WO2010008719A2 - Oral pharmaceutical formulations of vla-4 antagonists - Google Patents
Oral pharmaceutical formulations of vla-4 antagonists Download PDFInfo
- Publication number
- WO2010008719A2 WO2010008719A2 PCT/US2009/047353 US2009047353W WO2010008719A2 WO 2010008719 A2 WO2010008719 A2 WO 2010008719A2 US 2009047353 W US2009047353 W US 2009047353W WO 2010008719 A2 WO2010008719 A2 WO 2010008719A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- pharmaceutical formulation
- compound
- fatty acid
- vla
- dose
- Prior art date
Links
- 239000005557 antagonist Substances 0.000 title claims abstract description 32
- 239000008203 oral pharmaceutical composition Substances 0.000 title abstract description 7
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 87
- 108010008212 Integrin alpha4beta1 Proteins 0.000 claims abstract description 43
- 239000004094 surface-active agent Substances 0.000 claims abstract description 31
- 238000000034 method Methods 0.000 claims abstract description 22
- 208000027866 inflammatory disease Diseases 0.000 claims abstract description 18
- 230000002401 inhibitory effect Effects 0.000 claims abstract 2
- 229940125890 compound Ia Drugs 0.000 claims description 146
- -1 sorbitan fatty acid ester Chemical class 0.000 claims description 67
- 239000000203 mixture Substances 0.000 claims description 41
- 235000014113 dietary fatty acids Nutrition 0.000 claims description 37
- 239000000194 fatty acid Substances 0.000 claims description 37
- 229930195729 fatty acid Natural products 0.000 claims description 37
- 150000001875 compounds Chemical class 0.000 claims description 34
- 238000009472 formulation Methods 0.000 claims description 31
- 239000008187 granular material Substances 0.000 claims description 28
- 239000007884 disintegrant Substances 0.000 claims description 20
- 150000003839 salts Chemical class 0.000 claims description 18
- 238000011282 treatment Methods 0.000 claims description 18
- 239000011230 binding agent Substances 0.000 claims description 17
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 claims description 16
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 claims description 16
- 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 15
- 239000003085 diluting agent Substances 0.000 claims description 14
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 claims description 13
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 13
- 239000000314 lubricant Substances 0.000 claims description 12
- 150000004665 fatty acids Chemical class 0.000 claims description 11
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 claims description 10
- 229920002785 Croscarmellose sodium Polymers 0.000 claims description 10
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 claims description 10
- 229960001681 croscarmellose sodium Drugs 0.000 claims description 10
- 235000010947 crosslinked sodium carboxy methyl cellulose Nutrition 0.000 claims description 10
- 239000008101 lactose Substances 0.000 claims description 10
- 235000019333 sodium laurylsulphate Nutrition 0.000 claims description 10
- WHNWPMSKXPGLAX-UHFFFAOYSA-N N-Vinyl-2-pyrrolidone Chemical compound C=CN1CCCC1=O WHNWPMSKXPGLAX-UHFFFAOYSA-N 0.000 claims description 9
- 229920000881 Modified starch Polymers 0.000 claims description 8
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical group [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 claims description 8
- 235000019359 magnesium stearate Nutrition 0.000 claims description 8
- JRZJOMJEPLMPRA-UHFFFAOYSA-N olefin Natural products CCCCCCCC=C JRZJOMJEPLMPRA-UHFFFAOYSA-N 0.000 claims description 8
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 claims description 6
- 239000004359 castor oil Substances 0.000 claims description 6
- 235000019438 castor oil Nutrition 0.000 claims description 6
- 229960000913 crospovidone Drugs 0.000 claims description 6
- ZEMPKEQAKRGZGQ-XOQCFJPHSA-N glycerol triricinoleate Natural products CCCCCC[C@@H](O)CC=CCCCCCCCC(=O)OC[C@@H](COC(=O)CCCCCCCC=CC[C@@H](O)CCCCCC)OC(=O)CCCCCCCC=CC[C@H](O)CCCCCC ZEMPKEQAKRGZGQ-XOQCFJPHSA-N 0.000 claims description 6
- 235000013809 polyvinylpolypyrrolidone Nutrition 0.000 claims description 6
- 229920000523 polyvinylpolypyrrolidone Polymers 0.000 claims description 6
- 239000000377 silicon dioxide Substances 0.000 claims description 6
- 235000012239 silicon dioxide Nutrition 0.000 claims description 6
- 229920003109 sodium starch glycolate Polymers 0.000 claims description 6
- 239000008109 sodium starch glycolate Substances 0.000 claims description 6
- 229940079832 sodium starch glycolate Drugs 0.000 claims description 6
- 239000000454 talc Substances 0.000 claims description 6
- 229910052623 talc Inorganic materials 0.000 claims description 6
- 201000006417 multiple sclerosis Diseases 0.000 claims description 5
- 239000011734 sodium Substances 0.000 claims description 5
- 229910052708 sodium Inorganic materials 0.000 claims description 5
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 claims description 4
- 229920000168 Microcrystalline cellulose Polymers 0.000 claims description 4
- 229920002472 Starch Polymers 0.000 claims description 4
- 235000021355 Stearic acid Nutrition 0.000 claims description 4
- 229920013820 alkyl cellulose Polymers 0.000 claims description 4
- 125000005037 alkyl phenyl group Chemical group 0.000 claims description 4
- 150000008051 alkyl sulfates Chemical class 0.000 claims description 4
- 150000008052 alkyl sulfonates Chemical class 0.000 claims description 4
- 150000001412 amines Chemical class 0.000 claims description 4
- 125000005228 aryl sulfonate group Chemical group 0.000 claims description 4
- 229920001400 block copolymer Polymers 0.000 claims description 4
- 125000004181 carboxyalkyl group Chemical group 0.000 claims description 4
- 235000010980 cellulose Nutrition 0.000 claims description 4
- 229920002678 cellulose Polymers 0.000 claims description 4
- 239000001913 cellulose Substances 0.000 claims description 4
- LYCAIKOWRPUZTN-UHFFFAOYSA-N ethylene glycol Natural products OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 claims description 4
- 125000005456 glyceride group Chemical group 0.000 claims description 4
- 229940075507 glyceryl monostearate Drugs 0.000 claims description 4
- WGCNASOHLSPBMP-UHFFFAOYSA-N hydroxyacetaldehyde Natural products OCC=O WGCNASOHLSPBMP-UHFFFAOYSA-N 0.000 claims description 4
- 239000008108 microcrystalline cellulose Substances 0.000 claims description 4
- 229940016286 microcrystalline cellulose Drugs 0.000 claims description 4
- 235000019813 microcrystalline cellulose Nutrition 0.000 claims description 4
- 239000001788 mono and diglycerides of fatty acids Substances 0.000 claims description 4
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 claims description 4
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 claims description 4
- 229920002503 polyoxyethylene-polyoxypropylene Polymers 0.000 claims description 4
- 239000008107 starch Substances 0.000 claims description 4
- 229940032147 starch Drugs 0.000 claims description 4
- 235000019698 starch Nutrition 0.000 claims description 4
- 239000008117 stearic acid Substances 0.000 claims description 4
- LDVVTQMJQSCDMK-UHFFFAOYSA-N 1,3-dihydroxypropan-2-yl formate Chemical compound OCC(CO)OC=O LDVVTQMJQSCDMK-UHFFFAOYSA-N 0.000 claims description 3
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 claims description 3
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 claims description 3
- 229920000663 Hydroxyethyl cellulose Polymers 0.000 claims description 3
- 239000004354 Hydroxyethyl cellulose Substances 0.000 claims description 3
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 claims description 3
- 229930195725 Mannitol Natural products 0.000 claims description 3
- FUFJGUQYACFECW-UHFFFAOYSA-L calcium hydrogenphosphate Chemical compound [Ca+2].OP([O-])([O-])=O FUFJGUQYACFECW-UHFFFAOYSA-L 0.000 claims description 3
- CJZGTCYPCWQAJB-UHFFFAOYSA-L calcium stearate Chemical compound [Ca+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O CJZGTCYPCWQAJB-UHFFFAOYSA-L 0.000 claims description 3
- 235000013539 calcium stearate Nutrition 0.000 claims description 3
- 239000008116 calcium stearate Substances 0.000 claims description 3
- 235000019700 dicalcium phosphate Nutrition 0.000 claims description 3
- 229940095079 dicalcium phosphate anhydrous Drugs 0.000 claims description 3
- 235000019447 hydroxyethyl cellulose Nutrition 0.000 claims description 3
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 claims description 3
- 239000001863 hydroxypropyl cellulose Substances 0.000 claims description 3
- 229960001375 lactose Drugs 0.000 claims description 3
- 239000000594 mannitol Substances 0.000 claims description 3
- 235000010355 mannitol Nutrition 0.000 claims description 3
- 229960001855 mannitol Drugs 0.000 claims description 3
- 229920000036 polyvinylpyrrolidone Polymers 0.000 claims description 3
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 claims description 3
- 229940069328 povidone Drugs 0.000 claims description 3
- MVPICKVDHDWCJQ-UHFFFAOYSA-N ethyl 3-pyrrolidin-1-ylpropanoate Chemical compound CCOC(=O)CCN1CCCC1 MVPICKVDHDWCJQ-UHFFFAOYSA-N 0.000 claims description 2
- 229940045902 sodium stearyl fumarate Drugs 0.000 claims description 2
- KEAYESYHFKHZAL-UHFFFAOYSA-N Sodium Chemical class [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 abstract description 4
- JRGSXHUFFDTFCN-IBGZPJMESA-N (3s)-3-[[1-[(2-chlorophenyl)methyl]-4-hydroxy-5-methyl-2-oxopyridin-3-yl]carbamoylamino]-3-(4-methylphenyl)propanoic acid Chemical compound C1=CC(C)=CC=C1[C@H](CC(O)=O)NC(=O)NC(C1=O)=C(O)C(C)=CN1CC1=CC=CC=C1Cl JRGSXHUFFDTFCN-IBGZPJMESA-N 0.000 abstract 1
- 239000002775 capsule Substances 0.000 description 37
- 210000004698 lymphocyte Anatomy 0.000 description 30
- 238000004090 dissolution Methods 0.000 description 24
- 229940068196 placebo Drugs 0.000 description 20
- 239000000902 placebo Substances 0.000 description 20
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 15
- 210000000265 leukocyte Anatomy 0.000 description 14
- 239000003814 drug Substances 0.000 description 13
- 229940079593 drug Drugs 0.000 description 13
- 230000000694 effects Effects 0.000 description 12
- 230000004044 response Effects 0.000 description 11
- 206010053613 Type IV hypersensitivity reaction Diseases 0.000 description 10
- 238000004458 analytical method Methods 0.000 description 10
- 230000005951 type IV hypersensitivity Effects 0.000 description 10
- 208000027930 type IV hypersensitivity disease Diseases 0.000 description 10
- 210000004369 blood Anatomy 0.000 description 9
- 239000008280 blood Substances 0.000 description 9
- 210000004027 cell Anatomy 0.000 description 9
- 230000008859 change Effects 0.000 description 9
- 230000000630 rising effect Effects 0.000 description 9
- 229930182480 glucuronide Natural products 0.000 description 8
- 239000002207 metabolite Substances 0.000 description 8
- 230000002093 peripheral effect Effects 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 101000994375 Homo sapiens Integrin alpha-4 Proteins 0.000 description 7
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 description 7
- 102100032818 Integrin alpha-4 Human genes 0.000 description 7
- 102100025304 Integrin beta-1 Human genes 0.000 description 7
- 230000002411 adverse Effects 0.000 description 7
- 210000001616 monocyte Anatomy 0.000 description 7
- 229920000136 polysorbate Polymers 0.000 description 7
- 230000002265 prevention Effects 0.000 description 7
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 7
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 6
- 230000005764 inhibitory process Effects 0.000 description 6
- 238000009533 lab test Methods 0.000 description 6
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 5
- 102100033467 L-selectin Human genes 0.000 description 5
- RVGRUAULSDPKGF-UHFFFAOYSA-N Poloxamer Chemical compound C1CO1.CC1CO1 RVGRUAULSDPKGF-UHFFFAOYSA-N 0.000 description 5
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 5
- 239000013566 allergen Substances 0.000 description 5
- 239000007857 degradation product Substances 0.000 description 5
- 210000003979 eosinophil Anatomy 0.000 description 5
- 239000000706 filtrate Substances 0.000 description 5
- 230000003285 pharmacodynamic effect Effects 0.000 description 5
- 230000000144 pharmacologic effect Effects 0.000 description 5
- 229960000502 poloxamer Drugs 0.000 description 5
- 229920001983 poloxamer Polymers 0.000 description 5
- 238000003860 storage Methods 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 239000008186 active pharmaceutical agent Substances 0.000 description 4
- 239000003945 anionic surfactant Substances 0.000 description 4
- 230000000052 comparative effect Effects 0.000 description 4
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 4
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 4
- 238000005469 granulation Methods 0.000 description 4
- 230000003179 granulation Effects 0.000 description 4
- 208000005252 hepatitis A Diseases 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 238000007884 metabolite profiling Methods 0.000 description 4
- 239000002736 nonionic surfactant Substances 0.000 description 4
- 239000000843 powder Substances 0.000 description 4
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- 238000004007 reversed phase HPLC Methods 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 108010067306 Fibronectins Proteins 0.000 description 3
- 102000016359 Fibronectins Human genes 0.000 description 3
- 206010020751 Hypersensitivity Diseases 0.000 description 3
- 206010028980 Neoplasm Diseases 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 208000026935 allergic disease Diseases 0.000 description 3
- 230000007815 allergy Effects 0.000 description 3
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 238000001574 biopsy Methods 0.000 description 3
- 230000021164 cell adhesion Effects 0.000 description 3
- 230000006378 damage Effects 0.000 description 3
- 201000010099 disease Diseases 0.000 description 3
- 238000001035 drying Methods 0.000 description 3
- 239000000428 dust Substances 0.000 description 3
- 238000011156 evaluation Methods 0.000 description 3
- 230000002349 favourable effect Effects 0.000 description 3
- 229940046528 grass pollen Drugs 0.000 description 3
- 208000015181 infectious disease Diseases 0.000 description 3
- 102000006495 integrins Human genes 0.000 description 3
- 108010044426 integrins Proteins 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- 230000036470 plasma concentration Effects 0.000 description 3
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 3
- 229920000053 polysorbate 80 Polymers 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 108090000623 proteins and genes Proteins 0.000 description 3
- 238000007390 skin biopsy Methods 0.000 description 3
- 238000010181 skin prick test Methods 0.000 description 3
- 238000000825 ultraviolet detection Methods 0.000 description 3
- 238000002255 vaccination Methods 0.000 description 3
- 238000005550 wet granulation Methods 0.000 description 3
- 241000222122 Candida albicans Species 0.000 description 2
- LZZYPRNAOMGNLH-UHFFFAOYSA-M Cetrimonium bromide Chemical compound [Br-].CCCCCCCCCCCCCCCC[N+](C)(C)C LZZYPRNAOMGNLH-UHFFFAOYSA-M 0.000 description 2
- 239000002033 PVDF binder Substances 0.000 description 2
- 229920001219 Polysorbate 40 Polymers 0.000 description 2
- 229920001214 Polysorbate 60 Polymers 0.000 description 2
- 239000004147 Sorbitan trioleate Substances 0.000 description 2
- PRXRUNOAOLTIEF-ADSICKODSA-N Sorbitan trioleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@@H](OC(=O)CCCCCCC\C=C/CCCCCCCC)[C@H]1OC[C@H](O)[C@H]1OC(=O)CCCCCCC\C=C/CCCCCCCC PRXRUNOAOLTIEF-ADSICKODSA-N 0.000 description 2
- 206010047700 Vomiting Diseases 0.000 description 2
- 208000027418 Wounds and injury Diseases 0.000 description 2
- IJCWFDPJFXGQBN-RYNSOKOISA-N [(2R)-2-[(2R,3R,4S)-4-hydroxy-3-octadecanoyloxyoxolan-2-yl]-2-octadecanoyloxyethyl] octadecanoate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@@H](OC(=O)CCCCCCCCCCCCCCCCC)[C@H]1OC[C@H](O)[C@H]1OC(=O)CCCCCCCCCCCCCCCCC IJCWFDPJFXGQBN-RYNSOKOISA-N 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 210000003651 basophil Anatomy 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 229940095731 candida albicans Drugs 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 239000012738 dissolution medium Substances 0.000 description 2
- 230000008030 elimination Effects 0.000 description 2
- 238000003379 elimination reaction Methods 0.000 description 2
- 210000002889 endothelial cell Anatomy 0.000 description 2
- 230000003511 endothelial effect Effects 0.000 description 2
- 210000003038 endothelium Anatomy 0.000 description 2
- 239000000284 extract Substances 0.000 description 2
- 230000009246 food effect Effects 0.000 description 2
- 235000021471 food effect Nutrition 0.000 description 2
- 210000003714 granulocyte Anatomy 0.000 description 2
- IPCSVZSSVZVIGE-UHFFFAOYSA-N hexadecanoic acid Chemical compound CCCCCCCCCCCCCCCC(O)=O IPCSVZSSVZVIGE-UHFFFAOYSA-N 0.000 description 2
- 230000016784 immunoglobulin production Effects 0.000 description 2
- 230000028709 inflammatory response Effects 0.000 description 2
- 208000014674 injury Diseases 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 231100000682 maximum tolerated dose Toxicity 0.000 description 2
- 235000012054 meals Nutrition 0.000 description 2
- 230000005012 migration Effects 0.000 description 2
- 238000013508 migration Methods 0.000 description 2
- 238000002156 mixing Methods 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 239000008389 polyethoxylated castor oil Substances 0.000 description 2
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 2
- 229940068968 polysorbate 80 Drugs 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 230000003334 potential effect Effects 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 238000012552 review Methods 0.000 description 2
- 239000007909 solid dosage form Substances 0.000 description 2
- 235000019337 sorbitan trioleate Nutrition 0.000 description 2
- 229960000391 sorbitan trioleate Drugs 0.000 description 2
- 235000011078 sorbitan tristearate Nutrition 0.000 description 2
- 239000001589 sorbitan tristearate Substances 0.000 description 2
- 229960004129 sorbitan tristearate Drugs 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 210000002700 urine Anatomy 0.000 description 2
- CUNWUEBNSZSNRX-RKGWDQTMSA-N (2r,3r,4r,5s)-hexane-1,2,3,4,5,6-hexol;(z)-octadec-9-enoic acid Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO.OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO.CCCCCCCC\C=C/CCCCCCCC(O)=O.CCCCCCCC\C=C/CCCCCCCC(O)=O.CCCCCCCC\C=C/CCCCCCCC(O)=O CUNWUEBNSZSNRX-RKGWDQTMSA-N 0.000 description 1
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 1
- FFJCNSLCJOQHKM-CLFAGFIQSA-N (z)-1-[(z)-octadec-9-enoxy]octadec-9-ene Chemical compound CCCCCCCC\C=C/CCCCCCCCOCCCCCCCC\C=C/CCCCCCCC FFJCNSLCJOQHKM-CLFAGFIQSA-N 0.000 description 1
- RZRNAYUHWVFMIP-KTKRTIGZSA-N 1-oleoylglycerol Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(O)CO RZRNAYUHWVFMIP-KTKRTIGZSA-N 0.000 description 1
- FKOKUHFZNIUSLW-UHFFFAOYSA-N 2-Hydroxypropyl stearate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(C)O FKOKUHFZNIUSLW-UHFFFAOYSA-N 0.000 description 1
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 1
- CDOUZKKFHVEKRI-UHFFFAOYSA-N 3-bromo-n-[(prop-2-enoylamino)methyl]propanamide Chemical compound BrCCC(=O)NCNC(=O)C=C CDOUZKKFHVEKRI-UHFFFAOYSA-N 0.000 description 1
- HIQIXEFWDLTDED-UHFFFAOYSA-N 4-hydroxy-1-piperidin-4-ylpyrrolidin-2-one Chemical compound O=C1CC(O)CN1C1CCNCC1 HIQIXEFWDLTDED-UHFFFAOYSA-N 0.000 description 1
- XZIIFPSPUDAGJM-UHFFFAOYSA-N 6-chloro-2-n,2-n-diethylpyrimidine-2,4-diamine Chemical compound CCN(CC)C1=NC(N)=CC(Cl)=N1 XZIIFPSPUDAGJM-UHFFFAOYSA-N 0.000 description 1
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 1
- USFZMSVCRYTOJT-UHFFFAOYSA-N Ammonium acetate Chemical compound N.CC(O)=O USFZMSVCRYTOJT-UHFFFAOYSA-N 0.000 description 1
- 239000005695 Ammonium acetate Substances 0.000 description 1
- 201000001320 Atherosclerosis Diseases 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 108010067225 Cell Adhesion Molecules Proteins 0.000 description 1
- 102000016289 Cell Adhesion Molecules Human genes 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 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
- LVGKNOAMLMIIKO-UHFFFAOYSA-N Elaidinsaeure-aethylester Natural products CCCCCCCCC=CCCCCCCCC(=O)OCC LVGKNOAMLMIIKO-UHFFFAOYSA-N 0.000 description 1
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 1
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 1
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 1
- 206010061218 Inflammation Diseases 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 241000701460 JC polyomavirus Species 0.000 description 1
- 238000003109 Karl Fischer titration Methods 0.000 description 1
- 241000209082 Lolium Species 0.000 description 1
- 206010025280 Lymphocytosis Diseases 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 206010028813 Nausea Diseases 0.000 description 1
- IGFHQQFPSIBGKE-UHFFFAOYSA-N Nonylphenol Natural products CCCCCCCCCC1=CC=C(O)C=C1 IGFHQQFPSIBGKE-UHFFFAOYSA-N 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 1
- 238000002944 PCR assay Methods 0.000 description 1
- 235000021314 Palmitic acid Nutrition 0.000 description 1
- 229920001363 Polidocanol Polymers 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920002685 Polyoxyl 35CastorOil Polymers 0.000 description 1
- 229920002690 Polyoxyl 40 HydrogenatedCastorOil Polymers 0.000 description 1
- 229920002701 Polyoxyl 40 Stearate Polymers 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 229920002642 Polysorbate 65 Polymers 0.000 description 1
- 206010039491 Sarcoma Diseases 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
- NWGKJDSIEKMTRX-AAZCQSIUSA-N Sorbitan monooleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O NWGKJDSIEKMTRX-AAZCQSIUSA-N 0.000 description 1
- IYFATESGLOUGBX-YVNJGZBMSA-N Sorbitan monopalmitate Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O IYFATESGLOUGBX-YVNJGZBMSA-N 0.000 description 1
- HVUMOYIDDBPOLL-XWVZOOPGSA-N Sorbitan monostearate Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O HVUMOYIDDBPOLL-XWVZOOPGSA-N 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 108010000134 Vascular Cell Adhesion Molecule-1 Proteins 0.000 description 1
- 102100023543 Vascular cell adhesion protein 1 Human genes 0.000 description 1
- FGUZFFWTBWJBIL-XWVZOOPGSA-N [(1r)-1-[(2s,3r,4s)-3,4-dihydroxyoxolan-2-yl]-2-hydroxyethyl] 16-methylheptadecanoate Chemical compound CC(C)CCCCCCCCCCCCCCC(=O)O[C@H](CO)[C@H]1OC[C@H](O)[C@H]1O FGUZFFWTBWJBIL-XWVZOOPGSA-N 0.000 description 1
- LWZFANDGMFTDAV-BURFUSLBSA-N [(2r)-2-[(2r,3r,4s)-3,4-dihydroxyoxolan-2-yl]-2-hydroxyethyl] dodecanoate Chemical compound CCCCCCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O LWZFANDGMFTDAV-BURFUSLBSA-N 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 125000002252 acyl group Chemical group 0.000 description 1
- 229940045714 alkyl sulfonate alkylating agent Drugs 0.000 description 1
- 229940043376 ammonium acetate Drugs 0.000 description 1
- 235000019257 ammonium acetate Nutrition 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 231100001127 band 4 compound Toxicity 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 230000036772 blood pressure Effects 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 235000021152 breakfast Nutrition 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 230000007969 cellular immunity Effects 0.000 description 1
- 230000003399 chemotactic effect Effects 0.000 description 1
- 208000037976 chronic inflammation Diseases 0.000 description 1
- 230000006020 chronic inflammation Effects 0.000 description 1
- 229940075614 colloidal silicon dioxide Drugs 0.000 description 1
- 208000010247 contact dermatitis Diseases 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 235000019329 dioctyl sodium sulphosuccinate Nutrition 0.000 description 1
- 230000006806 disease prevention Effects 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- SFNALCNOMXIBKG-UHFFFAOYSA-N ethylene glycol monododecyl ether Chemical compound CCCCCCCCCCCCOCCO SFNALCNOMXIBKG-UHFFFAOYSA-N 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- YQEMORVAKMFKLG-UHFFFAOYSA-N glycerine monostearate Natural products CCCCCCCCCCCCCCCCCC(=O)OC(CO)CO YQEMORVAKMFKLG-UHFFFAOYSA-N 0.000 description 1
- RZRNAYUHWVFMIP-HXUWFJFHSA-N glycerol monolinoleate Natural products CCCCCCCCC=CCCCCCCCC(=O)OC[C@H](O)CO RZRNAYUHWVFMIP-HXUWFJFHSA-N 0.000 description 1
- SVUQHVRAGMNPLW-UHFFFAOYSA-N glycerol monostearate Natural products CCCCCCCCCCCCCCCCC(=O)OCC(O)CO SVUQHVRAGMNPLW-UHFFFAOYSA-N 0.000 description 1
- 229920001903 high density polyethylene Polymers 0.000 description 1
- 238000009478 high shear granulation Methods 0.000 description 1
- 239000004700 high-density polyethylene Substances 0.000 description 1
- 239000008240 homogeneous mixture Substances 0.000 description 1
- 230000004727 humoral immunity Effects 0.000 description 1
- 238000002991 immunohistochemical analysis Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 108010043603 integrin alpha4beta7 Proteins 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000000968 intestinal effect Effects 0.000 description 1
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 1
- 229950006462 lauromacrogol 400 Drugs 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 1
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 1
- 238000007449 liver function test Methods 0.000 description 1
- 206010025135 lupus erythematosus Diseases 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 230000000116 mitigating effect Effects 0.000 description 1
- 239000004570 mortar (masonry) Substances 0.000 description 1
- WQEPLUUGTLDZJY-UHFFFAOYSA-N n-Pentadecanoic acid Natural products CCCCCCCCCCCCCCC(O)=O WQEPLUUGTLDZJY-UHFFFAOYSA-N 0.000 description 1
- 230000008693 nausea Effects 0.000 description 1
- SNQQPOLDUKLAAF-UHFFFAOYSA-N nonylphenol Chemical compound CCCCCCCCCC1=CC=CC=C1O SNQQPOLDUKLAAF-UHFFFAOYSA-N 0.000 description 1
- UYDLBVPAAFVANX-UHFFFAOYSA-N octylphenoxy polyethoxyethanol Chemical compound CC(C)(C)CC(C)(C)C1=CC=C(OCCOCCOCCOCCO)C=C1 UYDLBVPAAFVANX-UHFFFAOYSA-N 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- 235000021313 oleic acid Nutrition 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920000259 polyoxyethylene lauryl ether Polymers 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 235000010483 polyoxyethylene sorbitan monopalmitate Nutrition 0.000 description 1
- 239000000249 polyoxyethylene sorbitan monopalmitate Substances 0.000 description 1
- 235000010989 polyoxyethylene sorbitan monostearate Nutrition 0.000 description 1
- 239000001818 polyoxyethylene sorbitan monostearate Substances 0.000 description 1
- 235000010988 polyoxyethylene sorbitan tristearate Nutrition 0.000 description 1
- 239000001816 polyoxyethylene sorbitan tristearate Substances 0.000 description 1
- 229940099429 polyoxyl 40 stearate Drugs 0.000 description 1
- 229940101027 polysorbate 40 Drugs 0.000 description 1
- 229940113124 polysorbate 60 Drugs 0.000 description 1
- 229940099511 polysorbate 65 Drugs 0.000 description 1
- 239000008057 potassium phosphate buffer Substances 0.000 description 1
- 229940093625 propylene glycol monostearate Drugs 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 238000005096 rolling process Methods 0.000 description 1
- 238000005070 sampling Methods 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- APSBXTVYXVQYAB-UHFFFAOYSA-M sodium docusate Chemical group [Na+].CCCCC(CC)COC(=O)CC(S([O-])(=O)=O)C(=O)OCC(CC)CCCC APSBXTVYXVQYAB-UHFFFAOYSA-M 0.000 description 1
- RYYKJJJTJZKILX-UHFFFAOYSA-M sodium octadecanoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCC([O-])=O RYYKJJJTJZKILX-UHFFFAOYSA-M 0.000 description 1
- 229940045870 sodium palmitate Drugs 0.000 description 1
- 159000000000 sodium salts Chemical class 0.000 description 1
- GGXKEBACDBNFAF-UHFFFAOYSA-M sodium;hexadecanoate Chemical compound [Na+].CCCCCCCCCCCCCCCC([O-])=O GGXKEBACDBNFAF-UHFFFAOYSA-M 0.000 description 1
- 229940057429 sorbitan isostearate Drugs 0.000 description 1
- 229950006451 sorbitan laurate Drugs 0.000 description 1
- 229940035044 sorbitan monolaurate Drugs 0.000 description 1
- 235000011067 sorbitan monolaureate Nutrition 0.000 description 1
- 229950004959 sorbitan oleate Drugs 0.000 description 1
- 229950003429 sorbitan palmitate Drugs 0.000 description 1
- 229960005078 sorbitan sesquioleate Drugs 0.000 description 1
- 229950011392 sorbitan stearate Drugs 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 229960004793 sucrose Drugs 0.000 description 1
- BDHFUVZGWQCTTF-UHFFFAOYSA-M sulfonate Chemical compound [O-]S(=O)=O BDHFUVZGWQCTTF-UHFFFAOYSA-M 0.000 description 1
- 238000004885 tandem mass spectrometry Methods 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 230000008673 vomiting Effects 0.000 description 1
- 238000001238 wet grinding Methods 0.000 description 1
Classifications
-
- 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
- A61K9/14—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
- A61K9/16—Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
- A61K9/1605—Excipients; Inactive ingredients
- A61K9/1617—Organic compounds, e.g. phospholipids, fats
- A61K9/1623—Sugars or sugar alcohols, e.g. lactose; Derivatives thereof; Homeopathic globules
-
- 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
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/4412—Non condensed pyridines; Hydrogenated derivatives thereof having oxo groups directly attached to the heterocyclic ring
-
- 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
- A61K9/14—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
- A61K9/16—Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
- A61K9/1605—Excipients; Inactive ingredients
- A61K9/1629—Organic macromolecular compounds
- A61K9/1652—Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
Definitions
- the invention also relates to methods of using such pharmaceutical formulations for the treatment and/or prevention of inflammatory disease.
- VLA-4 Very late antigen-4
- VCAM-I integrin receptor vascular cell adhesion molecule- 1
- VLA-4 integrin normally expressed on leukocyte plasma membranes, does not adhere to its appropriate ligands until the leukocytes are activated by chemotactic agents or other stimuli which are often produced by the endothelium or other cells at the site of injury. Only then do the integrins undergo the conformational change necessary to confer high binding affinity for the endothelial adhesion molecules.
- white blood cells also called leukocytes
- a tissue has been invaded by a microorganism or has been damaged
- white blood cells also called leukocytes
- One of the most important aspects of the inflammatory response involves the cell adhesion event.
- white blood cells are found circulating through the bloodstream.
- the white blood cells recognize the invaded or damaged tissue, bind to the wall of the capillary and migrate through the capillary into the affected tissue. These events are mediated by a family of proteins called cell adhesion molecules.
- VLA-4 also called is a heterodimeric cell surface receptor protein composed of CD49d (alpha) and CD29 (beta). VLA-4 is predominantly distributed on the surface of monocytes, lymphocytes and two subclasses of granulocytes: eosinophils and basophils. This protein plays a key role in cell adhesion through its ability to recognize and bind VCAM-I and fibronectin, proteins associated with the endothelial cells that line the interior wall of capillaries.
- endothelial cells express a series of adhesion molecules, including VCAM-I, that are critical for binding the white blood cells that are necessary for fighting infection.
- VCAM-I adhesion molecules
- the white blood cells Prior to binding to VCAM-I or fibronectin, the white blood cells initially bind to certain adhesion molecules to slow their flow and allow the cells to "roll" along the activated endothelium. Monocytes, lymphocytes, basophils and eosinophils are then able to firmly bind to VCAM-I or fibronectin on the blood vessel wall via the VLA-4 integrin. There is evidence that such interactions are also involved in transmigration of these white blood cells into the damaged tissue as well as the initial rolling event itself.
- VLA-4 inhibition has been shown to decrease the accumulation of white blood cells and eosinophils at the site of inflammation, subsequently lessening the severity of inflammatory diseases.
- VLA-4 antagonists and methods of making the same including (3S)-3-[( ⁇ [l-(2- chlorobenzyl)-4-hydroxy-5 -methyl-2-oxo- 1 ,2-dihydropyridin-3 -yl] amino ⁇ carbonyl)amino] -3 -(4- methylphenyl)propanoic acid (referred to herein as Compound I and pharmaceutically acceptable salts thereof, such as its mono-sodium salt referred to herein as Compound IA, are described in United States Patent No. 6,972,296, the entire disclosure of which is incorporated herein by reference. United States Patent No.
- 6,972,296 also generally describes pharmaceutical formulations of VLA-4 antagonists, including Compound I or pharmaceutically acceptable salts thereof, in combination with a pharmaceutically acceptable carrier.
- United States Patent Publication No. 2007/0060608 generally describes pharmaceutical formulations of VLA-4 antagonists, including Compound I or pharmaceutically acceptable salts thereof, in combination with one or more other therapeutically active compounds, and a pharmacologically acceptable diluent.
- United States Patent No. 6,972,296 and United States Patent Publication No. 2007/0060608 also describe methods of using the same to treat and/or prevent disease states in which VLA-4 is involved.
- VLA-4 antagonists including Compound I or a pharmaceutically acceptable salt thereof, that provide acceptable drug loading, dissolution, stability, and bioavailability for a treatment regimen wherein the number of doses administered per day to achieve the desired therapeutic plasma concentration could be reduced.
- Such formulations would reduce the dose, reduce the cost of goods for the product, and/or reduce the dosing regimen.
- Solid dosage forms of such VLA-4 antagonists would also provide greater convenience for patients and hence promote patient compliance.
- the pharmaceutical formulations of the present invention address the aforementioned needs.
- the pharmaceutical formulations of the present invention comprising a surfactant provide enhanced dissolution of a VLA-4 antagonist compared to that of a VLA-4 antagonist in the absence of surfactant.
- such pharmaceutical formulations also provide a favorable pharmacokinetic profile with less variability in humans for VLA-4 antagonists that are BCS class IV.
- wet granulation surprisingly provided a dosage form of VLA-4 antagonist with advantageous bioavailability that provides modest increases in peripheral lymphocyte counts at a single dose ⁇ 150 mg Compound IA.
- These granulation pharmaceutical formulations thereby provide relatively higher drug loading compared to dry-blended formulations and thus reduce number of doses administered per day to achieve the desired therapeutic plasma concentration.
- the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof and a surfactant.
- the granulate further comprises a binder, a disintegrant, a diluent, or a combination of two or more thereof.
- the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation.
- the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof, a surfactant, a binder, a disintegrant, and a diluent, wherein the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation.
- the VLA-4 antagonist is the mono-sodium salt of(3S)-3-[( ⁇ [l-(2-chlorobenzyl)-4-hydroxy-5-methyl-2-oxo-l,2-dihydropyridin-3- yl]amino ⁇ carbonyl)amino]-3-(4-methylphenyl)propanoic acid, referred to herein as Compound IA, the structure of which is:
- the surfactant is a non-ionic surfactant, an anionic surfactant, or a combination thereof.
- the surfactant is a polyoxyalkylene sorbitan fatty acid ester, a sorbitan fatty acid ester, an alkylene glycol fatty acid ester, a polyoxyalkylene fatty acid ester, a fatty acid ester, a polyoxyalkylene fatty acid ether, a C i 6 to C 24 fatty acid, a fatty acid mono-, di- or poly-glyceride, a polyoxyalkylene alkyl phenol, an alkyl phenyl ether, a polyoxyethylene polyoxypropylene block copolymer, a fatty amine oxide, a fatty acid alkanolamide, an alkyl cellulose, a carboxyalkyl cellulose, a polyoxyalkylene castor oil derivatives, an alkyl sulfate, an olefin sulfate, an ether sulfate, a monoglyceride sulfate, an alkyl s
- the binder is hydroxyethyl cellulose, hydroxypropyl cellulose, pregelatinized starch, povidone, or a combination of two or more thereof. In one preferred embodiment, the binder is pregelatinized starch. In certain preferred embodiments, the disintegrant is sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof. In one preferred embodiment, the disingegrant is croscarmellose sodium.
- the diluent is lactose, microcrystalline cellulose, mannitol, dibasic calcium phosphate, starch, or a combination of two or more thereof. In one preferred embodiment, the diluent is lactose.
- the surfactant is sodium lauryl sulfate and the pharmaceutical formulation further comprises pregelatinized starch, crocarmellose sodium, and lactose.
- the pharmaceutical formulation further comprises an extragranular excipient.
- the extragranular excipient is a disintegrant, a flow aid, a lubricant, or a combination of two or more thereof.
- the pharmaceutical formulation further comprises an extragranular disintegrant which is sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof. In one preferred embodiment, the extragranular disintegrant is croscarmellose sodium. In another preferred embodiment, the pharmaceutical formulation further comprises an extragranular flow aid which is silicon dioxide, talc, or a combination thereof. In certain preferred embodiments, the extragranular flow aid is silicon dioxide. In still other preferred embodiments, the pharmaceutical formulation further comprises an extragranular lubricant which is magnesium stearate, calcium stearate, glyceryl monostearate, sodium stearyl fumarate, stearic acid, talc, or a combination of two or more thereof. In certain preferred embodiments, the extragranular lubricant is magnesium stearate.
- the pharmaceutical formulation is contained in a capsule.
- the VLA-4 antagonist comprises 50% by weight of the pharmaceutical formulation.
- the present invention provides pharmaceutical formulations comprising a granulate of Compound IA which provides a mean steady-state AUC of Compound IA that is about 45 microgram-hr/ml when administered at a 200 mg dose of Compound IA once-a-day to a patient.
- the present invention also encompasses pharmaceutical formulations which are similarly bioavailable such that the relative mean steady-state AUC of Compound IA is within 80% to 125% of 45 microgram-hr/ml, that is within the range from about 36 microgram-hr/ml to about 57 microgram-hr/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient.
- the pharmaceutical formulation provides a mean steady-state AUC of Compound IA which is at least 80% of 45 microgram-hr/ml, that is at least 36 microgram-hr/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient. In one embodiment, the pharmaceutical formulation provides a mean steady-state AUC of Compound IA which is at least 45 microgram-hr/ml when administered at a 200 mg dose of Compound IA once- a-day to a patient.
- the present invention provides pharmaceutical formulations comprising a granulate of Compound IA which provides a mean steady-state Cmin of Compound IA that is at least 0.2 microgram/ml when administered at a 200 mg dose of Compound IA once-a-day to a patient.
- the pharmaceutical formulation further comprises sodium lauryl sulfate, pregelatinized starch, crocarmellose sodium, and lactose.
- the pharmaceutical formulation comprising a granulate of Compound IA provides a median Tmax that is in the range from about 1 hour to about 4 hours when administered at a 200 mg dose of Compound IA once-a-day to a patient.
- the pharmaceutical formulation comprising a granulate of Compound IA provides a mean steady-state Cmax of Compound IA that is about 6 microgram/ml when administered at a 200 mg dose of Compound IA once-a-day to a patient.
- the present invention also encompasses pharmaceutical formulations which are similarly bioavailable such that the relative mean steady-state Cmax of Compound IA is within 80% to 125% of 6 microgram/ml, that is within the range from about 4.8 microgram/ml to about 7.6 microgram/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient.
- the pharmaceutical formulation comprising a granulate of Compound IA provides a mean steady-state Cmax of Compound IA which is at least 80% of 6 microgram/ml, that is at least 4.8 microgram/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient.
- the pharmaceutical formulation provides a mean steady-state Cmax of Compound IA which is at least 6 microgram/ml when administered at a 200 mg dose of Compound IA once-a- day to a patient.
- a pharmaceutical formulation of the present invention comprising Compound IA provides a mean steady-state AUC of Compound IA that is at least 36 microgram hr/mL when administered at a dose that is 200 mg once-a-day to a patient and assessed as described infra herein (See Examples).
- a pharmaceutical formulation of the present invention provides a mean steady-state Cmax of Compound IA that is at least 4.8 microgram/mL as measured as described infra herein (See Examples).
- a pharmaceutical formulation of the present invention comprising a granulate of Compound IA provides a mean steady-state Cmnin of Compound IA that is at least 0.2 microgram hr/mL when administered at a dose that is 200 mg once-a-day to a patient and measured as described infra herein (see Examples).
- the present invention provides methods for the treatment or prevention or inhibition of progression of an inflammatory disease, which comprises administering to a patient in need thereof a therapeutically effective amount of any of the pharmaceutical formulations of the present invention.
- the inflammatory disease is multiple sclerosis. DETAILED DESCRIPTION OF THE DRAWINGS
- Figure 1 is a graph of the lymphocyte change from time-matched baseline over a time period up to 12 hours post-dose in subjects given a single dose of either placebo, or Compound IA that is 50 mg, 150 mg, 300 mg, 600 mg, or 1000 mg.
- Figure 2 is a bar graph of the lymphocyte change from time-matched baseline at various time points (specifically, 4 hours, maximum, and day average) post-dose in subjects given a single dose of either placebo, or Compound IA that is 50 mg, 150 mg, 300 mg, 600 mg, or 1000 mg.
- AUC is the area under the plasma concentration-time curve from time zero to a certain time period of the sample.
- AUC (4h) means the area under the plasma concentration-time curve from time zero to 4 hours.
- CL/F is the apparent total clearance of the drug from plasma after oral administration. CL/F is calculated by dividing the dose administered by the AUC.
- treating or “treatment” is intended to mean mitigating or alleviating the symptoms of the recited condition, disease or disorder in a mammal such as a human.
- the present invention provides pharmaceutical formulations of VLA-4 antagonist Compound I or a pharmaceutically acceptable salt thereof suitable for oral administration that meet the aforementioned need for acceptable drug loading, dissolution, stability, and bioavailability of Compound I.
- the VLA-4 antagonist is Compound IA.
- the pharmaceutical formulations of the present invention comprising a surfactant enhance the dissolution of VLA-4 antagonist Compound IA at pH 7 relative to a pharmaceutical formulation lacking a surfactant.
- the pharmaceutical formulations of the present invention provide favorable dissolution profiles and bioavailability of the Compound IA.
- Compound I can be prepared according to Example 25, at columns 75-76, of U.S. Pat. No. 6,972,296; also see Scheme 25 at columns 41-42, incorporated herein by reference.
- Pharmaceutically acceptable salts can be formed with an appropriate base.
- the mono-sodium salt of Compound I, herein referred to as Compound IA has the following structure:
- Compound IA can be formed from Compound I by reacting the free acid with an appropriate base, e.g., sodium hydroxide.
- an appropriate base e.g., sodium hydroxide.
- Compound IA is a Class IV compound, that is, a compound having low solubility and low permeability. Consequently, Compound IA has relatively low bioavailability.
- Compound IA has very low bulk density (0.08 g/cc) and very poor flow properties as the crystalline form is needle-like and thus not readily micronized on a commercial scale.
- pharmaceutical formulations that are sufficiently stable and provide enhanced bioavailability of Compound IA suitable for commercialization are needed.
- the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof and a surfactant.
- the granulate further comprises a binder, a disintegrant, a diluent, or a combination of two or more thereof.
- the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation.
- the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof, a surfactant, a binder, a disintegrant, and a diluent, wherein the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation.
- the VLA-4 antagonist is Compound IA.
- Binders are generally used to impart cohesive qualities to the solid dosage form.
- binders include, without limitation, hydroxyethyl cellulose, hydroxypropyl cellulose, pregelatinized starch, povidone, or a combination of two or more thereof. Binders may make up about 1% to about 15% by weight. In certain preferred embodiments, the binder is present at about 15% by weight. In one preferred embodiment, the binder is pregelatinized starch and is present in an amount that is at least 15% by weight.
- disintegrants include, without limitation, sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof. Disintegrants may make up about 1% to 25% by weight. In one preferred embodiment, the disintegrant is croscarmellose sodium present at about 15% by weight. In another preferred embodiment, the disintegrant is sodium starch glycolate present in an amount that is up to about 10% by weight. In yet another preferred embodiment, the disintegrant is crospovidone present in an amount that is up to about 5% by weight.
- Suitable surfactants for use in the present invention include nonionic surfactants, anionic surfactants, or a combination thereof.
- nonionic surfactants include, without limitation, polyoxyalkylene sorbitan fatty acid esters, sorbitan fatty acid esters, alkylene glycol fatty acid esters, polyoxyalkylene fatty acid esters, fatty acid esters, polyoxyalkylene fatty acid ethers, Ci 6 to C 24 fatty acids, fatty acid mono-, di- or poly-glycerides, polyoxyalkylene alkyl phenols, alkyl phenyl ethers, polyoxyethylene polyoxypropylene block copolymers, fatty amine oxides, fatty acid alkanolamides, alkyl cellulose, carboxyalkyl cellulose, and polyoxyalkylene castor oil derivatives.
- nonionic surfactants include polyoxyethylene(20) sorbitan monolaurate (Tween® 20), polyoxyethylene(4) sorbitan monolaurate (Tween® 21), polyoxyethylene(20) sorbitan monopalmitate (Tween® 40), polyoxyethylene(20) sorbitan monostearate (Tween® 60), polyoxyethylene(20) sorbitan tristearate (Tween® 65), polyoxyethylene(20) sorbitan monooleate (Tween® 80 or polysorbate 80), or polyoxyethylene(20) sorbitan trioleate (Tween®85), lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, glycerol monooleate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, sorbitan laurate, sorbitan oleate, sorbitan palmitate, sorbit
- Poloxamer®3 88, Poloxamer® 407 (BASF Wyandotte Corp.); methylcellulose and carboxymethyl cellulose.
- anionic surfactants include, without limitation, alkyl sulfates, olefin sulfates, ether sulfates, monoglyceride sulfates, alkyl sulfonates, aryl sulfonates, olefin sulfonates, alkyl sulfosuccinates, and aryl sulfosuccinates.
- anionic surfactants include sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate.
- the surfactant is a polyoxyalkylene sorbitan fatty acid ester, a sorbitan fatty acid ester, an alkylene glycol fatty acid ester, a polyoxyalkylene fatty acid ester, a fatty acid ester, a polyoxyalkylene fatty acid ether, a C 16 to C 24 fatty acid, a fatty acid mono-, di- or poly-glyceride, a polyoxyalkylene alkyl phenol, an alkyl phenyl ether, a polyoxyethylene polyoxypropylene block copolymer, a fatty amine oxide, a fatty acid alkanolamide, an alkyl cellulose, a carboxyalkyl cellulose, a polyoxyalkylene castor oil derivative, an alkyl
- the surfactant is sodium lauryl sulfate.
- Surfactants may make up about 0.5% to about 2% by weight. In one preferred embodiment, the surfactant is present at about 1% by weight. In one preferred embodiment, the surfactant is sodium lauryl sulfate present at about 1% by weight.
- diluents include, without limitation, lactose, microcrystalline cellulose, mannitol, dibasic calcium phosphate, starch, or a combination of two or more thereof.
- the diluent is lactose.
- the diluent is microcrystalline cellulose in combination with lactose.
- the pharmaceutical formulation of the present invention may further comprise an extragranular excipient which is a glidant, lubricant, disintegrant, or a combination of two or more thereof.
- glidants also known as flow agents, include, without limitation, silicon dioxide (including colloidal silicon dioxide), talc, or a combination thereof. Glidants may make up about 0.1% to about 0.5% by weight. In certain preferred embodiments, the glidant is present at about 0.5% by weight. In one preferred embodiment, the glidant is silicon dioxide.
- lubricants include, without limitation, magnesium stearate, calcium stearate, glyceryl monostearate, sodium stearyl fiimarate, stearic acid, talc, or a combination of two or more thereof. Lubricants may make up about 0.1% to about 1%. In certain preferred embodiments, the lubricant is present at about 0.5% by weight. In one preferred embodiment, the lubricant is magnesium stearate.
- Suitable oral pharmaceutical formulations include, but are not limited to, capsules, tablets, granules, powders, and unit dose packets.
- the oral pharmaceutical formulation is a capsule.
- the oral pharmaceutical formulation is a tablet.
- compositions of the present invention can be prepared by a variety of suitable wet granulation, including high shear wet granulation, techniques.
- suitable wet granulation including high shear wet granulation, techniques.
- such pharmaceutical formulations can be prepared using the process diagramed in Scheme I below:
- Compound I or a pharmaceutically acceptable salt thereof; and (ii) a granular excipient (e.g., surfactant, diluent, disintegrant, binder) to provide a first dry-blended powder
- a granular excipient e.g., surfactant, diluent, disintegrant, binder
- step "d” can be dried in any one of the following ways: a) within the high-shear granulator, using microwave power as the source of energy to evaporate the granulating liquid and vacuum to remove the vapor; b) in a fluidized bed dryer using heated air to dry the granules and remove the vapor; or c) in a tray dryer, in which the wet granules are spread on trays in a heated and ventilated drying oven.
- Another aspect of the invention provides methods for the treatment and/or prevention of disease states in which VLA-4 is involved in a patient comprising administering to the patient a pharmaceutical formulation according to the present invention.
- the pharmaceutical formulations comprise Compound I or a pharmaceutically acceptable salt thereof, preferably Compound IA.
- VLA-4 is also found on various cancer cells, including leukemia, melanoma, lymphoma and sarcoma cells. Cell adhesion involving VLA-4 may be involved in the metastasis of certain cancers. Inhibitors of VLA-4 binding may, therefore, also be useful in the treatment of some forms of cancer.
- Compound I or a pharmaceutically acceptable salt thereof, preferably Compound IA is administered for the treatment of an inflammatory disease.
- Compound I or a pharmaceutically acceptable salt thereof, preferably Compound IA is administered for the prevention or inhibition of an inflammatory disease.
- the inflammatory disease is multiple sclerosis.
- a typical recommended daily dosage regimen for the treatment of an inflammatory disease can range from about 25 mg/day to about 1000 mg/day. In certain preferred embodiments, the recommended daily dosage regimen for the treatment of an inflammatory disease can range from 25 mg/day to 400 mg/day. In one preferred embodiment, the daily dosage regimen for the treatment of an inflammatory disease is about 400 mg/day administered orally in a single daily dose. Similarly, a typical recommended daily dosage regimen for the prevention or inhibition of an inflammatory disease can range from about 25 mg/day to about 1000 mg/day.
- the recommended daily dosage regimen for the prevention or inhibition of an inflammatory disease can range from 25 mg/day to 400 mg/day. In one preferred embodiment, the daily dosage regimen for the prevention or inhibition of an inflammatory disease is about 400 mg/day administered orally in a single daily dose.
- Exemplary formulations A-D were made by wet granulation. Specifically, the formulations were manufactured by first wet-granulating Compound IA and the granular excipients to make a granulate. The granules were then dried, mixed with the extragranular excipients and then encapsulated in size 0 capsules. The outline of the method of manufacture is presented below.
- lubricant e.g., magnesium stearate
- capsules of exemplary formulation D were packaged as 10 capsules per bottle in induction-sealed HDPE bottle with child- resistant caps. Samples were stored at 25 0 C and 60% relative humidity up to 1 year or 40 0 C and 75% relative humidity up to 6 months. Following storage under the aforementioned conditions, samples were pulled at various time points (e.g., 1 month, 3 months, 6 months, 9 months, 12 months) and assessed for moisture content (using volumetric Karl Fischer titration method), label strength, and the presence of Compound IA degradation products.
- samples for assay and degradation product testing were prepared as follows. Capsules were emptied, contents ground using mortar and pestle; an aliquot of powder weighed and diluted with 1:1 water:85%acetonitrile, then shaken to dissolve drug and subsequently filtered to remove insoluble formulation components. The filtrate was then further diluted with 1:1 water:85% acetonitrile; and the sample analyzed using reverse-phase High Performance Liquid Chromatography.
- an ACE 3 C18 column (4.6 mm x 150 mm, 3.0 ⁇ m particles), a gradient elution with mobile phases consisting of acetonitrile, water and 85% phosphoric acid (60:40:0.05, v:v:v).
- Quantitation of Compound IA was performed by UV detection at 220 nm against an external Compound I reference standard.
- the detection limit for the presence of Compound IA degradation products using this assay is less than 0.05%, and the quantitation limit of 0.1%.
- B was determined at pH 7 to simulate intestinal pH.
- a sample dissolution profile was obtained for the release rate of Compound IA from capsules using a USP-II dissolution apparatus, with paddles operated at 50 rpm.
- Each formulation was tested in 900 mL of 0.02 M potassium phosphate buffer (pH 7) as dissolution medium. Aliquots of 10 ml were removed periodically (e.g., 15, 30, 45, and 60 minutes) and filtered through a 0.45 micrometer Millipore Millex-HV
- Exemplary formulation B provides enhanced dissolution of Compound IA in the presence of surfactant compared to that of Compound IA in the absence of surfactant. Consequently, the present inventors believe that formulations of Compound I or a salt thereof comprising surfactant will be beneficial to increase the extent of Compound I absorption in the gastrointestinal tract thereby leading to enhanced bioavailability of Compound I.
- dissolution studies were carried out for capsules of exemplary formulations C and D at pH 5.7.
- a sample dissolution profile was obtained for the release rate of Compound IA from capsules using a USP-II dissolution apparatus, with paddles operated at 100 rpm, maintained at 37 0 C ⁇ 0.5 0 C.
- Each formulation was tested in 900 mL of 0.02 M ammonium acetate buffer with 1.5% Cetyl Trimethyl Ammonium Bromide (CTAB) (pH 5.7) as dissolution medium.
- CAB Cetyl Trimethyl Ammonium Bromide
- dissolution profiles of capsules of exemplary formulations C, D, and comparative formulation E demonstrated 90% or greater release of Compound IA within 30 minutes. As such, all three pharmaceutical formulations provide favorable dissolution profiles for release of Compound IA.
- the dissolution profile obtained from samples at the beginning, middle, and end of the batch reflected adequate dissolution, and comparison of the 60-minutes percent-dissolution indicate acceptable content uniformity across the batch.
- ECGs were obtained on Days -1 and 1, and these ECGs were machine read with over-reading by the investigator.
- blood samples were collected predose (0 hour), 0.25, 0.5, 0.75, 1, 2, 3, 4, 6, 8, 12, 24, 36, 48, and 72 hours postdose.
- metabolite profiling blood samples were collected predose (0 hour), 2 and 6 hours postdose, and urine was collected from 0 to 24 hours postdose.
- Whole blood was collected predose (0 hour), 2 and 6 hours postdose to explore the effect of Compound IA on the expression of cell surface markers and the relative abundance of lymphocyte subtypes in peripheral blood using FACS analyses.
- Multiple blood samples were collected on Days -1 and 1 in a time-matched manner to determine the peripheral lymphocyte counts.
- Test Product, Dose, Mode of Administration Compound IA, 50 mg capsules (specifically, capsules of comparative formulation F described in Table IB above) and 200 mg capsules (specifically, capsules of exemplary formulation D described in Table IA above).
- Doses administered were: 1 x 50 mg (total 50 mg), 3 x 50 mg (total 150 mg), 6 x 50 mg (total 300 mg), 3 x 200 mg (total 600 mg), and 5 x 200 mg (total 1000 mg).
- the study drug was administered orally.
- Reference Therapy, Dose, Mode of Administration Matching placebo 50 mg and 200 mg capsules.
- the number of placebo capsules was the same as the number of Compound IA capsules required for each treatment (dose-level) group.
- the placebo capsule(s) were administered orally.
- Pharmacokinetics The following pharmacokinetic parameters were to be determined. Cmax, Tmax, AUC, K, t ⁇ ⁇ , Vd/F, and CL/F. Pharmacodynamics: The effect of Compound IA on lymphocyte count, the expression of cell surface markers, and the relative abundance of lymphocyte subtypes in peripheral blood were also explored by conventional hematology and FACS analyses.
- Compound IA pharmaceutical formulations described herein provided sufficient bioavailability following oral administration at a single dose >150 mg to elicit a modest increase in peripheral circulating lymphocytes. The exposure appeared to increase as dose increased, and a high-fat meal appeared to increase exposure to Compound IA. An appreciable amount of acyl glucuronide metabolite was observed in plasma.
- Compound IA pharmaceutical formulations described herein provided sufficient bioavailability following oral administration at a single dose >150 mg to elicit a modest increase in peripheral circulating lymphocytes.
- the first part assessed the safety, tolerability, pharmacokinetics, and pharmacological activity of rising multiple doses of Compound IA.
- the pharmacological activity of Compound IA was assessed primarily by delayed-type hypersensitivity (DTH) with skin biopsy. Three dose groups were treated for 14 days with once- daily doses of either 200, 400, or 800 mg Compound IA.
- the second part assessed the safety, tolerability, pharmacokinetics, and pharmacological activity of Compound IA at one dose level (maximum tolerated dose or highest dose from Part 1 of this study) in otherwise healthy subjects with known allergy as evidenced by a positive skin-prick test.
- the effect of Compound IA on allergen-induced late cutaneous response was assessed in Part 2.
- Blood samples were collected into EDTA tubes predose (0 hour) and at 0.5, 1, 2, 3. 4. 6, 8, 12, and 24 hours postdose on Days 1 and 14, predose on Days 11, 12, and 13, and at 36 hours (Day 15), 48 hours (Day 16), and 72 hours (Day 17) postdose on Day 14 for Compound IA analyses. All plasma samples were analyzed for Compound IA using a validated liquid chromatography/tandem mass spectrometry (LC/MS/MS) assay method with a LLOQ of 25 ng/mL. Blood samples were collected predose (0 hour) and 2, 6 and 24 hours postdose on Day 1 and predose (0 hour) and 2, 6, and 24 hours postdose on Day 14. for metabolite profiling. Urine was collected predose on Day 1 and from 0 to 24 hours postdose on Days 1 and 14, for metabolite profiling including analysis of Compound IA.
- LC/MS/MS liquid chromatography/tandem mass spectrometry
- Test Product, Dose, Mode of Administration Compound IA, 200 mg capsules
- Doses administered included: 1 x 200 mg, 2 x 200 mg. and 4 x 200 mg.
- Reference Product, Dose, Mode of Administration Matching placebo capsules. The number of placebo capsules were the same as Compound IA for each treatment group.
- Duration of Treatment Subjects were treated with multiple doses of Compound IA or matching placebo (14 doses, once per day) and were followed for 3 days as inpatients after the end of treatment. The subjects were followed up as outpatients until 28 days post-last-dose of Compound IA or matching placebo. Criteria for Evaluation Primary Endpoints: Safety and tolerability of Compound IA were assessed by collecting adverse events, vital signs, ECGs, physical examinations, and safety laboratory tests.
- Criteria for Evaluation Secondary Endpoints 1) The following pharmacokinetic parameters were determined: Cmax, Tmax, AUC, K, X 1 A, Vd/F, CI/F, and accumulation factor. 2) The following pharmacodynamic endpoints were evaluated: the DTH score and numbers of T cell infiltrate in the skin biopsy; peripheral lymphocyte counts, the expression of cell surface markers, and the relative abundance of lymphocyte subtypes determined by FACS analysis in peripheral blood. Scores of allergen-induced late cutaneous response and eosinophil abundance in the skin biopsy were determined. Serum antibody titers to hepatitis A were assessed at different times following vaccination. In addition, plasma JC virus was analyzed using PCR assay.
- Plasma Compound IA concentrations and pharmacokinetic parameters were listed and summarized using descriptive statistics.
- the primary pharmacokinetic parameters for Compound IA were the AUC and Cmax values.
- Log-transformed AUC and Cmax were statistically analyzed using a one-way analyses of variance model.
- Ninety percent confidence interval estimates of the mean concentrations and pharmacokinetic parameters for each dose were computed.
- the dose normalized Cmax and AUC were analyzed in a similar fashion and 90% confidence intervals for difference were computed for a preliminary assessment of dose proportionality. Trough concentrations at Days 11, 12, 13, 14, and 15 were used to assess steady-state for each dose level.
- Compound IA had a terminal half-life of approximately 10 to 14 hours. There was a dose-related, but apparently less than dose-proportional, increase of exposure (Table 9). Both Cmax and AUC increased when the dose was increased from 200 to 800 mg. Compound IB plus its isomers resulting from acyl migration and/or, perhaps, anomerization, were observed in plasma (Table 8), and the ratio of acyl glucuronide to parent drug in plasma was approximately 0.3 to 0.4. The elimination half-life of Compound IB paralleled that of Compound IA, suggesting that the elimination of Compound IB was formation rate-limited. The variability of pharmacokinetics is mild to moderate.
- Skin tissue lymphocytes from biopsy of the DTH test were quantified and descriptive statistics were provided. According to the protocol, eosinophils from biopsy of allergen induced late cutaneous responses were to be quantified, but the analysis was not performed due to lack of effect of Compound IA on induction of late cutaneous response.
- the potential effect of Compound IA on cellular and humoral immunity was assessed using the DTH response and antibody production to hepatitis A vaccination in this rising multiple-dose study.
- the DTH response to Candida albicans (Candin®) were assessed either before or following 11 days of Compound IA.
- Compound IA at doses up to 800 mg had no significant effect on the DTH response.
- Immunohistochemical analysis did not reveal any apparent effect of Compound IA on CD29 and/or CD49d expressing lymphocytes from tissue biopsy of Candin-induced DTH response.
- the potential effect on allergen-induced late cutaneous response was also assessed in a small number of subjects at the 800 mg dose level; no apparent effect was observed at this dose level.
- no apparent effect on antibody production against hepatitis A vaccination was observed with treatment of Compound IA at doses up to 800 mg.
- Compound IA had no clinically significant effect on blood pressure, pulse rate, or ECGs.
- Compound IA was not associated with clinically significant effects on safety laboratory tests, including liver function tests, electrolytes, and hematology. • Compound IA pharmaceutical formulations described herein provided sufficient bioavailability following oral administration at a single dose to elicit modest lymphocytosis.
- Compound IA was orally bioavailable and eliminated with a half-life of 10 to 14 hours.
Landscapes
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Life Sciences & Earth Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Medicinal Preparation (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The present invention provides oral pharmaceutical formulations comprising a VLA-4 antagonist and a surfactant. In one preferred embodiment, the VLA-4 antagonist is the mono-sodium salt of (3S)-3-[({[1-(2-chlorobenzyl)-4-hydroxy-5-methyl-2-oxo-1,2-dihydropyridin-3-yl]amino}carbonyl)amino]-3-(4-methylphenyl)propanoic acid. The present invention also provides methods for treating and/or preventing and/or inhibiting progress of an inflammatory disease using such pharmaceutical formulations.
Description
ORAL PHARMACEUTICAL FORMULATIONS OF VLA-4 ANTAGONISTS
FIELD OF THE INVENTION The present invention relates to novel oral pharmaceutical formulations comprising a VLA-
4 antagonist and a surfactant. The invention also relates to methods of using such pharmaceutical formulations for the treatment and/or prevention of inflammatory disease.
BACKGROUND OF THE INVENTION Citation of or reference to any application or publication in this Section or any Section of this application is not an admission that such document is available as prior art to the present invention.
Very late antigen-4 (VLA-4) is an alpha4betal integrin dimer. Primarily, integrin receptor vascular cell adhesion molecule- 1 (VCAM-I) is an endothelial ligand for VLA-4 and for integrin alpha4beta7. VLA-4 integrin, normally expressed on leukocyte plasma membranes, does not adhere to its appropriate ligands until the leukocytes are activated by chemotactic agents or other stimuli which are often produced by the endothelium or other cells at the site of injury. Only then do the integrins undergo the conformational change necessary to confer high binding affinity for the endothelial adhesion molecules. When a tissue has been invaded by a microorganism or has been damaged, white blood cells, also called leukocytes, play a major role in the inflammatory response. One of the most important aspects of the inflammatory response involves the cell adhesion event. Generally, white blood cells are found circulating through the bloodstream. However, when a tissue is infected or becomes damaged, the white blood cells recognize the invaded or damaged tissue, bind to the wall of the capillary and migrate through the capillary into the affected tissue. These events are mediated by a family of proteins called cell adhesion molecules.
There are three main types of white blood cells: granulocytes, monocytes and lymphocytes. The integrin VLA-4 (also called
is a heterodimeric cell surface receptor protein composed of CD49d (alpha) and CD29 (beta). VLA-4 is predominantly distributed on the surface of monocytes, lymphocytes and two subclasses of granulocytes: eosinophils and basophils. This
protein plays a key role in cell adhesion through its ability to recognize and bind VCAM-I and fibronectin, proteins associated with the endothelial cells that line the interior wall of capillaries.
Following infection or damage of tissue surrounding a capillary, endothelial cells express a series of adhesion molecules, including VCAM-I, that are critical for binding the white blood cells that are necessary for fighting infection. Prior to binding to VCAM-I or fibronectin, the white blood cells initially bind to certain adhesion molecules to slow their flow and allow the cells to "roll" along the activated endothelium. Monocytes, lymphocytes, basophils and eosinophils are then able to firmly bind to VCAM-I or fibronectin on the blood vessel wall via the VLA-4 integrin. There is evidence that such interactions are also involved in transmigration of these white blood cells into the damaged tissue as well as the initial rolling event itself.
Although white blood cell migration to the site of injury helps fight infection and destroy foreign material, in many instances this migration can become uncontrolled, with white blood cells flooding to the scene, causing widespread tissue damage as found in chronic inflammation and autoimmune disorders. VLA-4 inhibition has been shown to decrease the accumulation of white blood cells and eosinophils at the site of inflammation, subsequently lessening the severity of inflammatory diseases.
VLA-4 antagonists and methods of making the same, including (3S)-3-[({[l-(2- chlorobenzyl)-4-hydroxy-5 -methyl-2-oxo- 1 ,2-dihydropyridin-3 -yl] amino } carbonyl)amino] -3 -(4- methylphenyl)propanoic acid (referred to herein as Compound I and pharmaceutically acceptable salts thereof, such as its mono-sodium salt referred to herein as Compound IA, are described in United States Patent No. 6,972,296, the entire disclosure of which is incorporated herein by reference. United States Patent No. 6,972,296 also generally describes pharmaceutical formulations of VLA-4 antagonists, including Compound I or pharmaceutically acceptable salts thereof, in combination with a pharmaceutically acceptable carrier. United States Patent Publication No. 2007/0060608 generally describes pharmaceutical formulations of VLA-4 antagonists, including Compound I or pharmaceutically acceptable salts thereof, in combination with one or more other therapeutically active compounds, and a pharmacologically acceptable diluent. United States Patent No. 6,972,296 and United States Patent Publication No. 2007/0060608 also describe methods of using the same to treat and/or prevent disease states in which VLA-4 is involved.
Nevertheless, there is a need for pharmaceutical formulations of VLA-4 antagonists, including Compound I or a pharmaceutically acceptable salt thereof, that provide acceptable drug loading, dissolution, stability, and bioavailability for a treatment regimen wherein the number of
doses administered per day to achieve the desired therapeutic plasma concentration could be reduced. Such formulations would reduce the dose, reduce the cost of goods for the product, and/or reduce the dosing regimen. Solid dosage forms of such VLA-4 antagonists would also provide greater convenience for patients and hence promote patient compliance. These and other objectives are provided by the novel pharmaceutical formulations of the present invention.
SUMMARY OF THE INVENTION
The pharmaceutical formulations of the present invention address the aforementioned needs. In particular, the pharmaceutical formulations of the present invention comprising a surfactant provide enhanced dissolution of a VLA-4 antagonist compared to that of a VLA-4 antagonist in the absence of surfactant. Surprisingly, such pharmaceutical formulations also provide a favorable pharmacokinetic profile with less variability in humans for VLA-4 antagonists that are BCS class IV. In addition, wet granulation surprisingly provided a dosage form of VLA-4 antagonist with advantageous bioavailability that provides modest increases in peripheral lymphocyte counts at a single dose ≥ 150 mg Compound IA. These granulation pharmaceutical formulations thereby provide relatively higher drug loading compared to dry-blended formulations and thus reduce number of doses administered per day to achieve the desired therapeutic plasma concentration.
In one embodiment, the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof and a surfactant. In certain embodiments, the granulate further comprises a binder, a disintegrant, a diluent, or a combination of two or more thereof. In certain preferred embodiments, the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation. In another embodiment, the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof, a surfactant, a binder, a disintegrant, and a diluent, wherein the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation. In preferred embodiments, the VLA-4 antagonist is the mono-sodium salt of(3S)-3-[({[l-(2-chlorobenzyl)-4-hydroxy-5-methyl-2-oxo-l,2-dihydropyridin-3- yl]amino}carbonyl)amino]-3-(4-methylphenyl)propanoic acid, referred to herein as Compound IA, the structure of which is:
Pharmaceutically acceptable salts of Compound I, including the sodium salt of Compound IA are described in United States Patent No. 6,972,296 at column 52, line 48 to column 53, line 38, incorporated herein by reference. In certain preferred embodiments, the surfactant is a non-ionic surfactant, an anionic surfactant, or a combination thereof. In one preferred embodiment, the surfactant is a polyoxyalkylene sorbitan fatty acid ester, a sorbitan fatty acid ester, an alkylene glycol fatty acid ester, a polyoxyalkylene fatty acid ester, a fatty acid ester, a polyoxyalkylene fatty acid ether, a C i6 to C24 fatty acid, a fatty acid mono-, di- or poly-glyceride, a polyoxyalkylene alkyl phenol, an alkyl phenyl ether, a polyoxyethylene polyoxypropylene block copolymer, a fatty amine oxide, a fatty acid alkanolamide, an alkyl cellulose, a carboxyalkyl cellulose, a polyoxyalkylene castor oil derivatives, an alkyl sulfate, an olefin sulfate, an ether sulfate, a monoglyceride sulfate, an alkyl sulfonate, an aryl sulfonate, an olefin sulfonate, an alkyl sulfosuccinate, an aryl sulfosuccinate, or a combination of two or more thereof. In one preferred embodiment, the surfactant is sodium lauryl sulfate. Preferably, the surfactant is present at about 0.5% to about 2% by weight. In one preferred embodiment, the surfactant is present at about 1% by weight.
In certain preferred embodiments, the binder is hydroxyethyl cellulose, hydroxypropyl cellulose, pregelatinized starch, povidone, or a combination of two or more thereof. In one preferred embodiment, the binder is pregelatinized starch. In certain preferred embodiments, the disintegrant is sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof. In one preferred embodiment, the disingegrant is croscarmellose sodium.
In certain preferred embodiments, the diluent is lactose, microcrystalline cellulose, mannitol, dibasic calcium phosphate, starch, or a combination of two or more thereof. In one preferred embodiment, the diluent is lactose.
In certain preferred embodiments, the surfactant is sodium lauryl sulfate and the pharmaceutical formulation further comprises pregelatinized starch, crocarmellose sodium, and lactose.
In certain embodiments, the pharmaceutical formulation further comprises an extragranular excipient. In one preferred embodiment, the extragranular excipient is a disintegrant, a flow aid, a lubricant, or a combination of two or more thereof.
In certain preferred embodiments, the pharmaceutical formulation further comprises an extragranular disintegrant which is sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof. In one preferred embodiment, the extragranular disintegrant is croscarmellose sodium. In another preferred embodiment, the pharmaceutical formulation further comprises an extragranular flow aid which is silicon dioxide, talc, or a combination thereof. In certain preferred embodiments, the extragranular flow aid is silicon dioxide. In still other preferred embodiments, the pharmaceutical formulation further comprises an extragranular lubricant which is magnesium stearate, calcium stearate, glyceryl monostearate, sodium stearyl fumarate, stearic acid, talc, or a combination of two or more thereof. In certain preferred embodiments, the extragranular lubricant is magnesium stearate.
In one preferred embodiment, the pharmaceutical formulation is contained in a capsule. In one preferred embodiment, the VLA-4 antagonist comprises 50% by weight of the pharmaceutical formulation.
In another aspect the present invention provides pharmaceutical formulations comprising a granulate of Compound IA which provides a mean steady-state AUC of Compound IA that is about 45 microgram-hr/ml when administered at a 200 mg dose of Compound IA once-a-day to a patient. The present invention also encompasses pharmaceutical formulations which are similarly bioavailable such that the relative mean steady-state AUC of Compound IA is within 80% to 125% of 45 microgram-hr/ml, that is within the range from about 36 microgram-hr/ml to about 57 microgram-hr/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient. In one embodiment, the pharmaceutical formulation provides a mean steady-state AUC of Compound IA which is at least 80% of 45 microgram-hr/ml, that is at least 36 microgram-hr/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient. In one embodiment, the pharmaceutical formulation provides a mean steady-state AUC of Compound IA which is at least 45 microgram-hr/ml when administered at a 200 mg dose of Compound IA once- a-day to a patient. In another aspect the present invention provides pharmaceutical formulations comprising a granulate of Compound IA which provides a mean steady-state Cmin of Compound IA that is at least 0.2 microgram/ml when administered at a 200 mg dose of Compound IA once-a-day to a patient. In one embodiment, the pharmaceutical formulation further comprises sodium lauryl sulfate, pregelatinized starch, crocarmellose sodium, and lactose.
In one embodiment, the pharmaceutical formulation comprising a granulate of Compound IA provides a median Tmax that is in the range from about 1 hour to about 4 hours when administered at a 200 mg dose of Compound IA once-a-day to a patient. In one embodiment, the pharmaceutical formulation comprising a granulate of Compound IA provides a mean steady-state Cmax of Compound IA that is about 6 microgram/ml when administered at a 200 mg dose of Compound IA once-a-day to a patient. The present invention also encompasses pharmaceutical formulations which are similarly bioavailable such that the relative mean steady-state Cmax of Compound IA is within 80% to 125% of 6 microgram/ml, that is within the range from about 4.8 microgram/ml to about 7.6 microgram/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient. In a certain embodiment, the pharmaceutical formulation comprising a granulate of Compound IA provides a mean steady-state Cmax of Compound IA which is at least 80% of 6 microgram/ml, that is at least 4.8 microgram/ml, when administered at a 200 mg dose of Compound IA once-a-day to a patient. In one embodiment, the pharmaceutical formulation provides a mean steady-state Cmax of Compound IA which is at least 6 microgram/ml when administered at a 200 mg dose of Compound IA once-a- day to a patient.
Notably, steady state exposure of Compound IA was attained by Day 11 when the pharmaceutical formulations of the present invention were given once-a-day.
In one embodiment, a pharmaceutical formulation of the present invention comprising Compound IA provides a mean steady-state AUC of Compound IA that is at least 36 microgram hr/mL when administered at a dose that is 200 mg once-a-day to a patient and assessed as described infra herein (See Examples).
In an embodiment, a pharmaceutical formulation of the present invention provides a mean steady-state Cmax of Compound IA that is at least 4.8 microgram/mL as measured as described infra herein (See Examples).
In an embodiment, a pharmaceutical formulation of the present invention comprising a granulate of Compound IA provides a mean steady-state Cmnin of Compound IA that is at least 0.2 microgram hr/mL when administered at a dose that is 200 mg once-a-day to a patient and measured as described infra herein (see Examples). In another aspect, the present invention provides methods for the treatment or prevention or inhibition of progression of an inflammatory disease, which comprises administering to a patient in need thereof a therapeutically effective amount of any of the pharmaceutical formulations of the present invention. In one preferred embodiment, the inflammatory disease is multiple sclerosis.
DETAILED DESCRIPTION OF THE DRAWINGS
Figure 1 is a graph of the lymphocyte change from time-matched baseline over a time period up to 12 hours post-dose in subjects given a single dose of either placebo, or Compound IA that is 50 mg, 150 mg, 300 mg, 600 mg, or 1000 mg.
Figure 2 is a bar graph of the lymphocyte change from time-matched baseline at various time points (specifically, 4 hours, maximum, and day average) post-dose in subjects given a single dose of either placebo, or Compound IA that is 50 mg, 150 mg, 300 mg, 600 mg, or 1000 mg.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as those commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. The materials, methods and examples are illustrative only, and are not intended to be limiting. All publications, patents and other documents mentioned herein are incorporated by reference in their entirety.
As used herein, the term "AUC" is the area under the plasma concentration-time curve from time zero to a certain time period of the sample. For example, AUC (4h) means the area under the plasma concentration-time curve from time zero to 4 hours.
As used herein, the term "CL/F" is the apparent total clearance of the drug from plasma after oral administration. CL/F is calculated by dividing the dose administered by the AUC.
The term "treating" or "treatment" is intended to mean mitigating or alleviating the symptoms of the recited condition, disease or disorder in a mammal such as a human.
As used herein, the term "tl/2" refers to the half-life of the drug. PHARMACEUTICAL FORMULATIONS
In most preferred embodiments, the present invention provides pharmaceutical formulations of VLA-4 antagonist Compound I or a pharmaceutically acceptable salt thereof suitable for oral administration that meet the aforementioned need for acceptable drug loading, dissolution, stability, and bioavailability of Compound I. In preferred embodiments, the VLA-4
antagonist is Compound IA. The pharmaceutical formulations of the present invention comprising a surfactant enhance the dissolution of VLA-4 antagonist Compound IA at pH 7 relative to a pharmaceutical formulation lacking a surfactant. Likewise, the pharmaceutical formulations of the present invention provide favorable dissolution profiles and bioavailability of the Compound IA.
Compound I can be prepared according to Example 25, at columns 75-76, of U.S. Pat. No. 6,972,296; also see Scheme 25 at columns 41-42, incorporated herein by reference. Pharmaceutically acceptable salts can be formed with an appropriate base. The mono-sodium salt of Compound I, herein referred to as Compound IA, has the following structure:
and can be formed from Compound I by reacting the free acid with an appropriate base, e.g., sodium hydroxide. The development of Compound IA necessitates overcoming several physicochemical and pharmacokinetic challenges. Notably, Compound IA has low solubility at pH = 6.8 (< 1 microgram/mL). In addition, according to the Biopharmaceutics Classification System, Compound IA is a Class IV compound, that is, a compound having low solubility and low permeability. Consequently, Compound IA has relatively low bioavailability. Furthermore, Compound IA has very low bulk density (0.08 g/cc) and very poor flow properties as the crystalline form is needle-like and thus not readily micronized on a commercial scale. Thus, pharmaceutical formulations that are sufficiently stable and provide enhanced bioavailability of Compound IA suitable for commercialization are needed.
In one embodiment, the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof and a surfactant. In certain embodiments, the granulate further comprises a binder, a disintegrant, a diluent, or a combination of two or more thereof. In certain preferred embodiments, the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation. In another embodiment, the present invention provides pharmaceutical formulations suitable for oral administration comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof, a surfactant, a binder, a disintegrant,
and a diluent, wherein the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation. Preferably, the VLA-4 antagonist is Compound IA.
Binders are generally used to impart cohesive qualities to the solid dosage form. Examples of binders include, without limitation, hydroxyethyl cellulose, hydroxypropyl cellulose, pregelatinized starch, povidone, or a combination of two or more thereof. Binders may make up about 1% to about 15% by weight. In certain preferred embodiments, the binder is present at about 15% by weight. In one preferred embodiment, the binder is pregelatinized starch and is present in an amount that is at least 15% by weight.
Examples of disintegrants include, without limitation, sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof. Disintegrants may make up about 1% to 25% by weight. In one preferred embodiment, the disintegrant is croscarmellose sodium present at about 15% by weight. In another preferred embodiment, the disintegrant is sodium starch glycolate present in an amount that is up to about 10% by weight. In yet another preferred embodiment, the disintegrant is crospovidone present in an amount that is up to about 5% by weight.
Suitable surfactants for use in the present invention include nonionic surfactants, anionic surfactants, or a combination thereof. Examples of nonionic surfactants include, without limitation, polyoxyalkylene sorbitan fatty acid esters, sorbitan fatty acid esters, alkylene glycol fatty acid esters, polyoxyalkylene fatty acid esters, fatty acid esters, polyoxyalkylene fatty acid ethers, Ci6 to C24 fatty acids, fatty acid mono-, di- or poly-glycerides, polyoxyalkylene alkyl phenols, alkyl phenyl ethers, polyoxyethylene polyoxypropylene block copolymers, fatty amine oxides, fatty acid alkanolamides, alkyl cellulose, carboxyalkyl cellulose, and polyoxyalkylene castor oil derivatives. For example, nonionic surfactants include polyoxyethylene(20) sorbitan monolaurate (Tween® 20), polyoxyethylene(4) sorbitan monolaurate (Tween® 21), polyoxyethylene(20) sorbitan monopalmitate (Tween® 40), polyoxyethylene(20) sorbitan monostearate (Tween® 60), polyoxyethylene(20) sorbitan tristearate (Tween® 65), polyoxyethylene(20) sorbitan monooleate (Tween® 80 or polysorbate 80), or polyoxyethylene(20) sorbitan trioleate (Tween®85), lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, glycerol monooleate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, sorbitan laurate, sorbitan oleate, sorbitan palmitate, sorbitan stearate, sorbitan tristearate, sorbitan sesquioleate, sorbitan trioleate, sorbitan isostearate, propylene glycol monostearate, polyoxyethylene monostearate, polyoxyethylene distearate, glyceryl monostearate, polyoxyethylene lauryl ether, polyoxyethylene cetyl ether, polyoxyethylene stearyl ether, polyoxyethylene oleyl ether, palmitic acid, stearic acid, oleic acid, ethyl oleate,
isopropyl myristate, sodium palmitate, sodium stearate, sodium oleate, nonylphenol polyethoxyethanols, tributylphenoxy-polyethoxyethanol, octylphenoxy-polyethoxyethanol, polyoxyethylene glycerol triricinoleate or polyoxyl 35 castor oil (Cremophor® EL, BASF Corp.), polyoxyethylene glycerol oxystearate (Cremophor® RH 40), polyethylene glycol 60 hydrogenated castor oil (Cremophor® RH 60), Poloxamer® 124, Poloxamer® 188, Poloxamer® 237,
Poloxamer®3 88, Poloxamer® 407 (BASF Wyandotte Corp.); methylcellulose and carboxymethyl cellulose. Examples of anionic surfactants include, without limitation, alkyl sulfates, olefin sulfates, ether sulfates, monoglyceride sulfates, alkyl sulfonates, aryl sulfonates, olefin sulfonates, alkyl sulfosuccinates, and aryl sulfosuccinates. For example, anionic surfactants include sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate. In one preferred embodiment, the surfactant is a polyoxyalkylene sorbitan fatty acid ester, a sorbitan fatty acid ester, an alkylene glycol fatty acid ester, a polyoxyalkylene fatty acid ester, a fatty acid ester, a polyoxyalkylene fatty acid ether, a C16 to C24 fatty acid, a fatty acid mono-, di- or poly-glyceride, a polyoxyalkylene alkyl phenol, an alkyl phenyl ether, a polyoxyethylene polyoxypropylene block copolymer, a fatty amine oxide, a fatty acid alkanolamide, an alkyl cellulose, a carboxyalkyl cellulose, a polyoxyalkylene castor oil derivative, an alkyl sulfate, an olefin sulfate, an ether sulfate, a monoglyceride sulfate, an alkyl sulfonate, an aryl sulfonate, an olefin sulfonate, an alkyl sulfosuccinate, an aryl sulfosuccinate, or a combination of two or more thereof. In a preferred embodiment, the surfactant is sodium lauryl sulfate. Surfactants may make up about 0.5% to about 2% by weight. In one preferred embodiment, the surfactant is present at about 1% by weight. In one preferred embodiment, the surfactant is sodium lauryl sulfate present at about 1% by weight.
Examples of diluents include, without limitation, lactose, microcrystalline cellulose, mannitol, dibasic calcium phosphate, starch, or a combination of two or more thereof. In one preferred embodiment, the diluent is lactose. In other preferred embodiments, the diluent is microcrystalline cellulose in combination with lactose.
The pharmaceutical formulation of the present invention may further comprise an extragranular excipient which is a glidant, lubricant, disintegrant, or a combination of two or more thereof.
Examples of glidants, also known as flow agents, include, without limitation, silicon dioxide (including colloidal silicon dioxide), talc, or a combination thereof. Glidants may make up about 0.1% to about 0.5% by weight. In certain preferred embodiments, the glidant is present at about 0.5% by weight. In one preferred embodiment, the glidant is silicon dioxide.
Examples of lubricants include, without limitation, magnesium stearate, calcium stearate, glyceryl monostearate, sodium stearyl fiimarate, stearic acid, talc, or a combination of two or more
thereof. Lubricants may make up about 0.1% to about 1%. In certain preferred embodiments, the lubricant is present at about 0.5% by weight. In one preferred embodiment, the lubricant is magnesium stearate.
Suitable oral pharmaceutical formulations include, but are not limited to, capsules, tablets, granules, powders, and unit dose packets. In one embodiment, the oral pharmaceutical formulation is a capsule. In another embodiment, the oral pharmaceutical formulation is a tablet.
METHODS OF PREPARING PHARMACEUTICAL FORMULA TIONS
Pharmaceutical formulations of the present invention can be prepared by a variety of suitable wet granulation, including high shear wet granulation, techniques. For example, such pharmaceutical formulations can be prepared using the process diagramed in Scheme I below:
Scheme I
(a) Dry-blend:
(i) Compound I or a pharmaceutically acceptable salt thereof; and (ii) a granular excipient (e.g., surfactant, diluent, disintegrant, binder) to provide a first dry-blended powder
(b) Agglomerate the first dry-blended powder from Step "a" in a high shear granulator using a granulating solution comprising water and/or an organic solvent
(c) Form a wet milled granulate by wet-milling the agglomerate prepared in Step "b"
(d) Form a dried granulate by drying the wet milled granulate from Step "c
(e) Dry mill the dried granulate from Step "d"
(f)
Note that the binder may be added to the dry-blend in step "a" or dissolved in the granulating solution used in step "b". Also note that the wet granulate in step "d" can be dried in any one of the following ways: a) within the high-shear granulator, using microwave power as the source of energy to evaporate the granulating liquid and vacuum to remove the vapor; b) in a fluidized bed dryer using heated air to dry the granules and remove the vapor; or c) in a tray dryer, in which the wet granules are spread on trays in a heated and ventilated drying oven.
METHODS OF TREATMENT ' AND/ OR PREVENTION
Another aspect of the invention provides methods for the treatment and/or prevention of disease states in which VLA-4 is involved in a patient comprising administering to the patient a pharmaceutical formulation according to the present invention. In preferred embodiments, the pharmaceutical formulations comprise Compound I or a pharmaceutically acceptable salt thereof, preferably Compound IA.
Representative diseases which can be treated using the formulations of the present invention include, but are not limited to, atherosclerosis, rheumatoid arthritis, asthma, allergy, multiple sclerosis, lupus, inflammatory bowel disease, graft rejection, contact hypersensitivity, and type I diabetes. In addition to being found on some white blood cells, VLA-4 is also found on various cancer cells, including leukemia, melanoma, lymphoma and sarcoma cells. Cell adhesion involving VLA-4 may be involved in the metastasis of certain cancers. Inhibitors of VLA-4 binding may, therefore, also be useful in the treatment of some forms of cancer.
In one embodiment, Compound I or a pharmaceutically acceptable salt thereof, preferably Compound IA, is administered for the treatment of an inflammatory disease. In another embodiment, Compound I or a pharmaceutically acceptable salt thereof, preferably Compound IA, is administered for the prevention or inhibition of an inflammatory disease. In one preferred embodiment, the inflammatory disease is multiple sclerosis.
The amount and frequency of administration of Compound I or a pharmaceutically acceptable salt thereof will be regulated according to the judgment of the attending clinician considering such factors as age, condition, size of the patient as well as severity of the symptoms being treated. A typical recommended daily dosage regimen for the treatment of an inflammatory disease can range from about 25 mg/day to about 1000 mg/day. In certain preferred embodiments, the recommended daily dosage regimen for the treatment of an inflammatory disease can range from 25 mg/day to 400 mg/day. In one preferred embodiment, the daily dosage regimen for the treatment of an inflammatory disease is about 400 mg/day administered orally in a single daily dose.
Similarly, a typical recommended daily dosage regimen for the prevention or inhibition of an inflammatory disease can range from about 25 mg/day to about 1000 mg/day. In certain preferred embodiments, the recommended daily dosage regimen for the prevention or inhibition of an inflammatory disease can range from 25 mg/day to 400 mg/day. In one preferred embodiment, the daily dosage regimen for the prevention or inhibition of an inflammatory disease is about 400 mg/day administered orally in a single daily dose.
Other features and embodiments of the invention will become apparent by the following examples which are given for illustration of the invention rather than limiting its intended scope.
EXAMPLES
P KEP ARA TION OF EXEMPLARY FORMULA TIONS
Exemplary formulations of the present invention are detailed in Tables IA and IB.
Table IB: Comparative formulations of Compound IA
Exemplary formulations A-D were made by wet granulation. Specifically, the formulations were manufactured by first wet-granulating Compound IA and the granular excipients to make a granulate. The granules were then dried, mixed with the extragranular excipients and then encapsulated in size 0 capsules. The outline of the method of manufacture is presented below.
• Weigh Compound IA (active pharmaceutical ingredient (API)) and granular excipients including binder. • Pass granular excipients through an 18 mesh sieve.
• Add screened granular excipients and API to granulator (e.g., Fielder Granulator) alternating between granular excipients and API.
• Mix for about 5 minutes dry.
• Add water slowly while mixing until well blended. • Pass the wet granulate through a Comil 4 mesh screen.
• Dry the granulate on the Glatt fluid bed drier until a loss-on-drying (i.e., moisture content) of 1-3% is obtained.
• Pass the dried granulate through a Comil screen of about 16 mesh.
• Blend the dried granulation along with pre-screened extragranular excipients (other than lubricant, e.g., magnesium stearate) in an appropriate sized blender for about 15 minutes.
• Add the screened lubricant, e.g., magnesium stearate, and blend for another 3 minutes.
• Encapsulate the final blend into size 0 capsules on a Bosch capsule filling machine using a 16 mm dosing disk.
In contrast, comparative formulations E and F were made by dry-blending Compound IA with the excipients to form a homogenous mixture and then encapsulating the mixture in size 0 capsules.
STABILITY DATA FOR EXEMPLARY FORMULATIONS
Stability studies were conducted on capsules of exemplary formulation D. Specifically, capsules were packaged as 10 capsules per bottle in induction-sealed HDPE bottle with child- resistant caps. Samples were stored at 25 0C and 60% relative humidity up to 1 year or 40 0C and 75% relative humidity up to 6 months. Following storage under the aforementioned conditions, samples were pulled at various time points (e.g., 1 month, 3 months, 6 months, 9 months, 12 months) and assessed for moisture content (using volumetric Karl Fischer titration method), label strength, and the presence of Compound IA degradation products.
The presence of Compound IA and determination of degradation products in capsules following storage was assessed using reverse-phase High Performance Liquid Chromatography. In brief, samples for assay and degradation product testing were prepared as follows. Capsules were emptied, contents ground using mortar and pestle; an aliquot of powder weighed and diluted with 1:1 water:85%acetonitrile, then shaken to dissolve drug and subsequently filtered to remove insoluble formulation components. The filtrate was then further diluted with 1:1 water:85% acetonitrile; and the sample analyzed using reverse-phase High Performance Liquid Chromatography. Specifically, an ACE 3 C18 column (4.6 mm x 150 mm, 3.0 μm particles), a gradient elution with mobile phases consisting of acetonitrile, water and 85% phosphoric acid (60:40:0.05, v:v:v). Quantitation of Compound IA was performed by UV detection at 220 nm against an external Compound I reference standard. Notably, the detection limit for the presence of Compound IA degradation products using this assay is less than 0.05%, and the quantitation limit of 0.1%.
Samples stored under both conditions had acceptable moisture contents between 3.6% and 4.7%, as compared to an initial moisture content of 3.6%. Likewise, samples stored under both conditions displayed acceptable label strength between 97.6% and 99.1% that was comparable to the initial label strength of 95.9%. Lastly, the presence of Compound IA degradation products was neither quantifiable up to the 1 year time point examined following storage at 25 0C and 60% relative humidity nor up to the 6 months time point following storage at 40 0C and 75% relative humidity.
These results indicate that Compound IA is stable within the formulation tested for at least
1 year.
DISSOLUTION PROFILE OF EXEMPLARY FORMULATIONS The dissolution profile of Compound IA in exemplary formulations Capsule A and Capsule
B was determined at pH 7 to simulate intestinal pH. In brief, a sample dissolution profile was obtained for the release rate of Compound IA from capsules using a USP-II dissolution apparatus, with paddles operated at 50 rpm. Each formulation was tested in 900 mL of 0.02 M potassium phosphate buffer (pH 7) as dissolution medium. Aliquots of 10 ml were removed periodically (e.g., 15, 30, 45, and 60 minutes) and filtered through a 0.45 micrometer Millipore Millex-HV
PVDF filter. The first 3 mL of filtrate was discarded and a 2.5 mL portion of the remaining filtrate was diluted 1:4 by volume with a mobile phase solution containing acetonitrile:water: 85% phosphoric acid (60:40:0.05, v:v:v). The samples were analyzed by reverse-phase HPLC maintained at 30 0C ± 2 0C utilizing an ACE 3 C18 column (4.6 mm x 150 mm, 3.0 μm particles). Quantitation of Compound IA was performed by UV detection at 265 nm against an external Compound I reference standard. The dissolution profiles of Compound IA in capsules of exemplary formulations A (0% SLS) and B (0.5% SLS) are illustrated in Table 2.
Table 2: Dissolution profile of Compound IA in exemplary formulations A and B
Exemplary formulation B provides enhanced dissolution of Compound IA in the presence of surfactant compared to that of Compound IA in the absence of surfactant. Consequently, the present inventors believe that formulations of Compound I or a salt thereof comprising surfactant will be beneficial to increase the extent of Compound I absorption in the gastrointestinal tract thereby leading to enhanced bioavailability of Compound I.
Similarly, dissolution studies were carried out for capsules of exemplary formulations C and D at pH 5.7. In brief, a sample dissolution profile was obtained for the release rate of Compound IA from capsules using a USP-II dissolution apparatus, with paddles operated at 100 rpm, maintained at 37 0C ± 0.5 0C. Each formulation was tested in 900 mL of 0.02 M ammonium
acetate buffer with 1.5% Cetyl Trimethyl Ammonium Bromide (CTAB) (pH 5.7) as dissolution medium. Aliquots of 10 ml were removed periodically (e.g., 15, 30, 45, and 60 minutes) and filtered through a 0.45 micrometer Millipore Millex-HV PVDF filter. The first 3 mL of filtrate was discarded and a 2.5 mL portion of the remaining filtrate was diluted 1:4 by volume with a mobile phase solution containing acetonitrile:water: 85% phosphoric acid (60:40:0.05, v:v:v). The samples were analyzed by reverse-phase HPLC maintained at 30 0C ± 2 0C utilizing an ACE 3 C18 column (4.6 mm x 150 mm, 3.0 μm particles). Quantitation of Compound IA was performed by UV detection at 220 nm against an external Compound I reference standard. The dissolution profiles for Compound IA in capsules= of exemplary formulations C (25 mg per capsule granulation formulation), D (200 mg per capsule granulation formulation), and E (5 mg per capsule dry- blended formulation) are illustrated in Tables 3, 4, and 5 respectively.
The dissolution profiles of capsules of exemplary formulations C, D, and comparative formulation E demonstrated 90% or greater release of Compound IA within 30 minutes. As such, all three pharmaceutical formulations provide favorable dissolution profiles for release of Compound IA.
Further dissolution studies were carried out for capsules of exemplary formulation D at pH 7 (as described above) to ascertain whether the dissolution profile displayed uniformity across the
batch produced. Specifically, a sample from the beginning, middle, and end of the batch were examined. The resultant dissolution profiles of this study are illustrated in Table 6.
The dissolution profile obtained from samples at the beginning, middle, and end of the batch reflected adequate dissolution, and comparison of the 60-minutes percent-dissolution indicate acceptable content uniformity across the batch.
CLINICAL STUDIES OF COMPOUND IA PHARMACEUTICAL FORMULATIONS Rising single-dose assessment in healthy adult subjects
Objective: (i) Evaluate the safety and tolerability of Compound IA when administered orally at single doses of 50, 150, 300, 600, and 1000 mg. (ii) Determine the single dose pharmacokinetic profile of Compound IA in healthy volunteers. Explore the pharmacological activity of Compound IA. Assess food effect on the exposure to Compound IA in a preliminary manner.
Methodology: Single-center, randomized, third-party blind (within dose-level), placebo- controlled, rising, single-dose study conducted in conformance with Good Clinical Practices. Five dose groups were administered a single dose of 50, 150, 300, 600, and 1000 mg of Compound IA as described below. Within each dose group, six subjects were to receive the active drug, and two subjects were to receive placebo. The first dose level was 50 mg. Each of the higher doses in the progression was not administered until the safety and tolerability data of the previously administered lower dose, up to 72 hours postdose, had been evaluated. The decision to progress to high dose levels was determined by the review of safety laboratory tests, vital signs, electrocardiograms (ECGs), and adverse events and agreed to by sponsor representatives and the principal investigator. Multiple time-matched ECGs were obtained on Days -1 and 1, and these ECGs were machine read with over-reading by the investigator. For pharmacokinetic analyses, blood samples were collected predose (0 hour), 0.25, 0.5, 0.75, 1, 2, 3, 4, 6, 8, 12, 24, 36, 48, and 72 hours postdose. For metabolite profiling, blood samples were collected predose (0 hour), 2 and
6 hours postdose, and urine was collected from 0 to 24 hours postdose. Whole blood was collected predose (0 hour), 2 and 6 hours postdose to explore the effect of Compound IA on the expression of cell surface markers and the relative abundance of lymphocyte subtypes in peripheral blood using FACS analyses. Multiple blood samples were collected on Days -1 and 1 in a time-matched manner to determine the peripheral lymphocyte counts.
Food Effect on Pharmacokinetics of Compound IA: Subjects receiving 600 mg Compound IA or placebo returned to the study site after 7 days of washout to receive a second dose of Compound IA or placebo within 5 minutes after consuming a standardized high-fat breakfast. Blood samples were collected predose (0 hour), 0.25, 0.5, 0.75, 1, 2, 3, 4, 6, 8, 12, 24, 36, 48, and 72 hours postdose for pharmacokinetic analyses.
Number of Subjects: A total of 40 healthy volunteers participated in this study. Thirty subjects (6 at each dose level) received a single dose of Compound IA at a dose of 50, 150, 300, 600, or 1000 mg. Ten subjects (2 at each dose level) received a single dose of matching placebo. The determination of sample size was based on empirical rather than statistical consideration. Diagnosis and Criteria for Inclusion: A total of 40 healthy adult subjects (16 men and 24 women) between the ages of 18 and 44 years were treated. All of the subjects were Caucasian. Body weights ranged from 50.2 kg to 89.6 kg, and heights ranged from 151 cm to 187 cm. The subjects BMI ranged from 18.9 to 27.1. All but one subject completed the study. A subject who was administered a 1000 mg dose of Compound IA was discontinued from the study due to adverse events (nausea, vomiting). This subject vomited immediately following dosing and all of the capsules were intact in the vomitus.
Test Product, Dose, Mode of Administration: Compound IA, 50 mg capsules (specifically, capsules of comparative formulation F described in Table IB above) and 200 mg capsules (specifically, capsules of exemplary formulation D described in Table IA above). Doses administered were: 1 x 50 mg (total 50 mg), 3 x 50 mg (total 150 mg), 6 x 50 mg (total 300 mg), 3 x 200 mg (total 600 mg), and 5 x 200 mg (total 1000 mg). The study drug was administered orally.
Reference Therapy, Dose, Mode of Administration: Matching placebo 50 mg and 200 mg capsules. The number of placebo capsules was the same as the number of Compound IA capsules required for each treatment (dose-level) group. The placebo capsule(s) were administered orally.
Duration of Treatment: After a screening phase of up to 3 weeks, subjects were administered a single dose of Compound IA or matching placebo and were followed for 3 days
after dosing. Subjects from the 600 mg dose group (Compound IA or matching placebo) returned to the study site, after a washout period of 7 days, to receive a second dose of Compound IA or matching placebo within 5 minutes after consuming a standardized high-fat meal.
Criteria for Evaluation: Safety and tolerability of Compound IA were assessed by collecting adverse events, vital signs, ECGs, physical examinations, and safety laboratory tests. Adverse event, laboratory test results, and ECGs have been reviewed for signs of any serious adverse events.
Pharmacokinetics: The following pharmacokinetic parameters were to be determined. Cmax, Tmax, AUC, K, tιΛ, Vd/F, and CL/F. Pharmacodynamics: The effect of Compound IA on lymphocyte count, the expression of cell surface markers, and the relative abundance of lymphocyte subtypes in peripheral blood were also explored by conventional hematology and FACS analyses.
Safety: Adverse events were tabulated by treatment. ECG parameters, vital signs, and safety laboratory tests were summarized using descriptive statistics. Pharmacokinetics: Multiple plasma samples were collected for pharmacokinetic analyses, but the final pharmacokinetic parameters were not reported. Based on preclinical animal data, no significant amount of acyl glucuronide metabolite was anticipated in humans; therefore, the plasma samples were not collected under optimal condition (i.e., acidified condition) to accurately quantify the circulating acyl glucuronide metabolite. However, metabolite profiling from this study suggested an appreciable amount of circulating acyl glucuronide metabolite, which could be readily converted to parent drug during the process of sample handling. The plasma concentration determined under the non-optimized sampling/storage condition could lead to an overestimation of circulating parent drug and an underestimation of acyl glucuronide metabolite. Despite the above- mentioned limitations, the following conclusion about the pharmacokinetics of Compound IA can be drawn: Compound IA pharmaceutical formulations described herein provided sufficient bioavailability following oral administration at a single dose >150 mg to elicit a modest increase in peripheral circulating lymphocytes. The exposure appeared to increase as dose increased, and a high-fat meal appeared to increase exposure to Compound IA. An appreciable amount of acyl glucuronide metabolite was observed in plasma. Pharmacodynamics: Lymphocyte counts from Day -1 and up to 24 hours postdose were listed and the change from time-match baseline count were summarized using descriptive statistics. Both florescence intensity (CD62L, CD49d, and CD29) for subtypes of lymphocytes and the relative abundance of subtypes of lymphocytes and monocytes were also reported.
Descriptive statistics for the change of mean fluorescence intensity from predose (Day 1) for CD62L, CD49d, and CD29 for subtypes of lymphocytes were provided. Results from FACS analyses were also listed for each subject. The relative abundance (expressed as a percentage) of subtypes of lymphocytes and monocytes and its change from predose baseline were summarized using descriptive statistics, the results of which are illustrated graphically in Figures 1 and 2. As reflected in Figures 1 and 2, Compound IA administered at a single dose >150 mg results in a modest increase in peripheral circulating lymphocytes.
Conclusions: Based on this rising single-dose study of Compound IA pharmaceutical formulations, the following conclusions were drawn: • Compound IA is safe and well tolerated at single doses up to and including 1000 mg.
• Compound IA pharmaceutical formulations described herein provided sufficient bioavailability following oral administration at a single dose >150 mg to elicit a modest increase in peripheral circulating lymphocytes.
Rising multiple-dose assessment in healthy adult subjects
Objectives: (i) Evaluate the safety and tolerability of Compound IA when administered orally once daily for 14 days at doses of 200, 400, and 800 mg. (ii) Determine the single dose and multiple-dose pharmacokinetic profile of Compound IA in healthy volunteers. Explore the pharmacological activities of Compound IA. Methodology: This was a randomized, placebo-controlled, third-party blind (within dose-level), rising multiple-dose study to evaluate the safety, tolerability, and pharmacokinetics of Compound IA in healthy volunteers and in a group of otherwise healthy subjects with known allergy as evidenced by a positive skin-prick test (>5 mm to house dust mite or grass pollen extract). This study consisted of two parts. The first part assessed the safety, tolerability, pharmacokinetics, and pharmacological activity of rising multiple doses of Compound IA. The pharmacological activity of Compound IA was assessed primarily by delayed-type hypersensitivity (DTH) with skin biopsy. Three dose groups were treated for 14 days with once- daily doses of either 200, 400, or 800 mg Compound IA. The second part assessed the safety, tolerability, pharmacokinetics, and pharmacological activity of Compound IA at one dose level (maximum tolerated dose or highest dose from Part 1 of this study) in otherwise healthy subjects with known allergy as evidenced by a positive skin-prick test. In addition to a DTH response, the effect of Compound IA on allergen-induced late cutaneous response was assessed in Part 2. For Part 1 of the study, 8 subjects were randomized to active drug and 4 subjects to placebo within each dose
group. The first dose level was 200 mg. Each of the higher doses in the progression was assessed after the safety and tolerability data from the previously administered lower dose had been evaluated. The decision to progress to higher dose levels was determined following review of safety laboratories, vital signs, electrocardiograms (ECGs) and adverse events Subjects in each cohort were challenged with three different concentrations/dilutions of Candin® (antigen extracts from Candida albicans) for a DTH response at Baseline (Day - 3) and approximately at 1 hour postdose on Day 11. Subjects were vaccinated with hepatitis A on Day 2 and antibody titers determined at Baseline, Day 17 and Day 42. For Part 2 of the study, subjects with a positive skin-prick test to either dust mite or grass pollen were treated with the maximum tolerated dose of Compound IA or matching placebo for 14 days. An intradermal allergen (either house dust mite or rye grass pollen) challenge was performed at Baseline (Day - 2) and approximately at 1 hour postdose on Day 11.
Blood samples were collected into EDTA tubes predose (0 hour) and at 0.5, 1, 2, 3. 4. 6, 8, 12, and 24 hours postdose on Days 1 and 14, predose on Days 11, 12, and 13, and at 36 hours (Day 15), 48 hours (Day 16), and 72 hours (Day 17) postdose on Day 14 for Compound IA analyses. All plasma samples were analyzed for Compound IA using a validated liquid chromatography/tandem mass spectrometry (LC/MS/MS) assay method with a LLOQ of 25 ng/mL. Blood samples were collected predose (0 hour) and 2, 6 and 24 hours postdose on Day 1 and predose (0 hour) and 2, 6, and 24 hours postdose on Day 14. for metabolite profiling. Urine was collected predose on Day 1 and from 0 to 24 hours postdose on Days 1 and 14, for metabolite profiling including analysis of Compound IA.
Number of Subjects: A total of 47 healthy adult subjects (30 men and 17 women) between the ages of 18 and 48 years of age (mean, 25.4 years of age) were treated. Thirty-nine (83%) of the subjects were white; 7 (15%) were Asians; and one was multi-racial. Two subjects were withdrawn from the study; one was due to lab abnormalities and the other was due to a personal reason. Test Product, Dose, Mode of Administration: Compound IA, 200 mg capsules
(specifically, capsules of exemplary formulation D described in Table IA above). Doses administered included: 1 x 200 mg, 2 x 200 mg. and 4 x 200 mg.
Reference Product, Dose, Mode of Administration: Matching placebo capsules. The number of placebo capsules were the same as Compound IA for each treatment group. Duration of Treatment: Subjects were treated with multiple doses of Compound IA or matching placebo (14 doses, once per day) and were followed for 3 days as inpatients after the end of treatment. The subjects were followed up as outpatients until 28 days post-last-dose of Compound IA or matching placebo.
Criteria for Evaluation Primary Endpoints: Safety and tolerability of Compound IA were assessed by collecting adverse events, vital signs, ECGs, physical examinations, and safety laboratory tests.
Criteria for Evaluation Secondary Endpoints: 1) The following pharmacokinetic parameters were determined: Cmax, Tmax, AUC, K, X1A, Vd/F, CI/F, and accumulation factor. 2) The following pharmacodynamic endpoints were evaluated: the DTH score and numbers of T cell infiltrate in the skin biopsy; peripheral lymphocyte counts, the expression of cell surface markers, and the relative abundance of lymphocyte subtypes determined by FACS analysis in peripheral blood. Scores of allergen-induced late cutaneous response and eosinophil abundance in the skin biopsy were determined. Serum antibody titers to hepatitis A were assessed at different times following vaccination. In addition, plasma JC virus was analyzed using PCR assay.
Statistical Methods Pharmacokinetics: Plasma Compound IA concentrations and pharmacokinetic parameters were listed and summarized using descriptive statistics. The primary pharmacokinetic parameters for Compound IA were the AUC and Cmax values. Log-transformed AUC and Cmax were statistically analyzed using a one-way analyses of variance model. Ninety percent confidence interval estimates of the mean concentrations and pharmacokinetic parameters for each dose were computed. The dose normalized Cmax and AUC were analyzed in a similar fashion and 90% confidence intervals for difference were computed for a preliminary assessment of dose proportionality. Trough concentrations at Days 11, 12, 13, 14, and 15 were used to assess steady-state for each dose level.
Pharmacokinetics: Following oral administration, Compound IA was absorbed with a median Tmax of 3 hours. Steady-state concentration of Compound IA was achieved by Day 11. Pharmacokinetics parameters of Compound IA and its acyl glucuronide metabolite, Compound IB, from the rising multiple-dose study are shown in Table 7 and Table 8.
Compound IA had a terminal half-life of approximately 10 to 14 hours. There was a dose-related, but apparently less than dose-proportional, increase of exposure (Table 9). Both Cmax and AUC increased when the dose was increased from 200 to 800 mg. Compound IB plus its isomers resulting from acyl migration and/or, perhaps, anomerization, were observed in plasma (Table 8), and the ratio of acyl glucuronide to parent drug in plasma was approximately 0.3 to 0.4. The elimination half-life of Compound IB paralleled that of Compound IA, suggesting that the
elimination of Compound IB was formation rate-limited. The variability of pharmacokinetics is mild to moderate.
Statistical Methods Pharmacodynamics: Lymphocyte counts from Day -1 and at different times postdose were listed and the change from time-matched baseline counts was summarized using descriptive statistics. Results from FACS analyses were listed for each subject. Both fluorescence intensity (CD62L, CD49d, and CD29) for subtypes of lymphocytes and the relative abundance of subtypes of lymphocytes and monocytes were reported. Descriptive statistics for the change of mean fluorescence intensity from predose (Day 1) for CD62L, CD49d, and CD29 for subtypes of lymphocytes were provided. The relative abundance (expressed as a percentage) of subtypes of lymphocytes and monocytes and its change from predose baseline were summarized using descriptive statistics. Skin tissue lymphocytes from biopsy of the DTH test were quantified and descriptive statistics were provided. According to the protocol, eosinophils from biopsy of allergen induced late cutaneous responses were to be quantified, but the analysis was not performed due to lack of effect of Compound IA on induction of late cutaneous response.
Pharmacodynamics: To assess the effect of Compound IA on peripheral lymphocyte counts, multiple blood samples were collected both prior to (Day -1) and at steady state (on Day 14) exposures to Compound IA. As reflected in Table 10, Compound IA caused a modest increase on peripheral lymphocyte counts.
Once daily administration of Compound IA at a dose up to 800 mg for 14 days had no apparent effect on the subpopulation of lymphocytes and cell surface markers such as CD29, CD49d, and CD62L. Even though Compound IA appeared to cause a subtle increase of CD34 positive cells in the circulating blood, these data should be interpreted with caution due to small changes (mean 11% to 30% increase at 6 hours postdose on Day 14 in Compound IA-treated
subjects vs a mean 17% decrease in placebo-treated subjects) and small sample size and variability in data.
The potential effect of Compound IA on cellular and humoral immunity was assessed using the DTH response and antibody production to hepatitis A vaccination in this rising multiple-dose study. The DTH response to Candida albicans (Candin®) were assessed either before or following 11 days of Compound IA. Compound IA at doses up to 800 mg had no significant effect on the DTH response. Immunohistochemical analysis did not reveal any apparent effect of Compound IA on CD29 and/or CD49d expressing lymphocytes from tissue biopsy of Candin-induced DTH response. The potential effect on allergen-induced late cutaneous response was also assessed in a small number of subjects at the 800 mg dose level; no apparent effect was observed at this dose level. In addition, no apparent effect on antibody production against hepatitis A vaccination was observed with treatment of Compound IA at doses up to 800 mg.
Conclusions: Based on this rising multiple-dose study of Compound IA pharmaceutical formulations, the following conclusions were drawn: • Oral administration of Compound IA at daily doses up to 800 mg for 14 days was safe and generally well tolerated.
• Compound IA had no clinically significant effect on blood pressure, pulse rate, or ECGs.
• Compound IA was not associated with clinically significant effects on safety laboratory tests, including liver function tests, electrolytes, and hematology. • Compound IA pharmaceutical formulations described herein provided sufficient bioavailability following oral administration at a single dose to elicit modest lymphocytosis.
• Compound IA was orally bioavailable and eliminated with a half-life of 10 to 14 hours.
• Exposure to Compound IA increased as the dose of Compound IA increased.
• Acyl glucuronide metabolite (Compound IB) was observed as a major circulating metabolite of Compound IA and was eliminated in parallel with its parent drug.
The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims.
Claims
Claims:
1 A pharmaceutical formulation comprising a granulate comprising a VLA-4 antagonist that is Compound I or a pharmaceutically acceptable salt thereof and a surfactant, wherein the formulation is suitable for oral administration.
2. The pharmaceutical formulation of claim 1 wherein the granulate further comprises a binder, a disintegrant, a diluent, or a combination of two or more thereof.
3. The pharmaceutical formulation of claim 1, wherein the VLA-4 antagonist comprises about 5% to 50% by weight of the pharmaceutical formulation.
4. The pharmaceutical formulation of claim 1, wherein the VLA-4 antagonist is Compound IA.
5. The pharmaceutical formulation of claim 1, wherein the surfactant is a polyoxyalkylene sorbitan fatty acid ester, a sorbitan fatty acid ester, an alkylene glycol fatty acid ester, a polyoxyalkylene fatty acid ester, a fatty acid ester, a polyoxyalkylene fatty acid ether, a C16 TO C24 fatty acid, a fatty acid mono-, di- or poly-glyceride, a polyoxyalkylene alkyl phenol, an alkyl phenyl ether, a polyoxyethylene polyoxypropylene block copolymer, a fatty amine oxide, a fatty acid alkanolamide, an alkyl cellulose, a carboxyalkyl cellulose, a polyoxyalkylene castor oil derivatives, an alkyl sulfate, an olefin sulfate, an ether sulfate, a monoglyceride sulfate, an alkyl sulfonate, an aryl sulfonate, an olefin sulfonate, an alkyl sulfosuccinate, an aryl sulfosuccinate, or a combination of two or more thereof.
6. The pharmaceutical formulation of claim 1, wherein the surfactant is sodium lauryl sulfate.
7. The pharmaceutical formulation of claim 1, wherein the surfactant is present at about 0.5% to about 2% by weight.
8. The pharmaceutical formulation of claim 2, wherein the binder is hydroxyethyl cellulose, hydroxypropyl cellulose, povidone, prege latinized starch, or a combination of two or more thereof.
9. The pharmaceutical formulation of claim 2, wherein the binder is pregelatinized starch.
10. The pharmaceutical formulation of claim 2, wherein the disintegrant is sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof.
1 1. The pharmaceutical formulation of claim 2, wherein the disintegrant is croscarmellose sodium.
12. The pharmaceutical formulation of claim 2, wherein the diluent is lactose, microcrystalline cellulose, mannitol, dibasic calcium phosphate, starch, or a combination of two or more thereof.
13. The pharmaceutical formulation of claim 2, wherein the diluent is lactose.
14. The pharmaceutical formulation of claim 1, further comprising an extragranular excipient.
15. The pharmaceutical formulation of claim 1, further comprising an extragranular disintegrant which is sodium starch glycolate, crospovidone, croscarmellose sodium, or a combination of two or more thereof.
16. The pharmaceutical formulation of claim 1, further comprising an extragranular disintegrant which is croscarmellose sodium.
17. The pharmaceutical formulation of claim 1, further comprising an extragranular flow aid which is silicon dioxide, talc, or a combination thereof.
18. The pharmaceutical formulation of claim 1, further comprising an extragranular flow aid which is silicon dioxide.
19. The pharmaceutical formulation of claim 1, further comprising an extragranular lubricant which is magnesium stearate, calcium stearate, glyceryl monostearate, sodium stearyl fumarate, stearic acid, talc, or a combination of two or more thereof.
20. The pharmaceutical formulation of claim 1, further comprising an extragranular lubricant which is magnesium stearate.
21. The pharmaceutical formulation of claim 1, wherein the VLA-4 antagonist comprises 50% by weight of the pharmaceutical formulation.
22. A pharmaceutical formulation comprising a granulate of Compound IA which provides a median Tmax that is in the range of about 1 to about 4 hours post-dose.
23. A method for the treatment of an inflammatory disease, comprising administering to a patient in need thereof a therapeutically effective amount of the pharmaceutical formulation of claim 1.
24. The method of claim 23, wherein the inflammatory disease is multiple sclerosis.
25. A method for inhibiting the progression of an inflammatory disease, comprising administering to a patient in need thereof a therapeutically effective amount of the pharmaceutical formulation of claim 1.
26. The method of claim 25, wherein the inflammatory disease is multiple sclerosis.
27. The pharmaceutical formulation of claim 4 wherein the surfactant is sodium lauryl sulfate and the formulation further comprises pregelatinized starch, crocarmellose sodium, and lactose.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US6185708P | 2008-06-16 | 2008-06-16 | |
US61/061,857 | 2008-06-16 |
Publications (3)
Publication Number | Publication Date |
---|---|
WO2010008719A2 true WO2010008719A2 (en) | 2010-01-21 |
WO2010008719A3 WO2010008719A3 (en) | 2010-03-11 |
WO2010008719A9 WO2010008719A9 (en) | 2010-07-01 |
Family
ID=41395474
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2009/047353 WO2010008719A2 (en) | 2008-06-16 | 2009-06-15 | Oral pharmaceutical formulations of vla-4 antagonists |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2010008719A2 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2019246455A1 (en) | 2018-06-20 | 2019-12-26 | Progenity, Inc. | Treatment of a disease of the gastrointestinal tract with an integrin inhibitor |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
FR2761266A1 (en) * | 1997-03-28 | 1998-10-02 | Sanofi Sa | WET-GRANULATED PHARMACEUTICAL COMPOSITION FOR ORAL ADMINISTRATION OF AN N-PIPERIDINO-3-PYRAZOLECARBOXAMIDE DERIVATIVE, ITS SALTS AND SOLVATES |
US6238695B1 (en) * | 1998-04-07 | 2001-05-29 | Dupont Pharmaceuticals Company | Formulation of fast-dissolving efavirenz capsules or tablets using super-disintegrants |
WO2004044046A2 (en) * | 2002-11-08 | 2004-05-27 | Encysive Pharmaceuticals | Combination products with carboxylic acid derivatives that inhibit the binding of integrins to their receptors and other therapeutic compounds |
US20050003001A1 (en) * | 2001-11-07 | 2005-01-06 | Hisami Yamaguchi | Method for improving dissolution of poorly dispersible medicaments |
-
2009
- 2009-06-15 WO PCT/US2009/047353 patent/WO2010008719A2/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
FR2761266A1 (en) * | 1997-03-28 | 1998-10-02 | Sanofi Sa | WET-GRANULATED PHARMACEUTICAL COMPOSITION FOR ORAL ADMINISTRATION OF AN N-PIPERIDINO-3-PYRAZOLECARBOXAMIDE DERIVATIVE, ITS SALTS AND SOLVATES |
US6238695B1 (en) * | 1998-04-07 | 2001-05-29 | Dupont Pharmaceuticals Company | Formulation of fast-dissolving efavirenz capsules or tablets using super-disintegrants |
US20050003001A1 (en) * | 2001-11-07 | 2005-01-06 | Hisami Yamaguchi | Method for improving dissolution of poorly dispersible medicaments |
WO2004044046A2 (en) * | 2002-11-08 | 2004-05-27 | Encysive Pharmaceuticals | Combination products with carboxylic acid derivatives that inhibit the binding of integrins to their receptors and other therapeutic compounds |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2019246455A1 (en) | 2018-06-20 | 2019-12-26 | Progenity, Inc. | Treatment of a disease of the gastrointestinal tract with an integrin inhibitor |
Also Published As
Publication number | Publication date |
---|---|
WO2010008719A9 (en) | 2010-07-01 |
WO2010008719A3 (en) | 2010-03-11 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7627302B2 (en) | Palbociclib solid dosage forms | |
RU2616262C2 (en) | Compositions and methods for myelofibrosis treatment | |
TW201731509A (en) | Dosage schedule for 2-hydroxy-6-((2-(1-isopropyl-1H-pyrazol-5-yl)pyridin-3-yl)methoxy)-benzaldehyde | |
CN101267810A (en) | Capsule formulation of pirfenidone and pharmaceutically acceptable excipients | |
CN102056589A (en) | Solid pharmaceutical formulations comprising BIBW 2992 | |
US20240307398A1 (en) | Formulations, methods, kits, and dosage forms for treating atopic dermatitis and for improved stability of an active pharmaceutical ingredient | |
EA029586B1 (en) | Pharmaceutical composition for treatment of hiv infections | |
CN103156860A (en) | Olanzapine composition and preparation method thereof | |
AU2018279834A1 (en) | Liquid dosage forms to treat cancer | |
CN1839850A (en) | Dispersion tablet medicine containing oryzanol and its preparation method | |
CN108125918A (en) | Everolimus pharmaceutical composition | |
WO2010008719A9 (en) | Oral pharmaceutical formulations of vla-4 antagonists | |
CN103284953B (en) | Bicyclol solid preparation and preparation method thereof | |
CN118284415A (en) | Darotamine pharmaceutical composition and preparation method and application thereof | |
CN109381431B (en) | Huperzine A sustained-release pellet and preparation method thereof | |
US20240285532A1 (en) | A plurality of tasquinimod particles and use thereof | |
CN108785273B (en) | A kind of entecavir capsule pharmaceutical composition and preparation method thereof | |
CN103330944A (en) | Bicyclol oral solid pharmaceutical composition, and preparation and preparation method thereof | |
TW202245786A (en) | Applications of quinazoline compound and pharmaceutical composition | |
EA040951B1 (en) | DOSAGE FORMS CIS-4-[2-{[(3S,4R)-3-FLUOROXAN-4-YL]AMINO}-8-(2,4,6-TRICHLORORANINO)-9H-PURINE-9-YL]-1 -METHYLCYCLOHEXANE-1-CARBOXAMIDE AND THEIR USE IN A METHOD FOR CANCER TREATMENT | |
OA18938A (en) | Dosing regimens for 2-Hydroxy-6-((2-(1Isopropyl-1H-Pyrazol-5-Yl)Pyridin-3-Yl)Methoxy) Benzaldehyde |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 09741501 Country of ref document: EP Kind code of ref document: A2 |
|
NENP | Non-entry into the national phase in: |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 09741501 Country of ref document: EP Kind code of ref document: A2 |