated as
dead or morzbound mzcezn treated group ]
MORTALITY of DISEASE = [No.of
No.0f dead or moribound mice in control group
- The percent inhibition according to mortality was calculated as
Number of dead or mortbound treated group ]
INHIBITION (%) of MORTALITY __ (1 x100
Number of dead or moribound mice in control group
Calculation of duration of disease
. The mean duration ofdisease expressed in days was calculated as
)3 Duration 0f disease 0f each mouse
Mean Duration = (
No.0f mice in th e group
1 0 Calculation of mean delay in onset of disease
. The mean onset of disease expressed in days was calculated as
Onset of disease of each mouse]
Mean Onset = [E
No.0f mice in th e group
. The mean delay in onset of disease expressed in days was calculated by subtracting the
mean onset of disease in control group from test group.
1 5 Calculation of the mean maximal score and percent inhibition
. The mean maximal score (MMS) of each group was calculated as
Score of ea ch mouse)
[MMS _ (EMaximal
N0. of mice in th e group
. The percent tion ing to MMS was calculated as
Mu z~ r d
INHIBITION (%) of MMS =(PM) x 100
MMS of control group
Calculation of the group mean score and percent inhibition
. The daily scores of each mouse in the test group were summed and the individual mean
daily score (IMS) was ated as
2 Body score of mouse
[MS =[
Observation period (days)J
. The mean group score (GMS) was calculated as
EIMS of ea chmouse
GMS =[
No. of mice in th e group ]
. The percent inhibition was calculated as
M t d
INHIBITION (%) of GMS = l—W x100
GMS of control group
Results/Discussion
A summary of the incidence, mortality, mean maximal scores (MMS), group mean score (GMS),
duration of the disease, onset of the disease and the activity of each group compared to the e
treated control group is shown in Table 3 below.
WO 16686 PCT/U82012/048689
Table 3: Mortality, incidence, MMS, GMS, Duration and Onset and EAE inhibition compared to
Vehicle
InnMortalityIncidence % MMS % GMS % Mean Onset Mean
1 2 I .
Negative 0/13 13/13 3.2 j; .0 10.5 i 18.1 1’.
Control 1.0 + 1.3 4.63
Vehicle 10 1.0
Laquinimod 0/13 8/13 38.5 % 1.8 i , 3.8 % 0.8 60.0 % 21.3 _+_ 9.2 i
mg/kg 1.7 p= 0.04 + p = 0.003 8.2 8.3
I i "
I a "
Laquiniomd 0/15 5/15o1.5 % 75.0 % 0.3 85.0%p=<0.001 25.7 i 7.8 5.0 35
mg/kg 0.8 i p + 0.5 p = < 7.4
I II I M a -
IFN-B 0/13 11/13 15.4 % 2.6 i . 1.7 . 15.4 i
50,000 1.4 +
. p = 0.3 7.8
lU/mouse
lFN-B 0/13 12/13 7.7 %
500,000
IU/mouse
Laquinimod + 0/13 113
IFN-B
mg/kg +
50,000
IU/mouse
Laquinimod + 0/15 5/15
IFN-B
mg/kg +
500,000
lU/mouse
lFN-B (Day 1 0/13 13/13
to 7)
500,000
IU/mouse
IFN-B (Day8 0/13 10/13
to 30)
500,000
IU/mouse
WO 16686 PCT/U82012/048689
The activity of the IFN-B administered groups in combination with laquinimod (10 mg/kg) compared
to the group d with laquinimod (10 mg/kg) is shown in Table 4 below.
Table 4: Laquinimod alone and in combination with IFN-B compared to laquinimod (10 mg/kg).
Mortalit Incidenc % Mean
Treatme y inhibitio inhibitio 3 Duratio
nt n2 n
(daYS)
Laquinmod 0113 .
mg/kg .OH‘Q
Laquinmod 0/15
mg/kg 151+?
Laquinmod m3
+ IFN-B
mg/kg + .0”?
50,000
IU/mouse
Laquinmod #15
+ lFN-B
mg/kg +
500,000
The activity as compared to vehicle and laquinimod are shown in Tables 5 and 6 below:
Table 5: Activity compared to vehicle
—-_-:I
m—_—
-_—_
Table 6: Activity compared to Laquinimod
—GrouMean Score GMS
Vehicle (saline —_
La uinimod 10m - __
La uinimod 25m -
IFN beta 50,000 IU/mouse)
IFN beta 500,0001U/m0use__
Table 7: ty compared to interferon-B (500,000 TU/mouse)
upMean Score . .
IFN beta 500,000 IU/mouse) _-
Lauinimod 25 m-/k 0 3 66.7%
IFN beta 50,000 IU/mouse) + La uinimod l0 m k ) ——0.5
IFN beta (500,000 IU/mouse + Lauinimod (10 m k )
IFN beta 00 IU/mouse _
Under the conditions of the test IFN-B at dose levels of 50,000 IU/mouse and 500,000 IU/mouse
exhibited additive activity in the suppression of EAE when tested in combination with laquinimod at
dose level of 10 mg/kg.
The groups d with IFN—l3 at dose levels of 50,000 IU/mouse and 500,000 lU/mouse in
combination with laquinimod (10 mg/kg) exhibited 75% and 90% activity respectively according to
GMS compared to 15%, 55% and 60% activity in groups treated with IFN—[5 at dose levels of 50,000
IU/mouse and 500,000 IU/mouse and laquinimod at dose level of 10 mg/kg respectively when
compared to the vehicle administered control group.
The groups treated with IFN~B at dose levels of 50,000 se and 500,000 IU/mouse in
combination with laquinimod (10 mg/kg) exhibited 37.5% and 75% ty respectively according to
GMS when compared to group treated with laquinimod at dose level of 10 mg/kg.
It is ant to note the mouse dosing ted here cannot be used to determine human dosing by
simply adjusting for body weight, because a gram ofmouse tissue is not equivalent to a gram of human
tissue. For this reason, the National Institutes of Health (NIH) es a table of Equivalent Surface
Area Dosage Conversion Factors below (Table 8) which provides conversion factors that account for
surface area to weight ratios between species.
Table 8: lent Surface Area Dosage Conversion Factors
.Mouse 20 g Rat 150g Monkey 3kg Dog 8kg Man 60kg
FROM 1/4 1/6 1/12
1 3/5
Dog --4 1 12
12/3
——-7 3
EXAMPLE 3: Clinical Trial Phase II — Assessment Of Add—on Effect Of La uinimod In Rela sin
Multi le Sclerosis RMS Sub‘ects d With Glatiramer Acetate GA Or Interferon-beta IFN—
A multinational, multicenter, randomized, double—blind, el—group, placebo—controlled study,
followed by a double—blind active extension phase is conducted to assess the safety, tolerability and
efficacy of two daily doses of oral laquinimod (0.6mg or 1.2mg) in adjunct to glatiramer acetate (GA)
or interferon-beta (IFN- B)-la/ lb preparations in subjects with relapsing multiple sclerosis (RMS).
Study Duration
The total study duration for each le subject will be up to 19 months:
0 Screening phase: up to about 1 month.
0 Double-Blind Placebo Controlled (DBPC) ent phase: about 9 months of once—daily oral
administration of laquinimod 0.6 , day or placebo in addition to current therapy
(i.e., subcutaneous GA 20mg or any of the following lFN—fi preparations: ®
Avonex
Betaseron®/Betaferon®, Rebif® or Extavia®).
o Double-Blind Active Extension (DBAE) phase: all subjects who complete all 9 months of the
DBPC treatment phase are offered the opportunity to continue to a DBAE phase. During this
phase, all subjects continue the same background injectable treatment which they used in the
DBPC phase.
0 Subjects who were originally assigned to either of the active oral treatment arms nimod
0.6 mg or 1.2 mg) continue with their original oral treatment assignment. Subjects originally
PCT/U82012/048689
assigned to placebo are equally randomized to either laquinimod 0.6mg or 1.2mg. The
duration of this phase is 9 months.
Study tion
Relapsing Multiple Sclerosis (RMS).
Study Design
Eligible subjects are equally (121:1) randomized into one of the following treatment arms:
1. GA 20 mg or any IFN—B preparation + oral daily administration of laquinimod capsules 0.6
2. GA 20 mg or any IFN-B preparation + oral daily administration of laquinimod capsules 1.2
mg.
3. GA 20 mg or any IFN-B preparation + oral daily o .
The 0.6 mg laquinimod capsule can be manufactured according to the method disclosed in PCT
International Application Publication No. 7/ 146248, published December 21, 2007 (see, page
, line 5 to page 11, line 3).
Randomization is stratified in a way that in each arm the number of subjects treated by GA will be
equal to the number of subjects treated by IFN-B preparations (Avonex®, ron®/Betaferon®,
Rebit® or Extavia®).
During the DBAE phase, subjects ue the same background injectable treatment which they used
in the DBPC phase. ts who were originally ed to either of the active oral arms [either
laquinimod 0.6mg (arm I) or l.2mg (arm 2)] continue with their original oral treatment assignment.
Subjects originally assigned to placebo (arm 3) are equally ized to either laquinimod 0.6mg or
1.2mg.
During the DBPC phase, subjects are evaluated at study sites for ll scheduled visits at Months: —1
(screening), 0 (baseline) and every month thereafter until Month 9 (termination! early termination).
During the DBAE phase subjects are evaluated at study sites for 6 scheduled visits at months 9
[Baseline EXT; the termination visit of the DBPC phase], 10/ lAE, 11/2AE, 12/3AE, 15/4AE and
18/5AE (termination/ early termination visit of the DBAE phase).
The following assessments are med at the specified time points:
WO 2013016686 2012/048689
During both DBPC and DBAE phases, vital signs are measured at each study visit.
During the DBPC phase, a physical examination is performed at Month —1 (Screening) and
Months 0 (Baseline), 1, 3, 6 and 9 (Termination/Early Termination visit of the DBPC phase).
During the DBAE phase, a physical examination is performed at Month 9 (Baseline EXT;
termination visit of the DBPC , lO/lAE, 12/3AE and 18/5AE (Termination/Early
Termination visit of the DBAE .
The following safety clinical laboratory tests are performed:
a. Complete blood count (CBC) with differential — at all scheduled visits in both DBPC
and DBAE phases.
b. Serum chemistry (including electrolytes, liver enzymes, creatinine, direct and total
bilirubin and pancreatic amylase), and urinalysis is performed at all led visits
in both DBPC and DBAE phases. Lipase is tested in case of abnormal pancreatic
amylase results. Glomerular ion rate (GFR) is calculated at Month -1
(Screening) and prior to each MRI scan.
c. Lipid profile (total cholesterol, HDL, LDL and triglycerides) is performed at month ~
1 (Screening) or Month 0 (Baseline) of the DBPC phase, under fasting conditions.
d. During the DBPC phase, d on tests (TSH, T3 and free T4) are performed
at Months 0 (Baseline), 6 and 9 (Termination/ Early termination visit of the DBPC
phase). During the DBAE phase d function tests (TSH, T3 and free T4) are
performed at Months 9 (Baseline EXT; termination visit of the DBPC phase),
/4AE and 18/5AE (termination/ early termination visit of the DBAE phase).
e. Urinalysis is performed at the Screening visit.
f. Serum B—hCG (human choriogonadotropin beta) is performed in women of child-
bearing potential at each scheduled study visit in both DBPC and DBAE phases.
Urine dipstick B—hCG in women of child—bearing potential during both the DBPC and the
DBAE phases, at all post-Screening study visits and the early termination visit. In addition,
during the DBAE Phase, urine B-hCG test is performed at home twice between scheduled
visits:
a. At months 13AE and 14AE (30i4 days and 60i4 after Month 12AE visit,
respectively).
W0 2013.1016686 PCTfU82012/048689
b. At months 16AE and 17AE (30:84 days and 60i4 after Month 15AE visit,
respectively).
The subject is contacted by the site staff via telephone within 72 hours after the test is
led to be performed and asked specific questions regarding the test. In case of
suspected pregnancy (positive urine B—hCG test result), the caller instructs the subject to stop
taking the study drug and to arrive to the site as soon as possible (but within 10 days) with all
study drugs.
During the DBPC phase, electrocardiograms (ECG) is performed at months —1 (screening), 0
ine; three recordings 10 min apart, before first dose), 1, 2, 3, 6 and 9 (termination/early
termination visit of the DBPC phase). During the DBAE phase, ECGs are med at
Months 9 (Baseline EXT; ation visit of the DBPC phase), 10/lAE, ll/2AE, 12/3AE,
/4AE and 18/5AE (Termination/ Early Termination visit of the DBAE phase).
Chest X—ray is performed at month —1 (screening), if not performed within 6 months prior to
the screening visit.
Adverse Events (AEs) are monitored hout the study.
Concomitant Medications are monitored throughout the study (both phases).
During the DBPC phase, neurological evaluations, ing Expanded Disability Status
Scale (EDSS), Ambulation Index (Al) and onal system score (F8) are performed at
Months: —1 (screening), 0 (baseline), 3, 6, and 9 (Termination/Early Termination of the
DBPC phase). During the DBAE phase, neurological evaluations, including EDSS, Al and
FS scores are performed at Months 9 (Baseline; ation visit of the DBPC phase),
12/3AE, 15/4AE and lS/SAE (Termination/Early Termination of the DBAE phase).
. During the DBPC phase, Symbol Digit Modalities Test (SDMT) are performed at Months 0
(Baseline), 6 and 9 (Termination/Early Termination visit of the DBPC phase). During the
DBAE phase, SDMT is performed at Months 9 (Baseline EXT; ation visit of the
DBPC phase), 15/4AE and lS/SAE nation/Early Termination visit of the DBAE
phase).
11. During the DBPC phase, each subject undergoes 3 MRI scans at Months: 0 (baseline), 3 and
9 (Termination/Early Termination Visit of the DBPC phase). During the DBAE phase, each
3O subject undergoes 2 MRI scans at Months 9 (Baseline EXT; Termination visit scan of the
DBPC phase) and 18/5AE (Termination/Early Termination Visit of the DBAE phase).
PCT/U82012/048689
12. During the DBPC phase, Pharmacokinetic (PK) study: Blood samples for analysis of
laquinimod plasma concentrations are ted from all subjects at Months I, 3 and 6.
13. During the DBPC phase, whole blood samples are collected for lymphocyte
immunophenotyping at Months 0 (Baseline), 3 and 9 (Termination/Early Termination).
14. Health Economics and Quality of Life: During the DBPC phase, the Work tivity and
Activities Impairment Questionnaire-General Health GH) (US sites only) and the
European Quality of Life (EuroQoL) Questionnaire (EQSD) are filled out at months 0
(Baseline), and 9 (Termination/Early Termination). During the DBAE phase, the WPAI-GH
(US sites only) and the EQSD questionnaire are filled out at Months 9 (Baseline EXT;
Termination Visit of the DBPC phase) and l8/5AE (termination/early termination visit of the
DBAE phase).
. Relapses are confirmed/monitored throughout the study (both phases).
Relapse Treatment
The allowed ent for a e is intravenous Methylprednisolone lgr/day for up to 5
consecutive days.
Monitoring
The subjects are closely monitored through the study course by an external independent Data
Monitoring Committee (DMC).
MRI Activity Alert Criteria
2O In case 5 or more GdE—Tl lesions are demonstrated on an MRI scan, the MRI reading center issues a
notification letter to the Sponsor, investigator and the DMC. MRI parameters of activity are not
considered stopping rules and the decision regarding individual subject‘s participation in the trial is at
the tion of the treating physician.
Ancillag studies:
Pharmacogenetic (PGx) assessment: Blood samples for PGx ters are collected from all
subjects that signed the informed consent form (separate from that of the core study), pending Ethics
Committees al, during the DBPC phase, ably at Month 0 (Baseline) or any other visit
following Month 0.
Number ects
Approximately 600 subjects.
Inclusion/Exclusion Criteria
Inclusion Criteria
1. ts must have a documented MS diagnosis as defined by the Revised McDonald criteria
[Ann Neurol 201 l: 69:292—302], with a relapsing disease course.
Subjects must be relapse free, in a stable neurological condition and free of corticosteroid
treatment [intravenous (IV), intramuscular (1M) and/or oral] 60 days prior to randomization.
Subjects must be treated with GA (Copaxone®) or an IFN—B preparation (Avonex®,
Betaseron®/Betaferon®, Rebit® or Extavia®), at a stable dose for at least 6 months prior to
ization (switching between IFN—B preparations during the 6 months prior to
randomization is allowed; switching between any lFN—B preparation and GA, or vice versa, is
exclusionary), and there is no plan to change the subject’s injectable treatment (either
Copaxone® or IFN-B preparation) during the course of the study.
Subjects must have an EDSS score of 1.5-4.5 (inclusive) at randomization.
Subjects must be between 18 and 55 years of age, inclusive.
Women of child—bearing potential must practice an acceptable method of birth control.
Acceptable methods of birth l in this study include: surgical sterilization, intrauterine
devices, oral contraceptive, contraceptive patch, long-acting injectable contraceptive,
2O partner’s omy or double—barrier method (condom or diaphragm with spermicide).
Subjects must be able to sign and date a written informed t prior to ng the study.
Subjects must be g and able to comply with the protocol requirements for the duration
of the study.
Exclusion Criteria
Have a non-relapsing, progressive form of MS (e.g., PPMS) (as defined by Lublin and
Reingold, 1996).
An onset of a e, unstable neurological condition or any treatment with corticosteroids
[intravenous (iv), intramuscular (im) and/or per os (po)] or corticotropic hormone 6O
PCT/U52012/048689
days prior to randomization (last day of steroid treatment should be equal or greater than 60
days prior to ization).
3. Use of mental or investigational drugs, and/or participation in drug clinical studies
within the 6 months prior to randomization.
4. Use ofimmunosuppressive agents within 6 months prior to randomization.
. Use of zumab (Tysabri®), fingolimod (Gilenya®) or anti—B cell therapy within the 2
years prior to ization.
6. Previous use of any of the following: cytotoxic agents, Mitoxantrone (Novantrone®),
cladribine, laquinimod, total body irradiation, total lymphoid irradiation, stem cell treatment,
autologous bone marrow transplantation or allogenic bone marrow transplantation.
7. Previous treatment with intravenous immunoglobulin (IVIG) or plasmapheresis within 2
months prior to ization.
8. Use of moderate/strong inhibitors of CYP3A4 within 2 weeks prior to the randomization
9. Use of inducers of CYP3A4 within 2 weeks prior to randomization.
10. Pregnancy or breastfeeding.
ll. A ZZXULN serum elevation of either alanine transaminase (ALT) or ate minase
(AST) at screening.
12. Serum direct bilirubin which is EZXULN at ing
13. Subjects with a potentially clinically significant or unstable medical or surgical condition that
2O would preclude safe and complete study participation, as determined by medical history,
physical examinations, ECG, laboratory tests or chest X—ray. Such conditions may include:
a. A cardi0vascular or pulmonary disorder that cannot be well—controlled by standard
treatment permitted by the study protocol.
b. Renal es.
c. Any form of acute or chronic liver disease.
d. Known human immunodeficiency virus (HIV) positive status.
WO 2013016686 PCT/U52012/048689
e. A y of drug and/or alcohol abuse.
f. An unstable psychiatric disorder.
g. Any malignancies, excluding basal cell carcinoma (BCC), in the last 5 years.
14. A glomerular filtration rate (GFR) less than 60 ml/min at screening visit.
. A known history of sensitivity to gadolinium (Gd).
16. Inability to successfully undergo MRI seaming.
17. Previous endovascular ent for Chronic Cerebrospinal Venous Insufficiency (CCSVl).
l8. Known drug hypersensitivity that would preclude administration of laquinimod, such as
hypersensitivity to: mannitol, meglumine or sodium stearyl fumarate.
Route and Dosage Form
1. GA 20 mg or an preparation of interferon-beta (lFN—B) + oral daily administration of
laquinimod capsules 0.6 mg (one laquinimod capsule 0.6 mg and one placebo capsule for
laquinimod) (applicable to both DBPC and 'DBAE phases).
2. GA 20 mg/lmL or an IFN-B preparation + oral daily administration of imod 1.2 mg (2
capsules of laquinimod 0.6 mg) (applicable to both DBPC and DBAE phases)
3. GA 20 mg or an preparation of lFN-B + oral daily administration of placebo (2 placebo
capsules for laquinimod) (applicable only to DBPC phase).
Outcome Measures
The y objectives of the study are to assess the , tolerability and efficacy of two daily
2O doses of oral laquinimod (0.6mg or 1.2mg) in adjunct to GA or IFN-B preparation (Avonex®,
ron®/Betaferon®, Rebif® or Extavia®) in subjects with RMS.
Primam Efficacy Endpoint for DBPC Phase:
0 The percent brain volume change (PBVC) between month 0 ine) to Month 9
(Termination/Early Termination after Month 6 of the DBPC phase).
WO 16686 PCT/U82012/048689
Key Exploratog Efficacy Endpoints for DBPC phase:
Change in whole brain Magnetic Transfer Ratio (MTR) histogram between month 0
(Baseline) and Month 9 (Termination/Early Termination visit after Month 6 of the DBPC
phase).
Time to Confirmed Disease Progression (CDP). CDP is defined as a sustained se in
EDSS of 21 point from Baseline for at least 3 months. Progression cannot be confirmed
during a relapse.
Exploratory nts for DBPC Phase
The percent change in cortical thickness between month 0 (baseline) and month 9
(termination/early termination visit after month 6).
The cumulative number new Tl hypointense lesions at months 3 and 9 (termination/early
termination visit after month 6).
The number of active (new T2 or GdE-Tl) lesions at month 3 that evolved into black holes at
month 9 (termination/early ation visit after month 6).
The cumulative number of GdE—Tl lesions at months 3 and 9 (termination/early termination
visit after month 6).
Change in T2 lesion volume from O (baseline) to month 9 (termination/early termination visit
after month 6).
Change in GdE—Tl lesions volume from month 0 ine) to month 9 (termination/early
2O termination visit after month 6).
Change from baseline to month 9 (termination/early termination visit after month 6) in
SDMT score.
The general health status, as assessed by the EuroQoL (EQSD) questionnaire.
Assessment of the effect of general health and symptom severity on work, using the work
productivity and activities impairment l Health (WPAl-GH) questionnaire.
Annualized Relapse Rate (ARR).
The time to the first confirmed relapse.
WO 2013016686 PCT/U82012/048689
- Pharmacokinetics of laquinimod.
Exploratory Endpoints for DBAE Phase
A similar set of endpoints are analyzed for the DBAE phase.
Safety and Tolerabilig Endpoints for DPBC phase
0 The cumulative number of GdE—Tl lesions at months 3 and 9.
0 The cumulative number of Combined Unique Active (CUA) lesions at months 3 and 9.
0 Number of subjects with adverse events.
0 Number of subjects with potentially clinically significant abnormalities based on laboratory
tests and vital signs and ECGs during the study.
0 Proportion of subjects (%) who urely discontinue from the study, reason of
discontinuation and the time to withdrawal.
0 Proportion of subjects (%) who urely tinue from the study due to adverse events
(ABS) and the time to withdrawal.
Results/Discussion
This study assesses safety, bility and efficacy of laquinimod in adjunct to glatiramer acetate
(GA) or interferon—beta (IFN—B) in relapsing multiple sclerosis (RMS) subjects. Since the mechanisms
of action of laquinimod and IFN-B have not been fully elucidated, the effect of the combined y
cannot be predicted and must be evaluated mentally.
Daily administration of laquinimod (p.o., 0.6 mg/day and 1.2 mg/day) as an add-on therapy for a
patient already receiving interferon-beta (IFN—B) provides increased efficacy (provides an additive
effect or more than an additive effect) in relapsing multiple sclerosis (RMS) subjects without unduly
increasing adverse side effects or affecting the safety of the treatment. Daily administration of
laquinimod (p.o., 0.6 mg/day and 1.2 mg/day) as an add-on therapy to IFN-B is also safe for use in
treating relapsing le sis (RMS) patients.
Administration of laquinimod (p.o., 0.6 mg/day and 1.2 mg/day) as an add-on y to IFN—B
provides a clinically meaningful advantage and is more effective (provides an additive effect or more
than an additive ) in treating relapsing multiple sclerosis (RMS) ts than when lFN—B is
administered alone (at the same dose) in the following manner:
W0 2013IOI6686
1. The add-on therapy is more effective (provides an additive effect or more than an additive
effect) in ng the decrease in brain volume (determined by the percent brain volume
change (PBVC)), in relapsing multiple sclerosis (RMS) patients.
The add-on therapy is more effective (provides an additive effect or more than an additive
effect) in increasing the time to confirmed e progression (CDP), in relapsing multiple
sclerosis (RMS) ts, where CDP is defined as a sustained increase in EDSS of 21 point
from Baseline for at least 3 . Progression cannot be confirmed during a relapse.
The add-on therapy is more ive (provides an additive effect or more than an additive
effect) in reducing abnormalities observed in whole Brain MTR histogram, in relapsing
multiple sclerosis (RMS) ts during.
The add-on therapy is more ive (provides an additive effect or more than an additive
effect) in ng the number of confirmed relapses and therefore the relapse rate, in
relapsing multiple sclerosis (RMS) patients.
The add—on therapy is also more effective (provides an additive effect or more than an
additive effect) in reducing the accumulation of physical disability in relapsing multiple
sclerosis (RMS) patients, as measured by the time to confirmed progression of EDSS.
The add-on therapy is more ive (provides an additive effect or more than an additive
effect) in reducing nitored disease activity in relapsing multiple sclerosis (RMS)
patients, as measured by the cumulative number of T1 Gd-enhancing lesions on Tl-weighted
images, the cumulative number new Tl hypointense lesions, the cumulative number ofnew T2
lesions, the cumulative number of new Tl hypointense s on Tl-weight images (black
holes), the number of active (new T2 or ) lesions, presence or absence of GdE lesions,
change in total volume of Tl Gd-enhancing lesions, change in total volume of T2 lesions,
and/or cortical thickness.
The add—on therapy is more ive (provides an additive effect or more than an additive
effect) in reducing brain atrophy in relapsing le sclerosis (RMS) patients.
The add—on therapy is more effective (provides an additive effect or more than an additive
effect) in reducing the frequency of relapses, the frequency of clinical exacerbation, and the
risk for confirmed progression in relapsing multiple sis (RMS) patients.
WO 16686 PCT/U52012/048689
9. The add-on therapy is more effective (provides an additive effect or more than an ve
effect) in increasing the time to confirmed relapse in relapsing multiple sclerosis (RMS)
patients.
. The add-on therapy is more ive (provides an additive effect or more than an additive
effect) in improving the l health status (as assessed by the EuroQoL (EQSD)
questionnaire), symptom severity on work (as assessed by the work productivity and
activities impairment General Health (WPAI-GH) questionnaire) and quality of life, in
relapsing multiple sis (RMS) patients.
11. The add-on therapy is more effective (provides an additive effect or more than an additive
effect) in decreasing cerebral dysfunction/cognitive impairment (as assessed by Symbol Digit
Modalities Test (SDMT)), in relapsing multiple sclerosis (RMS) patients during the double
blind study .
Administration of laquinimod (p.o., 0.6 mg/day and 1.2 mg/day) as an add—on therapy to IFN—B
provides a clinically meaningful advantage and is more effective (provides an additive effect or more
than an additive effect) in delaying the conversion to clinically definite MS in patients presenting a
C18 tive ofMS than when IFN—B is administered alone (at the same dose).
Administration of laquinimod (p.o., 0.6 mg/day and 1.2 mg/day) as an add-on therapy to IFN-B
provides a clinically meaningful advantage and is more effective (provides an additive effect or more
than an additive effect) in reducing the rate of development of clinically definite MS, the occurrence
of new MRI—detected lesions in the brain, the accumulation of lesion area in the brain and brain
atrophy in persons at high risk for developing MS, and is more effective in reducing the ence of
clinically definite MS and preventing rsible brain damage in these persons than when IFN—B is
administered alone (at the same dose).
Based on the foregoing, analogous s are expected for therapy using laquinimod (p.o., 0.6
mg/day and 1.2 ) in combination with IFN—B. Specifically, daily administration of laquinimod
(p.o., 0.6 mg/day and 1.2 mg/day) in combination with IFN-B provides increased efficacy (provides
an additive effect or more than an ve effect) over the administration of each agent alone in
relapsing multiple sis (RMS) subjects without unduly increasing adverse side effects or
affecting the safety of the ent. Daily administration of imod (p.o., 0.6 mg/day) in
ation with interferon-beta (IFN-B) is also safe for use in treating relapsing multiple sclerosis
(RMS) patients.
Administration of laquinimod (p.o., 0.6 mg/day and 1.2 mg/day) in combination with IFN—B provides
a clinically meaningful advantage and is more effective (provides an additive effect or more than an
PCT/U52012/048689
additive effect) in treating relapsing multiple sclerosis (RMS) patients than when IFN—B is administered
alone (at the same dose) in the following manner:
12. The combination therapy is more effective des an additive effect or more than an
additive effect) in reducing the se in brain volume (determined by the percent brain
volume change (PBVC)), in relapsing multiple sclerosis (RMS) patients.
l3. The combination therapy is more effective (provides an additive effect or more than an
additive effect) in increasing the time to confirmed disease progression (CDP), in relapsing
multiple sclerosis (RMS) patients, where CDP is defined as a sustained increase in EDSS of
21 point from Baseline for at least 3 months. Progression cannot be ed during a
relapse.
14. The ation therapy is more effective (provides an additive effect or more than an
additive effect) in reducing abnormalities observed in whole Brain MTR histogram, in
relapsing multiple sclerosis (RMS) ts during.
. The combination therapy is more effective (provides an additive effect or more than an
ve effect) in reducing the number of confirmed relapses and therefore the relapse rate, in
relapsing multiple sclerosis (RMS) patients.
l6. The combination therapy is also more effective (provides an additive effect or more than an
additive effect) in reducing the accumulation of physical disability in relapsing multiple
sis (RMS) patients, as measured by the time to confirmed progression .
2O l7. The combination therapy is more effective (provides an additive effect or more than an
additive ) in reducing MRI-monitored disease activity in relapsing multiple sclerosis
(RMS) patients, as measured by the cumulative number of T1 (Ed—enhancing lesions on T1-
weighted images, the cumulative number new Tl hypointense lesions, the cumulative number
of new T2 lesions, the cumulative number ofnew Tl tense lesions on Tl-weight images
(black holes), the number of active (new T2 or GdE—Tl) s, presence or e of GdE
lesions, change in total volume of Tl Gd-enhancing lesions, change in total volume of T2
lesions, and/or cortical thickness.
18. The combination y is more effective (provides an additive effect or more than an
additive effect) in reducing brain atrophy in relapsing multiple sclerosis (RMS) patients.
W0 ‘016686 PCT/U82012/048689
19. The combination therapy is more effective (provides an additive effect or more than an
additive effect) in reducing the frequency of relapses, the frequency of clinical exacerbation,
and the risk for confirmed progression in relapsing multiple sclerosis (RMS) patients.
. The combination therapy is more effective (provides an additive effect or more than an
additive effect) in increasing the time to confirmed relapse in relapsing multiple sclerosis
(RMS) patients.
21. The combination therapy is more effective (provides an ve effect or more than an
additive effect) in improving the l health status (as assessed by the EuroQoL (EQSD)
questionnaire), symptom severity on work (as assessed by the work productivity and
activities impairment General Health (WPAI—GH) questionnaire) and quality of life, in
relapsing multiple sclerosis (RMS) patients.
22. The ation therapy is more effective (provides an additive effect or more than an
additive effect) in decreasing cerebral dysfunction/cognitive ment (as assessed by
Symbol Digit Modalities Test (SDMT)), in relapsing multiple sclerosis (RMS) patients during
the double blind study period.
Administration of laquinimod (p.o., 0.6 mg/day and 1.2 mg/day) in combination with IFN-B provides
a clinically meaningful age and is more effective (provides an additive effect or more than an
additive effect) in delaying the conversion to clinically definite MS in patients presenting a C18
suggestive ofMS than when IFN—B is administered alone (at the same dose).
Administration of laquinimod (p.0., 0.6 mg/day and 1.2 mg/day) in ation with IFN—B es
a ally meaningful advantage and is more effective (provides an additive effect or more than an
additive effect) in ng the rate of development of clinically definite MS, the occurrence of new
tected lesions in the brain, the accumulation of lesion area in the brain and brain atrophy in
persons at high risk for developing MS, and is more effective in reducing the occurrence of clinically
definite MS and preventing irreversible brain damage in these persons than when IFN—B is
administered alone (at the same dose).
W0 201314116686
References
“COPAXONE®” in Physician’s Desk Reference, Thompson Reuters — Physician’s Desk
Reference Inc, Montvale, NJ, 2008, 3110-3113.
Barkhof, F. (1999) “IVERI in Multiple Sclerosis: Correlation with Expanded Disability Status Scale
(EDSS)”, Multiple Sclerosis. 5(4):283-286 (Abstract).
Bjartmar C, Fox RI. (2002) “Pathological mechanisms and disease progression of multiple
sclerosis: therapeutic ation”, Drugs ofToday. 3827-29.
Brex et al., (2002) “A longitudinal study of alities on MRI and disability from
multiple sis”, N Engl J Med. Jan 17, 2002 346(3): 158-64.
Brod et al. (2000) Annals of Neurology, 47: 127-13 1.
Brfick (2011) “Insight into the mechanism of laquinimod action.” J Neurol Sci. 2011 Jul
;306(1-2):173—9.
Brunmark et al., (2002) “The new orally active immunoregulator imod (AER-215062)
effectively inhibits pment and relapses of mental autoimmune encephalomyelitis”,
I Neuroimmunology. 130: 1 63-1 72.
Chabot and Yong, eron—lb increases IL—10 in a model of T cell - microglia interaction:
Relevance to MS, Neurol. 2000, 55: 1497-1505.
Chabot et al., Cytokine production in T lymphocyte-microglia interaction is attenuated by
glatiramer acetate: A mechanism for therapeutic efficacy in multiple sclerosis, Mull. Scien, in
press.
. Comi et a1. (2007) LAQ/5062 Study Group. “The Effect of Two Doses of Laquinimod on
MRI—Monitored Disease ty in Patients with Relapsing—Remitting Multiple Sclerosis: A
Multi-Center, Randomized, Double—Blind, Placebo-Controlled Study”, Presented at: 59th
Annual g of the American y of Neurology; April 28—May 5, 2007; Boston,
MA.
ll. Compston, Genetic susceptibility to multiple sclerosis, in McAlpine’s Mutiple Sclerosis,
Matthews, B. ed., London: ill Livingstone, 1991, 301-319.
12. Conway and Cohen (2010) “Combination therapy in multiple sclerosis”, LancetNeurol,
9:299—308.
PCT/U82012/048689
13. Costello et a1. (2007) “Combination therapies for multiple sclerosis: scientific rationale,
clinical trails, and clinical practice, Current Opinion in Neurology, 20:281—285.
14. Dal Canto et a1. (1977) Multiple sclerosis. Animal model: Theiler’s virus infection in mice.
Am. J. Pat/2. 88:497-500.
. De Stefano et a1. (1999) “Evidence of early axonal damage in patients with multiple sclerosis”,
Neurology. 52(Supp12):A378.
I6. Dunitz, M. Multiple sclerosis therapeutics, Ed. Rudick and Goodkin. London: Taylor &
Francis, 1999.
17. Durelli et al. and the Independent ison of Interferon (INCOMIN) Trial Study Group.
(2002) “Every—other-day interferon beta-1b versus once—weekly interferon beta-la for multiple
sclerosis: results of a 2-year ctive randomised multicentre study (INCOMIN)”, .
359:1453-60.
18. EMEA Guideline on al Investigation of Medicinal Products for the Treatment of Multiple
Sclerosis EWP/561/98 Rev. 1, Nove.2006).
19. EPAR, Rebif®, Scientific Discussion.
. Filippi et al., Glatiramer acetate s the proportion of MS lesions evolving into black
holes, Neurol, 2001, 57:731—733.
21. Fischer et al., (1999) “The Multiple Sclerosis onal Composite measure (MSFC): an
integrated approach to MS al outcome assessment”, Multiple Sclerosis, 5(4):244—250.
22. Fisk et al., (1994) “Measuring the Functional Impact of Fatigue: Initial tion of Fatigue
Impact Scale”, Clin Inf Dis. 18 Suppl 1:S79-83.
23. Fisk et al., (1994) “The Impact of Fatigue on Patients with Multiple Sclerosis”, Can J Neurol
Sci. 2129-14.
24. Frenandez (2007) “Combination therapy in multiple sclerosis”, Journal of the neuroloolg'cal
sciences, —103.
. Frohman et al., (2003) “The utility of MRI in suspected MS: report of the Therapeutics and
Technology Assessment Subcommittee of the American Academy ofNeurology”, Neurology.
Sep 9,2003, 61(5):602—1l.
PCT/U52012/048689
26. Gold (2008) “Combination therapies in multiple sis”, J Neurol, 255 [Suppl l]:51—60.
27. Golder W, (2007) “Magnetic resonance spectroscopy in clinical oncology”, Onkologie. 27(3):
304—9.
28. Grossman et al., (1994) Magnetization transfer: theory and clinical ations in
neuroradiology”, RadioGraphics. 14:279—290.
29. Guidance for Industry. In vivo drug lism/drug interaction studies — study , data
analysis, and recommendations for closing and labeling, US Dept. Health and Human Svcs.,
FDA, Ctr. for Drug Eval. and Res, Ctr. for Biologics Eval. and Res, Clin. / Pharm, Nov.
1999 <http://www.fda.gov/Cber/gdlns/metabol.pdf>.
30. Gurevich et a1. (2010) “Laquinimod suppress antigen presentation in relapsing—remitting
multiple sclerosis: in-vitro high—throughput gene expression study” (J Neuroimmunol. 2010
Apr 15;221(1—2):87—94. Epub 2010 Mar 27.
31. Guyatt et a1. (1985) “The 6—minute walk: a new measure of exercise capacity in patients with
chronic heart failure”, Can Med Assoc J 1322919—823.
32. Hafler and Weiner, MS: A CNS and ic autoimmune disease, Immunol. Todav, 1989,
:104~107.
33. Hartung et al. (2005) “Significance ofneutralizing antibodies to interferon beta during treatment
of multiple sclerosis: expert opinions based on the Proceedings of an lntemational Consensus
Conference”, Eur J Neurol. 12:588—601.
2O 34. Hauser et al. (1983) “Intensive immunosuppression in ssive multiple sclerosis”, New
Engl JMed. 308:173—180.
. Hla T, Lee MJ, Ancellin N, Paik JH, Kluk MJ . "Lysophospholipids—receptor
revelations”. e 294 (5548): .
36. Hohlfeld et al. (2000) “The neuroprotective effect of ation: implications for the therapy of
multiple sclerosis”, J Neuroimmunol. 1072161-166.
37. Johnson et al., Copolymer 1 reduces e rate and improves disability in relapsingremitting
multiple sclerosis: results of a phase III multicenter, double-blind placebo-
controlled trial. The Copolymer 1 Multiple Sclerosis Study Group, Neurol., 1995, 45: 1268.
38. Kleinschmidt-DeMasters et al. (2005) New England Journal of Medicine, 353:369—379.
PCT/U82012/048689
39. Kurtzke JF, (1983) “Rating neurologic impairment in multiple sclerosis: an expanded
lity status scale (EDSS)”, Neurology :l444-l452.
40. Lampert, Autoimmune and virus-induced inating diseases. A review, Am. J. Path,
1978, 91:176-208.
41. Langer—Gould et al. (2005) New England Journal of Medicine, 353:369—379.
42. Lublin FD, Reingold SC (1996) “Defining the clinical course of multiple sclerosis”, .
46:907-911.
43. Martyn, The epidemiology of multiple sclerosis, in McAlpine’s Multiple Sclerosis, Matthews,
B., ed., : Churchil Livingstone, 1991, 3—40.
44. McDonald et al., Recommended diagnostic criteria for multiple sclerosis: guidelines from the
International Panel on the diagnosis of multiple sis. Ann. Neurol. 2001, 50:121—127.
45. Mehta et al. (1996) “Magnetization transfer magnetic resonance imaging: a clinical review”,
Topics in Magetic Resonance Imaging 8(4):214-30.
46. Miki et al. (1999) “Relapsing-Remitting Multiple Sclerosis: Longitudinal Analysis of MR
Images — Lack of Correlation between Changes in T2 Lesion Volume and Clinical gs”,
ogy. 213:395-399.
47. Milo and Panitch (2011) “Combination therapy in multiple sis”, Journal of
Neuroimmunology, 231(201 l):23-3 l.
48. Moraal et al. (2008) “Subtraction MR Images in a Multiple Sclerosis Multicenter Clinical Trial
Setting”, Radiology, 250(2):506-514.
49. le sclerosis: its diagnosis, ms, types and stages, 2003
<http://www.albany.net/~tjc/multiple—sclerosis.html>.
50. s et al. (2003) “Immunomodulation in multiple sclerosis: from immunosuppression to
neuroprotection”, Trends Pharmacol Sci. 24:131—138.
51. Noseworthy et al. (2000) “Multiple sis”, N Engl J Med. 343:938-952.
52. Olsson, Immunology of multiple sclerosis, Curr. Opin. Neurol. Neurosurg, 1992, 5:195-202.
53. Panitch et al. for the EVIDENCE (Evidence of Interferon Dose—response: European North
American Comparative Efficacy) Study Group and the University of British Columbia
PCT/U82012/048689
MS/MRI Research Group. (2002) “Randomized comparative study of interferon B-Ia treatment
regiments in MS”, The EVIDENCE Trial. Neurology. 59: 1496-1506.
54. Parkman, Graft-versus-host Disease, Ann. Rev. Med, 1991, 42: 189-197.
55. PCT International Application Publication No. , published April 26, 2007,
international filing date October 18, 2006.
56. PCT International Application Publication No. , published December 21,
2007, international filing date June 12, 2007.
57. Polman et al. (2011) “Diagnostic Criteria for Multiple sis: 2010 Revisions to the
McDonald Criteria”, Ann N eural, 69:292-302.
58. Polman et al., (2005) “Diagnostic criteria for le sclerosis: 2005 revisions to the
McDonald Criteria”, Annals of Neurology, Volume 58 Issue 6, Pages 840~846.
59. Polman et al., (2005) ment with laquinimod s development of active MRI lesions
in relapsing MS”, Neurology. 64:987-991.
60. Poser et al. (1983) “New Diagnostic Criteria for Multiple Sclerosis: Guidelines for Research
Protocols”, Annals of ogy, March 1983, 13(3):227-230.
61. Rodriguez et al. (1987) Theiler’s murine encephalomyelitis: a model of demyelination and
persistence of virus. Crit. Rev. l. 7:325.
62. Rosen Y, (2007) “The Recent advances in magnetic resonance neurospectroscopy”,
Neurotherapeutics. 27(3):330-45.
63. Rudick et al. (2006) New England Journal of Medicine, 354:91 1-923.
64. , R. (1999) “Disease—Modifying Drugs for ing-Remitting Multiple Sclerosis and
Future Directions for Multiple Sclerosis Therapeutics”, Neurotherpatueics. 56:1079—1084.
65. Runstrom et a1. (2002) “Laquinimod (AER-215062) a candidate drug for treatment of
Multiple Sclerosis inhibits the development of experimental autoimmune encephalomyelitis
in IFN-B knock—out mice”, (Abstract), Medicon Valley Academy, Malmoe, Sweden.
66. Runstrom et a1. (2006) “Inhibition of the pment of chronic experimental autoimmune
encephalomyelitis by laquinimod 15062) in IFN-B k.o. and wild type mice” l of
Neuroimmunology, 173(2006):69—78.
2012/048689
67. rg-Wollheim et a1. (2005) “48-week open safety study with high-dose oral laquinimod
in patients”, Mult Scler. 11:8154 (Abstract).
68. Sorenson PS. (2006) “Neutralising antibodies to interferon-B — measurement, clinical relevance,
and management”, J Neurol. 253 [Suppl 6]:VI/16-VI/22.
69. Teitelbaum et al., Suppression of Experimental Allergic Encephalomyelitis by a Synthetic
Polypeptide, Eur. J. l. 1971, 1: 8.
70. Teitelbaum et a1., Suppression of Experimental Allergic Encephalomyelitis with Basic
Polymers, Eur. J. Immunol, 1973, 3: 273-279.
71. The National MS Society (USA), The Disease 'ng Drug Brochure, October 19, 2006.
72. U.S. Patent ation Publication No. 2010—0322900, published December 23, 2010
(Tarcic et al.).
73. U.S. Patent Application ation No. 2011—0027219, published February 3, 2011 (Tarcic
et al.).
74. U.S. Patent Application Publication No. 2011-0034508, published February 10, 2011 (Liat
Hayardeny).
75. U.S. Patent Application Publication No. 2011—0217295, published September 8, 2011 (Haviv
and Tarcic).
76. U.S. Patent Application Publication No. 2011-0218179, published September 8, 2011 (Haviv
and Tarcic).
77. U.S. Patent Application Publication No. 2011—0218203, hed September 8, 2011 (Joel
Kaye et al.).
78. U.S. Patent Application Publication No. 2012-0010239, published January 12, 2012
(Piryatinsky et al.).
79. U.S. Patent Application Publication No. 2012-0010238, published January 12, 2012
(Fristedt).
80. U.S. Patent Application Publication No. 142730, published June 7, 2012 (Tarcic et
al.).
81. Vollmer et al. (2008) “Glatiramer acetate after induction therapy with ntrone in relapsing
multiple sis” Multiple Sclerosis, 00: 18
82. Yang et al., (2004) “Laquinimod (AER-215062) suppresses the development of experimental
mune alomyelitis, tes the Th1/Th2 balance and s the Th3 cytokine
TGF-B in Lewis rats”, J. Neuroimmunol. 15623-9.
83. Yong (2002) “Differential mechanisms of action of interferon-{3 and glatiramer acetate in MS”
Neurology, 5921—7.
84. Zou et al. (2002) “Suppression of experimental autoimmune neuritis by AER—215062 is
associated with altered Th1/Th2 e and inhibited migration of inflammatory cells into the
peripheral nerve tissue”, Neuropharmacology. 42:731.
85. Clinical Trials Website, article entitled “Study of Fampridine—ER Tablets in Patients With
Multiple Sclerosis“, retrieved July 10, 2012,
<hgtp ://c linicaltrials. gov/et2/show?term=fampridine&cond=multiple+sclerosis&phase=2&ran
ki7>-
86. “COPAXONE®” in Physician’s Desk Reference, Thompson Reuters — Physician’s Desk
Reference Inc., Montvale, NJ, 2008, 3110-3113.
87. U.S. Patent No. 3,849,550, issued November 19, 1974 (Teitelbaum et a1).
88. U.S. Patent No. 5,800,808, issued September 1, 1998 (Konfino et al).
89. U.S. Patent No. 964, issued January 12, 1999 (Aharoni et a1).
2O 90. U.S. Patent No. 5,981,589, issued November 9, 1999 (Konfino et a1).
91. U.S. Patent No. 6,048,898, issued April 11, 2000 (Konfino et a1).
92. U.S. Patent No. 6,054,430, issued April 25, 2000 (Konfino et al).
93. US. Patent No. 851, issued Jun 20, 2000 (Bjork et al).
94. U.S. Patent No. 6,214,791, issued April 10, 2001 (Arnon et al).
95. U.S. Patent No. 6,342,476, issued January 29, 2002 (Konfino, et al).
96. U.S. Patent No. 6,362,161, issued March 26, 2002 (Konfino et al).
97. Alejandro Horga; Xavier Montalban 06/04/2008; Expert Rev Neurother. 2008;8(5):699-714.
98. US. Patent No. 208, issued September 15, 2009 (Jansson et al).
99. US. Patent no. 7,884,208, issued February 8, 2011 (Frenkel et al.).
100. RTT News Article dated April 12, 11, entitled “Teva Pharma, Active Biotech Post ve
Laquinimod Phase 3 ALLEGRO Results”.
101. Teva Press Release dated August 1, 2011, ed “Results of Phase III BRAVO Trial
Reinforce Unique Profile of Laquinimod for Multiple Sclerosis Treatment”.
102. US. Patent No. 7,989,473, issued August 2, 2011 (Patashnik et al.).
103. US. Patent No. 8,178,127, issued May 15, 2012 i et a1.).
104. National MS Society Website, retrieved July 10, 2012 <http://www.nationalrnssociety.org/for—
professionals/researchers/olinical—study—measures/t25~
105. Salama et al. (2003) Multiple Sclerosis, 9228—3 1.
Claims (53)
1. Use of 0.6mg laquinimod in the cture of a medicament for the treatment of a human patient afflicted with multiple sclerosis or ting a clinically isolated syndrome, wherein the medicament is prepared as an add-on therapy to or in combination with an amount of interferon- 8, n the amounts when taken together is more effective to treat the human patient than when each agent at the same amount is administered alone.
2. Use of 0.6mg laquinimod and an amount of interferon-6 in the manufacture of a medicament for the treatment of a human patient afflicted with multiple sclerosis or presenting a clinically isolated me, wherein the amounts when taken together is more effective to treat the human patient than when each agent at the same amount is administered alone.
3. Use of an amount of laquinimod in the manufacture of a medicament for the treatment of a human patient afflicted with multiple sclerosis or presenting a clinically isolated syndrome, wherein the medicament is prepared as an add-on therapy to or in combination with an amount of interferon-6 (IFN-B), wherein the s when taken together are more effective to treat the human patient than when each agent at the same amount is administered alone.
4. Use of an amount of laquinimod and an amount of interferon-6 in the cture of a medicament for the ent of a human t afflicted with multiple sclerosis or presenting a clinically isolated syndrome, n the amounts when taken together are more effective to treat the human patient than when each agent at the same amount is administered alone.
5. The use of any one of claims 1—4, wherein the laquinimod is laquinimod sodium.
6. The use of any one of claims 1—5, n the multiple sclerosis is relapsing multiple sclerosis.
7. The use of any one of claims 1—6, wherein the multiple sclerosis is ing—remitting multiple sclerosis.
8. The use of any one of claims 1-7, wherein the amount of laquinimod and the amount of interferon-B when taken together is effective to reduce a symptom of multiple sclerosis in the human patient.
9. The use of claim 8, wherein the symptom is a MRI-monitored multiple sclerosis disease activity, relapse rate, accumulation of al lity, frequency of relapses, decreased time to confirmed disease progression, decreased time to confirmed relapse, frequency of clinical exacerbation, brain y, neuronal dysfunction, neuronal injury, neuronal degeneration, neuronal apoptosis, risk for confirmed progression, deterioration of visual on, fatigue, impaired mobility, cognitive impairment, reduction of brain , abnormalities observed in whole Brain MTR histogram, deterioration in general health status, functional status, quality of life, and/or symptom severity on work.
10. The use of claim 9, n the amount of laquinimod and the amount of interferon-B when taken together is effective to decrease or inhibit reduction ofbrain .
11. The use of claim 10, wherein brain volume is measured by percent brain volume change (PBVC).
12. The use of claim 9, wherein the amount of laquinimod and the amount of eron-B when taken together is effective to increase time to confirmed disease ssion.
13. The use of claim 12, n time to confirm disease progression is increased by 20-60%, or by at least 50%.
14. The use of claim 9, wherein the amount of laquinimod and the amount of interferon—B when taken together is effective to decrease abnormalities observed in whole brain MTR histogram.
15. The use of claim 9, wherein the accumulation of physical disability is measured by Kurtzke Expanded Disability Status Scale (EDSS) score, and /or wherein the accumulation of physical disability is assessed by the time to confirmed disease progression as measured by Kurtzke Expanded Disability Status Scale (EDSS) score.
16. The use of claim 15, n the patient had an EDSS score of 0-5 .5, an EDSS score of 1.5- 4.5, or an EDSS score of 5.5 or greater prior to administration of laquinimod and/or confirmed disease progression is a 1 point increase of the EDSS score or 0.5 point increase of the EDSS score.
17. The use of claim 9, wherein impaired mobility is assessed by the 25 Foot Walk test, the 12-Item Multiple Sclerosis Walking Scale (MSWS—lZ) self-report questionnaire, the Ambulation Index (AI), the Six—Minute Walk (6MW) Test, or the Lower Extremity Manual Muscle Test ) Test.
l8. The use of claim 9, wherein the amount of laquinimod and the amount of interferon-B when taken together is effective to reduce cognitive impairment.
l9. The use of claim 18, wherein cognitive impairment is assessed by the Symbol Digit Modalities Test (SDMT) score.
20. The use of claim 9, wherein general health status is assessed by the EuroQol (EQSD) onnaire, Subject Global Impression (SGI), or Clinician Global Impression of Change (CGIC).
21. The use of claim 9, wherein functional status is measured by the patient’s Short-Form General Health survey (SF—36) Subject Reported Questionnaire score.
22. The use of claim 9, wherein quality of life is assessed by SF-36, EQSD, Subject Global Impression (SGI), or ian Global Impression of Change (CGIC).
23. The use of claims 21 or 22, wherein the patient’s SF—36 mental component summary score (MSC) is improved and/or wherein the patient’s SF-36 physical component summary sore (PSC) is improved.
24. The use of claim 9, wherein e is ed by the EQSD, the patient’s d Fatigue Impact Scale (MFIS) score or the French valid version of the Fatigue Impact Scale (EMlF-SEP) SCOI‘C .
25. The use of claim 9, wherein symptom severity on work is measured by the work productivity and activities impairment General Health (WPAI-GH) questionnaire.
26. The use of any one of claims 1-25, wherein the medicament is prepared for oral administration.
27. The use of any one of claims 1—26, wherein the medicament is prepared for administration daily, more often than once daily, or less often than once daily.
28. The use of any one of claims 3-27 wherein the ment comprises a unit dose of laquinimod of less than 0.6 mg.
29. The use of any one of claims 3—27, wherein the medicament comprises a unit dose of laquinimod of 0. 1 -40.0 mg.
30. The use of claim 29, wherein the medicament comprises a unit dose of laquinimod of 0.1-2.5 mg/day, O.25-2.0 mg or 0.5-1.2 mg.
31. The use of any one of claims 3-30, wherein the medicament comprises a unit dose of imod of0.25 mg, 0.3 mg, 0.5 mg, 0.6 mg, 1.0 mg, 1.2 mg, 1.5 mg or 2.0 mg.
32. The use of any one of claims 1—31, wherein the medicament is prepared for administration via aneous ion or intramuscular injection.
33. The use of claim 32, wherein a) the interferon-B is interferon beta-la and the medicament is prepared for administration intramuscularly at 30 mcg, once weekly; b) the interferon-B is interferon b and the medicament is prepared for administration subcutaneously at 0.25 mg, every other day; or c) the interferon-B is interferon beta-1a and the medicament is ed for administration subcutaneously at 22—44 mcg, three times a week.
34. The use of any one of claims 1—33, where the ment further comprises or is prepared for use in combination with nonsteroidal anti-inflammatory drugs (NSAIDS), salicylates, slow- acting drugs, gold compounds, hydroxychloroquine, sulfasalazine, combinations of slow— acting drugs, corticosteroids, cytotoxic drugs, immunosuppressive drugs and/or antibodies.
35. The use of any one of claims 1-34, wherein the medicament is prepared for periodic administration for at least 3 days, more than 30 days, more than 42 days, 8 weeks or more, at least 12 weeks, at least 24 weeks, more than 24 weeks or 6 months or more.
36. The use of any one of claims 1-35, wherein the medicament inhibits a m of ing multiple sis by at least 20%, at least 30%, at least 50%, at least 70%, more than 100%, more than 300% or more than 1000% .
37. The use of any one of claims 1—36, wherein each of the amount of laquinimod when taken alone, and the amount of eron—B when taken alone is effective to treat the human patient, or wherein either the amount of laquinimod when taken alone, the amount of interferon-B when taken alone, or each such amount when taken alone is not effective to treat the human patient.
38. A package comprising: a) a first pharmaceutical composition comprising an amount of laquinimod and a pharmaceutically acceptable carrier; b) a second pharmaceutical composition comprising an amount of interferon—B and a pharmaceutically able carrier; and c) instructions for use of the first and second pharmaceutical compositions together to treat a human patient afflicted with le sclerosis or presenting a clinically isolated syndrome, wherein the amount of laquinimod and the amount of interferon-B when taken er are more effective to treat the human patient than each agent at the same amount is administered alone.
39. The package of claim 38, wherein the first pharmaceutical composition is in liquid form, in solid form, in capsule form or in tablet form.
40. The package of claim 39, wherein the tablets are coated with a coating which inhibits oxygen from contacting the core, and/or the coating comprises a cellulosic polymer, a detackifier, a gloss enhancer, and/or pigment.
41. The e of any one of claims 38-40, wherein the first pharmaceutical composition further comprises mannitol, an alkalinizing agent, an ion reducing agent, a lubricant, and/or a filler, wherein: a) if the first pharmaceutical composition further comprises an alkalinizing agent, it is preferably meglumine, b) if the first pharmaceutical composition r comprises a lubricant, i) it is preferably present in the composition as solid particles and/or ii) it is preferably sodium stearyl fumarate or magnesium stearate, c) if the first pharmaceutical composition further comprises a filler, i) it is preferably present in the composition as solid particles and/or ii) it is preferably lactose, lactose monohydrate, , isomalt, mannitol, sodium starch glycolate, sorbitol, lactose spray dried, e anhydrouse, or a combination thereof, preferably the filler is mannitol or lactose drate.
42. The e of any one of claims 38-41, n the first pharmaceutical composition is stable and free of an alkalinizing agent or an oxidation reducing agent, preferably the first pharmaceutical composition is free of an alkalinizing agent and free of an oxidation reducing agent.
43. The package of any one of claims 38-42, wherein the first pharmaceutical composition is stable and free of disintegrant.
44. The package of any one of claims 38-43, further comprising a desiccant, preferably the desiccant is silica gel.
45. The package of any one of claims 38-44, wherein the first ceutical composition is stable has a moisture content of no more than 4%.
46. The package of any one of claims 38-45, wherein laquinimod is present in the ition as solid particles.
47. The package of any one of claims 38-46, wherein the package is a sealed packaging having a moisture bility of not more than 15 mg/day per liter, preferably the sealed package is a blister pack in which the m moisture permeability is no more than 0.005 mg/day.
48. The package of claim 47, wherein the sealed package is a bottle, preferably the bottle is closed with a heat induction liner.
49. The package of claims 47 or 48, wherein the sealed package ses an HDPE bottle.
50. The package of any one of claims 47-49, wherein the sealed package comprises an oxygen absorbing agent, ably the oxygen absorbing agent is iron.
51. The package of any one of claims 38-50, wherein the amount of laquinimod in the first composition is less than 0.6 mg or 01-400 mg, preferably 0.1-2.5 mg, 0.25—20 mg or 0.5-1.2
52. The package of claim 51, wherein the amount of laquinimod in the first composition is 0.25 mg, 0.3 mg, 0.5 mg, 0.6 mg, 1.0 mg, 1.2 mg, 1.5 mg or 2.0 mg.
53. Use according to claim 1, substantially as herein described with reference to any one of the Examples and/or
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161512817P | 2011-07-28 | 2011-07-28 | |
US61/512,817 | 2011-07-28 | ||
PCT/US2012/048689 WO2013016686A1 (en) | 2011-07-28 | 2012-07-27 | Treatment of multiple sclerosis with combination of laquinimod and interferon-beta |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ621215A NZ621215A (en) | 2015-11-27 |
NZ621215B2 true NZ621215B2 (en) | 2016-03-01 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20160166648A1 (en) | Treatment of Multiple Sclerosis With Combination of Laquinimod and Interferon-Beta | |
US20160361352A1 (en) | Treatment of Multiple Sclerosis With Combination of Laquinimod and Glatiramer Acetate | |
US8889627B2 (en) | Treatment of multiple sclerosis with combination of laquinimod and fingolimod | |
WO2017048457A1 (en) | Combination of laquinimod and pridopidine to treat multiple sclerosis | |
US20170007596A1 (en) | Treatment of multiple sclerosis with combination of laquinimod and flupirtine | |
US20180036302A1 (en) | Treatment of multiple sclerosis with combination of laquinimod and a statin | |
US20160271094A1 (en) | Laquinimod Combination Therapy For Treatment Of Multiple Sclerosis | |
US20160296513A1 (en) | Treatment of multiple sclerosis by alemtuzumab induction followed by laquinimod therapy | |
NZ621215B2 (en) | Treatment of multiple sclerosis with combination of laquinimod and interferon-beta | |
US20160317525A1 (en) | Treatment of multiple sclerosis with combination of laquinimod and teriflunomide | |
TW201404394A (en) | Treatment of multiple sclerosis with combination of laquinimod and interferon-beta |