WO2014174073A1 - Sustained release formulations of tofacitinib - Google Patents
Sustained release formulations of tofacitinib Download PDFInfo
- Publication number
- WO2014174073A1 WO2014174073A1 PCT/EP2014/058448 EP2014058448W WO2014174073A1 WO 2014174073 A1 WO2014174073 A1 WO 2014174073A1 EP 2014058448 W EP2014058448 W EP 2014058448W WO 2014174073 A1 WO2014174073 A1 WO 2014174073A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- formulation
- tofacitinib
- formulations
- release
- mpa
- Prior art date
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 161
- 238000009472 formulation Methods 0.000 title claims abstract description 127
- UJLAWZDWDVHWOW-YPMHNXCESA-N tofacitinib Chemical compound C[C@@H]1CCN(C(=O)CC#N)C[C@@H]1N(C)C1=NC=NC2=C1C=CN2 UJLAWZDWDVHWOW-YPMHNXCESA-N 0.000 title claims abstract description 65
- 239000004012 Tofacitinib Substances 0.000 title claims abstract description 64
- 229960001350 tofacitinib Drugs 0.000 title claims abstract description 64
- 238000013268 sustained release Methods 0.000 title claims abstract description 27
- 239000012730 sustained-release form Substances 0.000 title claims abstract description 27
- 239000003814 drug Substances 0.000 claims abstract description 63
- 150000003839 salts Chemical class 0.000 claims abstract description 14
- 239000003795 chemical substances by application Substances 0.000 claims description 48
- 230000003113 alkalizing effect Effects 0.000 claims description 47
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 claims description 43
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 claims description 43
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 claims description 43
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical group OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 claims description 40
- 229920000663 Hydroxyethyl cellulose Polymers 0.000 claims description 35
- 229920001477 hydrophilic polymer Polymers 0.000 claims description 35
- 235000019447 hydroxyethyl cellulose Nutrition 0.000 claims description 35
- 239000004354 Hydroxyethyl cellulose Substances 0.000 claims description 28
- 235000010418 carrageenan Nutrition 0.000 claims description 23
- 229920001525 carrageenan Polymers 0.000 claims description 23
- 229960004247 tofacitinib citrate Drugs 0.000 claims description 23
- SYIKUFDOYJFGBQ-YLAFAASESA-N tofacitinib citrate Chemical group OC(=O)CC(O)(C(O)=O)CC(O)=O.C[C@@H]1CCN(C(=O)CC#N)C[C@@H]1N(C)C1=NC=NC2=C1C=CN2 SYIKUFDOYJFGBQ-YLAFAASESA-N 0.000 claims description 23
- 239000000679 carrageenan Substances 0.000 claims description 22
- 229940113118 carrageenan Drugs 0.000 claims description 22
- UHVMMEOXYDMDKI-JKYCWFKZSA-L zinc;1-(5-cyanopyridin-2-yl)-3-[(1s,2s)-2-(6-fluoro-2-hydroxy-3-propanoylphenyl)cyclopropyl]urea;diacetate Chemical compound [Zn+2].CC([O-])=O.CC([O-])=O.CCC(=O)C1=CC=C(F)C([C@H]2[C@H](C2)NC(=O)NC=2N=CC(=CC=2)C#N)=C1O UHVMMEOXYDMDKI-JKYCWFKZSA-L 0.000 claims description 22
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 20
- 235000010980 cellulose Nutrition 0.000 claims description 19
- 229920002678 cellulose Polymers 0.000 claims description 19
- 239000001913 cellulose Substances 0.000 claims description 19
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 claims description 18
- 239000001509 sodium citrate Substances 0.000 claims description 18
- NLJMYIDDQXHKNR-UHFFFAOYSA-K sodium citrate Chemical compound O.O.[Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O NLJMYIDDQXHKNR-UHFFFAOYSA-K 0.000 claims description 18
- 239000011248 coating agent Substances 0.000 claims description 10
- 229910000019 calcium carbonate Inorganic materials 0.000 claims description 9
- 238000000576 coating method Methods 0.000 claims description 9
- 239000000395 magnesium oxide Substances 0.000 claims description 9
- CPLXHLVBOLITMK-UHFFFAOYSA-N magnesium oxide Inorganic materials [Mg]=O CPLXHLVBOLITMK-UHFFFAOYSA-N 0.000 claims description 9
- AXZKOIWUVFPNLO-UHFFFAOYSA-N magnesium;oxygen(2-) Chemical compound [O-2].[Mg+2] AXZKOIWUVFPNLO-UHFFFAOYSA-N 0.000 claims description 9
- 239000006185 dispersion Substances 0.000 claims description 7
- 239000008194 pharmaceutical composition Substances 0.000 claims description 7
- 239000000378 calcium silicate Substances 0.000 claims description 6
- 229910052918 calcium silicate Inorganic materials 0.000 claims description 6
- OYACROKNLOSFPA-UHFFFAOYSA-N calcium;dioxido(oxo)silane Chemical compound [Ca+2].[O-][Si]([O-])=O OYACROKNLOSFPA-UHFFFAOYSA-N 0.000 claims description 6
- 239000001508 potassium citrate Substances 0.000 claims description 5
- 229960002635 potassium citrate Drugs 0.000 claims description 5
- QEEAPRPFLLJWCF-UHFFFAOYSA-K potassium citrate (anhydrous) Chemical compound [K+].[K+].[K+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O QEEAPRPFLLJWCF-UHFFFAOYSA-K 0.000 claims description 5
- 235000011082 potassium citrates Nutrition 0.000 claims description 5
- 239000012458 free base Substances 0.000 claims description 3
- 206010039073 rheumatoid arthritis Diseases 0.000 claims description 3
- 229940079593 drug Drugs 0.000 abstract description 62
- 230000002829 reductive effect Effects 0.000 abstract description 7
- 230000009246 food effect Effects 0.000 abstract description 4
- 235000021471 food effect Nutrition 0.000 abstract description 4
- 239000003826 tablet Substances 0.000 description 69
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 40
- 239000011159 matrix material Substances 0.000 description 40
- 229940071826 hydroxyethyl cellulose Drugs 0.000 description 27
- 239000010410 layer Substances 0.000 description 22
- 229920000642 polymer Polymers 0.000 description 22
- 235000019359 magnesium stearate Nutrition 0.000 description 20
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 18
- 239000000945 filler Substances 0.000 description 18
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 18
- 239000008108 microcrystalline cellulose Substances 0.000 description 18
- 229940016286 microcrystalline cellulose Drugs 0.000 description 18
- 239000002609 medium Substances 0.000 description 16
- 235000011083 sodium citrates Nutrition 0.000 description 16
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 14
- 239000008101 lactose Substances 0.000 description 14
- 229960001375 lactose Drugs 0.000 description 14
- 239000000243 solution Substances 0.000 description 13
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 12
- 230000036470 plasma concentration Effects 0.000 description 12
- 238000004090 dissolution Methods 0.000 description 11
- 238000007922 dissolution test Methods 0.000 description 11
- 230000000694 effects Effects 0.000 description 11
- OSGAYBCDTDRGGQ-UHFFFAOYSA-L calcium sulfate Chemical compound [Ca+2].[O-]S([O-])(=O)=O OSGAYBCDTDRGGQ-UHFFFAOYSA-L 0.000 description 10
- 239000008187 granular material Substances 0.000 description 10
- 239000000314 lubricant Substances 0.000 description 10
- 238000000034 method Methods 0.000 description 10
- 235000010216 calcium carbonate Nutrition 0.000 description 8
- 239000012738 dissolution medium Substances 0.000 description 8
- 239000012530 fluid Substances 0.000 description 8
- 210000001035 gastrointestinal tract Anatomy 0.000 description 8
- 229960000869 magnesium oxide Drugs 0.000 description 8
- 235000012245 magnesium oxide Nutrition 0.000 description 8
- 239000012071 phase Substances 0.000 description 8
- 238000012360 testing method Methods 0.000 description 8
- 229920003094 Methocel™ K4M Polymers 0.000 description 7
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 6
- 229920003096 Methocel™ K100M Polymers 0.000 description 6
- 239000008186 active pharmaceutical agent Substances 0.000 description 6
- 230000001419 dependent effect Effects 0.000 description 6
- 210000002784 stomach Anatomy 0.000 description 6
- 230000002378 acidificating effect Effects 0.000 description 5
- 235000012241 calcium silicate Nutrition 0.000 description 5
- 235000011132 calcium sulphate Nutrition 0.000 description 5
- 238000007906 compression Methods 0.000 description 5
- 230000006835 compression Effects 0.000 description 5
- 239000007857 degradation product Substances 0.000 description 5
- 235000019700 dicalcium phosphate Nutrition 0.000 description 5
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 238000013265 extended release Methods 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 239000012729 immediate-release (IR) formulation Substances 0.000 description 4
- 239000013563 matrix tablet Substances 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 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 description 3
- 238000010521 absorption reaction Methods 0.000 description 3
- XAAHAAMILDNBPS-UHFFFAOYSA-L calcium hydrogenphosphate dihydrate Chemical compound O.O.[Ca+2].OP([O-])([O-])=O XAAHAAMILDNBPS-UHFFFAOYSA-L 0.000 description 3
- 238000012377 drug delivery Methods 0.000 description 3
- 230000037406 food intake Effects 0.000 description 3
- 239000011521 glass Substances 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 239000008185 minitablet Substances 0.000 description 3
- 239000008363 phosphate buffer Substances 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 230000002459 sustained effect Effects 0.000 description 3
- 238000005550 wet granulation Methods 0.000 description 3
- WSVLPVUVIUVCRA-KPKNDVKVSA-N Alpha-lactose monohydrate Chemical compound O.O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O WSVLPVUVIUVCRA-KPKNDVKVSA-N 0.000 description 2
- 102000006500 Janus Kinase 3 Human genes 0.000 description 2
- 108010019421 Janus Kinase 3 Proteins 0.000 description 2
- 229920003091 Methocel™ Polymers 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M Potassium chloride Chemical compound [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 2
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 2
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 2
- XLOMVQKBTHCTTD-UHFFFAOYSA-N Zinc monoxide Chemical compound [Zn]=O XLOMVQKBTHCTTD-UHFFFAOYSA-N 0.000 description 2
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- SNAAJJQQZSMGQD-UHFFFAOYSA-N aluminum magnesium Chemical compound [Mg].[Al] SNAAJJQQZSMGQD-UHFFFAOYSA-N 0.000 description 2
- 239000011324 bead Substances 0.000 description 2
- FUFJGUQYACFECW-UHFFFAOYSA-L calcium hydrogenphosphate Chemical compound [Ca+2].OP([O-])([O-])=O FUFJGUQYACFECW-UHFFFAOYSA-L 0.000 description 2
- 239000001175 calcium sulphate Substances 0.000 description 2
- 229920003123 carboxymethyl cellulose sodium Polymers 0.000 description 2
- 229940063834 carboxymethylcellulose sodium Drugs 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 210000001072 colon Anatomy 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 229910003460 diamond Inorganic materials 0.000 description 2
- 239000010432 diamond Substances 0.000 description 2
- 238000011978 dissolution method Methods 0.000 description 2
- 230000008030 elimination Effects 0.000 description 2
- 238000003379 elimination reaction Methods 0.000 description 2
- 238000010828 elution Methods 0.000 description 2
- 230000003628 erosive effect Effects 0.000 description 2
- 235000012631 food intake Nutrition 0.000 description 2
- 230000002496 gastric effect Effects 0.000 description 2
- -1 hydroxide anions Chemical class 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 229960001021 lactose monohydrate Drugs 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 238000004088 simulation Methods 0.000 description 2
- 239000002002 slurry Substances 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000000126 substance Substances 0.000 description 2
- 229920001285 xanthan gum Polymers 0.000 description 2
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2r,3r,4s,5r,6s)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2s,3r,4s,5r,6r)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2r,3r,4s,5r,6r)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 1
- SMZOUWXMTYCWNB-UHFFFAOYSA-N 2-(2-methoxy-5-methylphenyl)ethanamine Chemical compound COC1=CC=C(C)C=C1CCN SMZOUWXMTYCWNB-UHFFFAOYSA-N 0.000 description 1
- GUBGYTABKSRVRQ-UHFFFAOYSA-N 2-(hydroxymethyl)-6-[4,5,6-trihydroxy-2-(hydroxymethyl)oxan-3-yl]oxyoxane-3,4,5-triol Chemical compound OCC1OC(OC2C(O)C(O)C(O)OC2CO)C(O)C(O)C1O GUBGYTABKSRVRQ-UHFFFAOYSA-N 0.000 description 1
- NIXOWILDQLNWCW-UHFFFAOYSA-N 2-Propenoic acid Natural products OC(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-N 0.000 description 1
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 1
- CYDQOEWLBCCFJZ-UHFFFAOYSA-N 4-(4-fluorophenyl)oxane-4-carboxylic acid Chemical compound C=1C=C(F)C=CC=1C1(C(=O)O)CCOCC1 CYDQOEWLBCCFJZ-UHFFFAOYSA-N 0.000 description 1
- GJCOSYZMQJWQCA-UHFFFAOYSA-N 9H-xanthene Chemical compound C1=CC=C2CC3=CC=CC=C3OC2=C1 GJCOSYZMQJWQCA-UHFFFAOYSA-N 0.000 description 1
- 229910002012 Aerosil® Inorganic materials 0.000 description 1
- 239000005995 Aluminium silicate Substances 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
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 1
- 229920001685 Amylomaize Polymers 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- 229920001353 Dextrin Polymers 0.000 description 1
- 239000004375 Dextrin Substances 0.000 description 1
- 208000030453 Drug-Related Side Effects and Adverse reaction Diseases 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 229920003134 Eudragit® polymer Polymers 0.000 description 1
- 206010017943 Gastrointestinal conditions Diseases 0.000 description 1
- 239000001828 Gelatine Substances 0.000 description 1
- 229920002907 Guar gum Polymers 0.000 description 1
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 208000037026 Invasive Fungal Infections Diseases 0.000 description 1
- 229920000161 Locust bean gum Polymers 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- VVQNEPGJFQJSBK-UHFFFAOYSA-N Methyl methacrylate Chemical compound COC(=O)C(C)=C VVQNEPGJFQJSBK-UHFFFAOYSA-N 0.000 description 1
- HDSBZMRLPLPFLQ-UHFFFAOYSA-N Propylene glycol alginate Chemical compound OC1C(O)C(OC)OC(C(O)=O)C1OC1C(O)C(O)C(C)C(C(=O)OCC(C)O)O1 HDSBZMRLPLPFLQ-UHFFFAOYSA-N 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- 229920002385 Sodium hyaluronate Polymers 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 235000010443 alginic acid Nutrition 0.000 description 1
- 229920000615 alginic acid Polymers 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 235000012211 aluminium silicate Nutrition 0.000 description 1
- 235000019257 ammonium acetate Nutrition 0.000 description 1
- 229940043376 ammonium acetate Drugs 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 229960000892 attapulgite Drugs 0.000 description 1
- 239000000440 bentonite Substances 0.000 description 1
- 229910000278 bentonite Inorganic materials 0.000 description 1
- 235000012216 bentonite Nutrition 0.000 description 1
- SVPXDRXYRYOSEX-UHFFFAOYSA-N bentoquatam Chemical compound O.O=[Si]=O.O=[Al]O[Al]=O SVPXDRXYRYOSEX-UHFFFAOYSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 235000000332 black box Nutrition 0.000 description 1
- 229910021538 borax Inorganic materials 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 229960002713 calcium chloride Drugs 0.000 description 1
- 235000011148 calcium chloride Nutrition 0.000 description 1
- MKJXYGKVIBWPFZ-UHFFFAOYSA-L calcium lactate Chemical compound [Ca+2].CC(O)C([O-])=O.CC(O)C([O-])=O MKJXYGKVIBWPFZ-UHFFFAOYSA-L 0.000 description 1
- 239000001527 calcium lactate Substances 0.000 description 1
- 235000011086 calcium lactate Nutrition 0.000 description 1
- 229960002401 calcium lactate Drugs 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 229920001577 copolymer Polymers 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 235000019425 dextrin Nutrition 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- 150000004683 dihydrates Chemical class 0.000 description 1
- FSBVERYRVPGNGG-UHFFFAOYSA-N dimagnesium dioxido-bis[[oxido(oxo)silyl]oxy]silane hydrate Chemical compound O.[Mg+2].[Mg+2].[O-][Si](=O)O[Si]([O-])([O-])O[Si]([O-])=O FSBVERYRVPGNGG-UHFFFAOYSA-N 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- ZPWVASYFFYYZEW-UHFFFAOYSA-L dipotassium hydrogen phosphate Chemical compound [K+].[K+].OP([O-])([O-])=O ZPWVASYFFYYZEW-UHFFFAOYSA-L 0.000 description 1
- 235000019797 dipotassium phosphate Nutrition 0.000 description 1
- 229910000396 dipotassium phosphate Inorganic materials 0.000 description 1
- 238000007907 direct compression Methods 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 238000009506 drug dissolution testing Methods 0.000 description 1
- 229940126534 drug product Drugs 0.000 description 1
- 229940088679 drug related substance Drugs 0.000 description 1
- 238000007923 drug release testing Methods 0.000 description 1
- 238000007908 dry granulation Methods 0.000 description 1
- 210000001198 duodenum Anatomy 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 238000009477 fluid bed granulation Methods 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 238000012395 formulation development Methods 0.000 description 1
- 230000030136 gastric emptying Effects 0.000 description 1
- 210000003736 gastrointestinal content Anatomy 0.000 description 1
- 230000005176 gastrointestinal motility Effects 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 235000010417 guar gum Nutrition 0.000 description 1
- 239000000665 guar gum Substances 0.000 description 1
- 229960002154 guar gum Drugs 0.000 description 1
- 238000009478 high shear granulation Methods 0.000 description 1
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 1
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 1
- 229960003943 hypromellose Drugs 0.000 description 1
- 210000003405 ileum Anatomy 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 210000000936 intestine Anatomy 0.000 description 1
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 1
- 235000010420 locust bean gum Nutrition 0.000 description 1
- 239000000711 locust bean gum Substances 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 238000009476 low shear granulation Methods 0.000 description 1
- 239000004337 magnesium citrate Substances 0.000 description 1
- 229960005336 magnesium citrate Drugs 0.000 description 1
- 235000002538 magnesium citrate Nutrition 0.000 description 1
- VTHJTEIRLNZDEV-UHFFFAOYSA-L magnesium dihydroxide Chemical compound [OH-].[OH-].[Mg+2] VTHJTEIRLNZDEV-UHFFFAOYSA-L 0.000 description 1
- 239000001755 magnesium gluconate Substances 0.000 description 1
- 235000015778 magnesium gluconate Nutrition 0.000 description 1
- 229960003035 magnesium gluconate Drugs 0.000 description 1
- 239000000347 magnesium hydroxide Substances 0.000 description 1
- 229910001862 magnesium hydroxide Inorganic materials 0.000 description 1
- 229960000816 magnesium hydroxide Drugs 0.000 description 1
- 235000012254 magnesium hydroxide Nutrition 0.000 description 1
- 239000000391 magnesium silicate Substances 0.000 description 1
- 229910052919 magnesium silicate Inorganic materials 0.000 description 1
- 235000019792 magnesium silicate Nutrition 0.000 description 1
- 229960002366 magnesium silicate Drugs 0.000 description 1
- IAKLPCRFBAZVRW-XRDLMGPZSA-L magnesium;(2r,3s,4r,5r)-2,3,4,5,6-pentahydroxyhexanoate;hydrate Chemical compound O.[Mg+2].OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O.OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C([O-])=O IAKLPCRFBAZVRW-XRDLMGPZSA-L 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 229910000000 metal hydroxide Inorganic materials 0.000 description 1
- 150000004692 metal hydroxides Chemical class 0.000 description 1
- 229910044991 metal oxide Inorganic materials 0.000 description 1
- 150000004706 metal oxides Chemical class 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 150000004682 monohydrates Chemical class 0.000 description 1
- LPUQAYUQRXPFSQ-DFWYDOINSA-M monosodium L-glutamate Chemical compound [Na+].[O-]C(=O)[C@@H](N)CCC(O)=O LPUQAYUQRXPFSQ-DFWYDOINSA-M 0.000 description 1
- 239000004223 monosodium glutamate Substances 0.000 description 1
- 235000013923 monosodium glutamate Nutrition 0.000 description 1
- 230000003183 myoelectrical effect Effects 0.000 description 1
- 229920001206 natural gum Polymers 0.000 description 1
- 238000002414 normal-phase solid-phase extraction Methods 0.000 description 1
- 239000006186 oral dosage form Substances 0.000 description 1
- 229940126701 oral medication Drugs 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 229910052625 palygorskite Inorganic materials 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 229920001277 pectin Polymers 0.000 description 1
- 235000010987 pectin Nutrition 0.000 description 1
- 239000001814 pectin Substances 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 235000015497 potassium bicarbonate Nutrition 0.000 description 1
- 239000011736 potassium bicarbonate Substances 0.000 description 1
- 229910000028 potassium bicarbonate Inorganic materials 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- 235000011164 potassium chloride Nutrition 0.000 description 1
- 229960002816 potassium chloride Drugs 0.000 description 1
- TYJJADVDDVDEDZ-UHFFFAOYSA-M potassium hydrogencarbonate Chemical compound [K+].OC([O-])=O TYJJADVDDVDEDZ-UHFFFAOYSA-M 0.000 description 1
- 229940086066 potassium hydrogencarbonate Drugs 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 235000010409 propane-1,2-diol alginate Nutrition 0.000 description 1
- 239000000770 propane-1,2-diol alginate Substances 0.000 description 1
- 210000001187 pylorus Anatomy 0.000 description 1
- 230000011514 reflex Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 239000012488 sample solution Substances 0.000 description 1
- 238000005070 sampling Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 239000002356 single layer Substances 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 235000015424 sodium Nutrition 0.000 description 1
- 229940083542 sodium Drugs 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- HELHAJAZNSDZJO-OLXYHTOASA-L sodium L-tartrate Chemical compound [Na+].[Na+].[O-]C(=O)[C@H](O)[C@@H](O)C([O-])=O HELHAJAZNSDZJO-OLXYHTOASA-L 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 229940010747 sodium hyaluronate Drugs 0.000 description 1
- 239000001540 sodium lactate Substances 0.000 description 1
- 235000011088 sodium lactate Nutrition 0.000 description 1
- 229940005581 sodium lactate Drugs 0.000 description 1
- RYYKJJJTJZKILX-UHFFFAOYSA-M sodium octadecanoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCC([O-])=O RYYKJJJTJZKILX-UHFFFAOYSA-M 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 235000011008 sodium phosphates Nutrition 0.000 description 1
- 239000001433 sodium tartrate Substances 0.000 description 1
- 229960002167 sodium tartrate Drugs 0.000 description 1
- 235000011004 sodium tartrates Nutrition 0.000 description 1
- 235000010339 sodium tetraborate Nutrition 0.000 description 1
- YWIVKILSMZOHHF-QJZPQSOGSA-N sodium;(2s,3s,4s,5r,6r)-6-[(2s,3r,4r,5s,6r)-3-acetamido-2-[(2s,3s,4r,5r,6r)-6-[(2r,3r,4r,5s,6r)-3-acetamido-2,5-dihydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-2-carboxy-4,5-dihydroxyoxan-3-yl]oxy-5-hydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-3,4,5-trihydroxyoxane-2- Chemical compound [Na+].CC(=O)N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@H](O3)C(O)=O)O)[C@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](C(O)=O)O1 YWIVKILSMZOHHF-QJZPQSOGSA-N 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 238000012430 stability testing Methods 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 229940032147 starch Drugs 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 229920002994 synthetic fiber Polymers 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 230000001225 therapeutic effect Effects 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- PLSARIKBYIPYPF-UHFFFAOYSA-H trimagnesium dicitrate Chemical compound [Mg+2].[Mg+2].[Mg+2].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O.[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O PLSARIKBYIPYPF-UHFFFAOYSA-H 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- BSVBQGMMJUBVOD-UHFFFAOYSA-N trisodium borate Chemical compound [Na+].[Na+].[Na+].[O-]B([O-])[O-] BSVBQGMMJUBVOD-UHFFFAOYSA-N 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- 229960000281 trometamol Drugs 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 235000010493 xanthan gum Nutrition 0.000 description 1
- 239000000230 xanthan gum Substances 0.000 description 1
- 229940082509 xanthan gum Drugs 0.000 description 1
- 239000011787 zinc oxide Substances 0.000 description 1
- 229960001296 zinc oxide Drugs 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
- A61K9/20—Pills, tablets, discs, rods
- A61K9/2004—Excipients; Inactive ingredients
- A61K9/2022—Organic macromolecular compounds
- A61K9/205—Polysaccharides, e.g. alginate, gums; Cyclodextrin
-
- 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
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- 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
- A61K9/20—Pills, tablets, discs, rods
- A61K9/2004—Excipients; Inactive ingredients
- A61K9/2022—Organic macromolecular compounds
- A61K9/205—Polysaccharides, e.g. alginate, gums; Cyclodextrin
- A61K9/2054—Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
-
- 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
- A61K9/20—Pills, tablets, discs, rods
- A61K9/2072—Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
- A61K9/2086—Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
Definitions
- the present invention relates to sustained release formulations for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof.
- Tofacitinib 3- ⁇ (3R,4R)-4-methyl-3-[methyl-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- amino]-piperidin-l -yl ⁇ 3-oxo-propionitrile, is an inhibitor of the enzyme Janus Kinase 3 (JAK3) indicated for treatment of rheumatoid arthritis. Its synthetic preparation is disclosed in WO 02/096909 Al, WO 03/048162 Al and WO 2007/012953 A2.
- the recommended dose is 5 mg given orally twice daily.
- the FDA label contains a black-box warning alerting about the risk of serious side effects, such as serious infections (tuberculosis, invasive fungal and viral infections), lymphoma and other malignancies. Serious concerns about the risk of malignancy with 10 mg dose were raised by the FDA and eventually this dosage was not approved.
- the risk of serious side effects increases in a dose and time-dependant fashion. An increased frequency of side effects of drugs is usually observed when peak plasma concentrations exceed a certain level.
- Tofacitinib exhibits linear pharmacokinetics, with the systemic exposure (AUCM) and peak plasma concentration (Cmax) increasing in proportion to the dose in the dose range of 1 to 100 mg.
- peak plasma concentrations of the 10 mg tofacitinib dose are approximately two times higher than those of the 5 mg dose and they may be too high for a safe drug administration.
- Plasma concentration of Tofacitinib after oral administration is directly related to the release rate of the drug in the gastrointestinal tract.
- IR immediate release
- Sustained peak plasma concentrations can theoretically be achieved by means of sustained release matrix systems. However, when such systems are made of hydrophilic polymers, they seldom provide pH independent drug release of pH-dependent soluble drugs and they are normally incapable of attaining zero-order release except for practically insoluble drugs. From FDA published data it can be seen that the aqueous solubility of Tofacitinib citrate is pH-dependent with higher solubility at low pH and drastically decreased solubility at pH above 3.9. pH dependent solubility can lead to pH-dependent in vivo drug release from sustained release matrices. Drug release varies as a function of movement through segments of the gastrointestinal tract with different pH. This can lead to inefficient drug delivery and bigger inter-subject variability, since pH in the gastrointestinal tract varies significantly between subjects. Sustained release formulations that would provide pH independent drug release throughout whole gastrointestinal tract are very desirable.
- the robustness of drug release is also critical, since mechanical stress applied to matrix tablets during gastrointestinal transit may lead to faster disruption of the gel layer and influence the plasma drug concentrations over time. This can change the therapeutic efficacy and safety.
- Different mechanical stress can be exerted on matrix tablets due to variety of gastrointestinal conditions.
- MMC migrating myoelectric complex
- the movement of tablets during this phase is rapid and tablets are subject to increased mechanical stress, especially during gastric emptying.
- gastrointestinal motility is elevated during and after food intake. This causes even greater mechanical stress on pharmaceutical compositions.
- food intake stimulates the transport of gastrointestinal content from the terminal small intestine into the colon, a mechanism that is known as gastro-ileal or gastro-ileocecal reflex, which can again exert elevated mechanical stress on the matrix tablet.
- WO 2012/100949 Al discloses oral dosage forms for modified release comprising Tofacitinib. Such formulations are disclosed in very general terms and give no direction to the skilled man for preparing sustained formulations with adequate release profile and properties. 10 examples of specific formulations are also provided, but no dissolution test results. When reproduced, such formulations showed poor release properties, liberating the active ingredient too fast for any practical application, and proved not to be resistant to mechanical stress.
- the present disclosure provides a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof, a hydrophilic polymer and an alkalizing agent, wherein the pH at 25 °C of a 1% weight/volume water solution or dispersion of the formulation is between 5.5 and 11. It further provides a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof and a polymer, wherein the hydrophilic polymer is either a cellulose derivative with a viscosity between 1000 and 150000 mPa-s, a carrageenan with a viscosity of 5 mPa-s or higher or a mixture thereof.
- a once daily dosage regimen of the formulations of the invention preferably for the treatment of rheumatoid arthritis, is also disclosed.
- FIG. 1 illustrates simulated plasma profiles after twice daily administration of immediate release formulations containing 5 mg and 10 mg of tofacitinib (full and dashed line) and once daily administration of a sustained release formulation containing 11 mg of Tofacitinib (dotted line).
- FIG. 2 illustrates the stability profile of Tofacitinib citrate at various pH.
- FIG. 3 illustrates the dissolution test results of the formulations of example 1 (25% HPMC without alkalizing agent) - top - and example 15 (60 % of HEC without alkalizing agent) - bottom - using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
- FIG. 4 illustrates the dissolution test results of the formulation of example 2 (25% HPMC with insoluble alkalizing agents) using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
- FIG. 5 illustrates the dissolution test results of the formulations of example 3 (25% HPMC with 9 % sodium citrate) - top - and of example 5 (25%> HPMC with 48 % sodium citrate) - bottom -using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
- FIG. 6 illustrates the dissolution test results of the formulation of example 8b (60% HEC with 25 % sodium citrate) using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
- FIG. 7 illustrates the dissolution test results of the formulations of example 9 (25%> HPMC with calcium hydrogen phosphate dihydrate) - top - and 10 (25% HPMC with calcium sulphate) - bottom - using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
- FIG. 8 illustrates the dissolution test results of the formulations of examples 11-13 (15 % circle, 25 % triangle and 35 > diamond HPMC) with (dotted lines) or without mechanical stress (full lines) using apparatus 1, 100 RPM, 900 ml of pH 6.8 medium.
- FIG. 9 illustrates the dissolution test results of the formulations of examples 14 and 15 (30 % circle, 60% diamond HEC) with (dotted lines) or without mechanical stress (full lines) using apparatus 1, 100 RPM, 900 ml of pH 6.8 medium.
- FIG. 10 illustrates the dissolution test results of the formulations of example 2 (25 % HPMC with insoluble alkalizing agents) with (dotted lines) or without mechanical stress (full lines) using apparatus 1 , 100 RPM, 900 ml of pH 6.8 medium.
- FIG. 11 illustrates the dissolution test results of the formulations of example 17 (HEC + carrageenan) using apparatus 2 at 100 rpm and dissolution media with different pH (dotted line pH 1.2 + 6.8; full line 6.8 ) - top - and with (dotted line) or without mechanical stress (full line) using Apparatus 1, 100 RPM, 900 ml of pH 6.8 medium - bottom -.
- FIG. 12 illustrates a schematic view of a hydrophilic matrix bilayer tablet comprising the active pharmaceutical ingredient (API) tofacitinib and a sustained release (SR) polymer.
- API active pharmaceutical ingredient
- SR sustained release
- hydrophilic polymer refers to water soluble polymeric materials that in contact with an aqueous environment swell and form a gel matrix. They are characterized by the presence of polar groups attached to the main polymer backbone of the molecule (see C.A. Finch: Hydrophilic polymers. In: R.W.Dyson (ed.) Specialty Polymers; Blackie&Son Ltd 1987).
- alkalizing agent refers to any agent that counteracts or neutralizes acidity and/or increases the concentration of hydroxide anions (OH ) in water.
- An ideal sustained release formulation releases the drug with a constant rate between 4 and 18 hours, preferably over 16 hours (zero-order release), and provides effective drug plasma concentration levels over a longer period of time, while at the same time not exceeding the peak plasma concentration of the immediate release formulation, as shown in FIG.l . This results in improved safety and efficacy of the drug product.
- the plasma concentration is then obtained by dividing the amount of drug A by the volume of distribution V d .
- the drug is completely absorbed with about 20% of first-pass elimination.
- the absorption rate for the immediate release formulation is dictated by the transit of drug from the stomach into the intestine, where the absorption takes place.
- the dissolution rate for the sustained release formulation was chosen to mimic the preferred constant (i.e
- sustained release formulation has to be carefully developed to provide drug release that would give optimal plasma concentrations and at the same time be robust enough to prevent dose dumping or a high burst effect. Both phenomena cause the premature and exaggerated release of a drug that can greatly increase its concentration in the body and thereby produce adverse effects. Dose dumping can be a consequence of poor sustained release formulation and several in vivo factors such as mechanical stress in the gastrointestinal tract (i.e. passage of formulation from the stomach into the duodenum and from the ileum to the colon), presence of food or alcohol in the stomach, differences in pH along the
- Burst release is a phenomenon where an initial large bolus of drug is released before the release rate reaches a stable profile. It is related to some formulations (e.g. hydrophilic matrix systems) and well soluble drugs (S. B. Tiwari, A. R. Rajabi-Siahboomi, "Extended-release oral drug delivery technologies: Monolithic Matrix systems ", Methods in Molecular Biology, Drug Delivery Systems, Vol. 437, 217-243.).
- one object of the present invention is the provision of Tofacitinib formulations causing reduced drug level fluctuation in plasma.
- Another object of the present invention is the provision of formulations of Tofacitinib with reduced side effects.
- Another object of the present invention is the provision of Tofacitinib formulations with pH independent release avoiding a burst at the acidic pH of the stomach.
- Another object of the present invention is the provision of Tofacitinib formulations that are mechanically robust.
- Another object of the present invention is the provision of Tofacitinib formulations that prevent dose dumping or a high burst effect.
- Another object of the present invention is the provision of Tofacitinib formulations that do not show a food effect.
- Another object of the present invention is the provision of Tofacitinib formulations with a zero order release profile.
- Another object of the present invention is the provision of a dosage regimen for Tofacitinib with improved patient compliance.
- One embodiment of the present invention is a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof, a hydrophilic polymer and an alkalizing agent, wherein the pH at 25°C of a 1% weight/volume water solution or dispersion of the formulation is between 5.5 and 11.
- Another embodiment of the present invention is a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof and a hydrophilic polymer, wherein the hydrophilic polymer is either a cellulose derivative with a viscosity between 1000 and 150000 mPa-s, a carrageenan with a viscosity greater than 5 mPa-s or a mixture thereof.
- Formulations of the invention are matrix formulations where a rate-controlling hydrophilic polymer forms a matrix through which the drug is dissolved or dispersed.
- Such formulations can be in the form of tablets, minitablets (that can be further inserted in capsules), pellets and granules, preferably in the form of tablets.
- Tablets can be monolayer or multilayer, with tofacitinib included in one or more layers. Tablets can be further coated with a functional or a non-functional coating.
- the formulations of the invention contain tofacitinib citrate.
- the formulations of the invention comprise between 5 and 25 mg, more preferably between 20 and 22 mg or between 10 and 12 mg of tofacitinib or a pharmaceutically acceptable salt thereof, the amount being calculated based on tofacitinib free base.
- the formulations of the invention comprise tofacitinib or a pharmaceutically acceptable salt thereof in an amount between 10 and 12 mg expressed as tofacitinib free base and, wherein such formulations are administered once a day, the Cmax of tofacitinib measured in plasma is below 35 ng/ml, preferably between 25 and 35 ng/ml.
- Hydrophilic polymers include natural gums and polysaccharides (such as alginates, xanthan, carrageenan, locust bean gum, chitosan, guar gum, pectin, crossed-linked high amylose starch, gelatine), semisynthetic materials (such as cellulose derivatives, propylene glycol alginate) and synthetic materials (such as polyethylene oxide and homo- and copolymers of acrylic acid).
- Preferred hydrophilic polymers are cellulose derivatives (such methylcellulose, hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, carboxymethyl cellulose sodium), carrageenan and mixtures thereof.
- Especially preferred hydrophilic polymers are hydroxypropyl methylcellulose (HPMC), hydroxyethyl cellulose (HEC), carrageenan and mixtures thereof.
- the amount of hydrophilic polymer is preferably between 15 and 80%, preferably between 20 and 70%, more preferably between 25 and 60%> w/w, based on the total weight of the pharmaceutical composition excluding any coating. If the hydrophilic polymer is present in insufficient amounts the dissolution profile is either too fast or the drug release is more easily influenced by mechanical stress. If the hydrophilic polymer is present in excessive amounts, the drug release is too slow, e.g. it could take more than 24 hours to release the drug; in addition the manufacturing of the composition may be very difficult (see figures 8 and 9).
- the hydrophilic polymer is hydroxypropyl methylcellulose (HPMC) in an amount between 20 and 60%> w/w, more preferably between 25 and 50% w/w, based on the total weight of the pharmaceutical composition excluding any coating.
- HPMC hydroxypropyl methylcellulose
- the hydrophilic polymer is hydroxyethyl cellulose (HEC) in an amount between 30 and 70% w/w, more preferably between 40 and 60%> w/w, based on the total weight of the pharmaceutical composition excluding any coating.
- HEC hydroxyethyl cellulose
- the hydrophilic polymer is a carrageenan in an amount between 20 and 70 %, more preferably between 40 and 60% w/w, based on the total weight of the pharmaceutical composition excluding any coating.
- the formulations of the invention comprise an alkalizing agent in order to control the solubility of the active substance and thus obtain a pH independent drug release.
- the alkalizing agents of the invention lower the dissolution rate at pH 1.2, thus preventing the burst release due to tofacitinib ' s greater solubility in acidic media.
- tofacitinib citrate is a very stable substance in the solid state, its solution is sensitive to alkaline pH, especially at pH 10 and above ( Figure 2). Due to such instability, alkalizing agents for tofacitinib formulations must be chosen very carefully during formulation development.
- the pH at 25°C of a 1% weight/volume water solution or dispersion of the formulation must be between 5.5 and 11, preferably between 6 and 10.5.
- the alkalizing agents may be soluble as well as insoluble.
- alkalizing agents are aluminum hydroxide, bentonite, kaolin, calcium carbonate, calcium silicate, calcium hydrogen phosphate, calcium sulphate, calcium chloride, calcium lactate, sodium hydrogen carbonate, sodium stearate, sodium citrate, sodium phosphate, sodium borate, sodium lactate, sodium hyaluronate, sodium acetate, monosodium glutamate, sodium tartrate, attapulgite, potassium citrate, potassium hydrogen carbonate, potassium chloride, potassium hydrogen phosphate, tromethamine, zinc oxide, magnesium silicate, magnesium aluminum silicate, magnesium oxide, magnesium hydroxide, magnesium citrate, magnesium gluconate, dextrin, carboxymethyl cellulose sodium, carrageenan, xanthan gum and starch.
- Preferred alkalizing agents are selected from the group consisting of salts of inorganic and organic acids. More preferred alkalizing agents are selected from the group consisting of metal oxides, metal hydroxides, salts of weak acids and some polymers that give pH of solution/dispersion in stated range. Even more preferred alkalizing agents are selected from the group consisting of magnesium oxide, calcium carbonate, calcium silicate, potassium citrate and sodium citrate and mixtures thereof. Most preferred alkalizing agents are selected from the group consisting of magnesium oxide, calcium carbonate, sodium citrate and combinations thereof. In a particularly preferred embodiment, alkalizing agents are selected from the group consisting of potassium citrate, sodium citrate and combinations thereof.
- the amount of alkalizing agent should be sufficient to increase pH of a tablet above 5.5 and thus decrease the drug release in acidic media while at the same time providing a stable formulation.
- Preferably such amount is between 5 and 60% w/w, more preferably between 8 and 50 %>, based on the total weight of the pharmaceutical composition excluding any coating.
- Tofacitinib release form hydroxypropyl methylcellulose matrix tablet is faster at pH 1.2 than at pH 6.8 (figure 3 - top). Similar drug release trend was observed also for Tofacitinib formulation with
- hydroxyethylcellulose as a matrix forming polymer (figure 3 - bottom).
- This pH dependent drug release is most likely a consequence of pH dependent solubility of tofacitinib.
- Sustained release formulations that show pH independent drug release of tofacitinib citrate are prepared by incorporation of the alkalizing agents of the invention. By this approach the local pH within the tablet is modified and similar dissolution profiles are obtained in phosphate buffer at 6.8 pH and hydrochloric acid dissolution medium at pH 1.2.
- Table 1 pH and stability of tofacitinib formulations of the examples
- alkalizing agents of the invention successfully diminishes the initial burst release of tofacitinib from matrix formulations at pH 1.2 and a similar drug release is obtained regardless of the pH of the medium.
- calcium carbonate, magnesium oxide and calcium silicate alone or in combination successfully prevent the burst release in acidic medium (figure 4, example 2).
- sodium citrate is used (figure 5 and 6).
- Formulations with different quantities (9-48 %) of sodium citrate enable comparable drug release regardless of the pH of the medium ( Figure 5).
- some formulations with calcium hydrogen phosphate or calcium sulfate do not show pH independent drug release (examples 9-10, Figure 7).
- the viscosity of gel layer formed on the matrix in contact with water affects the erosion rate and consequently the drug release from the matrix formulation as well as determines the gel-layer robustness.
- the viscosity of the hydrophilic polymers determines the viscosity of the formed gel layer.
- the hydrophilic polymer is a cellulose polymer with a viscosity between 1000 and 150000 mPa-s, preferably between 2000 and 120000 mPa-s, most preferably between 2000 and 100000 mPa-s.
- the hydrophilic polymer is hydroxypropyl methylcellulose with a viscosity between 1000 and 140000 mPa-s.
- HPMC is available, for example, as Methocel® K4M or Kl 00M grade from Dow Chemical.
- the hydrophilic polymer is hydroxyethylcellulose with a viscosity between 1000 and 150000 mPa-s.
- HEC is available, for example, as Natrosol® 250 M, H or HH grade from Aqualon.
- the hydrophilic polymer is a carrageenan with a viscosity above 5 mPa-s, more preferably between 5 and 750 mPa-s. Most preferably lambda and iota carrageenans are used.
- Table 2 Tofacitinib release from matrix tablets with the composition of example 12 where only the viscosity grade of HPMC is varied, with (MS) or without mechanical stress (No MS) using Apparatus 1 , 100 RPM, 900 ml of pH 6.8 medium.
- Table 3 Tofacitinib release from hydrophilic cellulose matrix tablets with the composition of example 15 where only the viscosity grade of HEC is varied, with (MS) or without mechanical stress (No MS) using Apparatus 1, 100 RPM, 900 ml of pH 6.8 medium.
- the formulations of the invention show a pH independent drug release and are at the same time mechanically robust.
- the formulation of example 2 when tested according to discriminatory dissolution method for mechanical robustness shows that the addition of the alkalizing agents of the invention does not negatively effects the resistance of the formulation to mechanical stress (Figure 10).
- Formulations prepared with mixtures of cellulose and carrageenan polymers are especially preferred.
- a particularly preferred embodiment of the present invention relates to formulations comprising hydroxyethylcellulose and carrageenan, which shows characteristics superior to those of other sustained release formulations, i.e. pH independent drug release, zero-order drug release and high robustness to mechanical stress.
- the solubility of the drug in the polymeric matrix is a critical property that defines the drug release mechanism. For more soluble drugs the release is more diffusion controlled, for drugs with lower solubility the release is erosion controlled.
- Drug release profiles for hydrophilic matrices are first order for highly soluble drug or zero-order for practically insoluble drugs. To obtain more zero-like drug release also for more soluble drugs like tofacitinib is more challenging with matrix formulations.
- a particularly preferred embodiment consists of bilayer tablets. A hydrophilic polymer and Tofacitinib are combined in a first layer and an additional API free layer is applied on only one side of the first layer. The drug release from the first layer is restricted by the second layer and an initial burst effect is reduced.
- Several formulations of the invention were prepared with different hydrophilic polymers incorporated in the second layer to achieve a zero-order drug release.
- Q (t) is the percentage of drug released at a given time point t, k is release rate constant.
- n can be used as a criterion to evaluate the release-mechanism kinetics and usually assumed values between 0.45 and 0.89 (for cylindrical shapes).
- n is equal to 0.89
- the release kinetic is of zero order, and hence, with values for n approaching 0.89, the dissolution profile of a drug becomes progressively more linear. From calculated values n it is clearly evident that the release mechanism in case of bilayer tablets is more linear compared to standard HPMC matrix tablet, as in all bilayer tablets n is greater compared to basic HPMC matrix tablet.
- Drug release rate is also slower which is reflected in the calculated release rate constant k (Table 4).
- Bilayer tablets were also tested for mechanical robustness. It is crucial from the safety aspect for patient that both layers are adhered well to each other and do not separate during tablet manipulation (packaging operation, taking form blister packaging or even in the gastrointestinal tract). Separation of both layers could lead to faster drug release and higher Cmax plasma concentrations. We have showed that mechanical stress during dissolution testing that simulates mechanical stress in the gastrointestinal tract does not affect the release of tofacitinib form tested tablets Table 5.
- Table 5 Tofacitinib release from bilayer tablets of example 16 (25% HPMC in first layer) with different polymers in the second layer (A - HPMC, B - PEO, C - HEC) with (MS) or without mechanical stress (No MS) using apparatus 1, 100 RPM, 900 ml of pH 6.8 media
- the formulations of the invention can be prepared either with wet or dry granulation or, in the case of tablets, with direct compression.
- wet granulation low shear, high shear as well as fluid bed granulation techniques can be used.
- aqueous solution or pure water may be utilized to prepare granulating solution.
- the tablets were compressed using ordinary tableting equipment.
- the size of a tablet can range from 2 to 20 mm, preferably from 8 to 13 mm for normal tablets and from 2 to 5 mm for mini tablets. Tablets can be further coated. Coating can either modulate the drug release or just provide better tablet recognition by patient (non-functional coating). In case of mini tablet they can be optionally encapsulated.
- Formulations of the invention can further comprise further excipients, preferably selected from the group consisting of fillers, disintegrants, binders, lubricants, glidants, coloring agents and coating agents.
- the formulations of the invention attain pH independent drug release avoiding a burst at the acidic pH of the stomach, are mechanically robust and do not show a food effect. They also prevent dose dumping or a high burst effect. They cause reduced drug level fluctuation in plasma and thus present reduced side effects. They are suitable for once-daily administration regimen and allow for improved patient compliance.
- the viscosity of a hydrophilic polymer refers to the viscosity of a water solution containing 2 % of the hydrophilic polymer calculated on a dry basis and measured with a Brookfield viscometer at 20°C, according to the method disclosed in the USP35-NF30 S2, Chapter Hypromellose.
- the viscosity of a carrageenan refers to the viscosity of a water solution containing 1.5 % of the carrageenan and measured using a rotational viscometer at 75°C, according to the method disclosed in the USP35-NF30 S2, Chapter Carrageenan.
- a discriminatory dissolution method was used, which simulates gastrointestinal mechanical stress and transition through pylorus and is thus suitable to detect burst release for extended release formulations.
- the test was performed using Apparatus 1 (basket method), 100 rpm and 900 ml phosphate buffer medium at pH 6.8. After lh 30 minutes from the beginning of the test formulations were transferred into plastic tubes containing 5 ml of medium and 15 glass beads of 8 mm diameter and were then shaken for 10 minutes on a laboratory shaker at 300 strokes per minute. Afterwards formulations were transferred back to baskets and the dissolution test continued through the remaining sampling time points. Dissolution profiles obtained from this test were compared to profiles obtained from an analogous test without the glass beads manipulation phase. To evaluate the influence of alkalizing agents, formulations were tested with a pH simulation test (pH 1.2
- Simulation test was performed using Apparatus 2 (paddle method) at 100 rpm with JP basket sinkers to prevent floating.
- Tofacitinib degradation products were followed by high performance liquid chromatography.
- Formulations were dissolved in a mixture of 40 w/w% methanol in water to achieve a concentration of 0.2 mg/ml of tofacitinib.
- the sample solution was injected into an HPLC system with a HSS T3 column (1.7 micrometer particles) using binary gradient elution.
- Mobile phase A consisted of 0.1% trifluoroacetic acid in water and mobile phase B consisted of 50% vol. mixture of acetonitrile and mobile phase A.
- Gradient elution was performed according to the following program: mobile phase A (%) / time (min): 80%>/0min; 0%>/8min; 80%>/8.5min.
- the detector was set to a wavelength of 290 nm and impurities quantitated as area percentage with no response factors applied.
- pH stability profile of Tofacitinib citrate was determined in the following manner: one ml of tofacitinib citrate solution (2 mg/ml) was pipetted into a 10 ml beaker. To this solution, 2 ml of a buffer solution was added and left to stand overnight (20-25°C, dark). Samples were then neutralized (if needed), filled to the 10 ml mark with solvent and analyzed by HPLC under the conditions described for monitoring degradation products.
- Tofacitinib is extracted from sodium heparinized human plasma by 96-well solid phase extraction. Before the extraction, radiolabeled tofacitinib is added as an internal standard. The samples are eluted with 13% NH 4 OH in methanol, evaporated to dryness and reconstituted with 50% methanol in water. The reconstituted sample is injected into an LC/MS/MS system using a Synergi Polar-RP column with a mobile phase of 40% 10 mM ammonium acetate and 60%> methanol (with 0.05%> formic acid).
- Core function example 1 example 2 mg/tablet mg/tablet
- magnesium stearate All ingredients, except magnesium stearate, were mixed together and sieved through 0.75 mm mesh sieve. Then sieved (0.5 mm mesh size) magnesium stearate was added and the mixture was additionally blended. The final blend was compressed into tablets at compression forces ranging from 10 to 25 kN using round 10 or 11 mm punches.
- Sodium citrate was first sieved through a 0.5 mm sieve. Sprayed dried lactose, microcrystalline cellulose, hydroxypropyl methylcellulose and tofacitinib citrate were mixed together and sieved through a 0.75 mm mesh sieve. Sieved sodium citrate was added and blended for 3 minutes. Then sieved (0.5 mm mesh size) magnesium stearate was added and the mixture was additionally blended. The final blend was compressed into tablets at compression forces ranging from 9 to 17 kN using round 11 mm punches.
- Core function example 6 example 7 mg/tablet mg/tablet
- HEC Hydrophilic cellulose
- Core function example 9 example 10 mg/tablet mg/tablet
- Table 1 1 Hydrophilic cellulose matrix tablets with different levels of HPMC
- Core function example 11 example 12 example 13 mg/tablet mg/tablet mg/tablet
- Hydroxypropyl methylcellulose matrix polymer 60.00 100.00 140.80
- Tofacitinib citrate is mixed with part of the hydroxypropyl methylcellulose, lactose and part of the microcrystalline cellulose.
- the mixture is granulated with water in a high shear mixer or fluid bed.
- the granulate is dried in oven or a fluid bed dryer.
- the dry mixture is sieved or milled through appropriate mesh size.
- the remaining microcrystalline cellulose and hydroxypropyl methylcellulose are mixed with the granulate.
- At the end sieved magnesium stearate is added and mixed together and tablets are compressed.
- Core function example 14 example 15 mg/tablet mg/tablet
- Example 14 Sprayed dried lactose, microcrystalline cellulose, hydroxyethylcellulose and tofacitinib citrate were mixed together and sieved through 0.75 mm mesh sieve. Then sieved (0.5 mm mesh size) magnesium stearate was added and the mixture was additionally blended. The final blend was compressed at compression forces ranging from 15 to 23 kN into tablets using round 10 mm punches.
- Example 15 Tofacitinib citrate is mixed with part of the hydroxyethylcellulose, lactose monohydrate and part of the microcrystalline cellulose. The mixture is granulated with water in a high shear mixer or fluid bed. The granulate is dried in oven or a fluid bed dryer. Dry mixture is sieved or milled through appropriate mesh size. Microcrystalline cellulose and the remaining hydroxyethylcellulose are mixed with the granulate. At the end sieved magnesium stearate is added and mixed together and tablets are compressed.
- Hydroxypropyl methylcellulose (type 2208 USP; matrix polymer 100.00
- hydroxyethylcellulose (Natrosol 250 HHX grade)
- Tofacitinib is mixed with part of hydro xypropyl methylcellulose, lactose and part of microcrystalline cellulose. The mixture is granulated with water in a high shear mixer or fluid bed. The granulate is dried in oven or a fluid bed dryer. Dry mixture is sieved or milled through appropriate mesh size. Microcrystalline cellulose and the remaining hydroxypropyl methylcellulose and mixed with the granulate.
- Second layer is added into the die and the bilayer tablet is compressed with compression force ranging from 8 - 23 kN.
- Lactose, microcrystalline cellulose, hydroxyethylcellulose and tofacitinib citrate were mixed together and sieved through 0.75 mm mesh sieve.
- the mixture was granulated with water in a high shear mixer of fluid bed.
- the granulate was dried in oven or fluid bed dryer.
- the resulting dry mixture was sieved or milled through an appropriate sieve.
- Carrageenan and magnesium stearate were added and the mixture was additionally blended and tablets were compressed.
- Table 15 Reference examples 1 - 4 corresponding respectively to example 1, 4, 5 and 9 of WO
- Reference examples 1 - 4 were tested according to discriminatory dissolution to evaluate mechanical susceptibility of matrix tablets. As it can be appreciated from table 16, drug release from reference samples was much faster under mechanical stress. Drug release from the formulation of reference example 3 was very fast even without mechanical stress so that such formulation cannot even be considered to be a sustained release formulation. Table 16: Drug release in dissolution medium with (MS) or without mechanical stress (No MS) using apparatus I, 100 RPM, 900 ml of pH 6.8 medium
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Preparation (AREA)
Abstract
Sustained release formulations comprising tofacitinib or a pharmaceutically acceptable salt thereof show reduced drug level fluctuation in plasma, pH independent release, mechanical robustness and no food effect and are suitable for once daily administration.
Description
SUSTAINED RELEASE FORMULATIONS OF TOFACITINIB
FIELD OF INDUSTRIAL APPLICABILITY
The present invention relates to sustained release formulations for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof.
BACKGROUND OF THE DISCLOSURE
Tofacitinib, 3- {(3R,4R)-4-methyl-3-[methyl-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)- amino]-piperidin-l -yl}3-oxo-propionitrile, is an inhibitor of the enzyme Janus Kinase 3 (JAK3) indicated for treatment of rheumatoid arthritis. Its synthetic preparation is disclosed in WO 02/096909 Al, WO 03/048162 Al and WO 2007/012953 A2.
The recommended dose is 5 mg given orally twice daily. The FDA label contains a black-box warning alerting about the risk of serious side effects, such as serious infections (tuberculosis, invasive fungal and viral infections), lymphoma and other malignancies. Serious concerns about the risk of malignancy with 10 mg dose were raised by the FDA and eventually this dosage was not approved. According to the prescription label the risk of serious side effects increases in a dose and time-dependant fashion. An increased frequency of side effects of drugs is usually observed when peak plasma concentrations exceed a certain level. Tofacitinib exhibits linear pharmacokinetics, with the systemic exposure (AUCM) and peak plasma concentration (Cmax) increasing in proportion to the dose in the dose range of 1 to 100 mg.
Therefore peak plasma concentrations of the 10 mg tofacitinib dose are approximately two times higher than those of the 5 mg dose and they may be too high for a safe drug administration.
Plasma concentration of Tofacitinib after oral administration is directly related to the release rate of the drug in the gastrointestinal tract. With an immediate release (IR) formulation a fast drug release is achieved giving rise to high peak plasma concentrations which are not sustained through a longer period of time.
Sustained peak plasma concentrations can theoretically be achieved by means of sustained release matrix systems. However, when such systems are made of hydrophilic polymers, they seldom provide pH independent drug release of pH-dependent soluble drugs and they are normally incapable of attaining zero-order release except for practically insoluble drugs. From FDA published data it can be seen that the aqueous solubility of Tofacitinib citrate is pH-dependent with higher solubility at low pH and drastically decreased solubility at pH above 3.9.
pH dependent solubility can lead to pH-dependent in vivo drug release from sustained release matrices. Drug release varies as a function of movement through segments of the gastrointestinal tract with different pH. This can lead to inefficient drug delivery and bigger inter-subject variability, since pH in the gastrointestinal tract varies significantly between subjects. Sustained release formulations that would provide pH independent drug release throughout whole gastrointestinal tract are very desirable.
For extended release formulations, the robustness of drug release is also critical, since mechanical stress applied to matrix tablets during gastrointestinal transit may lead to faster disruption of the gel layer and influence the plasma drug concentrations over time. This can change the therapeutic efficacy and safety. Different mechanical stress can be exerted on matrix tablets due to variety of gastrointestinal conditions. In a fasting state, the stress on the tablet mainly depends on the migrating myoelectric complex (MMC), which cycles every 90 to 120 min and normally starts in the stomach. The movement of tablets during this phase is rapid and tablets are subject to increased mechanical stress, especially during gastric emptying. Additionally, gastrointestinal motility is elevated during and after food intake. This causes even greater mechanical stress on pharmaceutical compositions. There is also evidence that food intake stimulates the transport of gastrointestinal content from the terminal small intestine into the colon, a mechanism that is known as gastro-ileal or gastro-ileocecal reflex, which can again exert elevated mechanical stress on the matrix tablet.
WO 2012/100949 Al discloses oral dosage forms for modified release comprising Tofacitinib. Such formulations are disclosed in very general terms and give no direction to the skilled man for preparing sustained formulations with adequate release profile and properties. 10 examples of specific formulations are also provided, but no dissolution test results. When reproduced, such formulations showed poor release properties, liberating the active ingredient too fast for any practical application, and proved not to be resistant to mechanical stress.
There is thus still the need for Tofacitinib formulations that are mechanically robust and do not show a food effect. There is also the need for Tofacitinib formulations that attain a zero order release profile.
In addition there is still the need for Tofacitinib formulations that provide reduced drug level fluctuation in plasma, resulting in diminished side effects. There is also the need for an improved dosage regimen to increase patient compliance.
SUMMARY OF THE DISCLOSURE
The present disclosure provides a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof, a hydrophilic polymer and an alkalizing agent, wherein the pH at 25 °C of a 1% weight/volume water solution or dispersion of the formulation is between 5.5 and 11. It further provides a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof and a polymer, wherein the hydrophilic polymer is either a cellulose derivative with a viscosity between 1000 and 150000 mPa-s, a carrageenan with a viscosity of 5 mPa-s or higher or a mixture thereof. A once daily dosage regimen of the formulations of the invention, preferably for the treatment of rheumatoid arthritis, is also disclosed.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 illustrates simulated plasma profiles after twice daily administration of immediate release formulations containing 5 mg and 10 mg of tofacitinib (full and dashed line) and once daily administration of a sustained release formulation containing 11 mg of Tofacitinib (dotted line).
FIG. 2 illustrates the stability profile of Tofacitinib citrate at various pH.
FIG. 3 illustrates the dissolution test results of the formulations of example 1 (25% HPMC without alkalizing agent) - top - and example 15 (60 % of HEC without alkalizing agent) - bottom - using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
FIG. 4 illustrates the dissolution test results of the formulation of example 2 (25% HPMC with insoluble alkalizing agents) using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
FIG. 5 illustrates the dissolution test results of the formulations of example 3 (25% HPMC with 9 % sodium citrate) - top - and of example 5 (25%> HPMC with 48 % sodium citrate) - bottom -using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
FIG. 6 illustrates the dissolution test results of the formulation of example 8b (60% HEC with 25 % sodium citrate) using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
FIG. 7 illustrates the dissolution test results of the formulations of example 9 (25%> HPMC with calcium hydrogen phosphate dihydrate) - top - and 10 (25% HPMC with calcium sulphate) - bottom - using apparatus 2 at 100 rpm and dissolution media with different pH: dashed line at pH 1.2 + 6.8; full line at pH 6.8.
FIG. 8 illustrates the dissolution test results of the formulations of examples 11-13 (15 % circle, 25 % triangle and 35 > diamond HPMC) with (dotted lines) or without mechanical stress (full lines) using
apparatus 1, 100 RPM, 900 ml of pH 6.8 medium.
FIG. 9 illustrates the dissolution test results of the formulations of examples 14 and 15 (30 % circle, 60% diamond HEC) with (dotted lines) or without mechanical stress (full lines) using apparatus 1, 100 RPM, 900 ml of pH 6.8 medium.
FIG. 10 illustrates the dissolution test results of the formulations of example 2 (25 % HPMC with insoluble alkalizing agents) with (dotted lines) or without mechanical stress (full lines) using apparatus 1 , 100 RPM, 900 ml of pH 6.8 medium.
FIG. 11 illustrates the dissolution test results of the formulations of example 17 (HEC + carrageenan) using apparatus 2 at 100 rpm and dissolution media with different pH (dotted line pH 1.2 + 6.8; full line 6.8 ) - top - and with (dotted line) or without mechanical stress (full line) using Apparatus 1, 100 RPM, 900 ml of pH 6.8 medium - bottom -.
FIG. 12 illustrates a schematic view of a hydrophilic matrix bilayer tablet comprising the active pharmaceutical ingredient (API) tofacitinib and a sustained release (SR) polymer.
DEFINITIONS
As used herein sustained release formulation refers to extended-release formulations as defined by the USP35-NF30 S2, Chapter 1151. Sustained release formulations make the active substance available over an extended period of time following ingestion. Sustained release formulations according to the invention release not more than 30% of Tofacitinib within 1 hour and at least 80 %> of tofacitinib between 4 and 18 hours using apparatus 2 USP at 100 rpm for 2 hours in 500 ml of 0.1 M hydrochloric acid medium with 2 g/1 of sodium chloride (pH= 1.2), followed by 900 ml of pH 6.8 medium.
As used herein hydrophilic polymer refers to water soluble polymeric materials that in contact with an aqueous environment swell and form a gel matrix. They are characterized by the presence of polar groups attached to the main polymer backbone of the molecule (see C.A. Finch: Hydrophilic polymers. In: R.W.Dyson (ed.) Specialty Polymers; Blackie&Son Ltd 1987).
As used herein alkalizing agent refers to any agent that counteracts or neutralizes acidity and/or increases the concentration of hydroxide anions (OH ) in water.
DETAILED DESCRIPTION OF THE DISCLOSURE
An ideal sustained release formulation releases the drug with a constant rate between 4 and 18 hours, preferably over 16 hours (zero-order release), and provides effective drug plasma concentration levels
over a longer period of time, while at the same time not exceeding the peak plasma concentration of the immediate release formulation, as shown in FIG.l . This results in improved safety and efficacy of the drug product.
The plasma profiles in FIG.l were simulated by solving one-compartment pharmacokinetic differential equation - dA/dt = ra(t)-keiA - where A is the amount of drug in plasma, ra(t) in vivo dissolution/absorption rate and kd elimination constant of drug from plasma. The plasma concentration is then obtained by dividing the amount of drug A by the volume of distribution Vd. The pharmacokinetic parameters were obtained from "NDA 203214 (Tofacitinib), Clinical Pharmacology and Biopharmaceutics Review (s), FDA": Vd=87 L, kd=0.23/h Im = 3 h). The drug is completely absorbed with about 20% of first-pass elimination. The absorption rate for the immediate release formulation is dictated by the transit of drug from the stomach into the intestine, where the absorption takes place. The dissolution rate for the sustained release formulation was chosen to mimic the preferred constant (i.e. zero-order) release over 16 hours.
Such sustained release formulation has to be carefully developed to provide drug release that would give optimal plasma concentrations and at the same time be robust enough to prevent dose dumping or a high burst effect. Both phenomena cause the premature and exaggerated release of a drug that can greatly increase its concentration in the body and thereby produce adverse effects. Dose dumping can be a consequence of poor sustained release formulation and several in vivo factors such as mechanical stress in the gastrointestinal tract (i.e. passage of formulation from the stomach into the duodenum and from the ileum to the colon), presence of food or alcohol in the stomach, differences in pH along the
gastrointestinal tract. Burst release is a phenomenon where an initial large bolus of drug is released before the release rate reaches a stable profile. It is related to some formulations (e.g. hydrophilic matrix systems) and well soluble drugs (S. B. Tiwari, A. R. Rajabi-Siahboomi, "Extended-release oral drug delivery technologies: Monolithic Matrix systems ", Methods in Molecular Biology, Drug Delivery Systems, Vol. 437, 217-243.).
Therefore one object of the present invention is the provision of Tofacitinib formulations causing reduced drug level fluctuation in plasma.
Another object of the present invention is the provision of formulations of Tofacitinib with reduced side effects.
Another object of the present invention is the provision of Tofacitinib formulations with pH independent
release avoiding a burst at the acidic pH of the stomach.
Another object of the present invention is the provision of Tofacitinib formulations that are mechanically robust.
Another object of the present invention is the provision of Tofacitinib formulations that prevent dose dumping or a high burst effect.
Another object of the present invention is the provision of Tofacitinib formulations that do not show a food effect.
Another object of the present invention is the provision of Tofacitinib formulations with a zero order release profile.
Another object of the present invention is the provision of a dosage regimen for Tofacitinib with improved patient compliance.
One embodiment of the present invention is a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof, a hydrophilic polymer and an alkalizing agent, wherein the pH at 25°C of a 1% weight/volume water solution or dispersion of the formulation is between 5.5 and 11.
Another embodiment of the present invention is a sustained release formulation for oral administration comprising tofacitinib or a pharmaceutically acceptable salt thereof and a hydrophilic polymer, wherein the hydrophilic polymer is either a cellulose derivative with a viscosity between 1000 and 150000 mPa-s, a carrageenan with a viscosity greater than 5 mPa-s or a mixture thereof.
Formulations of the invention are matrix formulations where a rate-controlling hydrophilic polymer forms a matrix through which the drug is dissolved or dispersed. Such formulations can be in the form of tablets, minitablets (that can be further inserted in capsules), pellets and granules, preferably in the form of tablets. Tablets can be monolayer or multilayer, with tofacitinib included in one or more layers. Tablets can be further coated with a functional or a non-functional coating.
In a preferred embodiment the formulations of the invention contain tofacitinib citrate.
In a preferred embodiment the formulations of the invention comprise between 5 and 25 mg, more preferably between 20 and 22 mg or between 10 and 12 mg of tofacitinib or a pharmaceutically acceptable salt thereof, the amount being calculated based on tofacitinib free base.
In a preferred embodiment the formulations of the invention comprise tofacitinib or a pharmaceutically acceptable salt thereof in an amount between 10 and 12 mg expressed as tofacitinib free base and, wherein such formulations are administered once a day, the Cmax of tofacitinib measured in plasma is below 35 ng/ml, preferably between 25 and 35 ng/ml.
Hydrophilic polymers include natural gums and polysaccharides (such as alginates, xanthan, carrageenan, locust bean gum, chitosan, guar gum, pectin, crossed-linked high amylose starch, gelatine), semisynthetic materials (such as cellulose derivatives, propylene glycol alginate) and synthetic materials (such as polyethylene oxide and homo- and copolymers of acrylic acid). Preferred hydrophilic polymers are cellulose derivatives (such methylcellulose, hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethyl cellulose, carboxymethyl cellulose sodium), carrageenan and mixtures thereof. Especially preferred hydrophilic polymers are hydroxypropyl methylcellulose (HPMC), hydroxyethyl cellulose (HEC), carrageenan and mixtures thereof.
The amount of hydrophilic polymer is preferably between 15 and 80%, preferably between 20 and 70%, more preferably between 25 and 60%> w/w, based on the total weight of the pharmaceutical composition excluding any coating. If the hydrophilic polymer is present in insufficient amounts the dissolution profile is either too fast or the drug release is more easily influenced by mechanical stress. If the hydrophilic polymer is present in excessive amounts, the drug release is too slow, e.g. it could take more than 24 hours to release the drug; in addition the manufacturing of the composition may be very difficult (see figures 8 and 9).
In a preferred embodiment the hydrophilic polymer is hydroxypropyl methylcellulose (HPMC) in an amount between 20 and 60%> w/w, more preferably between 25 and 50% w/w, based on the total weight of the pharmaceutical composition excluding any coating.
In another preferred embodiment the hydrophilic polymer is hydroxyethyl cellulose (HEC) in an amount between 30 and 70% w/w, more preferably between 40 and 60%> w/w, based on the total weight of the pharmaceutical composition excluding any coating.
In another preferred embodiment the hydrophilic polymer is a carrageenan in an amount between 20 and
70 %, more preferably between 40 and 60% w/w, based on the total weight of the pharmaceutical composition excluding any coating.
In a preferred embodiment the formulations of the invention comprise an alkalizing agent in order to control the solubility of the active substance and thus obtain a pH independent drug release. The alkalizing agents of the invention lower the dissolution rate at pH 1.2, thus preventing the burst release due to tofacitinib 's greater solubility in acidic media. Although tofacitinib citrate is a very stable substance in the solid state, its solution is sensitive to alkaline pH, especially at pH 10 and above (Figure 2). Due to such instability, alkalizing agents for tofacitinib formulations must be chosen very carefully during formulation development. In order to provide at the same time pH independent drug release and adequate stability of the drug substance, the pH at 25°C of a 1% weight/volume water solution or dispersion of the formulation must be between 5.5 and 11, preferably between 6 and 10.5.
The alkalizing agents may be soluble as well as insoluble. Examples of alkalizing agents are aluminum hydroxide, bentonite, kaolin, calcium carbonate, calcium silicate, calcium hydrogen phosphate, calcium sulphate, calcium chloride, calcium lactate, sodium hydrogen carbonate, sodium stearate, sodium citrate, sodium phosphate, sodium borate, sodium lactate, sodium hyaluronate, sodium acetate, monosodium glutamate, sodium tartrate, attapulgite, potassium citrate, potassium hydrogen carbonate, potassium chloride, potassium hydrogen phosphate, tromethamine, zinc oxide, magnesium silicate, magnesium aluminum silicate, magnesium oxide, magnesium hydroxide, magnesium citrate, magnesium gluconate, dextrin, carboxymethyl cellulose sodium, carrageenan, xanthan gum and starch.
Preferred alkalizing agents are selected from the group consisting of salts of inorganic and organic acids. More preferred alkalizing agents are selected from the group consisting of metal oxides, metal hydroxides, salts of weak acids and some polymers that give pH of solution/dispersion in stated range. Even more preferred alkalizing agents are selected from the group consisting of magnesium oxide, calcium carbonate, calcium silicate, potassium citrate and sodium citrate and mixtures thereof. Most preferred alkalizing agents are selected from the group consisting of magnesium oxide, calcium carbonate, sodium citrate and combinations thereof. In a particularly preferred embodiment, alkalizing agents are selected from the group consisting of potassium citrate, sodium citrate and combinations thereof.
The amount of alkalizing agent should be sufficient to increase pH of a tablet above 5.5 and thus decrease the drug release in acidic media while at the same time providing a stable formulation. Preferably such amount is between 5 and 60% w/w, more preferably between 8 and 50 %>, based on the total weight of the pharmaceutical composition excluding any coating.
Tofacitinib release form hydroxypropyl methylcellulose matrix tablet is faster at pH 1.2 than at pH 6.8 (figure 3 - top). Similar drug release trend was observed also for Tofacitinib formulation with
hydroxyethylcellulose as a matrix forming polymer (figure 3 - bottom). This pH dependent drug release is most likely a consequence of pH dependent solubility of tofacitinib. Sustained release formulations that show pH independent drug release of tofacitinib citrate are prepared by incorporation of the alkalizing agents of the invention. By this approach the local pH within the tablet is modified and similar dissolution profiles are obtained in phosphate buffer at 6.8 pH and hydrochloric acid dissolution medium at pH 1.2.
Table 1 : pH and stability of tofacitinib formulations of the examples
The addition of the alkalizing agents of the invention successfully diminishes the initial burst release of tofacitinib from matrix formulations at pH 1.2 and a similar drug release is obtained regardless of the pH of the medium. In a particularly preferred embodiment, calcium carbonate, magnesium oxide and calcium silicate alone or in combination successfully prevent the burst release in acidic medium (figure 4, example 2). In another particularly preferred embodiment sodium citrate is used (figure 5 and 6). Formulations with different quantities (9-48 %) of sodium citrate (examples 3-5) enable comparable drug release regardless of the pH of the medium (Figure 5).
On the other hand some formulations with calcium hydrogen phosphate or calcium sulfate do not show pH independent drug release (examples 9-10, Figure 7). This is unexpected since the pH of a 10% slurry of calcium sulfate is 7.3 for the dihydrate and 10.4 for anhydrous material and the pH of a calcium hydrogen phosphate dihydrate slurry is above 7 (both data according to Handbook of pharmaceutical excipients, 7th Ed.). Surprisingly, when dissolving the above mentioned formulations in water to reach a concentration of 1 % w/v on the solution/dispersion at 25 °C and measuring the pH of the resulting solution/dispersion, the pH of the formulation of example 10 is similar to that of an HPMC tablet without alkalizing agents (4.2 and 4.0 respectively) and the pH of the formulation of example 9 has a somewhat higher pH, but still below the one needed to obtain a pH independent release.
However a high pH within the tablet can negatively affect tofacitinib stability; therefore the stability of formulations with alkalizing agents was tested and compared to that of formulations without alkalizing agent (Table 1). Amounts of degradation products were measured at two stress conditions: 80°C and 60°C / 50% RH for one week. The stability of formulations containing sodium citrate was comparable to that of formulations without any alkalizing agents, thus making sodium citrate the most preferred alkalizing agent of the invention. Stability of formulations containing calcium carbonate, calcium silicate and magnesium oxide is slightly poorer, but acceptable long term stability can be expected notwithstanding a high pH of the tablet (10.5).
The viscosity of gel layer formed on the matrix in contact with water affects the erosion rate and consequently the drug release from the matrix formulation as well as determines the gel-layer robustness. The viscosity of the hydrophilic polymers determines the viscosity of the formed gel layer.
In a preferred embodiment the hydrophilic polymer is a cellulose polymer with a viscosity between 1000 and 150000 mPa-s, preferably between 2000 and 120000 mPa-s, most preferably between 2000 and 100000 mPa-s.
In a particularly preferred embodiment, the hydrophilic polymer is hydroxypropyl methylcellulose with a viscosity between 1000 and 140000 mPa-s. Such HPMC is available, for example, as Methocel® K4M or Kl 00M grade from Dow Chemical.
In a particularly preferred embodiment, the hydrophilic polymer is hydroxyethylcellulose with a viscosity between 1000 and 150000 mPa-s. Such HEC is available, for example, as Natrosol® 250 M, H or HH grade from Aqualon.
In a particularly preferred embodiment the hydrophilic polymer is a carrageenan with a viscosity above 5 mPa-s, more preferably between 5 and 750 mPa-s. Most preferably lambda and iota carrageenans are used.
Table 2: Tofacitinib release from matrix tablets with the composition of example 12 where only the viscosity grade of HPMC is varied, with (MS) or without mechanical stress (No MS) using Apparatus 1 , 100 RPM, 900 ml of pH 6.8 medium.
It should be noted that in the reference example with 100 mPa-s HPMC (Methocel Kl 00 LV) more than 80 % of tofacitinib is released in 4 h.
Table 3: Tofacitinib release from hydrophilic cellulose matrix tablets with the composition of example 15 where only the viscosity grade of HEC is varied, with (MS) or without mechanical stress (No MS) using Apparatus 1, 100 RPM, 900 ml of pH 6.8 medium.
In a particularly preferred embodiment, the formulations of the invention show a pH independent drug release and are at the same time mechanically robust. The formulation of example 2 when tested according to discriminatory dissolution method for mechanical robustness shows that the addition of the alkalizing agents of the invention does not negatively effects the resistance of the formulation to mechanical stress (Figure 10).
Formulations prepared with mixtures of cellulose and carrageenan polymers are especially preferred. A particularly preferred embodiment of the present invention relates to formulations comprising hydroxyethylcellulose and carrageenan, which shows characteristics superior to those of other sustained release formulations, i.e. pH independent drug release, zero-order drug release and high robustness to mechanical stress.
The solubility of the drug in the polymeric matrix is a critical property that defines the drug release mechanism. For more soluble drugs the release is more diffusion controlled, for drugs with lower solubility the release is erosion controlled. Drug release profiles for hydrophilic matrices are first order for highly soluble drug or zero-order for practically insoluble drugs. To obtain more zero-like drug release also for more soluble drugs like tofacitinib is more challenging with matrix formulations.
A particularly preferred embodiment consists of bilayer tablets. A hydrophilic polymer and Tofacitinib are combined in a first layer and an additional API free layer is applied on only one side of the first layer. The drug release from the first layer is restricted by the second layer and an initial burst effect is reduced. Several formulations of the invention were prepared with different hydrophilic polymers incorporated in the second layer to achieve a zero-order drug release.
For comparison and to evaluate the release mechanism we calculated Korsmeyer-Peppas exponent n and release rate constant k by fitting the dissolution curves into equation:
Q (t) = ktn
Q (t) is the percentage of drug released at a given time point t, k is release rate constant.
The exponent n can be used as a criterion to evaluate the release-mechanism kinetics and usually assumed values between 0.45 and 0.89 (for cylindrical shapes). When n is equal to 0.89, the release kinetic is of zero order, and hence, with values for n approaching 0.89, the dissolution profile of a drug becomes progressively more linear. From calculated values n it is clearly evident that the release mechanism in case of bilayer tablets is more linear compared to standard HPMC matrix tablet, as in all bilayer tablets n is greater compared to basic HPMC matrix tablet. Drug release rate is also slower which is reflected in the calculated release rate constant k (Table 4).
Table 4: Exponent n and release rate constant k for different formulations
Formulation n k
Example 16 A 0.581 15.49
Example 16 B 0.707 12.60
Example 16 C 0.615 14.16
Example 12 0.501 24.98
Bilayer tablets were also tested for mechanical robustness. It is crucial from the safety aspect for patient that both layers are adhered well to each other and do not separate during tablet manipulation (packaging operation, taking form blister packaging or even in the gastrointestinal tract). Separation of both layers could lead to faster drug release and higher Cmax plasma concentrations. We have showed that mechanical stress during dissolution testing that simulates mechanical stress in the gastrointestinal tract does not affect the release of tofacitinib form tested tablets Table 5.
Table 5: Tofacitinib release from bilayer tablets of example 16 (25% HPMC in first layer) with different polymers in the second layer (A - HPMC, B - PEO, C - HEC) with (MS) or without mechanical stress (No MS) using apparatus 1, 100 RPM, 900 ml of pH 6.8 media
The formulations of the invention can be prepared either with wet or dry granulation or, in the case of tablets, with direct compression. In case of wet granulation low shear, high shear as well as fluid bed granulation techniques can be used. For wet granulation organic solvents, aqueous solution or pure water may be utilized to prepare granulating solution. The tablets were compressed using ordinary tableting
equipment. The size of a tablet can range from 2 to 20 mm, preferably from 8 to 13 mm for normal tablets and from 2 to 5 mm for mini tablets. Tablets can be further coated. Coating can either modulate the drug release or just provide better tablet recognition by patient (non-functional coating). In case of mini tablet they can be optionally encapsulated.
Formulations of the invention can further comprise further excipients, preferably selected from the group consisting of fillers, disintegrants, binders, lubricants, glidants, coloring agents and coating agents.
Advantages
The formulations of the invention attain pH independent drug release avoiding a burst at the acidic pH of the stomach, are mechanically robust and do not show a food effect. They also prevent dose dumping or a high burst effect. They cause reduced drug level fluctuation in plasma and thus present reduced side effects. They are suitable for once-daily administration regimen and allow for improved patient compliance.
Viscosity measurement
As used herein the viscosity of a hydrophilic polymer refers to the viscosity of a water solution containing 2 % of the hydrophilic polymer calculated on a dry basis and measured with a Brookfield viscometer at 20°C, according to the method disclosed in the USP35-NF30 S2, Chapter Hypromellose.
As used herein the viscosity of a carrageenan refers to the viscosity of a water solution containing 1.5 % of the carrageenan and measured using a rotational viscometer at 75°C, according to the method disclosed in the USP35-NF30 S2, Chapter Carrageenan.
Drug release testing
To evaluate the mechanical susceptibility of matrix formulations, a discriminatory dissolution method was used, which simulates gastrointestinal mechanical stress and transition through pylorus and is thus suitable to detect burst release for extended release formulations. The test was performed using Apparatus 1 (basket method), 100 rpm and 900 ml phosphate buffer medium at pH 6.8. After lh 30 minutes from the beginning of the test formulations were transferred into plastic tubes containing 5 ml of medium and 15 glass beads of 8 mm diameter and were then shaken for 10 minutes on a laboratory shaker at 300 strokes per minute. Afterwards formulations were transferred back to baskets and the dissolution test continued through the remaining sampling time points. Dissolution profiles obtained from this test were compared to profiles obtained from an analogous test without the glass beads manipulation phase.
To evaluate the influence of alkalizing agents, formulations were tested with a pH simulation test (pH 1.2
- pH 6.8) and the resulting dissolution profiles compared to the dissolution profiles of tests performed at pH 6.8 only. The smaller the difference between these 2 profiles, the bigger the alkalizing agent efficacy. Simulation test was performed using Apparatus 2 (paddle method) at 100 rpm with JP basket sinkers to prevent floating. The acid phase was performed for 2 hours in 500 ml of 0.1 M hydrochloric acid medium with 2 g/1 of sodium chloride (pH= 1.2). After 2 hours the medium was replaced with 900 ml of phosphate buffer at pH 6.8 and the test continued through the remaining time points.
Stability testing
Tofacitinib degradation products were followed by high performance liquid chromatography.
Formulations were dissolved in a mixture of 40 w/w% methanol in water to achieve a concentration of 0.2 mg/ml of tofacitinib. The sample solution was injected into an HPLC system with a HSS T3 column (1.7 micrometer particles) using binary gradient elution. Mobile phase A consisted of 0.1% trifluoroacetic acid in water and mobile phase B consisted of 50% vol. mixture of acetonitrile and mobile phase A. Gradient elution was performed according to the following program: mobile phase A (%) / time (min): 80%>/0min; 0%>/8min; 80%>/8.5min. The detector was set to a wavelength of 290 nm and impurities quantitated as area percentage with no response factors applied.
Stability of formulations was monitored by exposing them to two stress conditions:
- in a closed glass vial at 80°C for 7 days,
- in an open vial at 60°C and 50 % relative humidity for 7 days.
After storage in the above mentioned conditions, formulations were analyzed and the amounts of degradation products measured by HPLC. The extent of degradation was determined by subtracting the total amount of degradation products of a non-stressed (control) sample from the total amount of degradation of a stressed sample. pH stability profile of Tofacitinib citrate was determined in the following manner: one ml of tofacitinib citrate solution (2 mg/ml) was pipetted into a 10 ml beaker. To this solution, 2 ml of a buffer solution was added and left to stand overnight (20-25°C, dark). Samples were then neutralized (if needed), filled to the 10 ml mark with solvent and analyzed by HPLC under the conditions described for monitoring degradation products.
Tofacitinib plasma concentration measurement
Tofacitinib is extracted from sodium heparinized human plasma by 96-well solid phase extraction. Before the extraction, radiolabeled tofacitinib is added as an internal standard. The samples are eluted with 13% NH4OH in methanol, evaporated to dryness and reconstituted with 50% methanol in water. The
reconstituted sample is injected into an LC/MS/MS system using a Synergi Polar-RP column with a mobile phase of 40% 10 mM ammonium acetate and 60%> methanol (with 0.05%> formic acid).
The following non- limiting examples are illustrative of the disclosure.
EXAMPLES
Examples 1-2
Table 6: Hydrophilic cellulose (HPMC) matrix tablets without (example 1) and with (example 2) insoluble alkalizing agents
Core function example 1 example 2 mg/tablet mg/tablet
Tofacitinib citrate active 35.53 35.53
Hydroxypropyl methylcellulose matrix polymer 100.00 100.00
(type 2208 USP)*
Sprayed dried lactose filler 160.47 90.47
Microcrystalline cellulose filler 100.00 25.00
Calcium carbonate alkalizing agent - 112.00
Magnesium oxide alkalizing agent - 10.00
Calcium silicate alkalizing agent - 23.00
Magnesium stearate lubricant 4.00 4.00
Total 400.00 400.00
* 90 w/w % of 100000 mPa-s viscosity grade (Methocel K100M Premium CRT) and 10 w/w % of 3550 mPa-s viscosity grade (Methocel K4M Premium CR®) were mixed.
All ingredients, except magnesium stearate, were mixed together and sieved through 0.75 mm mesh sieve. Then sieved (0.5 mm mesh size) magnesium stearate was added and the mixture was additionally blended. The final blend was compressed into tablets at compression forces ranging from 10 to 25 kN using round 10 or 11 mm punches.
Examples 3-5
Table 7: Hydrophilic cellulose (HPMC) matrix tablets with sodium citrate
Core function example 3 example 4 example 5 mg/tablet mg/tablet mg/tablet
Tofacitinib citrate active 35.53 35.53 35.53
Hydroxypropyl methylcellulose matrix polymer 100.00 100.00 100.00
(type 2208 USP)*
Sprayed dried lactose filler 176.22 90.47 45.47
Microcrystalline cellulose filler 48.00 25.00 25.00
Sodium citrate alkalizing agent 36.25 145.00 190.00
Magnesium stearate lubricant 4.00 4.00 4.00
Total 400.00 400.00 400.00
* 90% w/w of 100000 mPa-s viscosity grade (Methocel K100M Premium CR®) and 10% w/w of 3550 mPa-s viscosity grade (Methocel K4M Premium CR®) were mixed.
Sodium citrate was first sieved through a 0.5 mm sieve. Sprayed dried lactose, microcrystalline cellulose, hydroxypropyl methylcellulose and tofacitinib citrate were mixed together and sieved through a 0.75 mm mesh sieve. Sieved sodium citrate was added and blended for 3 minutes. Then sieved (0.5 mm mesh size) magnesium stearate was added and the mixture was additionally blended. The final blend was compressed into tablets at compression forces ranging from 9 to 17 kN using round 11 mm punches.
Examples 6-7
Table 8: Hydrophilic cellulose (HPMC) matrix tablets with insoluble alkalizing agents
Core function example 6 example 7 mg/tablet mg/tablet
Tofacitinib citrate active 35.53 35.53
Hydroxypropyl methylcellulose matrix polymer 100.00 100.00
(type 2208 USP)*
Sprayed dried lactose filler 90.47 94.47
Microcrystalline cellulose filler 88.00 26.00
Calcium carbonate alkalizing agent 80.00 -
Magnesium oxide alkalizing agent 2.00 -
Magnesium aluminum silicate alkalizing agent - 140.00
Magnesium stearate lubricant 4.00 4.00
Total 400.00 400.00
* 90% w/w of 100000 mPa-s viscosity grade (Methocel K100M Premium CR ) and 10% w/w of 3550 mPa-s viscosity grade (Methocel K4M Premium CR®) were mixed.
Formulations were prepared according to the procedure described for examples 1 -2.
Examples 8a and 8b
Table 9: Hydrophilic cellulose (HEC) matrix tablets with alkalizing ; agents
Core function example 8a example 8b mg/tablet mg/tablet
Tofacitinib citrate active 35.53 35.53
Hydroxyethylcellulose (Natrosol® matrix polymer 240.00 240.00
250 HHX grade, 3500-5500 mPa-s)
Microcrystalline cellulose filler - 20.00
Calcium carbonate alkalizing agent 110.47 -
Magnesium oxide alkalizing agent 10.00 -
Sodium citrate alkalizing agent - 100.47
Magnesium stearate lubricant 4.00 4.00
Total 400.00 400.00
Formulations were prepared according to the procedure described for examples 1-2.
Examples 9-10
Table 10: Hydrophilic cellulose (HPMC) matrix tablets with inorganic alkalizing agents
Core function example 9 example 10 mg/tablet mg/tablet
Tofacitinib citrate active 35.53 35.53
Hydroxypropyl methyl cellulose matrix polymer 100.00 100.00
(type 2208 USP)*
Sprayed dried lactose filler - 78.97
Microcrystalline cellulose filler 100.00 21.50
Calcium hydrogen phosphate dihydrate alkalizing agent 160.47 -
Calcium sulfate alkalizing agent - 160.00
Magnesium stearate lubricant 4.00 4.00
Total 400.00
* 90% w/w of 100000 mPa-s viscosity grade (Methocel K100M Premium CR®) and 10% w/w of 3550 mPa-s viscosity grade (Methocel K4M Premium CR®) were mixed.
Formulations were prepared according to the procedure described for examples 1-2.
Examples 11-13
Table 1 1 : Hydrophilic cellulose matrix tablets with different levels of HPMC
Core function example 11 example 12 example 13 mg/tablet mg/tablet mg/tablet
Tofacitinib citrate active 35.53 35.53 35.53
Hydroxypropyl methylcellulose matrix polymer 60.00 100.00 140.80
(type 2208 USP)*
Lactose filler 180.47 160.47 149.67
Microcrystalline cellulose filler 120.00 100.00 70.00
Magnesium stearate lubricant 4.00 4.00 4.00
Total 400.00 400.00 400.00
* 90 w/w % of 100000 mPa-s viscosity grade (Methocel K100M Premium CRT) and 10 w/w % of 3550 mPa- s viscosity grade (Methocel K4M Premium CR®) were mixed.
Tofacitinib citrate is mixed with part of the hydroxypropyl methylcellulose, lactose and part of the microcrystalline cellulose. The mixture is granulated with water in a high shear mixer or fluid bed. The granulate is dried in oven or a fluid bed dryer. The dry mixture is sieved or milled through appropriate mesh size. The remaining microcrystalline cellulose and hydroxypropyl methylcellulose are mixed with the granulate. At the end sieved magnesium stearate is added and mixed together and tablets are compressed.
Examples 14-15
Table 12: Hydrophilic cellulose matrix tablets with different levels and viscosities of HEC
Core function example 14 example 15 mg/tablet mg/tablet
Tofacitinib citrate active 35.53 35.53
Hydroxyethylcellulose (Natrosol® 250)* matrix polymer 120.00 240.00
Sprayed dried lactose or milled lactose filler 150.47 90.47 monohydrate
Microcrystalline cellulose filler 90.00 30.00
Magnesium stearate lubricant 4.00 4.00
Total 400.00 400.00
* In example 14 Natrosol 250 HX grade with viscosity 1500 - 2500 mPa s and in example 15 Natrosol 250 HHX grade with viscosity 3500 - 5500 mPa were used.
Example 14: Sprayed dried lactose, microcrystalline cellulose, hydroxyethylcellulose and tofacitinib citrate were mixed together and sieved through 0.75 mm mesh sieve. Then sieved (0.5 mm mesh size) magnesium stearate was added and the mixture was additionally blended. The final blend was compressed at compression forces ranging from 15 to 23 kN into tablets using round 10 mm punches.
Example 15: Tofacitinib citrate is mixed with part of the hydroxyethylcellulose, lactose monohydrate and part of the microcrystalline cellulose. The mixture is granulated with water in a high shear mixer or fluid bed. The granulate is dried in oven or a fluid bed dryer. Dry mixture is sieved or milled through appropriate mesh size. Microcrystalline cellulose and the remaining hydroxyethylcellulose are mixed with the granulate. At the end sieved magnesium stearate is added and mixed together and tablets are compressed.
Example 16
Table 13 : Hydrophilic cellulose matrix bilayer tablets with different polymers in the second layer: A - HPMC, B - PEO and C - HEC
Core function Example 16 mg/tablet
First layer - API + hydrophilic polymer
Tofacitinib citrate active 35.53
Hydroxypropyl methylcellulose matrix polymer 100.00
(type 2208 USP)*
Lactose monohydrate filler 160.47
Microcrystalline cellulose filler 100.00
Magnesium stearate lubricant 4.00
Total first layer 400.00
Second layer - hydrophilic polymer
Hydroxypropyl methylcellulose (type 2208 USP; matrix polymer 100.00
Methocel K4M Premium CR, viscosity 3550
mPa-s), polyethylene oxide (Polyox WSR N-12K
400-800 mPa-s, 2 % solution) or
hydroxyethylcellulose (Natrosol 250 HHX grade)
Total tablet 500.00 * 90 w/w % of 100000 mPa-s viscosity grade (Methocel K100M Premium CR®) and 10 w/w % of 3550 mPa-s viscosity grade (Methocel K4M Premium CR®) were mixed.
Tofacitinib is mixed with part of hydro xypropyl methylcellulose, lactose and part of microcrystalline cellulose. The mixture is granulated with water in a high shear mixer or fluid bed. The granulate is dried in oven or a fluid bed dryer. Dry mixture is sieved or milled through appropriate mesh size. Microcrystalline cellulose and the remaining hydroxypropyl methylcellulose and mixed with the granulate. At the end sieved magnesium stearate is added and mixed together to prepare final tableting mixture of first layer. Bilayer tablets are compressed. First API layer is filled into the die and slightly compressed at relatively low compression forces (for example 2 - 4 kN). Then the second layer is added into the die and the bilayer tablet is compressed with compression force ranging from 8 - 23 kN.
Example 17
Table 14: Hydrophilic cellulose matrix tablets with HEC + carrageenan
Core function example 17 mg/tablet
Tofacitinib citrate active 35.53
Lactose filler 90.47
Microcrystalline cellulose filler 30.00
Hydroxyethylcellulose matrix polymer 80.00
(Natrosol 250 HHX grade, 3500-5500 mPa-s
viscosity)
Carrageenan λ type (Viscarine® GP-209NF) matrix polymer 80.00
Carrageenan ι type (Gelcarine® GP-379NF) matrix polymer 80.00
Magnesium stearate lubricant 4.00
Total 400.00
Lactose, microcrystalline cellulose, hydroxyethylcellulose and tofacitinib citrate were mixed together and sieved through 0.75 mm mesh sieve. The mixture was granulated with water in a high shear mixer of fluid bed. The granulate was dried in oven or fluid bed dryer. The resulting dry mixture was sieved or milled through an appropriate sieve. Carrageenan and magnesium stearate were added and the mixture was additionally blended and tablets were compressed.
Reference examples 1-4
In order to show the advantages of the formulations of the invention, four formulations disclosed in WO 2012/100949 Alwere reproduced and analyzed according to the methods described above.
Table 15 : Reference examples 1 - 4 corresponding respectively to example 1, 4, 5 and 9 of WO
2012/100949 Al
All ingredients in reference examples 1 and 2, except magnesium stearate, were sieved and blended. Then sieved magnesium stearate was added and mixture was additionally blended and compressed into tablets. In examples 3 and 4 tofacitinib citrate, Eudragit® and lactose or cellulose microcrystalline were mixed and granulated with water. Granulate was dried at 40°C. The resulting granulate was sieved and Aerosil® and magnesium stearate were mixed. The resulting mixture was compressed into tablets.
Reference examples 1 - 4 were tested according to discriminatory dissolution to evaluate mechanical susceptibility of matrix tablets. As it can be appreciated from table 16, drug release from reference samples was much faster under mechanical stress. Drug release from the formulation of reference example 3 was very fast even without mechanical stress so that such formulation cannot even be considered to be a sustained release formulation.
Table 16: Drug release in dissolution medium with (MS) or without mechanical stress (No MS) using apparatus I, 100 RPM, 900 ml of pH 6.8 medium
All the tested reference formulations release tofacitinib too fast and would not provide a suitable plasma profile for a modified release formulation. Additionally all examples prepared did not show any mechanical robustness and thus possibly expose the patients to safety risks.
Claims
1. A sustained release formulation for oral administration comprising tofacitinib or a
pharmaceutically acceptable salt thereof, a hydrophilic polymer and an alkalizing agent, wherein the pH at 25°C of a 1% weight/volume water solution or dispersion of the formulation is between 5.5 and 11.
2. The formulation of claim 1, wherein the pharmaceutically acceptable salt of Tofacitinib is
Tofacitinib citrate.
3. The formulation of claims 1-2, wherein the pH of a 1% weight/volume water solution of the
formulation is between 6 and 10.5.
4. The formulation of claims 1-3, wherein the alkalizing agent is selected from the group consisting of magnesium oxide, calcium carbonate, calcium silicate, potassium citrate, sodium citrate and combinations thereof.
5. The formulation of claims 1 -4, wherein the alkalizing agent is selected from the group consisting of potassium citrate, sodium citrate and combinations thereof.
6. The formulation of claims 1-5, wherein the hydrophilic polymer is a cellulose derivative with a viscosity between 1000 and 150000 mPa-s, or a carrageenan with a viscosity greater than 5 mPa-s, or a mixture thereof.
7. A sustained release formulation for oral administration comprising tofacitinib or a
pharmaceutically acceptable salt thereof and a hydrophilic polymer, wherein the hydrophilic polymer is a cellulose derivative with a viscosity between 1000 and 150000 mPa-s, a carrageenan with a viscosity greater than 5 mPa-s, or a mixture thereof.
8. The formulation of any of the preceding claims, wherein the amount of hydrophilic polymer is between 15 and 80% based on the total weight of the pharmaceutical composition excluding any coating.
9. The formulation of any of the preceding claims, wherein the hydrophilic polymer is selected from the group consisting of hydroxypropyl methylcellulose, hydroxyethyl cellulose, carrageenan and mixtures thereof.
10. The formulation of claims 6-8, wherein such formulation contains a mixture of the cellulose
derivative and of the carrageenan.
11. The formulation of any of the preceding claims, wherein such formulation is a tablet.
12. The formulation of any of the preceding claims, wherein such formulation is a bilayer tablet.
13. The formulation of claims 1-12 for use as a medicament, wherein such formulation is
administered once a day.
14. The formulation of claims 1-12 for use in the treatment of rheumatoid arthritis, wherein such
formulation is administered once a day.
15. A sustained release formulation for oral administration comprising tofacitinib or a
pharmaceutically acceptable salt thereof in an amount between 10 and 12 mg expressed as tofacitinib free base, wherein such formulation is administered once a day and the Cmax of tofacitinib measured in plasma is below 35 ng/ml, preferably between 25 and 35 ng/ml.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP13165577 | 2013-04-26 | ||
EP13165577.1 | 2013-04-26 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2014174073A1 true WO2014174073A1 (en) | 2014-10-30 |
Family
ID=48190278
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2014/058448 WO2014174073A1 (en) | 2013-04-26 | 2014-04-25 | Sustained release formulations of tofacitinib |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2014174073A1 (en) |
Cited By (24)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104459004A (en) * | 2014-12-16 | 2015-03-25 | 南京艾德凯腾生物医药有限责任公司 | Content determination and related substance detection method of tofacitinib citrate |
CN106176640A (en) * | 2014-11-28 | 2016-12-07 | 四川海思科制药有限公司 | Pharmaceutical composition containing tofacitinib citrate and preparation method thereof |
CN106389371A (en) * | 2016-11-16 | 2017-02-15 | 杭州朱养心药业有限公司 | Tofacitinib citrate pharmaceutical composition |
CN106420648A (en) * | 2016-11-22 | 2017-02-22 | 山东淄博新达制药有限公司 | Tofacitinib citrate tablet and preparation method thereof |
WO2017029587A1 (en) * | 2015-08-19 | 2017-02-23 | Sun Pharmaceutical Industries Limited | Sustained release oral pharmaceutical compositions of tofacitinib |
US9937181B2 (en) | 2013-03-16 | 2018-04-10 | Pfizer Inc. | Tofacitinib oral sustained release dosage forms |
CN108066319A (en) * | 2016-11-10 | 2018-05-25 | 江苏先声药业有限公司 | Tofacitinib citrate enteric sustained-release pellet and preparation method thereof |
WO2019224058A1 (en) | 2018-05-24 | 2019-11-28 | Synthon B.V. | Controlled release tofacitinib compositions |
CN110787145A (en) * | 2019-12-17 | 2020-02-14 | 南京康川济医药科技有限公司 | Tofacitinib citrate sustained-release tablet and preparation method thereof |
CN110891551A (en) * | 2017-07-20 | 2020-03-17 | 印塔斯制药有限公司 | Non-pulse sustained release betahistine oral solid composition |
CN111150711A (en) * | 2018-11-07 | 2020-05-15 | 上海博志研新药物技术有限公司 | Tofacitinib controlled-release tablet, preparation method and application thereof |
CN111184696A (en) * | 2020-02-26 | 2020-05-22 | 江苏艾立康药业股份有限公司 | Tofacitinib citrate sustained-release tablet and preparation method thereof |
WO2020138791A3 (en) * | 2018-12-28 | 2020-08-20 | 주식회사 대웅제약 | Sustained-release preparation comprising tofacitinib or pharmaceutically acceptable salt thereof and manufacturing method therefor |
WO2020194081A1 (en) * | 2019-03-27 | 2020-10-01 | Unichem Laboratories Limited | Extended release composition of tofacitinib |
WO2021038014A1 (en) | 2019-08-29 | 2021-03-04 | Synthon B.V. | Controlled release tofacitinib compositions |
WO2021169724A1 (en) * | 2020-02-26 | 2021-09-02 | 上海博志研新药物技术有限公司 | Sustained-release dosage form of tofacitinib, preparation method therefor and application thereof |
WO2022254017A1 (en) | 2021-06-04 | 2022-12-08 | Synthon B.V. | Prolonged release tofacitinib compositions |
US20230270666A1 (en) * | 2020-07-28 | 2023-08-31 | Fang Zhou | Novel gastroretentive extended release dosage form |
WO2024042218A1 (en) | 2022-08-26 | 2024-02-29 | Synthon B.V. | Prolonged release tofacitinib compositions without functional coating |
US11976077B2 (en) | 2015-10-16 | 2024-05-07 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-α]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms therof |
US11993606B2 (en) | 2015-10-16 | 2024-05-28 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US11993605B2 (en) | 2015-10-16 | 2024-05-28 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US12077545B2 (en) | 2015-10-16 | 2024-09-03 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US12365689B2 (en) | 2015-10-16 | 2025-07-22 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2002096909A1 (en) | 2001-05-31 | 2002-12-05 | Pfizer Products Inc. | Optical resolution of (1-benzyl-4-methylpiperidin-3-yl) -methylamine and the use thereof for the preparation of pyrrolo 2,3-pyrimidine derivatives as protein kinases inhibitors |
WO2003048162A1 (en) | 2001-12-06 | 2003-06-12 | Pfizer Products Inc. | Novel crystalline compound |
WO2007012953A2 (en) | 2005-07-29 | 2007-02-01 | Pfizer Products Inc. | Pyrrolo[2,3-d]pyrimidine derivatives; their intermediates and synthesis |
WO2012100949A1 (en) | 2011-01-27 | 2012-08-02 | Ratiopharm Gmbh | Oral dosage forms for modified release comprising tasocitinib |
US20120195966A1 (en) * | 2011-01-27 | 2012-08-02 | Frank Sievert | Oral dosage form for modified release comprising a jak3 inhibitor |
-
2014
- 2014-04-25 WO PCT/EP2014/058448 patent/WO2014174073A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2002096909A1 (en) | 2001-05-31 | 2002-12-05 | Pfizer Products Inc. | Optical resolution of (1-benzyl-4-methylpiperidin-3-yl) -methylamine and the use thereof for the preparation of pyrrolo 2,3-pyrimidine derivatives as protein kinases inhibitors |
WO2003048162A1 (en) | 2001-12-06 | 2003-06-12 | Pfizer Products Inc. | Novel crystalline compound |
WO2007012953A2 (en) | 2005-07-29 | 2007-02-01 | Pfizer Products Inc. | Pyrrolo[2,3-d]pyrimidine derivatives; their intermediates and synthesis |
WO2012100949A1 (en) | 2011-01-27 | 2012-08-02 | Ratiopharm Gmbh | Oral dosage forms for modified release comprising tasocitinib |
US20120195966A1 (en) * | 2011-01-27 | 2012-08-02 | Frank Sievert | Oral dosage form for modified release comprising a jak3 inhibitor |
Non-Patent Citations (4)
Title |
---|
C.A. FINCH: "Hydrophilic polymers", 1987, BLACKIE&SON LTD |
M. SIMON: "Ulcerative colitis: medical management", CÔLON & RECTUM, vol. 7, no. 1, 1 February 2013 (2013-02-01), pages 11 - 17, XP055126727, ISSN: 1951-6371, DOI: 10.1007/s11725-013-0434-6 * |
NDA 203214 (TOFACITINIB), CLINICAL PHARMACOLOGY AND BIOPHARMACEUTICS REVIEW (S), FDA |
S. B. TIWARI; A. R. RAJABI-SIAHBOOMI: "Extended-release oral drug delivery technologies: Monolithic Matrix systems", METHODS IN MOLECULAR BIOLOGY, DRUG DELIVERY SYSTEMS, vol. 437, pages 217 - 243, XP009165010, DOI: doi:10.1007/978-1-59745-210-6_11 |
Cited By (37)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11253523B2 (en) | 2013-03-16 | 2022-02-22 | Pfizer Inc. | Tofacitinib oral sustained release dosage forms |
US10639309B2 (en) | 2013-03-16 | 2020-05-05 | Pfizer Inc. | Tofacitinib oral sustained release dosage forms |
US9937181B2 (en) | 2013-03-16 | 2018-04-10 | Pfizer Inc. | Tofacitinib oral sustained release dosage forms |
CN106176640B (en) * | 2014-11-28 | 2019-12-20 | 四川海思科制药有限公司 | Pharmaceutical composition containing tofacitinib citrate and preparation method thereof |
CN106176640A (en) * | 2014-11-28 | 2016-12-07 | 四川海思科制药有限公司 | Pharmaceutical composition containing tofacitinib citrate and preparation method thereof |
CN104459004A (en) * | 2014-12-16 | 2015-03-25 | 南京艾德凯腾生物医药有限责任公司 | Content determination and related substance detection method of tofacitinib citrate |
WO2017029587A1 (en) * | 2015-08-19 | 2017-02-23 | Sun Pharmaceutical Industries Limited | Sustained release oral pharmaceutical compositions of tofacitinib |
US12110297B2 (en) | 2015-10-16 | 2024-10-08 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US12116373B2 (en) | 2015-10-16 | 2024-10-15 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US12103933B2 (en) | 2015-10-16 | 2024-10-01 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US12091415B2 (en) | 2015-10-16 | 2024-09-17 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US12134621B2 (en) | 2015-10-16 | 2024-11-05 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US12077545B2 (en) | 2015-10-16 | 2024-09-03 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-N-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US11993605B2 (en) | 2015-10-16 | 2024-05-28 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US11993606B2 (en) | 2015-10-16 | 2024-05-28 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
US11976077B2 (en) | 2015-10-16 | 2024-05-07 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-α]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms therof |
US12365689B2 (en) | 2015-10-16 | 2025-07-22 | Abbvie Inc. | Processes for the preparation of (3S,4R)-3-ethyl-4-(3H-imidazo[1,2-a]pyrrolo[2,3-e]-pyrazin-8-yl)-n-(2,2,2-trifluoroethyl)pyrrolidine-1-carboxamide and solid state forms thereof |
CN108066319B (en) * | 2016-11-10 | 2022-01-14 | 江苏先声药业有限公司 | Tofacitinib citrate enteric sustained-release pellet and preparation method thereof |
CN108066319A (en) * | 2016-11-10 | 2018-05-25 | 江苏先声药业有限公司 | Tofacitinib citrate enteric sustained-release pellet and preparation method thereof |
CN106389371A (en) * | 2016-11-16 | 2017-02-15 | 杭州朱养心药业有限公司 | Tofacitinib citrate pharmaceutical composition |
CN106420648A (en) * | 2016-11-22 | 2017-02-22 | 山东淄博新达制药有限公司 | Tofacitinib citrate tablet and preparation method thereof |
CN110891551A (en) * | 2017-07-20 | 2020-03-17 | 印塔斯制药有限公司 | Non-pulse sustained release betahistine oral solid composition |
WO2019224058A1 (en) | 2018-05-24 | 2019-11-28 | Synthon B.V. | Controlled release tofacitinib compositions |
CN111150711A (en) * | 2018-11-07 | 2020-05-15 | 上海博志研新药物技术有限公司 | Tofacitinib controlled-release tablet, preparation method and application thereof |
CN111150711B (en) * | 2018-11-07 | 2023-04-07 | 上海博志研新药物技术有限公司 | Tofacitinib controlled-release tablet, preparation method and application thereof |
WO2020138791A3 (en) * | 2018-12-28 | 2020-08-20 | 주식회사 대웅제약 | Sustained-release preparation comprising tofacitinib or pharmaceutically acceptable salt thereof and manufacturing method therefor |
WO2020194081A1 (en) * | 2019-03-27 | 2020-10-01 | Unichem Laboratories Limited | Extended release composition of tofacitinib |
WO2021038014A1 (en) | 2019-08-29 | 2021-03-04 | Synthon B.V. | Controlled release tofacitinib compositions |
CN110787145A (en) * | 2019-12-17 | 2020-02-14 | 南京康川济医药科技有限公司 | Tofacitinib citrate sustained-release tablet and preparation method thereof |
WO2021169724A1 (en) * | 2020-02-26 | 2021-09-02 | 上海博志研新药物技术有限公司 | Sustained-release dosage form of tofacitinib, preparation method therefor and application thereof |
CN111184696A (en) * | 2020-02-26 | 2020-05-22 | 江苏艾立康药业股份有限公司 | Tofacitinib citrate sustained-release tablet and preparation method thereof |
EP4188366A4 (en) * | 2020-07-28 | 2024-09-04 | Novelstar Pharmaceuticals, Inc. | NEW GASTRORETENTIVE DOSAGE FORM WITH EXTENDED RELEASE |
US11844866B2 (en) * | 2020-07-28 | 2023-12-19 | Novelstar Pharmaceuticals, Inc. | Gastroretentive extended release dosage form |
US20230270666A1 (en) * | 2020-07-28 | 2023-08-31 | Fang Zhou | Novel gastroretentive extended release dosage form |
US20240382427A1 (en) * | 2020-07-28 | 2024-11-21 | Novelstar Pharmaceuticals, Inc. | Novel gastroretentive extended release dosage form |
WO2022254017A1 (en) | 2021-06-04 | 2022-12-08 | Synthon B.V. | Prolonged release tofacitinib compositions |
WO2024042218A1 (en) | 2022-08-26 | 2024-02-29 | Synthon B.V. | Prolonged release tofacitinib compositions without functional coating |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2014174073A1 (en) | Sustained release formulations of tofacitinib | |
US6881420B2 (en) | Compositions and dosage forms for gastric delivery of irinotecan and methods of treatment that use it to inhibit cancer cell proliferation | |
AU2001268722B8 (en) | Rapidly expanding composition for gastric retention and controlled release of therapeutic agents, and dosage forms including the composition | |
JP5808670B2 (en) | Composition containing weakly basic drug and sustained release dosage form | |
JP5634882B2 (en) | Drug delivery system comprising weakly basic drug and organic acid | |
WO2002070021A1 (en) | Gastric delivery of antineoplastic agents and methods of treatment via inhibition of cancer cell using them | |
WO2006082523A2 (en) | Pharmaceutical sustained release composition of metformin | |
AU2018202652A1 (en) | Gastro-retentive drug delivery system | |
CA2790164A1 (en) | Method of treating a disease condition susceptible to baclofen therapy | |
US11744803B2 (en) | PH-controlled pulsatile delivery system, methods for preparation and use thereof | |
BRPI0615014A2 (en) | solid pharmaceutical composition comprising 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine and a ph modifier and use thereof | |
WO2004019901A2 (en) | Sustained release pharmaceutical composition | |
CN107530337B (en) | treatment method | |
JP2024501051A (en) | Extended release upadacitinib formulation | |
ES2321908T3 (en) | PHARMACEUTICAL PREPARATIONS OF INDEPENDENTLY PROLONGED RELEASE OF PH. | |
US20230181479A1 (en) | Polymer based formulation for release of drugs and bioactives at specific git sites | |
KR20110130382A (en) | Manufacturing method of bioadhesive compact matrix | |
KERALIYA et al. | Formulation and evaluation of atenolol pulsatile press coated tablets using rupturable and erodible polymers | |
US20230201123A1 (en) | Pharmaceutical composition | |
JP6757596B2 (en) | Oral solid tablets | |
Chaudhari et al. | Pelletization techniques: Novel approach for drug delivery | |
KR102039345B1 (en) | A high swellable sustained-release triple-layer tablet containing pregabalin | |
RANOLAZINE | Reg. No. 26106507 | |
Saharan | Novel Fast Dissolving/Disintegrating Dosage Forms | |
Bharathkumar | Formulation and evaluation of sustained release tablet of diltiazem HCI using different polymers |
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: 14720105 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 14720105 Country of ref document: EP Kind code of ref document: A1 |