WO2017216584A1 - New compositions - Google Patents
New compositions Download PDFInfo
- Publication number
- WO2017216584A1 WO2017216584A1 PCT/GB2017/051782 GB2017051782W WO2017216584A1 WO 2017216584 A1 WO2017216584 A1 WO 2017216584A1 GB 2017051782 W GB2017051782 W GB 2017051782W WO 2017216584 A1 WO2017216584 A1 WO 2017216584A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- microcrystalline cellulose
- ffa
- active pharmaceutical
- pharmaceutical ingredient
- mcc
- Prior art date
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 216
- 229920000168 Microcrystalline cellulose Polymers 0.000 claims abstract description 176
- 235000019813 microcrystalline cellulose Nutrition 0.000 claims abstract description 148
- 229940016286 microcrystalline cellulose Drugs 0.000 claims abstract description 144
- 239000008108 microcrystalline cellulose Substances 0.000 claims abstract description 144
- 239000008186 active pharmaceutical agent Substances 0.000 claims abstract description 127
- 239000000546 pharmaceutical excipient Substances 0.000 claims abstract description 76
- 239000003814 drug Substances 0.000 claims abstract description 72
- 229940021182 non-steroidal anti-inflammatory drug Drugs 0.000 claims abstract description 62
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 28
- 206010013935 Dysmenorrhoea Diseases 0.000 claims abstract description 26
- 239000000041 non-steroidal anti-inflammatory agent Substances 0.000 claims abstract description 22
- 208000005171 Dysmenorrhea Diseases 0.000 claims abstract description 19
- 206010027599 migraine Diseases 0.000 claims abstract description 16
- 208000019695 Migraine disease Diseases 0.000 claims abstract description 12
- 229960004369 flufenamic acid Drugs 0.000 claims description 150
- LPEPZBJOKDYZAD-UHFFFAOYSA-N flufenamic acid Chemical compound OC(=O)C1=CC=CC=C1NC1=CC=CC(C(F)(F)F)=C1 LPEPZBJOKDYZAD-UHFFFAOYSA-N 0.000 claims description 150
- 238000000034 method Methods 0.000 claims description 65
- 239000011148 porous material Substances 0.000 claims description 45
- 239000002904 solvent Substances 0.000 claims description 41
- 239000001257 hydrogen Substances 0.000 claims description 28
- 229910052739 hydrogen Inorganic materials 0.000 claims description 28
- 239000006185 dispersion Substances 0.000 claims description 26
- 230000008569 process Effects 0.000 claims description 26
- 239000000126 substance Substances 0.000 claims description 26
- 125000003118 aryl group Chemical group 0.000 claims description 25
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 19
- 238000002156 mixing Methods 0.000 claims description 18
- 238000001125 extrusion Methods 0.000 claims description 16
- 238000001694 spray drying Methods 0.000 claims description 16
- RJKFOVLPORLFTN-LEKSSAKUSA-N Progesterone Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)C)[C@@]1(C)CC2 RJKFOVLPORLFTN-LEKSSAKUSA-N 0.000 claims description 15
- 230000015572 biosynthetic process Effects 0.000 claims description 15
- 238000010438 heat treatment Methods 0.000 claims description 14
- -1 pyroxicam Chemical compound 0.000 claims description 14
- 230000002829 reductive effect Effects 0.000 claims description 13
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 claims description 12
- 239000002243 precursor Substances 0.000 claims description 11
- CGIGDMFJXJATDK-UHFFFAOYSA-N indomethacin Chemical compound CC1=C(CC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 CGIGDMFJXJATDK-UHFFFAOYSA-N 0.000 claims description 10
- 238000004519 manufacturing process Methods 0.000 claims description 9
- PJJGZPJJTHBVMX-UHFFFAOYSA-N 5,7-Dihydroxyisoflavone Chemical compound C=1C(O)=CC(O)=C(C2=O)C=1OC=C2C1=CC=CC=C1 PJJGZPJJTHBVMX-UHFFFAOYSA-N 0.000 claims description 8
- 238000005903 acid hydrolysis reaction Methods 0.000 claims description 8
- 229940034982 antineoplastic agent Drugs 0.000 claims description 8
- 239000002246 antineoplastic agent Substances 0.000 claims description 8
- 229960003464 mefenamic acid Drugs 0.000 claims description 8
- 230000009477 glass transition Effects 0.000 claims description 7
- 238000007789 sealing Methods 0.000 claims description 7
- 230000003068 static effect Effects 0.000 claims description 7
- ZWEHNKRNPOVVGH-UHFFFAOYSA-N 2-Butanone Chemical compound CCC(C)=O ZWEHNKRNPOVVGH-UHFFFAOYSA-N 0.000 claims description 6
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 claims description 6
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 claims description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 claims description 6
- CMWTZPSULFXXJA-UHFFFAOYSA-N Naproxen Natural products C1=C(C(C)C(O)=O)C=CC2=CC(OC)=CC=C21 CMWTZPSULFXXJA-UHFFFAOYSA-N 0.000 claims description 6
- RWZYAGGXGHYGMB-UHFFFAOYSA-N anthranilic acid Chemical class NC1=CC=CC=C1C(O)=O RWZYAGGXGHYGMB-UHFFFAOYSA-N 0.000 claims description 6
- 125000004122 cyclic group Chemical group 0.000 claims description 6
- 229960002390 flurbiprofen Drugs 0.000 claims description 6
- SYTBZMRGLBWNTM-UHFFFAOYSA-N flurbiprofen Chemical compound FC1=CC(C(C(O)=O)C)=CC=C1C1=CC=CC=C1 SYTBZMRGLBWNTM-UHFFFAOYSA-N 0.000 claims description 6
- 239000000416 hydrocolloid Substances 0.000 claims description 6
- DKYWVDODHFEZIM-UHFFFAOYSA-N ketoprofen Chemical compound OC(=O)C(C)C1=CC=CC(C(=O)C=2C=CC=CC=2)=C1 DKYWVDODHFEZIM-UHFFFAOYSA-N 0.000 claims description 6
- 229960000991 ketoprofen Drugs 0.000 claims description 6
- 229960002009 naproxen Drugs 0.000 claims description 6
- CMWTZPSULFXXJA-VIFPVBQESA-N naproxen Chemical compound C1=C([C@H](C)C(O)=O)C=CC2=CC(OC)=CC=C21 CMWTZPSULFXXJA-VIFPVBQESA-N 0.000 claims description 6
- 239000000186 progesterone Substances 0.000 claims description 6
- 229960003387 progesterone Drugs 0.000 claims description 6
- BSYNRYMUTXBXSQ-UHFFFAOYSA-N Aspirin Chemical compound CC(=O)OC1=CC=CC=C1C(O)=O BSYNRYMUTXBXSQ-UHFFFAOYSA-N 0.000 claims description 5
- HEFNNWSXXWATRW-UHFFFAOYSA-N Ibuprofen Chemical compound CC(C)CC1=CC=C(C(C)C(O)=O)C=C1 HEFNNWSXXWATRW-UHFFFAOYSA-N 0.000 claims description 5
- 239000001768 carboxy methyl cellulose Substances 0.000 claims description 5
- BHQCQFFYRZLCQQ-OELDTZBJSA-N cholic acid Chemical compound C([C@H]1C[C@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(O)=O)C)[C@@]2(C)[C@@H](O)C1 BHQCQFFYRZLCQQ-OELDTZBJSA-N 0.000 claims description 5
- 229960001680 ibuprofen Drugs 0.000 claims description 5
- 229960000905 indomethacin Drugs 0.000 claims description 5
- 229940043355 kinase inhibitor Drugs 0.000 claims description 5
- 238000012545 processing Methods 0.000 claims description 5
- 150000003431 steroids Chemical class 0.000 claims description 5
- 229960000894 sulindac Drugs 0.000 claims description 5
- MLKXDPUZXIRXEP-MFOYZWKCSA-N sulindac Chemical compound CC1=C(CC(O)=O)C2=CC(F)=CC=C2\C1=C/C1=CC=C(S(C)=O)C=C1 MLKXDPUZXIRXEP-MFOYZWKCSA-N 0.000 claims description 5
- 229960002905 tolfenamic acid Drugs 0.000 claims description 5
- YEZNLOUZAIOMLT-UHFFFAOYSA-N tolfenamic acid Chemical compound CC1=C(Cl)C=CC=C1NC1=CC=CC=C1C(O)=O YEZNLOUZAIOMLT-UHFFFAOYSA-N 0.000 claims description 5
- RUDATBOHQWOJDD-UHFFFAOYSA-N (3beta,5beta,7alpha)-3,7-Dihydroxycholan-24-oic acid Natural products OC1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)CC2 RUDATBOHQWOJDD-UHFFFAOYSA-N 0.000 claims description 4
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 claims description 4
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 claims description 4
- 229960001138 acetylsalicylic acid Drugs 0.000 claims description 4
- 229960001671 azapropazone Drugs 0.000 claims description 4
- WOIIIUDZSOLAIW-NSHDSACASA-N azapropazone Chemical compound C1=C(C)C=C2N3C(=O)[C@H](CC=C)C(=O)N3C(N(C)C)=NC2=C1 WOIIIUDZSOLAIW-NSHDSACASA-N 0.000 claims description 4
- 238000000227 grinding Methods 0.000 claims description 4
- BDERNNFJNOPAEC-UHFFFAOYSA-N propan-1-ol Chemical compound CCCO BDERNNFJNOPAEC-UHFFFAOYSA-N 0.000 claims description 4
- 239000003909 protein kinase inhibitor Substances 0.000 claims description 4
- RUDATBOHQWOJDD-UZVSRGJWSA-N ursodeoxycholic acid Chemical compound C([C@H]1C[C@@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(O)=O)C)[C@@]2(C)CC1 RUDATBOHQWOJDD-UZVSRGJWSA-N 0.000 claims description 4
- 229960001661 ursodiol Drugs 0.000 claims description 4
- BHQCQFFYRZLCQQ-UHFFFAOYSA-N (3alpha,5alpha,7alpha,12alpha)-3,7,12-trihydroxy-cholan-24-oic acid Natural products OC1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 BHQCQFFYRZLCQQ-UHFFFAOYSA-N 0.000 claims description 3
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 claims description 3
- VOXZDWNPVJITMN-ZBRFXRBCSA-N 17β-estradiol Chemical compound OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3CCC2=C1 VOXZDWNPVJITMN-ZBRFXRBCSA-N 0.000 claims description 3
- XILVEPYQJIOVNB-UHFFFAOYSA-N 2-[3-(trifluoromethyl)anilino]benzoic acid 2-(2-hydroxyethoxy)ethyl ester Chemical compound OCCOCCOC(=O)C1=CC=CC=C1NC1=CC=CC(C(F)(F)F)=C1 XILVEPYQJIOVNB-UHFFFAOYSA-N 0.000 claims description 3
- MLDQJTXFUGDVEO-UHFFFAOYSA-N BAY-43-9006 Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=CC(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 MLDQJTXFUGDVEO-UHFFFAOYSA-N 0.000 claims description 3
- 229920002134 Carboxymethyl cellulose Polymers 0.000 claims description 3
- 239000004380 Cholic acid Substances 0.000 claims description 3
- ZBNZXTGUTAYRHI-UHFFFAOYSA-N Dasatinib Chemical compound C=1C(N2CCN(CCO)CC2)=NC(C)=NC=1NC(S1)=NC=C1C(=O)NC1=C(C)C=CC=C1Cl ZBNZXTGUTAYRHI-UHFFFAOYSA-N 0.000 claims description 3
- 229920002907 Guar gum Polymers 0.000 claims description 3
- 239000005517 L01XE01 - Imatinib Substances 0.000 claims description 3
- 239000005551 L01XE03 - Erlotinib Substances 0.000 claims description 3
- 239000005511 L01XE05 - Sorafenib Substances 0.000 claims description 3
- 239000002067 L01XE06 - Dasatinib Substances 0.000 claims description 3
- 239000005536 L01XE08 - Nilotinib Substances 0.000 claims description 3
- SBDNJUWAMKYJOX-UHFFFAOYSA-N Meclofenamic Acid Chemical compound CC1=CC=C(Cl)C(NC=2C(=CC=CC=2)C(O)=O)=C1Cl SBDNJUWAMKYJOX-UHFFFAOYSA-N 0.000 claims description 3
- JZFPYUNJRRFVQU-UHFFFAOYSA-N Niflumic acid Chemical compound OC(=O)C1=CC=CN=C1NC1=CC=CC(C(F)(F)F)=C1 JZFPYUNJRRFVQU-UHFFFAOYSA-N 0.000 claims description 3
- MKUXAQIIEYXACX-UHFFFAOYSA-N aciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCO)C=N2 MKUXAQIIEYXACX-UHFFFAOYSA-N 0.000 claims description 3
- 230000002411 adverse Effects 0.000 claims description 3
- 235000010948 carboxy methyl cellulose Nutrition 0.000 claims description 3
- 239000008112 carboxymethyl-cellulose Substances 0.000 claims description 3
- 229940105329 carboxymethylcellulose Drugs 0.000 claims description 3
- 235000010418 carrageenan Nutrition 0.000 claims description 3
- 239000000679 carrageenan Substances 0.000 claims description 3
- 229940113118 carrageenan Drugs 0.000 claims description 3
- 229920001525 carrageenan Polymers 0.000 claims description 3
- 235000019416 cholic acid Nutrition 0.000 claims description 3
- 229960002471 cholic acid Drugs 0.000 claims description 3
- 229960002448 dasatinib Drugs 0.000 claims description 3
- KXGVEGMKQFWNSR-UHFFFAOYSA-N deoxycholic acid Natural products C1CC2CC(O)CCC2(C)C2C1C1CCC(C(CCC(O)=O)C)C1(C)C(O)C2 KXGVEGMKQFWNSR-UHFFFAOYSA-N 0.000 claims description 3
- 229960001433 erlotinib Drugs 0.000 claims description 3
- AAKJLRGGTJKAMG-UHFFFAOYSA-N erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 claims description 3
- 229930182833 estradiol Natural products 0.000 claims description 3
- 229960005309 estradiol Drugs 0.000 claims description 3
- 239000000262 estrogen Substances 0.000 claims description 3
- 229940011871 estrogen Drugs 0.000 claims description 3
- 229960001493 etofenamate Drugs 0.000 claims description 3
- NOOCSNJCXJYGPE-UHFFFAOYSA-N flunixin Chemical compound C1=CC=C(C(F)(F)F)C(C)=C1NC1=NC=CC=C1C(O)=O NOOCSNJCXJYGPE-UHFFFAOYSA-N 0.000 claims description 3
- 229960000588 flunixin Drugs 0.000 claims description 3
- 239000000665 guar gum Substances 0.000 claims description 3
- 235000010417 guar gum Nutrition 0.000 claims description 3
- 229960002154 guar gum Drugs 0.000 claims description 3
- KTUFNOKKBVMGRW-UHFFFAOYSA-N imatinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 claims description 3
- 229960002411 imatinib Drugs 0.000 claims description 3
- 229960003803 meclofenamic acid Drugs 0.000 claims description 3
- 229960003251 morniflumate Drugs 0.000 claims description 3
- LDXSPUSKBDTEKA-UHFFFAOYSA-N morniflumate Chemical compound FC(F)(F)C1=CC=CC(NC=2C(=CC=CN=2)C(=O)OCCN2CCOCC2)=C1 LDXSPUSKBDTEKA-UHFFFAOYSA-N 0.000 claims description 3
- 229960000916 niflumic acid Drugs 0.000 claims description 3
- HHZIURLSWUIHRB-UHFFFAOYSA-N nilotinib Chemical compound C1=NC(C)=CN1C1=CC(NC(=O)C=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)=CC(C(F)(F)F)=C1 HHZIURLSWUIHRB-UHFFFAOYSA-N 0.000 claims description 3
- 229960001346 nilotinib Drugs 0.000 claims description 3
- 239000000583 progesterone congener Substances 0.000 claims description 3
- 235000010413 sodium alginate Nutrition 0.000 claims description 3
- 239000000661 sodium alginate Substances 0.000 claims description 3
- 229940005550 sodium alginate Drugs 0.000 claims description 3
- 229960003787 sorafenib Drugs 0.000 claims description 3
- 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 3
- SPMVMDHWKHCIDT-UHFFFAOYSA-N 1-[2-chloro-4-[(6,7-dimethoxy-4-quinolinyl)oxy]phenyl]-3-(5-methyl-3-isoxazolyl)urea Chemical compound C=12C=C(OC)C(OC)=CC2=NC=CC=1OC(C=C1Cl)=CC=C1NC(=O)NC=1C=C(C)ON=1 SPMVMDHWKHCIDT-UHFFFAOYSA-N 0.000 claims description 2
- XXJWYDDUDKYVKI-UHFFFAOYSA-N 4-[(4-fluoro-2-methyl-1H-indol-5-yl)oxy]-6-methoxy-7-[3-(1-pyrrolidinyl)propoxy]quinazoline Chemical compound COC1=CC2=C(OC=3C(=C4C=C(C)NC4=CC=3)F)N=CN=C2C=C1OCCCN1CCCC1 XXJWYDDUDKYVKI-UHFFFAOYSA-N 0.000 claims description 2
- SRSGVKWWVXWSJT-ATVHPVEESA-N 5-[(z)-(5-fluoro-2-oxo-1h-indol-3-ylidene)methyl]-2,4-dimethyl-n-(2-pyrrolidin-1-ylethyl)-1h-pyrrole-3-carboxamide Chemical compound CC=1NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C(C)C=1C(=O)NCCN1CCCC1 SRSGVKWWVXWSJT-ATVHPVEESA-N 0.000 claims description 2
- AILRADAXUVEEIR-UHFFFAOYSA-N 5-chloro-4-n-(2-dimethylphosphorylphenyl)-2-n-[2-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl]pyrimidine-2,4-diamine Chemical compound COC1=CC(N2CCC(CC2)N2CCN(C)CC2)=CC=C1NC(N=1)=NC=C(Cl)C=1NC1=CC=CC=C1P(C)(C)=O AILRADAXUVEEIR-UHFFFAOYSA-N 0.000 claims description 2
- 239000005411 L01XE02 - Gefitinib Substances 0.000 claims description 2
- 239000002147 L01XE04 - Sunitinib Substances 0.000 claims description 2
- 239000002136 L01XE07 - Lapatinib Substances 0.000 claims description 2
- 239000003798 L01XE11 - Pazopanib Substances 0.000 claims description 2
- 239000002118 L01XE12 - Vandetanib Substances 0.000 claims description 2
- 239000002145 L01XE14 - Bosutinib Substances 0.000 claims description 2
- 239000002146 L01XE16 - Crizotinib Substances 0.000 claims description 2
- 239000002144 L01XE18 - Ruxolitinib Substances 0.000 claims description 2
- 239000002138 L01XE21 - Regorafenib Substances 0.000 claims description 2
- 239000002139 L01XE22 - Masitinib Substances 0.000 claims description 2
- 239000002137 L01XE24 - Ponatinib Substances 0.000 claims description 2
- 239000002176 L01XE26 - Cabozantinib Substances 0.000 claims description 2
- 239000002177 L01XE27 - Ibrutinib Substances 0.000 claims description 2
- UIARLYUEJFELEN-LROUJFHJSA-N LSM-1231 Chemical compound C12=C3N4C5=CC=CC=C5C3=C3C(=O)NCC3=C2C2=CC=CC=C2N1[C@]1(C)[C@](CO)(O)C[C@H]4O1 UIARLYUEJFELEN-LROUJFHJSA-N 0.000 claims description 2
- 239000005462 Mubritinib Substances 0.000 claims description 2
- 239000005464 Radotinib Substances 0.000 claims description 2
- 239000003819 Toceranib Substances 0.000 claims description 2
- 229960001686 afatinib Drugs 0.000 claims description 2
- ULXXDDBFHOBEHA-CWDCEQMOSA-N afatinib Chemical compound N1=CN=C2C=C(O[C@@H]3COCC3)C(NC(=O)/C=C/CN(C)C)=CC2=C1NC1=CC=C(F)C(Cl)=C1 ULXXDDBFHOBEHA-CWDCEQMOSA-N 0.000 claims description 2
- 229960001611 alectinib Drugs 0.000 claims description 2
- KDGFLJKFZUIJMX-UHFFFAOYSA-N alectinib Chemical compound CCC1=CC=2C(=O)C(C3=CC=C(C=C3N3)C#N)=C3C(C)(C)C=2C=C1N(CC1)CCC1N1CCOCC1 KDGFLJKFZUIJMX-UHFFFAOYSA-N 0.000 claims description 2
- 125000005233 alkylalcohol group Chemical group 0.000 claims description 2
- 229960003005 axitinib Drugs 0.000 claims description 2
- RITAVMQDGBJQJZ-FMIVXFBMSA-N axitinib Chemical compound CNC(=O)C1=CC=CC=C1SC1=CC=C(C(\C=C\C=2N=CC=CC=2)=NN2)C2=C1 RITAVMQDGBJQJZ-FMIVXFBMSA-N 0.000 claims description 2
- 229950003054 binimetinib Drugs 0.000 claims description 2
- ACWZRVQXLIRSDF-UHFFFAOYSA-N binimetinib Chemical compound OCCONC(=O)C=1C=C2N(C)C=NC2=C(F)C=1NC1=CC=C(Br)C=C1F ACWZRVQXLIRSDF-UHFFFAOYSA-N 0.000 claims description 2
- UBPYILGKFZZVDX-UHFFFAOYSA-N bosutinib Chemical compound C1=C(Cl)C(OC)=CC(NC=2C3=CC(OC)=C(OCCCN4CCN(C)CC4)C=C3N=CC=2C#N)=C1Cl UBPYILGKFZZVDX-UHFFFAOYSA-N 0.000 claims description 2
- 229960003736 bosutinib Drugs 0.000 claims description 2
- 229950004272 brigatinib Drugs 0.000 claims description 2
- 229960001292 cabozantinib Drugs 0.000 claims description 2
- ONIQOQHATWINJY-UHFFFAOYSA-N cabozantinib Chemical compound C=12C=C(OC)C(OC)=CC2=NC=CC=1OC(C=C1)=CC=C1NC(=O)C1(C(=O)NC=2C=CC(F)=CC=2)CC1 ONIQOQHATWINJY-UHFFFAOYSA-N 0.000 claims description 2
- 229960002412 cediranib Drugs 0.000 claims description 2
- 229960001602 ceritinib Drugs 0.000 claims description 2
- 229940099352 cholate Drugs 0.000 claims description 2
- 229960002271 cobimetinib Drugs 0.000 claims description 2
- RESIMIUSNACMNW-BXRWSSRYSA-N cobimetinib fumarate Chemical compound OC(=O)\C=C\C(O)=O.C1C(O)([C@H]2NCCCC2)CN1C(=O)C1=CC=C(F)C(F)=C1NC1=CC=C(I)C=C1F.C1C(O)([C@H]2NCCCC2)CN1C(=O)C1=CC=C(F)C(F)=C1NC1=CC=C(I)C=C1F RESIMIUSNACMNW-BXRWSSRYSA-N 0.000 claims description 2
- 229960005061 crizotinib Drugs 0.000 claims description 2
- KTEIFNKAUNYNJU-GFCCVEGCSA-N crizotinib Chemical compound O([C@H](C)C=1C(=C(F)C=CC=1Cl)Cl)C(C(=NC=1)N)=CC=1C(=C1)C=NN1C1CCNCC1 KTEIFNKAUNYNJU-GFCCVEGCSA-N 0.000 claims description 2
- 229950000521 entrectinib Drugs 0.000 claims description 2
- SBNKFTQSBPKMBZ-UHFFFAOYSA-N ethenzamide Chemical compound CCOC1=CC=CC=C1C(N)=O SBNKFTQSBPKMBZ-UHFFFAOYSA-N 0.000 claims description 2
- 229960000514 ethenzamide Drugs 0.000 claims description 2
- 229950005309 fostamatinib Drugs 0.000 claims description 2
- GKDRMWXFWHEQQT-UHFFFAOYSA-N fostamatinib Chemical compound COC1=C(OC)C(OC)=CC(NC=2N=C(NC=3N=C4N(COP(O)(O)=O)C(=O)C(C)(C)OC4=CC=3)C(F)=CN=2)=C1 GKDRMWXFWHEQQT-UHFFFAOYSA-N 0.000 claims description 2
- 229960002584 gefitinib Drugs 0.000 claims description 2
- XGALLCVXEZPNRQ-UHFFFAOYSA-N gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 claims description 2
- 229960001507 ibrutinib Drugs 0.000 claims description 2
- XYFPWWZEPKGCCK-GOSISDBHSA-N ibrutinib Chemical compound C1=2C(N)=NC=NC=2N([C@H]2CN(CCC2)C(=O)C=C)N=C1C(C=C1)=CC=C1OC1=CC=CC=C1 XYFPWWZEPKGCCK-GOSISDBHSA-N 0.000 claims description 2
- 229960004891 lapatinib Drugs 0.000 claims description 2
- BCFGMOOMADDAQU-UHFFFAOYSA-N lapatinib Chemical compound O1C(CNCCS(=O)(=O)C)=CC=C1C1=CC=C(N=CN=C2NC=3C=C(Cl)C(OCC=4C=C(F)C=CC=4)=CC=3)C2=C1 BCFGMOOMADDAQU-UHFFFAOYSA-N 0.000 claims description 2
- 229960003784 lenvatinib Drugs 0.000 claims description 2
- WOSKHXYHFSIKNG-UHFFFAOYSA-N lenvatinib Chemical compound C=12C=C(C(N)=O)C(OC)=CC2=NC=CC=1OC(C=C1Cl)=CC=C1NC(=O)NC1CC1 WOSKHXYHFSIKNG-UHFFFAOYSA-N 0.000 claims description 2
- 229950001845 lestaurtinib Drugs 0.000 claims description 2
- 229960004655 masitinib Drugs 0.000 claims description 2
- WJEOLQLKVOPQFV-UHFFFAOYSA-N masitinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3SC=C(N=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 WJEOLQLKVOPQFV-UHFFFAOYSA-N 0.000 claims description 2
- ZVHNDZWQTBEVRY-UHFFFAOYSA-N momelotinib Chemical compound C1=CC(C(NCC#N)=O)=CC=C1C1=CC=NC(NC=2C=CC(=CC=2)N2CCOCC2)=N1 ZVHNDZWQTBEVRY-UHFFFAOYSA-N 0.000 claims description 2
- 229950008814 momelotinib Drugs 0.000 claims description 2
- 229950002212 mubritinib Drugs 0.000 claims description 2
- ZTFBIUXIQYRUNT-MDWZMJQESA-N mubritinib Chemical compound C1=CC(C(F)(F)F)=CC=C1\C=C\C1=NC(COC=2C=CC(CCCCN3N=NC=C3)=CC=2)=CO1 ZTFBIUXIQYRUNT-MDWZMJQESA-N 0.000 claims description 2
- FDMQDKQUTRLUBU-UHFFFAOYSA-N n-[3-[2-[4-(4-methylpiperazin-1-yl)anilino]thieno[3,2-d]pyrimidin-4-yl]oxyphenyl]prop-2-enamide Chemical compound C1CN(C)CCN1C(C=C1)=CC=C1NC1=NC(OC=2C=C(NC(=O)C=C)C=CC=2)=C(SC=C2)C2=N1 FDMQDKQUTRLUBU-UHFFFAOYSA-N 0.000 claims description 2
- HUFOZJXAKZVRNJ-UHFFFAOYSA-N n-[3-[[2-[4-(4-acetylpiperazin-1-yl)-2-methoxyanilino]-5-(trifluoromethyl)pyrimidin-4-yl]amino]phenyl]prop-2-enamide Chemical compound COC1=CC(N2CCN(CC2)C(C)=O)=CC=C1NC(N=1)=NC=C(C(F)(F)F)C=1NC1=CC=CC(NC(=O)C=C)=C1 HUFOZJXAKZVRNJ-UHFFFAOYSA-N 0.000 claims description 2
- HAYYBYPASCDWEQ-UHFFFAOYSA-N n-[5-[(3,5-difluorophenyl)methyl]-1h-indazol-3-yl]-4-(4-methylpiperazin-1-yl)-2-(oxan-4-ylamino)benzamide Chemical compound C1CN(C)CCN1C(C=C1NC2CCOCC2)=CC=C1C(=O)NC(C1=C2)=NNC1=CC=C2CC1=CC(F)=CC(F)=C1 HAYYBYPASCDWEQ-UHFFFAOYSA-N 0.000 claims description 2
- 229950008835 neratinib Drugs 0.000 claims description 2
- 229960004378 nintedanib Drugs 0.000 claims description 2
- XZXHXSATPCNXJR-ZIADKAODSA-N nintedanib Chemical compound O=C1NC2=CC(C(=O)OC)=CC=C2\C1=C(C=1C=CC=CC=1)\NC(C=C1)=CC=C1N(C)C(=O)CN1CCN(C)CC1 XZXHXSATPCNXJR-ZIADKAODSA-N 0.000 claims description 2
- 229950000778 olmutinib Drugs 0.000 claims description 2
- 229960003278 osimertinib Drugs 0.000 claims description 2
- DUYJMQONPNNFPI-UHFFFAOYSA-N osimertinib Chemical compound COC1=CC(N(C)CCN(C)C)=C(NC(=O)C=C)C=C1NC1=NC=CC(C=2C3=CC=CC=C3N(C)C=2)=N1 DUYJMQONPNNFPI-UHFFFAOYSA-N 0.000 claims description 2
- 229950011410 pacritinib Drugs 0.000 claims description 2
- HWXVIOGONBBTBY-ONEGZZNKSA-N pacritinib Chemical compound C=1C=C(C=2)NC(N=3)=NC=CC=3C(C=3)=CC=CC=3COC\C=C\COCC=2C=1OCCN1CCCC1 HWXVIOGONBBTBY-ONEGZZNKSA-N 0.000 claims description 2
- 229960000639 pazopanib Drugs 0.000 claims description 2
- CUIHSIWYWATEQL-UHFFFAOYSA-N pazopanib Chemical compound C1=CC2=C(C)N(C)N=C2C=C1N(C)C(N=1)=CC=NC=1NC1=CC=C(C)C(S(N)(=O)=O)=C1 CUIHSIWYWATEQL-UHFFFAOYSA-N 0.000 claims description 2
- PHXJVRSECIGDHY-UHFFFAOYSA-N ponatinib Chemical compound C1CN(C)CCN1CC(C(=C1)C(F)(F)F)=CC=C1NC(=O)C1=CC=C(C)C(C#CC=2N3N=CC=CC3=NC=2)=C1 PHXJVRSECIGDHY-UHFFFAOYSA-N 0.000 claims description 2
- 229960001131 ponatinib Drugs 0.000 claims description 2
- 229950004043 radotinib Drugs 0.000 claims description 2
- DUPWHXBITIZIKZ-UHFFFAOYSA-N radotinib Chemical compound C1=NC(C)=CN1C1=CC(NC(=O)C=2C=C(NC=3N=C(C=CN=3)C=3N=CC=NC=3)C(C)=CC=2)=CC(C(F)(F)F)=C1 DUPWHXBITIZIKZ-UHFFFAOYSA-N 0.000 claims description 2
- 229960004836 regorafenib Drugs 0.000 claims description 2
- FNHKPVJBJVTLMP-UHFFFAOYSA-N regorafenib Chemical compound C1=NC(C(=O)NC)=CC(OC=2C=C(F)C(NC(=O)NC=3C=C(C(Cl)=CC=3)C(F)(F)F)=CC=2)=C1 FNHKPVJBJVTLMP-UHFFFAOYSA-N 0.000 claims description 2
- 229950009855 rociletinib Drugs 0.000 claims description 2
- 238000002390 rotary evaporation Methods 0.000 claims description 2
- 229960000215 ruxolitinib Drugs 0.000 claims description 2
- HFNKQEVNSGCOJV-OAHLLOKOSA-N ruxolitinib Chemical compound C1([C@@H](CC#N)N2N=CC(=C2)C=2C=3C=CNC=3N=CN=2)CCCC1 HFNKQEVNSGCOJV-OAHLLOKOSA-N 0.000 claims description 2
- CYOHGALHFOKKQC-UHFFFAOYSA-N selumetinib Chemical compound OCCONC(=O)C=1C=C2N(C)C=NC2=C(F)C=1NC1=CC=C(Br)C=C1Cl CYOHGALHFOKKQC-UHFFFAOYSA-N 0.000 claims description 2
- 229950010746 selumetinib Drugs 0.000 claims description 2
- 229950003647 semaxanib Drugs 0.000 claims description 2
- WUWDLXZGHZSWQZ-WQLSENKSSA-N semaxanib Chemical compound N1C(C)=CC(C)=C1\C=C/1C2=CC=CC=C2NC\1=O WUWDLXZGHZSWQZ-WQLSENKSSA-N 0.000 claims description 2
- 229960001796 sunitinib Drugs 0.000 claims description 2
- WINHZLLDWRZWRT-ATVHPVEESA-N sunitinib Chemical compound CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C WINHZLLDWRZWRT-ATVHPVEESA-N 0.000 claims description 2
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 claims description 2
- 229960000940 tivozanib Drugs 0.000 claims description 2
- 229960005048 toceranib Drugs 0.000 claims description 2
- 229960004066 trametinib Drugs 0.000 claims description 2
- LIRYPHYGHXZJBZ-UHFFFAOYSA-N trametinib Chemical compound CC(=O)NC1=CC=CC(N2C(N(C3CC3)C(=O)C3=C(NC=4C(=CC(I)=CC=4)F)N(C)C(=O)C(C)=C32)=O)=C1 LIRYPHYGHXZJBZ-UHFFFAOYSA-N 0.000 claims description 2
- 229960000241 vandetanib Drugs 0.000 claims description 2
- UHTHHESEBZOYNR-UHFFFAOYSA-N vandetanib Chemical compound COC1=CC(C(/N=CN2)=N/C=3C(=CC(Br)=CC=3)F)=C2C=C1OCC1CCN(C)CC1 UHTHHESEBZOYNR-UHFFFAOYSA-N 0.000 claims description 2
- 229960003862 vemurafenib Drugs 0.000 claims description 2
- GPXBXXGIAQBQNI-UHFFFAOYSA-N vemurafenib Chemical compound CCCS(=O)(=O)NC1=CC=C(F)C(C(=O)C=2C3=CC(=CN=C3NC=2)C=2C=CC(Cl)=CC=2)=C1F GPXBXXGIAQBQNI-UHFFFAOYSA-N 0.000 claims description 2
- VERWOWGGCGHDQE-UHFFFAOYSA-N ceritinib Chemical compound CC=1C=C(NC=2N=C(NC=3C(=CC=CC=3)S(=O)(=O)C(C)C)C(Cl)=CN=2)C(OC(C)C)=CC=1C1CCNCC1 VERWOWGGCGHDQE-UHFFFAOYSA-N 0.000 claims 1
- ZNHPZUKZSNBOSQ-BQYQJAHWSA-N neratinib Chemical compound C=12C=C(NC\C=C\CN(C)C)C(OCC)=CC2=NC=C(C#N)C=1NC(C=C1Cl)=CC=C1OCC1=CC=CC=N1 ZNHPZUKZSNBOSQ-BQYQJAHWSA-N 0.000 claims 1
- 229940079593 drug Drugs 0.000 abstract description 67
- 239000004480 active ingredient Substances 0.000 abstract description 28
- 238000009472 formulation Methods 0.000 abstract description 22
- 238000011282 treatment Methods 0.000 abstract description 18
- 238000001727 in vivo Methods 0.000 abstract description 2
- 229920002678 cellulose Polymers 0.000 description 79
- 235000010980 cellulose Nutrition 0.000 description 77
- 239000001913 cellulose Substances 0.000 description 60
- 238000004090 dissolution Methods 0.000 description 23
- 239000002253 acid Substances 0.000 description 22
- 238000000113 differential scanning calorimetry Methods 0.000 description 19
- 238000002844 melting Methods 0.000 description 19
- 230000008018 melting Effects 0.000 description 19
- 239000011874 heated mixture Substances 0.000 description 18
- 230000003993 interaction Effects 0.000 description 17
- 230000000694 effects Effects 0.000 description 15
- 238000001179 sorption measurement Methods 0.000 description 15
- 239000000370 acceptor Substances 0.000 description 14
- 238000004458 analytical method Methods 0.000 description 14
- 239000000843 powder Substances 0.000 description 13
- 150000001875 compounds Chemical class 0.000 description 12
- 239000006069 physical mixture Substances 0.000 description 12
- 239000007789 gas Substances 0.000 description 10
- 238000001953 recrystallisation Methods 0.000 description 10
- 238000003860 storage Methods 0.000 description 10
- 238000002441 X-ray diffraction Methods 0.000 description 9
- 230000008901 benefit Effects 0.000 description 9
- 230000000875 corresponding effect Effects 0.000 description 9
- 238000005259 measurement Methods 0.000 description 9
- 150000003839 salts Chemical group 0.000 description 9
- 238000005033 Fourier transform infrared spectroscopy Methods 0.000 description 8
- 230000001225 therapeutic effect Effects 0.000 description 8
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 7
- 230000009471 action Effects 0.000 description 7
- 229940111136 antiinflammatory and antirheumatic drug fenamates Drugs 0.000 description 7
- 239000000463 material Substances 0.000 description 7
- 239000000047 product Substances 0.000 description 7
- 238000001157 Fourier transform infrared spectrum Methods 0.000 description 6
- 206010019233 Headaches Diseases 0.000 description 6
- 238000009826 distribution Methods 0.000 description 6
- 229940088679 drug related substance Drugs 0.000 description 6
- 238000001704 evaporation Methods 0.000 description 6
- 230000008020 evaporation Effects 0.000 description 6
- 229910052500 inorganic mineral Inorganic materials 0.000 description 6
- 239000011707 mineral Substances 0.000 description 6
- 238000001228 spectrum Methods 0.000 description 6
- 208000002193 Pain Diseases 0.000 description 5
- 238000010521 absorption reaction Methods 0.000 description 5
- 238000004630 atomic force microscopy Methods 0.000 description 5
- 239000003433 contraceptive agent Substances 0.000 description 5
- 238000003795 desorption Methods 0.000 description 5
- 229910001873 dinitrogen Inorganic materials 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 239000010931 gold Substances 0.000 description 5
- 231100000869 headache Toxicity 0.000 description 5
- 239000002245 particle Substances 0.000 description 5
- 239000000243 solution Substances 0.000 description 5
- 239000000725 suspension Substances 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 230000009466 transformation Effects 0.000 description 5
- 238000003775 Density Functional Theory Methods 0.000 description 4
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 4
- 150000001242 acetic acid derivatives Chemical class 0.000 description 4
- YZXBAPSDXZZRGB-DOFZRALJSA-N arachidonic acid Chemical compound CCCCC\C=C/C\C=C/C\C=C/C\C=C/CCCC(O)=O YZXBAPSDXZZRGB-DOFZRALJSA-N 0.000 description 4
- 230000002254 contraceptive effect Effects 0.000 description 4
- 239000012530 fluid Substances 0.000 description 4
- 210000001035 gastrointestinal tract Anatomy 0.000 description 4
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 4
- 229910052737 gold Inorganic materials 0.000 description 4
- 238000000265 homogenisation Methods 0.000 description 4
- 239000004615 ingredient Substances 0.000 description 4
- 230000002175 menstrual effect Effects 0.000 description 4
- 230000000144 pharmacologic effect Effects 0.000 description 4
- 230000008707 rearrangement Effects 0.000 description 4
- 229920006395 saturated elastomer Polymers 0.000 description 4
- 239000011550 stock solution Substances 0.000 description 4
- LNPDTQAFDNKSHK-UHFFFAOYSA-N valdecoxib Chemical compound CC=1ON=C(C=2C=CC=CC=2)C=1C1=CC=C(S(N)(=O)=O)C=C1 LNPDTQAFDNKSHK-UHFFFAOYSA-N 0.000 description 4
- RZVAJINKPMORJF-UHFFFAOYSA-N Acetaminophen Chemical compound CC(=O)NC1=CC=C(O)C=C1 RZVAJINKPMORJF-UHFFFAOYSA-N 0.000 description 3
- UHOVQNZJYSORNB-UHFFFAOYSA-N Benzene Chemical compound C1=CC=CC=C1 UHOVQNZJYSORNB-UHFFFAOYSA-N 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 150000007513 acids Chemical class 0.000 description 3
- 238000005280 amorphization Methods 0.000 description 3
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 3
- 229910052799 carbon Inorganic materials 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 238000012512 characterization method Methods 0.000 description 3
- 238000007385 chemical modification Methods 0.000 description 3
- 238000006731 degradation reaction Methods 0.000 description 3
- 239000012738 dissolution medium Substances 0.000 description 3
- 230000005284 excitation Effects 0.000 description 3
- 238000010348 incorporation Methods 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 230000005906 menstruation Effects 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 229910052757 nitrogen Inorganic materials 0.000 description 3
- 229910052760 oxygen Inorganic materials 0.000 description 3
- 239000000825 pharmaceutical preparation Substances 0.000 description 3
- 230000026731 phosphorylation Effects 0.000 description 3
- 238000006366 phosphorylation reaction Methods 0.000 description 3
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 3
- 239000004033 plastic Substances 0.000 description 3
- 238000001878 scanning electron micrograph Methods 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 238000000527 sonication Methods 0.000 description 3
- 239000007921 spray Substances 0.000 description 3
- 238000010561 standard procedure Methods 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 229920001169 thermoplastic Polymers 0.000 description 3
- 238000000844 transformation Methods 0.000 description 3
- RLFWWDJHLFCNIJ-UHFFFAOYSA-N Aminoantipyrine Natural products CN1C(C)=C(N)C(=O)N1C1=CC=CC=C1 RLFWWDJHLFCNIJ-UHFFFAOYSA-N 0.000 description 2
- 238000004438 BET method Methods 0.000 description 2
- KXDAEFPNCMNJSK-UHFFFAOYSA-N Benzamide Chemical compound NC(=O)C1=CC=CC=C1 KXDAEFPNCMNJSK-UHFFFAOYSA-N 0.000 description 2
- 125000002353 D-glucosyl group Chemical group C1([C@H](O)[C@@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 2
- 241001492220 Echovirus E2 Species 0.000 description 2
- 241001492209 Echovirus E4 Species 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- BFPYWIDHMRZLRN-SLHNCBLASA-N Ethinyl estradiol Chemical compound OC1=CC=C2[C@H]3CC[C@](C)([C@](CC4)(O)C#C)[C@@H]4[C@@H]3CCC2=C1 BFPYWIDHMRZLRN-SLHNCBLASA-N 0.000 description 2
- 208000032843 Hemorrhage Diseases 0.000 description 2
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 description 2
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- MITFXPHMIHQXPI-UHFFFAOYSA-N Oraflex Chemical compound N=1C2=CC(C(C(O)=O)C)=CC=C2OC=1C1=CC=C(Cl)C=C1 MITFXPHMIHQXPI-UHFFFAOYSA-N 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 208000024780 Urticaria Diseases 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
- WBDLSXCAFVEULB-UHFFFAOYSA-N acetonitrile;methylsulfinylmethane Chemical compound CC#N.CS(C)=O WBDLSXCAFVEULB-UHFFFAOYSA-N 0.000 description 2
- 230000000202 analgesic effect Effects 0.000 description 2
- 229940035676 analgesics Drugs 0.000 description 2
- 239000000730 antalgic agent Substances 0.000 description 2
- 230000001754 anti-pyretic effect Effects 0.000 description 2
- 239000001961 anticonvulsive agent Substances 0.000 description 2
- 229940045988 antineoplastic drug protein kinase inhibitors Drugs 0.000 description 2
- 239000002221 antipyretic Substances 0.000 description 2
- 239000003125 aqueous solvent Substances 0.000 description 2
- 229940114079 arachidonic acid Drugs 0.000 description 2
- 235000021342 arachidonic acid Nutrition 0.000 description 2
- 239000002585 base Substances 0.000 description 2
- 230000006399 behavior Effects 0.000 description 2
- 230000000740 bleeding effect Effects 0.000 description 2
- 238000009835 boiling Methods 0.000 description 2
- RYYVLZVUVIJVGH-UHFFFAOYSA-N caffeine Chemical compound CN1C(=O)N(C)C(=O)C2=C1N=CN2C RYYVLZVUVIJVGH-UHFFFAOYSA-N 0.000 description 2
- 238000004364 calculation method Methods 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 125000002915 carbonyl group Chemical group [*:2]C([*:1])=O 0.000 description 2
- 125000002843 carboxylic acid group Chemical group 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 239000002812 cholic acid derivative Substances 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- OROGSEYTTFOCAN-DNJOTXNNSA-N codeine Chemical compound C([C@H]1[C@H](N(CC[C@@]112)C)C3)=C[C@H](O)[C@@H]1OC1=C2C3=CC=C1OC OROGSEYTTFOCAN-DNJOTXNNSA-N 0.000 description 2
- 238000002425 crystallisation Methods 0.000 description 2
- 230000008025 crystallization Effects 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 239000008367 deionised water Substances 0.000 description 2
- 229910021641 deionized water Inorganic materials 0.000 description 2
- 238000001938 differential scanning calorimetry curve Methods 0.000 description 2
- 239000000539 dimer Substances 0.000 description 2
- 230000008034 disappearance Effects 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 238000001035 drying Methods 0.000 description 2
- PWVXXGRKLHYWKM-LJQANCHMSA-N eletriptan Chemical compound CN1CCC[C@@H]1CC(C1=C2)=CNC1=CC=C2CCS(=O)(=O)C1=CC=CC=C1 PWVXXGRKLHYWKM-LJQANCHMSA-N 0.000 description 2
- 230000002708 enhancing effect Effects 0.000 description 2
- 239000002389 essential drug Substances 0.000 description 2
- 229960002568 ethinylestradiol Drugs 0.000 description 2
- 239000011521 glass Substances 0.000 description 2
- 230000003301 hydrolyzing effect Effects 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 230000000968 intestinal effect Effects 0.000 description 2
- 230000007774 longterm Effects 0.000 description 2
- 239000000155 melt Substances 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- 238000000302 molecular modelling Methods 0.000 description 2
- 201000006417 multiple sclerosis Diseases 0.000 description 2
- 238000006386 neutralization reaction Methods 0.000 description 2
- 239000012299 nitrogen atmosphere Substances 0.000 description 2
- 238000002429 nitrogen sorption measurement Methods 0.000 description 2
- HGASFNYMVGEKTF-UHFFFAOYSA-N octan-1-ol;hydrate Chemical compound O.CCCCCCCCO HGASFNYMVGEKTF-UHFFFAOYSA-N 0.000 description 2
- 229960005489 paracetamol Drugs 0.000 description 2
- 239000008188 pellet Substances 0.000 description 2
- 230000035699 permeability Effects 0.000 description 2
- 229960002895 phenylbutazone Drugs 0.000 description 2
- VYMDGNCVAMGZFE-UHFFFAOYSA-N phenylbutazonum Chemical compound O=C1C(CCCC)C(=O)N(C=2C=CC=CC=2)N1C1=CC=CC=C1 VYMDGNCVAMGZFE-UHFFFAOYSA-N 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 238000000197 pyrolysis Methods 0.000 description 2
- ULFRLSNUDGIQQP-UHFFFAOYSA-N rizatriptan Chemical compound C1=C2C(CCN(C)C)=CNC2=CC=C1CN1C=NC=N1 ULFRLSNUDGIQQP-UHFFFAOYSA-N 0.000 description 2
- YGSDEFSMJLZEOE-UHFFFAOYSA-N salicylic acid Chemical compound OC(=O)C1=CC=CC=C1O YGSDEFSMJLZEOE-UHFFFAOYSA-N 0.000 description 2
- 238000004626 scanning electron microscopy Methods 0.000 description 2
- 230000007017 scission Effects 0.000 description 2
- 239000002356 single layer Substances 0.000 description 2
- 239000002002 slurry Substances 0.000 description 2
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 2
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 2
- 230000003595 spectral effect Effects 0.000 description 2
- 210000002784 stomach Anatomy 0.000 description 2
- 229940124530 sulfonamide Drugs 0.000 description 2
- 150000003456 sulfonamides Chemical class 0.000 description 2
- 235000011149 sulphuric acid Nutrition 0.000 description 2
- KQKPFRSPSRPDEB-UHFFFAOYSA-N sumatriptan Chemical compound CNS(=O)(=O)CC1=CC=C2NC=C(CCN(C)C)C2=C1 KQKPFRSPSRPDEB-UHFFFAOYSA-N 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- RJMIEHBSYVWVIN-LLVKDONJSA-N (2r)-2-[4-(3-oxo-1h-isoindol-2-yl)phenyl]propanoic acid Chemical compound C1=CC([C@H](C(O)=O)C)=CC=C1N1C(=O)C2=CC=CC=C2C1 RJMIEHBSYVWVIN-LLVKDONJSA-N 0.000 description 1
- RDJGLLICXDHJDY-NSHDSACASA-N (2s)-2-(3-phenoxyphenyl)propanoic acid Chemical compound OC(=O)[C@@H](C)C1=CC=CC(OC=2C=CC=CC=2)=C1 RDJGLLICXDHJDY-NSHDSACASA-N 0.000 description 1
- GUHPRPJDBZHYCJ-SECBINFHSA-N (2s)-2-(5-benzoylthiophen-2-yl)propanoic acid Chemical compound S1C([C@H](C(O)=O)C)=CC=C1C(=O)C1=CC=CC=C1 GUHPRPJDBZHYCJ-SECBINFHSA-N 0.000 description 1
- MDKGKXOCJGEUJW-VIFPVBQESA-N (2s)-2-[4-(thiophene-2-carbonyl)phenyl]propanoic acid Chemical compound C1=CC([C@@H](C(O)=O)C)=CC=C1C(=O)C1=CC=CS1 MDKGKXOCJGEUJW-VIFPVBQESA-N 0.000 description 1
- HSINOMROUCMIEA-FGVHQWLLSA-N (2s,4r)-4-[(3r,5s,6r,7r,8s,9s,10s,13r,14s,17r)-6-ethyl-3,7-dihydroxy-10,13-dimethyl-2,3,4,5,6,7,8,9,11,12,14,15,16,17-tetradecahydro-1h-cyclopenta[a]phenanthren-17-yl]-2-methylpentanoic acid Chemical compound C([C@@]12C)C[C@@H](O)C[C@H]1[C@@H](CC)[C@@H](O)[C@@H]1[C@@H]2CC[C@]2(C)[C@@H]([C@H](C)C[C@H](C)C(O)=O)CC[C@H]21 HSINOMROUCMIEA-FGVHQWLLSA-N 0.000 description 1
- PXGPLTODNUVGFL-BRIYLRKRSA-N (E,Z)-(1R,2R,3R,5S)-7-(3,5-Dihydroxy-2-((3S)-(3-hydroxy-1-octenyl))cyclopentyl)-5-heptenoic acid Chemical compound CCCCC[C@H](O)C=C[C@H]1[C@H](O)C[C@H](O)[C@@H]1CC=CCCCC(O)=O PXGPLTODNUVGFL-BRIYLRKRSA-N 0.000 description 1
- SYTBZMRGLBWNTM-SNVBAGLBSA-N (R)-flurbiprofen Chemical compound FC1=CC([C@H](C(O)=O)C)=CC=C1C1=CC=CC=C1 SYTBZMRGLBWNTM-SNVBAGLBSA-N 0.000 description 1
- NWUYHJFMYQTDRP-UHFFFAOYSA-N 1,2-bis(ethenyl)benzene;1-ethenyl-2-ethylbenzene;styrene Chemical compound C=CC1=CC=CC=C1.CCC1=CC=CC=C1C=C.C=CC1=CC=CC=C1C=C NWUYHJFMYQTDRP-UHFFFAOYSA-N 0.000 description 1
- GNUXVOXXWGNPIV-UHFFFAOYSA-N 1-methyl-5-[[methyl(1-phenylpropan-2-yl)amino]methyl]-2-phenyl-4-propan-2-yl-3-pyrazolone Chemical compound CN1N(C=2C=CC=CC=2)C(=O)C(C(C)C)=C1CN(C)C(C)CC1=CC=CC=C1 GNUXVOXXWGNPIV-UHFFFAOYSA-N 0.000 description 1
- VZUGVMQFWFVFBX-UHFFFAOYSA-N 2-(4-cyclohexyl-1-naphthyl)propanoic acid Chemical compound C12=CC=CC=C2C(C(C(O)=O)C)=CC=C1C1CCCCC1 VZUGVMQFWFVFBX-UHFFFAOYSA-N 0.000 description 1
- KRQUFUKTQHISJB-YYADALCUSA-N 2-[(E)-N-[2-(4-chlorophenoxy)propoxy]-C-propylcarbonimidoyl]-3-hydroxy-5-(thian-3-yl)cyclohex-2-en-1-one Chemical compound CCC\C(=N/OCC(C)OC1=CC=C(Cl)C=C1)C1=C(O)CC(CC1=O)C1CCCSC1 KRQUFUKTQHISJB-YYADALCUSA-N 0.000 description 1
- JIEKMACRVQTPRC-UHFFFAOYSA-N 2-[4-(4-chlorophenyl)-2-phenyl-5-thiazolyl]acetic acid Chemical compound OC(=O)CC=1SC(C=2C=CC=CC=2)=NC=1C1=CC=C(Cl)C=C1 JIEKMACRVQTPRC-UHFFFAOYSA-N 0.000 description 1
- NEAQRZUHTPSBBM-UHFFFAOYSA-N 2-hydroxy-3,3-dimethyl-7-nitro-4h-isoquinolin-1-one Chemical compound C1=C([N+]([O-])=O)C=C2C(=O)N(O)C(C)(C)CC2=C1 NEAQRZUHTPSBBM-UHFFFAOYSA-N 0.000 description 1
- BFCMUBZJTPADOD-UHFFFAOYSA-N 6-aminohexane-1-thiol;hydrochloride Chemical compound Cl.NCCCCCCS BFCMUBZJTPADOD-UHFFFAOYSA-N 0.000 description 1
- 208000004998 Abdominal Pain Diseases 0.000 description 1
- 208000035285 Allergic Seasonal Rhinitis Diseases 0.000 description 1
- RMMXTBMQSGEXHJ-UHFFFAOYSA-N Aminophenazone Chemical compound O=C1C(N(C)C)=C(C)N(C)N1C1=CC=CC=C1 RMMXTBMQSGEXHJ-UHFFFAOYSA-N 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- 208000008439 Biliary Liver Cirrhosis Diseases 0.000 description 1
- 208000033222 Biliary cirrhosis primary Diseases 0.000 description 1
- MNIPYSSQXLZQLJ-UHFFFAOYSA-N Biofenac Chemical compound OC(=O)COC(=O)CC1=CC=CC=C1NC1=C(Cl)C=CC=C1Cl MNIPYSSQXLZQLJ-UHFFFAOYSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 229920003043 Cellulose fiber Polymers 0.000 description 1
- 206010053684 Cerebrohepatorenal syndrome Diseases 0.000 description 1
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- 229910016523 CuKa Inorganic materials 0.000 description 1
- GUBGYTABKSRVRQ-CUHNMECISA-N D-Cellobiose 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)OC(O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-CUHNMECISA-N 0.000 description 1
- 206010012438 Dermatitis atopic Diseases 0.000 description 1
- 206010012735 Diarrhoea Diseases 0.000 description 1
- MYMOFIZGZYHOMD-UHFFFAOYSA-N Dioxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- RBBWCVQDXDFISW-UHFFFAOYSA-N Feprazone Chemical compound O=C1C(CC=C(C)C)C(=O)N(C=2C=CC=CC=2)N1C1=CC=CC=C1 RBBWCVQDXDFISW-UHFFFAOYSA-N 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- 208000012671 Gastrointestinal haemorrhages Diseases 0.000 description 1
- 206010061459 Gastrointestinal ulcer Diseases 0.000 description 1
- 208000007465 Giant cell arteritis Diseases 0.000 description 1
- 238000004566 IR spectroscopy Methods 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
- 206010061218 Inflammation Diseases 0.000 description 1
- LPHGQDQBBGAPDZ-UHFFFAOYSA-N Isocaffeine Natural products CN1C(=O)N(C)C(=O)C2=C1N(C)C=N2 LPHGQDQBBGAPDZ-UHFFFAOYSA-N 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- ZRVUJXDFFKFLMG-UHFFFAOYSA-N Meloxicam Chemical compound OC=1C2=CC=CC=C2S(=O)(=O)N(C)C=1C(=O)NC1=NC=C(C)S1 ZRVUJXDFFKFLMG-UHFFFAOYSA-N 0.000 description 1
- 208000007101 Muscle Cramp Diseases 0.000 description 1
- BLXXJMDCKKHMKV-UHFFFAOYSA-N Nabumetone Chemical compound C1=C(CCC(C)=O)C=CC2=CC(OC)=CC=C21 BLXXJMDCKKHMKV-UHFFFAOYSA-N 0.000 description 1
- 206010060860 Neurological symptom Diseases 0.000 description 1
- BRZANEXCSZCZCI-UHFFFAOYSA-N Nifenazone Chemical compound O=C1N(C=2C=CC=CC=2)N(C)C(C)=C1NC(=O)C1=CC=CN=C1 BRZANEXCSZCZCI-UHFFFAOYSA-N 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- 208000008469 Peptic Ulcer Diseases 0.000 description 1
- 208000020547 Peroxisomal disease Diseases 0.000 description 1
- 206010054956 Phonophobia Diseases 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 206010034960 Photophobia Diseases 0.000 description 1
- 208000007048 Polymyalgia Rheumatica Diseases 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 208000012654 Primary biliary cholangitis Diseases 0.000 description 1
- 102000001253 Protein Kinase Human genes 0.000 description 1
- 206010039085 Rhinitis allergic Diseases 0.000 description 1
- 206010039677 Scintillating scotoma Diseases 0.000 description 1
- 206010039729 Scotoma Diseases 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 208000007107 Stomach Ulcer Diseases 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 201000006704 Ulcerative Colitis Diseases 0.000 description 1
- 235000019888 Vivapur Nutrition 0.000 description 1
- 206010047700 Vomiting Diseases 0.000 description 1
- 201000004525 Zellweger Syndrome Diseases 0.000 description 1
- 208000036813 Zellweger spectrum disease Diseases 0.000 description 1
- 206010000059 abdominal discomfort Diseases 0.000 description 1
- 230000001133 acceleration Effects 0.000 description 1
- 229960004420 aceclofenac Drugs 0.000 description 1
- 229960004892 acemetacin Drugs 0.000 description 1
- FSQKKOOTNAMONP-UHFFFAOYSA-N acemetacin Chemical compound CC1=C(CC(=O)OCC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 FSQKKOOTNAMONP-UHFFFAOYSA-N 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000010933 acylation Effects 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 239000002390 adhesive tape Substances 0.000 description 1
- 229960005142 alclofenac Drugs 0.000 description 1
- ARHWPKZXBHOEEE-UHFFFAOYSA-N alclofenac Chemical compound OC(=O)CC1=CC=C(OCC=C)C(Cl)=C1 ARHWPKZXBHOEEE-UHFFFAOYSA-N 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 201000010105 allergic rhinitis Diseases 0.000 description 1
- 239000004411 aluminium Substances 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- SOYCMDCMZDHQFP-UHFFFAOYSA-N amfenac Chemical compound NC1=C(CC(O)=O)C=CC=C1C(=O)C1=CC=CC=C1 SOYCMDCMZDHQFP-UHFFFAOYSA-N 0.000 description 1
- 229950008930 amfenac Drugs 0.000 description 1
- QZNJPJDUBTYMRS-UHFFFAOYSA-M amfenac sodium hydrate Chemical compound O.[Na+].NC1=C(CC([O-])=O)C=CC=C1C(=O)C1=CC=CC=C1 QZNJPJDUBTYMRS-UHFFFAOYSA-M 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 235000001014 amino acid Nutrition 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 229960000212 aminophenazone Drugs 0.000 description 1
- LSNWBKACGXCGAJ-UHFFFAOYSA-N ampiroxicam Chemical compound CN1S(=O)(=O)C2=CC=CC=C2C(OC(C)OC(=O)OCC)=C1C(=O)NC1=CC=CC=N1 LSNWBKACGXCGAJ-UHFFFAOYSA-N 0.000 description 1
- 229950011249 ampiroxicam Drugs 0.000 description 1
- 229940051880 analgesics and antipyretics pyrazolones Drugs 0.000 description 1
- 239000012491 analyte Substances 0.000 description 1
- 230000008485 antagonism Effects 0.000 description 1
- 230000000507 anthelmentic effect Effects 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000398 anti-amebic effect Effects 0.000 description 1
- 230000002280 anti-androgenic effect Effects 0.000 description 1
- 230000003257 anti-anginal effect Effects 0.000 description 1
- 230000001088 anti-asthma Effects 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 230000001773 anti-convulsant effect Effects 0.000 description 1
- 230000003178 anti-diabetic effect Effects 0.000 description 1
- 230000003556 anti-epileptic effect Effects 0.000 description 1
- 230000000843 anti-fungal effect Effects 0.000 description 1
- 230000001387 anti-histamine Effects 0.000 description 1
- 230000001315 anti-hyperlipaemic effect Effects 0.000 description 1
- 230000003276 anti-hypertensive effect Effects 0.000 description 1
- 229940124599 anti-inflammatory drug Drugs 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000003561 anti-manic effect Effects 0.000 description 1
- 230000002460 anti-migrenic effect Effects 0.000 description 1
- 230000003579 anti-obesity Effects 0.000 description 1
- 230000003262 anti-osteoporosis Effects 0.000 description 1
- 230000000648 anti-parkinson Effects 0.000 description 1
- 230000000561 anti-psychotic effect Effects 0.000 description 1
- 230000000767 anti-ulcer Effects 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 239000000051 antiandrogen Substances 0.000 description 1
- 239000000924 antiasthmatic agent Substances 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000003146 anticoagulant agent Substances 0.000 description 1
- 229940127219 anticoagulant drug Drugs 0.000 description 1
- 239000003472 antidiabetic agent Substances 0.000 description 1
- 229960003965 antiepileptics Drugs 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000000739 antihistaminic agent Substances 0.000 description 1
- 229940111133 antiinflammatory and antirheumatic drug oxicams Drugs 0.000 description 1
- 229940111131 antiinflammatory and antirheumatic product propionic acid derivative Drugs 0.000 description 1
- 239000000939 antiparkinson agent Substances 0.000 description 1
- 229940125716 antipyretic agent Drugs 0.000 description 1
- VEQOALNAAJBPNY-UHFFFAOYSA-N antipyrine Chemical compound CN1C(C)=CC(=O)N1C1=CC=CC=C1 VEQOALNAAJBPNY-UHFFFAOYSA-N 0.000 description 1
- 229950008049 apricoxib Drugs 0.000 description 1
- JTMITOKKUMVWRT-UHFFFAOYSA-N apricoxib Chemical compound C1=CC(OCC)=CC=C1C1=CC(C)=CN1C1=CC=C(S(N)(=O)=O)C=C1 JTMITOKKUMVWRT-UHFFFAOYSA-N 0.000 description 1
- 239000007900 aqueous suspension Substances 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 239000012298 atmosphere Substances 0.000 description 1
- 238000000089 atomic force micrograph Methods 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 201000008937 atopic dermatitis Diseases 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 238000005284 basis set Methods 0.000 description 1
- 229960005149 bendazac Drugs 0.000 description 1
- BYFMCKSPFYVMOU-UHFFFAOYSA-N bendazac Chemical compound C12=CC=CC=C2C(OCC(=O)O)=NN1CC1=CC=CC=C1 BYFMCKSPFYVMOU-UHFFFAOYSA-N 0.000 description 1
- 229960005430 benoxaprofen Drugs 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- 239000003613 bile acid Substances 0.000 description 1
- QRZAKQDHEVVFRX-UHFFFAOYSA-N biphenyl-4-ylacetic acid Chemical compound C1=CC(CC(=O)O)=CC=C1C1=CC=CC=C1 QRZAKQDHEVVFRX-UHFFFAOYSA-N 0.000 description 1
- 238000007664 blowing Methods 0.000 description 1
- 229960003655 bromfenac Drugs 0.000 description 1
- ZBPLOVFIXSTCRZ-UHFFFAOYSA-N bromfenac Chemical compound NC1=C(CC(O)=O)C=CC=C1C(=O)C1=CC=C(Br)C=C1 ZBPLOVFIXSTCRZ-UHFFFAOYSA-N 0.000 description 1
- 230000031709 bromination Effects 0.000 description 1
- 238000005893 bromination reaction Methods 0.000 description 1
- 229960000962 bufexamac Drugs 0.000 description 1
- MXJWRABVEGLYDG-UHFFFAOYSA-N bufexamac Chemical compound CCCCOC1=CC=C(CC(=O)NO)C=C1 MXJWRABVEGLYDG-UHFFFAOYSA-N 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 244000309464 bull Species 0.000 description 1
- 229960003354 bumadizone Drugs 0.000 description 1
- FLWFHHFTIRLFPV-UHFFFAOYSA-N bumadizone Chemical compound C=1C=CC=CC=1N(C(=O)C(C(O)=O)CCCC)NC1=CC=CC=C1 FLWFHHFTIRLFPV-UHFFFAOYSA-N 0.000 description 1
- 229960001948 caffeine Drugs 0.000 description 1
- VJEONQKOZGKCAK-UHFFFAOYSA-N caffeine Natural products CN1C(=O)N(C)C(=O)C2=C1C=CN2C VJEONQKOZGKCAK-UHFFFAOYSA-N 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 238000004422 calculation algorithm Methods 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 125000004432 carbon atom Chemical group C* 0.000 description 1
- 229960003184 carprofen Drugs 0.000 description 1
- IVUMCTKHWDRRMH-UHFFFAOYSA-N carprofen Chemical compound C1=CC(Cl)=C[C]2C3=CC=C(C(C(O)=O)C)C=C3N=C21 IVUMCTKHWDRRMH-UHFFFAOYSA-N 0.000 description 1
- 229960000590 celecoxib Drugs 0.000 description 1
- RZEKVGVHFLEQIL-UHFFFAOYSA-N celecoxib Chemical compound C1=CC(C)=CC=C1C1=CC(C(F)(F)F)=NN1C1=CC=C(S(N)(=O)=O)C=C1 RZEKVGVHFLEQIL-UHFFFAOYSA-N 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- WRXDGGCKOUEOPW-UHFFFAOYSA-N ceritinib Chemical compound CC=1C=C(NC=2N=C(NC=3C(=CC=CC=3)NS(=O)(=O)C(C)C)C(Cl)=CN=2)C(OC(C)C)=CC=1C1CCNCC1 WRXDGGCKOUEOPW-UHFFFAOYSA-N 0.000 description 1
- 238000005660 chlorination reaction Methods 0.000 description 1
- 229950010851 cimicoxib Drugs 0.000 description 1
- KYXDNECMRLFQMZ-UHFFFAOYSA-N cimicoxib Chemical compound C1=C(F)C(OC)=CC=C1C1=C(Cl)N=CN1C1=CC=C(S(N)(=O)=O)C=C1 KYXDNECMRLFQMZ-UHFFFAOYSA-N 0.000 description 1
- 230000007882 cirrhosis Effects 0.000 description 1
- 208000019425 cirrhosis of liver Diseases 0.000 description 1
- 230000007012 clinical effect Effects 0.000 description 1
- 229960003140 clofezone Drugs 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 229960004126 codeine Drugs 0.000 description 1
- 230000001427 coherent effect Effects 0.000 description 1
- 238000009833 condensation Methods 0.000 description 1
- 230000005494 condensation Effects 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 229940124558 contraceptive agent Drugs 0.000 description 1
- 238000001816 cooling Methods 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 239000003246 corticosteroid Substances 0.000 description 1
- 229960001334 corticosteroids Drugs 0.000 description 1
- 229940111134 coxibs Drugs 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 230000001186 cumulative effect Effects 0.000 description 1
- 239000003255 cyclooxygenase 2 inhibitor Substances 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 229960003314 deracoxib Drugs 0.000 description 1
- WAZQAZKAZLXFMK-UHFFFAOYSA-N deracoxib Chemical compound C1=C(F)C(OC)=CC=C1C1=CC(C(F)F)=NN1C1=CC=C(S(N)(=O)=O)C=C1 WAZQAZKAZLXFMK-UHFFFAOYSA-N 0.000 description 1
- 229960003428 dexibuprofen Drugs 0.000 description 1
- HEFNNWSXXWATRW-JTQLQIEISA-N dexibuprofen Chemical compound CC(C)CC1=CC=C([C@H](C)C(O)=O)C=C1 HEFNNWSXXWATRW-JTQLQIEISA-N 0.000 description 1
- 229960002783 dexketoprofen Drugs 0.000 description 1
- DKYWVDODHFEZIM-NSHDSACASA-N dexketoprofen Chemical compound OC(=O)[C@@H](C)C1=CC=CC(C(=O)C=2C=CC=CC=2)=C1 DKYWVDODHFEZIM-NSHDSACASA-N 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 229960001259 diclofenac Drugs 0.000 description 1
- DCOPUUMXTXDBNB-UHFFFAOYSA-N diclofenac Chemical compound OC(=O)CC1=CC=CC=C1NC1=C(Cl)C=CC=C1Cl DCOPUUMXTXDBNB-UHFFFAOYSA-N 0.000 description 1
- 229960001536 difenpiramide Drugs 0.000 description 1
- PWHROYKAGRUWDQ-UHFFFAOYSA-N difenpiramide Chemical compound C=1C=CC=NC=1NC(=O)CC(C=C1)=CC=C1C1=CC=CC=C1 PWHROYKAGRUWDQ-UHFFFAOYSA-N 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 229910001882 dioxygen Inorganic materials 0.000 description 1
- 229940120889 dipyrone Drugs 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 239000002934 diuretic Substances 0.000 description 1
- 230000001882 diuretic effect Effects 0.000 description 1
- 229960001850 droxicam Drugs 0.000 description 1
- OEHFRZLKGRKFAS-UHFFFAOYSA-N droxicam Chemical compound C12=CC=CC=C2S(=O)(=O)N(C)C(C2=O)=C1OC(=O)N2C1=CC=CC=N1 OEHFRZLKGRKFAS-UHFFFAOYSA-N 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 229940000406 drug candidate Drugs 0.000 description 1
- 229940126534 drug product Drugs 0.000 description 1
- 230000009977 dual effect Effects 0.000 description 1
- 229960002472 eletriptan Drugs 0.000 description 1
- 238000000295 emission spectrum Methods 0.000 description 1
- 230000002357 endometrial effect Effects 0.000 description 1
- 206010015037 epilepsy Diseases 0.000 description 1
- 230000032050 esterification Effects 0.000 description 1
- 238000005886 esterification reaction Methods 0.000 description 1
- 238000006266 etherification reaction Methods 0.000 description 1
- 150000002170 ethers Chemical class 0.000 description 1
- 229960005293 etodolac Drugs 0.000 description 1
- XFBVBWWRPKNWHW-UHFFFAOYSA-N etodolac Chemical compound C1COC(CC)(CC(O)=O)C2=N[C]3C(CC)=CC=CC3=C21 XFBVBWWRPKNWHW-UHFFFAOYSA-N 0.000 description 1
- 229960004945 etoricoxib Drugs 0.000 description 1
- MNJVRJDLRVPLFE-UHFFFAOYSA-N etoricoxib Chemical compound C1=NC(C)=CC=C1C1=NC=C(Cl)C=C1C1=CC=C(S(C)(=O)=O)C=C1 MNJVRJDLRVPLFE-UHFFFAOYSA-N 0.000 description 1
- 238000000695 excitation spectrum Methods 0.000 description 1
- 229950009324 famprofazone Drugs 0.000 description 1
- 229960000192 felbinac Drugs 0.000 description 1
- 229960001395 fenbufen Drugs 0.000 description 1
- ZPAKPRAICRBAOD-UHFFFAOYSA-N fenbufen Chemical compound C1=CC(C(=O)CCC(=O)O)=CC=C1C1=CC=CC=C1 ZPAKPRAICRBAOD-UHFFFAOYSA-N 0.000 description 1
- 229960001419 fenoprofen Drugs 0.000 description 1
- 229960002679 fentiazac Drugs 0.000 description 1
- 229960000489 feprazone Drugs 0.000 description 1
- 230000004720 fertilization Effects 0.000 description 1
- 238000000445 field-emission scanning electron microscopy Methods 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000000706 filtrate Substances 0.000 description 1
- 238000001914 filtration Methods 0.000 description 1
- 229960002524 firocoxib Drugs 0.000 description 1
- FULAPETWGIGNMT-UHFFFAOYSA-N firocoxib Chemical compound C=1C=C(S(C)(=O)=O)C=CC=1C=1C(C)(C)OC(=O)C=1OCC1CC1 FULAPETWGIGNMT-UHFFFAOYSA-N 0.000 description 1
- 229960001321 flunoxaprofen Drugs 0.000 description 1
- ARPYQKTVRGFPIS-VIFPVBQESA-N flunoxaprofen Chemical compound N=1C2=CC([C@@H](C(O)=O)C)=CC=C2OC=1C1=CC=C(F)C=C1 ARPYQKTVRGFPIS-VIFPVBQESA-N 0.000 description 1
- 238000002189 fluorescence spectrum Methods 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 230000002496 gastric effect Effects 0.000 description 1
- 208000030304 gastrointestinal bleeding Diseases 0.000 description 1
- 230000001330 gastroprokinetic effect Effects 0.000 description 1
- 239000003163 gonadal steroid hormone Substances 0.000 description 1
- 235000013882 gravy Nutrition 0.000 description 1
- 125000001475 halogen functional group Chemical group 0.000 description 1
- 230000026030 halogenation Effects 0.000 description 1
- 238000005658 halogenation reaction Methods 0.000 description 1
- 231100001261 hazardous Toxicity 0.000 description 1
- 125000000623 heterocyclic group Chemical group 0.000 description 1
- 239000007970 homogeneous dispersion Substances 0.000 description 1
- OROGSEYTTFOCAN-UHFFFAOYSA-N hydrocodone Natural products C1C(N(CCC234)C)C2C=CC(O)C3OC2=C4C1=CC=C2OC OROGSEYTTFOCAN-UHFFFAOYSA-N 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 208000013403 hyperactivity Diseases 0.000 description 1
- 229960002595 ibuproxam Drugs 0.000 description 1
- BYPIURIATSUHDW-UHFFFAOYSA-N ibuproxam Chemical compound CC(C)CC1=CC=C(C(C)C(=O)NO)C=C1 BYPIURIATSUHDW-UHFFFAOYSA-N 0.000 description 1
- 229940090436 imitrex Drugs 0.000 description 1
- 239000012729 immediate-release (IR) formulation Substances 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 229960003444 immunosuppressant agent Drugs 0.000 description 1
- 230000001861 immunosuppressant effect Effects 0.000 description 1
- 239000003018 immunosuppressive agent Substances 0.000 description 1
- 229960004187 indoprofen Drugs 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000009878 intermolecular interaction Effects 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 230000026045 iodination Effects 0.000 description 1
- 238000006192 iodination reaction Methods 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- YYUAYBYLJSNDCX-UHFFFAOYSA-N isoxicam Chemical compound OC=1C2=CC=CC=C2S(=O)(=O)N(C)C=1C(=O)NC=1C=C(C)ON=1 YYUAYBYLJSNDCX-UHFFFAOYSA-N 0.000 description 1
- 229950002252 isoxicam Drugs 0.000 description 1
- 210000001503 joint Anatomy 0.000 description 1
- LGYTZKPVOAIUKX-UHFFFAOYSA-N kebuzone Chemical compound O=C1C(CCC(=O)C)C(=O)N(C=2C=CC=CC=2)N1C1=CC=CC=C1 LGYTZKPVOAIUKX-UHFFFAOYSA-N 0.000 description 1
- 229960000194 kebuzone Drugs 0.000 description 1
- 150000002576 ketones Chemical class 0.000 description 1
- 229960004752 ketorolac Drugs 0.000 description 1
- OZWKMVRBQXNZKK-UHFFFAOYSA-N ketorolac Chemical compound OC(=O)C1CCN2C1=CC=C2C(=O)C1=CC=CC=C1 OZWKMVRBQXNZKK-UHFFFAOYSA-N 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 230000003907 kidney function Effects 0.000 description 1
- 208000037805 labour Diseases 0.000 description 1
- 231100001231 less toxic Toxicity 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 238000012417 linear regression Methods 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000003908 liver function Effects 0.000 description 1
- 229960003768 lonazolac Drugs 0.000 description 1
- XVUQHFRQHBLHQD-UHFFFAOYSA-N lonazolac Chemical compound OC(=O)CC1=CN(C=2C=CC=CC=2)N=C1C1=CC=C(Cl)C=C1 XVUQHFRQHBLHQD-UHFFFAOYSA-N 0.000 description 1
- OXROWJKCGCOJDO-JLHYYAGUSA-N lornoxicam Chemical compound O=C1C=2SC(Cl)=CC=2S(=O)(=O)N(C)\C1=C(\O)NC1=CC=CC=N1 OXROWJKCGCOJDO-JLHYYAGUSA-N 0.000 description 1
- 229960002202 lornoxicam Drugs 0.000 description 1
- 231100000053 low toxicity Toxicity 0.000 description 1
- 229960002373 loxoprofen Drugs 0.000 description 1
- BAZQYVYVKYOAGO-UHFFFAOYSA-M loxoprofen sodium hydrate Chemical compound O.O.[Na+].C1=CC(C(C([O-])=O)C)=CC=C1CC1C(=O)CCC1 BAZQYVYVKYOAGO-UHFFFAOYSA-M 0.000 description 1
- 229960000994 lumiracoxib Drugs 0.000 description 1
- KHPKQFYUPIUARC-UHFFFAOYSA-N lumiracoxib Chemical compound OC(=O)CC1=CC(C)=CC=C1NC1=C(F)C=CC=C1Cl KHPKQFYUPIUARC-UHFFFAOYSA-N 0.000 description 1
- 206010025135 lupus erythematosus Diseases 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 229950007241 mavacoxib Drugs 0.000 description 1
- TTZNQDOUNXBMJV-UHFFFAOYSA-N mavacoxib Chemical compound C1=CC(S(=O)(=O)N)=CC=C1N1C(C=2C=CC(F)=CC=2)=CC(C(F)(F)F)=N1 TTZNQDOUNXBMJV-UHFFFAOYSA-N 0.000 description 1
- 229940103177 maxalt Drugs 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 238000002483 medication Methods 0.000 description 1
- 229960001929 meloxicam Drugs 0.000 description 1
- 208000007106 menorrhagia Diseases 0.000 description 1
- 210000004914 menses Anatomy 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- LVWZTYCIRDMTEY-UHFFFAOYSA-N metamizole Chemical compound O=C1C(N(CS(O)(=O)=O)C)=C(C)N(C)N1C1=CC=CC=C1 LVWZTYCIRDMTEY-UHFFFAOYSA-N 0.000 description 1
- GDOPTJXRTPNYNR-UHFFFAOYSA-N methyl-cyclopentane Natural products CC1CCCC1 GDOPTJXRTPNYNR-UHFFFAOYSA-N 0.000 description 1
- 238000001000 micrograph Methods 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- OJGQFYYLKNCIJD-UHFFFAOYSA-N miroprofen Chemical compound C1=CC(C(C(O)=O)C)=CC=C1C1=CN(C=CC=C2)C2=N1 OJGQFYYLKNCIJD-UHFFFAOYSA-N 0.000 description 1
- 229950006616 miroprofen Drugs 0.000 description 1
- REOJLIXKJWXUGB-UHFFFAOYSA-N mofebutazone Chemical compound O=C1C(CCCC)C(=O)NN1C1=CC=CC=C1 REOJLIXKJWXUGB-UHFFFAOYSA-N 0.000 description 1
- 229960005285 mofebutazone Drugs 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- OOGNFQMTGRZRAB-UHFFFAOYSA-N morazone Chemical compound CC1C(C=2C=CC=CC=2)OCCN1CC(C1=O)=C(C)N(C)N1C1=CC=CC=C1 OOGNFQMTGRZRAB-UHFFFAOYSA-N 0.000 description 1
- 229960004610 morazone Drugs 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- VGJDSNZOPPZCTB-UHFFFAOYSA-N n-methyl-2-(4-nitrophenyl)ethanamine;hydrochloride Chemical compound Cl.CNCCC1=CC=C([N+]([O-])=O)C=C1 VGJDSNZOPPZCTB-UHFFFAOYSA-N 0.000 description 1
- 229960004270 nabumetone Drugs 0.000 description 1
- 229960003759 naproxcinod Drugs 0.000 description 1
- AKFJWRDCWYYTIG-ZDUSSCGKSA-N naproxcinod Chemical compound C1=C([C@H](C)C(=O)OCCCCO[N+]([O-])=O)C=CC2=CC(OC)=CC=C21 AKFJWRDCWYYTIG-ZDUSSCGKSA-N 0.000 description 1
- 229920005615 natural polymer Polymers 0.000 description 1
- JWNPDZNEKVCWMY-VQHVLOKHSA-N neratinib Chemical compound C=12C=C(NC(=O)\C=C\CN(C)C)C(OCC)=CC2=NC=C(C#N)C=1NC(C=C1Cl)=CC=C1OCC1=CC=CC=N1 JWNPDZNEKVCWMY-VQHVLOKHSA-N 0.000 description 1
- 229960002187 nifenazone Drugs 0.000 description 1
- 229960000965 nimesulide Drugs 0.000 description 1
- HYWYRSMBCFDLJT-UHFFFAOYSA-N nimesulide Chemical compound CS(=O)(=O)NC1=CC=C([N+]([O-])=O)C=C1OC1=CC=CC=C1 HYWYRSMBCFDLJT-UHFFFAOYSA-N 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- 125000004433 nitrogen atom Chemical group N* 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 239000007935 oral tablet Substances 0.000 description 1
- 229940096978 oral tablet Drugs 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 238000012261 overproduction Methods 0.000 description 1
- 229960000273 oxametacin Drugs 0.000 description 1
- AJRNYCDWNITGHF-UHFFFAOYSA-N oxametacin Chemical compound CC1=C(CC(=O)NO)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 AJRNYCDWNITGHF-UHFFFAOYSA-N 0.000 description 1
- 229960002739 oxaprozin Drugs 0.000 description 1
- OFPXSFXSNFPTHF-UHFFFAOYSA-N oxaprozin Chemical compound O1C(CCC(=O)O)=NC(C=2C=CC=CC=2)=C1C1=CC=CC=C1 OFPXSFXSNFPTHF-UHFFFAOYSA-N 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 229960000649 oxyphenbutazone Drugs 0.000 description 1
- HFHZKZSRXITVMK-UHFFFAOYSA-N oxyphenbutazone Chemical compound O=C1C(CCCC)C(=O)N(C=2C=CC=CC=2)N1C1=CC=C(O)C=C1 HFHZKZSRXITVMK-UHFFFAOYSA-N 0.000 description 1
- 238000010979 pH adjustment Methods 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N papa-hydroxy-benzoic acid Natural products OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- 229960004662 parecoxib Drugs 0.000 description 1
- TZRHLKRLEZJVIJ-UHFFFAOYSA-N parecoxib Chemical compound C1=CC(S(=O)(=O)NC(=O)CC)=CC=C1C1=C(C)ON=C1C1=CC=CC=C1 TZRHLKRLEZJVIJ-UHFFFAOYSA-N 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000011236 particulate material Substances 0.000 description 1
- 238000005192 partition Methods 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 229960005222 phenazone Drugs 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 229960002702 piroxicam Drugs 0.000 description 1
- QYSPLQLAKJAUJT-UHFFFAOYSA-N piroxicam Chemical compound OC=1C2=CC=CC=C2S(=O)(=O)N(C)C=1C(=O)NC1=CC=CC=N1 QYSPLQLAKJAUJT-UHFFFAOYSA-N 0.000 description 1
- 229960000851 pirprofen Drugs 0.000 description 1
- PIDSZXPFGCURGN-UHFFFAOYSA-N pirprofen Chemical compound ClC1=CC(C(C(O)=O)C)=CC=C1N1CC=CC1 PIDSZXPFGCURGN-UHFFFAOYSA-N 0.000 description 1
- 230000036470 plasma concentration Effects 0.000 description 1
- 239000002798 polar solvent Substances 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 238000006116 polymerization reaction Methods 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 230000035935 pregnancy Effects 0.000 description 1
- 238000002203 pretreatment Methods 0.000 description 1
- 150000003138 primary alcohols Chemical class 0.000 description 1
- 238000003672 processing method Methods 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 229940002612 prodrug Drugs 0.000 description 1
- 229960000825 proglumetacin Drugs 0.000 description 1
- PTXGHCGBYMQQIG-UHFFFAOYSA-N proglumetacin Chemical compound C=1C=CC=CC=1C(=O)NC(C(=O)N(CCC)CCC)CCC(=O)OCCCN(CC1)CCN1CCOC(=O)CC(C1=CC(OC)=CC=C11)=C(C)N1C(=O)C1=CC=C(Cl)C=C1 PTXGHCGBYMQQIG-UHFFFAOYSA-N 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 150000005599 propionic acid derivatives Chemical class 0.000 description 1
- 229960002189 propyphenazone Drugs 0.000 description 1
- PXWLVJLKJGVOKE-UHFFFAOYSA-N propyphenazone Chemical compound O=C1C(C(C)C)=C(C)N(C)N1C1=CC=CC=C1 PXWLVJLKJGVOKE-UHFFFAOYSA-N 0.000 description 1
- 108060006633 protein kinase Proteins 0.000 description 1
- 235000018102 proteins Nutrition 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 230000000541 pulsatile effect Effects 0.000 description 1
- JEXVQSWXXUJEMA-UHFFFAOYSA-N pyrazol-3-one Chemical class O=C1C=CN=N1 JEXVQSWXXUJEMA-UHFFFAOYSA-N 0.000 description 1
- 150000003218 pyrazolidines Chemical class 0.000 description 1
- YREYEVIYCVEVJK-UHFFFAOYSA-N rabeprazole Chemical compound COCCCOC1=CC=NC(CS(=O)C=2NC3=CC=CC=C3N=2)=C1C YREYEVIYCVEVJK-UHFFFAOYSA-N 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 230000000306 recurrent effect Effects 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 231100000628 reference dose Toxicity 0.000 description 1
- 238000000611 regression analysis Methods 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 229940070979 relpax Drugs 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 229960000425 rizatriptan Drugs 0.000 description 1
- ZEXGDYFACFXQPF-UHFFFAOYSA-N robenacoxib Chemical compound OC(=O)CC1=CC(CC)=CC=C1NC1=C(F)C(F)=CC(F)=C1F ZEXGDYFACFXQPF-UHFFFAOYSA-N 0.000 description 1
- 229960000205 robenacoxib Drugs 0.000 description 1
- 229960000371 rofecoxib Drugs 0.000 description 1
- RZJQGNCSTQAWON-UHFFFAOYSA-N rofecoxib Chemical compound C1=CC(S(=O)(=O)C)=CC=C1C1=C(C=2C=CC=CC=2)C(=O)OC1 RZJQGNCSTQAWON-UHFFFAOYSA-N 0.000 description 1
- 238000010079 rubber tapping Methods 0.000 description 1
- 229960004889 salicylic acid Drugs 0.000 description 1
- 229940058287 salicylic acid derivative anticestodals Drugs 0.000 description 1
- 150000003872 salicylic acid derivatives Chemical class 0.000 description 1
- 150000003333 secondary alcohols Chemical class 0.000 description 1
- 239000000932 sedative agent Substances 0.000 description 1
- 230000001624 sedative effect Effects 0.000 description 1
- 238000004088 simulation Methods 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000012453 solvate Substances 0.000 description 1
- 239000011877 solvent mixture Substances 0.000 description 1
- 239000012798 spherical particle Substances 0.000 description 1
- 239000012086 standard solution Substances 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 239000000021 stimulant Substances 0.000 description 1
- 238000003756 stirring Methods 0.000 description 1
- 238000005556 structure-activity relationship Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- MBGGBVCUIVRRBF-UHFFFAOYSA-N sulfinpyrazone Chemical compound O=C1N(C=2C=CC=CC=2)N(C=2C=CC=CC=2)C(=O)C1CCS(=O)C1=CC=CC=C1 MBGGBVCUIVRRBF-UHFFFAOYSA-N 0.000 description 1
- 229960003329 sulfinpyrazone Drugs 0.000 description 1
- 238000006277 sulfonation reaction Methods 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000001117 sulphuric acid Substances 0.000 description 1
- 229960003708 sumatriptan Drugs 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 229960004492 suprofen Drugs 0.000 description 1
- ONWXNHPOAGOMTG-UHFFFAOYSA-N suxibuzone Chemical compound O=C1C(CCCC)(COC(=O)CCC(O)=O)C(=O)N(C=2C=CC=CC=2)N1C1=CC=CC=C1 ONWXNHPOAGOMTG-UHFFFAOYSA-N 0.000 description 1
- 229960003755 suxibuzone Drugs 0.000 description 1
- 238000002636 symptomatic treatment Methods 0.000 description 1
- 229950005628 tarenflurbil Drugs 0.000 description 1
- 239000006068 taste-masking agent Substances 0.000 description 1
- 206010043207 temporal arteritis Diseases 0.000 description 1
- 210000002435 tendon Anatomy 0.000 description 1
- 229960002871 tenoxicam Drugs 0.000 description 1
- WZWYJBNHTWCXIM-UHFFFAOYSA-N tenoxicam Chemical compound O=C1C=2SC=CC=2S(=O)(=O)N(C)C1=C(O)NC1=CC=CC=N1 WZWYJBNHTWCXIM-UHFFFAOYSA-N 0.000 description 1
- XYKWNRUXCOIMFZ-UHFFFAOYSA-N tepoxalin Chemical compound C1=CC(OC)=CC=C1N1C(C=2C=CC(Cl)=CC=2)=CC(CCC(=O)N(C)O)=N1 XYKWNRUXCOIMFZ-UHFFFAOYSA-N 0.000 description 1
- 229950009638 tepoxalin Drugs 0.000 description 1
- 125000003831 tetrazolyl group Chemical group 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 238000002076 thermal analysis method Methods 0.000 description 1
- 238000001757 thermogravimetry curve Methods 0.000 description 1
- 239000004416 thermosoftening plastic Substances 0.000 description 1
- 229960001312 tiaprofenic acid Drugs 0.000 description 1
- 229960001017 tolmetin Drugs 0.000 description 1
- UPSPUYADGBWSHF-UHFFFAOYSA-N tolmetin Chemical compound C1=CC(C)=CC=C1C(=O)C1=CC=C(CC(O)=O)N1C UPSPUYADGBWSHF-UHFFFAOYSA-N 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 231100000397 ulcer Toxicity 0.000 description 1
- 238000002604 ultrasonography Methods 0.000 description 1
- 230000002568 urticarial effect Effects 0.000 description 1
- 229960002004 valdecoxib Drugs 0.000 description 1
- 229950010082 vedaprofen Drugs 0.000 description 1
- 229910052724 xenon Inorganic materials 0.000 description 1
- FHNFHKCVQCLJFQ-UHFFFAOYSA-N xenon atom Chemical compound [Xe] FHNFHKCVQCLJFQ-UHFFFAOYSA-N 0.000 description 1
- 229960003414 zomepirac Drugs 0.000 description 1
- ZXVNMYWKKDOREA-UHFFFAOYSA-N zomepirac Chemical compound C1=C(CC(O)=O)N(C)C(C(=O)C=2C=CC(Cl)=CC=2)=C1C ZXVNMYWKKDOREA-UHFFFAOYSA-N 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
- 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
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/30—Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
- A61K47/36—Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
- A61K47/38—Cellulose; Derivatives thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/14—Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
- A61K9/16—Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
- A61K9/1605—Excipients; Inactive ingredients
- A61K9/1629—Organic macromolecular compounds
- A61K9/1652—Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
-
- C—CHEMISTRY; METALLURGY
- C08—ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
- C08L—COMPOSITIONS OF MACROMOLECULAR COMPOUNDS
- C08L1/00—Compositions of cellulose, modified cellulose or cellulose derivatives
- C08L1/02—Cellulose; Modified cellulose
Definitions
- This invention relates to new cellulose-based pharmaceutical compositions that provide for enhanced solubility of active ingredients, such as Type 2 and 4 BCS drugs, featured with low solubility in the gastrointestinal tract.
- the invention also relates to methods of manufacturing such pharmaceutical compositions.
- a drug substance is considered HIGHLY SOLUBLE when the highest dose strength is soluble in ⁇ 250 ml water over a pH range of 1 to 7.5.
- a drug substance is considered HIGHLY PERMEABLE when the extent of absorption in humans is determined to be > 90% of an administered dose, based on mass-balance or in comparison to an intravenous reference dose.
- a drug product is considered to be RAPIDLY DISSOLVING when > 85% of the labeled amount of drug substance dissolves within 30 minutes using USP apparatus I or II in a volume of ⁇ 900 ml buffer solutions.
- a dose number Do may also be useful as a solubility indicator:
- the present invention pertains to enhancing the solubility and bioavailability of Type 2 and 4 BCS drugs.
- Various APIs which are classified as Type 2 or 4 BCS drugs are disclosed in Takagi et al. Mol. Pharm. 2006; 3(6): 631-643; Lindenberg et al., Eur. J. Pharm. Biopharm. 2004; 58: 265-278; and Pham T. et al., Mol. Pharm. 2013; 10: 2445-2461.
- These references mainly deal with APIs on the WHO essential drugs list.
- the invention is useful also for developmental substances since it is known that at least more than half and as much as 90% of drug candidates are poorly soluble substances.
- the molecular descriptors for typical BCS Type 2 and 4 substances can be used as summarized by Pham T. et al. Mol. Pharm. 2013; 10: 2445-2461 , provided that they contain at least one aromatic ring or polycondensed cyclic structure.
- Table 1 Typical molecular descriptors of various classes BCS drugs. Table adapted from Pham T. et al. Mol. Pharm. 2013; 10: 2445-2461.
- nHA+B range 1 -15 1-40 2-27 1 -28 nHA+B 4.97/4 6.24/6 8.81/8 10.81/10 mean/median
- RBN range 0-13 0-19 0-16 1 -20
- NSAIDs are normally Type 2 or 4 BCS substances with low solubility although some of them, such as salicylic acid derivatives e.g. ASA, salicylic acid, ethenzamide, etc., or acetaminophen, are classed as Type 1 (or Type 3) substances.
- NSAIDs are a practical model of poorly soluble drugs because they are affordable and have relatively low toxicity as compared to other pharmacological classes of drugs and therefore are less hazardous for research personnel and environment.
- NSAIDs mimic arachidonic acid (AA) to be able to inhibit COX, i.e. (i) they contain a centre of acidity or a free carboxylic group and (ii) are largely lipophilic and feature at least one aromatic ring.
- NSAIDs have a centre of acidity, typically represented by a carboxylic acid group or alternatively by enolic group, hydroxamic acid, sulfonamide, or a tetrazole ring.
- the acidic group of NSAIDs is important for H-bonding with the polar region at Arg120 and oppositely located Tyr355 of COX (Hawkey 1999. Lancet 353:307- 314).
- NSAIDs possess at least one aromatic ring, which renders NSAIDs lipophilic, which is essential for inhibiting COX enzymes and ensures unhindered passive diffusion across biological membranes.
- the overall lipophilic character of NSAIDs results in poor solubility-limited bioavailability, i.e. Type 2 and 4 BCS drugs.
- Another important parameter is rate of absorption. Because most NSAIDs have poor solubility, their rate of absorption is limited and this results in poor bioavailability. To enhance the bioavailability and achieve a therapeutic effect, NSAIDs are typically administered in high doses and the peak plasma concentration is reached usually in 2 hours.
- NSAIDs are partly the reason for the side effects, such as peptic ulcers and ultimately bleeding or perforation of the stomach, associated with these drugs.
- Some 10% to 50% of patients are unable to tolerate treatment with NSAIDs because of side effects, including abdominal pain, diarrhoea, and upset stomach.
- Oral route of drug administration is the most frequently used and convenient route for administration of many drugs.
- Microcrystalline cellulose is an important tableting excipient for oral administration.
- MCC microcrystalline cellulose
- DP 100-350 level-off degree of polymerization
- MCC is an essentially non-porous powder featuring a surface area around 0.5-1 m 2 /g as measured by nitrogen gas adsorption (ibid). MCC is normally produced by spray-drying of the cellulose slurry that is obtained following the acidic hydrolysis.
- PCC porous crystalline cellulose
- Nakai Y. et al., Chem. Pharm. Bull. 26(10) 2983-2989 (1978) discloses compositions of MCC with naphthalene (non-pharmaceutical ingredient), camphor and o-cresol, non- pharmaceutical ingredient.
- Nakai Y., Drug Dev. Ind. Pharm. 1986; 12(7): 1017-1039. discloses formulations containing ground MCC mixtures having enhanced solubility. It is clear from Nakai (1986) that the drugs are not stable in the formulations disclosed therein. Furthermore, it is evident from the XRD data that the structure of the MCC gradually changes resulting in the MCC becoming progressively more amorphous over time.
- compositions comprising a modified microcrystalline cellulose excipient and an active pharmaceutical ingredient, wherein the modified microcrystalline cellulose excipient has a surface area of from about 2 to about 60 m 2 /g.
- compositions comprising such features are hereinafter referred to together as "the compositions of the invention”.
- compositions of the invention provide for rapid release of active pharmaceutical ingredients, particularly Type 2 and 4 BCS active ingredients, and also provide for an enhancement of their bioavailability in the gastrointestinal tract (GIT) following peroral administration of the composition.
- substantially all (e.g. at least about 85%) of the drug is released from the formulation in the first hour following administration of the formulation.
- the release may also be such that amount of drug released from the composition reaches a plateau within a short time (e.g. within about 1 hour, such as within about 15 minutes). That is, substantially all (e.g. at least about 90%) of the total amount of drug that is released during the first 24 hours may be released during the first two hours (e.g. during the first 60 minutes).
- the terms "rapid release” and “immediate release” are used interchangeably.
- the compositions of the invention are not limited to oral administration, and can be delivered by other routes, as is discussed elsewhere herein.
- rapid release will be understood by the skilled person to refer to compositions that provide for, and/or are adapted to provide for, a “quick” and/or an “immediate” release of drug (in which drug is released at a sufficiently high rate for a sufficient period of time to produce a therapeutic response in a very short time) from the formulation.
- Release from the formulation refers to release of the drug from the network structure of the modified microcrystalline cellulose excipient.
- compositions of the invention are also advantageous as they exhibit an increased physical stability of the active pharmaceutical ingredient (e.g. an increased stability of its physical form) within the formulation.
- the active pharmaceutical ingredient which is typically a Type 2 or 4 BCS substance, is preferably present in a predominantly amorphous form.
- the compositions allow the active pharmaceutical ingredient to remain in such a predominantly amorphous form for an extended period of time (e.g. up to at least 1 month, such as up to at least six months).
- the active pharmaceutical ingredient Upon release from the cellulose carrier, the active pharmaceutical ingredient is provided in a form having a high degree of amorphicity compared to other formulations. This in turn enhances the rate of dissolution of the active pharmaceutical ingredient in surrounding media, and accelerates uptake by the body.
- compositions of the invention may contain a plurality of active pharmaceutical ingredients, provided that at least one of said active pharmaceutical ingredients, which is typically a Type 2 or 4 BCS substance, is present in a predominantly amorphous form.
- Enhanced dissolution is particularly important for Type 2 and 4 BCS active ingredients as these substances have a low solubility profile.
- the overall dose of drug that is provided to the subject may be reduced while still achieving the intended therapeutic benefits.
- Microcrystalline cellulose is a well-defined and thoroughly studied natural polymer that is profoundly important in the pharmaceutical industry, especially in oral tablet formulations. MCC is particularly useful as a pharmaceutical tableting aid. A complete description of MCC is provided in European Pharmacopoeia.
- Microcrystalline cellulose is produced by mineral acid hydrolysis of refined cellulose to a level-off degree of polymerisation (DP 100-350). Under controlled conditions, the acid induces hydrolytic cleavage of glycosidic bonds in the amorphous regions of cellulose in the fibres leaving crystalline regions intact. The free crystallites flocculate and form compact structures of the cellulose fibre aggregates in the dispersion. The produced slurry is then washed and spray-dried to finally produce dry solid powder.
- the surface area of such conventional MCC is typically about 0.5-1 m 2 /g, measured by N2 gas adsorption.
- compositions of the invention comprise a modified microcrystalline cellulose excipient, which is obtainable from microcrystalline cellulose.
- the modified microcrystalline cellulose excipient has a surface area of from about 2 to about 60 m 2 /g which is significantly higher than that associated with conventional microcrystalline cellulose (which typically has a surface area in the region of 0.5-1 m 2 /g, measured by N2 gas adsorption), and significantly lower than that associated with "porous crystalline cellulose", PCC (which typically has a surface area in excess of 90 m 2 /g).
- the modified microcrystalline cellulose excipient disclosed herein can inhibit recrystallization of amorphous drug even if it features pores predominantly of about 50 nm in size (diameter), while previously it was suggested that pores smaller than 30 nm, and more preferably smaller than 10 nm, are needed to observe such effect.
- the active pores are predominantly in the region around 4 nm in size, see Oguchi et al. Yakuzaigaku, Vol. 57, No. 3 (1997) 168.
- the modified microcrystalline cellulose excipient comprises pores having a median pore size of greater than or about 40 nm (e.g. from about 40 nm to about 60 nm), or particularly greater than or about 50 nm.
- the modified microcrystalline cellulose excipient has a surface area of from about 2.5 to about 50 m 2 /g, for example from about 5 to about 50 m 2 /g. In a further embodiment, the modified microcrystalline cellulose excipient has a surface area of from about 5 to about 40 m 2 /g, e.g. from about 5 to about 30 m 2 /g, such from about 5 to about 20 m 2 /g. References herein to the surface area of the modified microcrystalline cellulose excipient are to the surface area of that excipient in the absence of any active pharmaceutical ingredient, i.e. prior to the introduction of an active pharmaceutical ingredient.
- Surface area values mentioned herein refer to values determined through measuring adsorption and desorption of a gas (typically nitrogen gas, at 77 K), and calculating the specific surface area according to the BET method during adsorption.
- gases typically nitrogen gas, at 77 K
- the modified microcrystalline cellulose excipients used in the compositions of the invention may also have total pore volumes that are particularly suited to the uses disclosed herein. Suitable pore volumes that may be mentioned in this respect include pore volumes of at least about 0.005 cm 3 /g (e.g.
- the modified microcrystalline cellulose excipient has a total pore volume of at least about 0.005 cm 3 /g. In a further embodiment, the modified microcrystalline cellulose excipient has a total pore volume of from about 0.005 to about 0.2 cm 3 /g, such as from about 0.01 to about 0.1 cm 3 /g, preferably from about 0.01 to about 0.05 cm 3 /g.
- the pore size distribution may be determined using any standard techniques known to those skilled in the art. One such suitable method includes calculation via the DFT method based on N2 adsorption data.
- the pore volume of the modified microcrystalline cellulose excipients disclosed herein may also vary in a characteristic manner with the size of said pores. Compared to ordinary MCC, which is essentially non-porous, the modified microcrystalline cellulose excipient disclosed here shows specific surface area and total pore volume values which are about an order of magnitude larger. Furthermore, the pore size distribution is broad and predominantly centred at around 50 nm. It has been unexpectedly found that an excipient having such larges pores is capable of being used to enhance the solubility of the BCS Class 2 and 4 drugs mentioned herein. PCC on the other hand typically has very much smaller pore sizes (with a median value in the region around 4 to 5 nm) and so there would be a greatly reduced contribution to the total pore volume from pores having a diameter in the region of 50 nm.
- the chemical structure of said cellulose may be provided either in the form in which it is naturally produced, or as a derivative of such a form.
- Derivatives that may be mentioned in this respect are produced by surface limited modification of cellulose without its dissolution.
- the chemical routes of modification may include any of the known surface limited reactions involving primary and secondary alcohols such as but not limited to TEMPO-mediated oxidation, acylation, esterification, etherification, epoxylation, sulfonation, phosphorylation, and halogenation, e.g. chlorination, bromination, or iodination.
- Such derivatives are typically only present at the 2 nd , 3 rd and 6 th carbon atoms of the D-glucose units.
- the cellulose when it is provided in the form of a derivative, it is a surface modified cellulose without dissolution.
- the cellulose has been modified primarily at only the exposed surfaces of the network structure rather than at every modifiable position on every D-glucose unit within the cellulose network.
- the large scale structure of the cellulose that is present prior to chemical modification can be preserved during and after chemical modification.
- the surface- and bulk-modified celluloses can be discerned by their degree of crystallinity, i.e. the degree of crystallinity of surface modified cellulose will be essentially unaltered following chemical modification, whereas that of bulk modified cellulose will be significantly reduced, compared to the unmodified cellulose.
- modified microcrystalline cellulose excipients that are useful in the compositions of the invention may be prepared by way of a variety of techniques, using standard equipment, such as by spray-drying a low surface area microcrystalline cellulose (i.e. MCC having a surface area of from about 0.5 to about 1 m 2 /g).
- MCC low surface area microcrystalline cellulose
- compositions of the invention typically involve spray- drying a dispersion comprising microcrystalline cellulose precursor (e.g. a low surface area microcrystalline cellulose) in a suitable solvent.
- a dispersion comprising microcrystalline cellulose precursor and a suitable solvent system.
- Solvent systems that may be used in this respect include aqueous solvents, e.g. mixtures of water with one or more additional water-miscible solvents (preferably one or more additional volatile water-miscible solvents).
- Particular solvent systems that are useful in the formation of modified microcrystalline cellulose excipients having the desired surface area include mixtures of water with one or more additional water-miscible solvents.
- Water-miscible solvents that may be used in this context are non-aqueous solvents with a relatively high volatility.
- Suitable solvents for use in this respect include water-miscible solvents having a vapour pressure of at least about 1 kPa, e.g. at least about 5 kPa, at 25°C.
- solvents include alcohols (such as methanol, ethanol and propanol (e.g. n- propanol and isopropanol)), ketones (such as acetone and methyl ethyl ketone), ethers (such as tetrahydrofuran), acetonitrile and mixtures thereof.
- solvent systems include solvent systems in which the one or more volatile water-miscible solvents are present in an amount ranging from about 10 to about 70% by volume of the solvent system. In one embodiment, the one or more volatile water-miscible solvents are present in an amount ranging from about 30 to about 60% by volume of the solvent system.
- the precursor of microcrystalline cellulose that may be used in such dispersions may be a particulate material, preferably one which has a D50 value (prior to the spray drying step) of up to 1 ⁇ , e.g. up to 500 nm.
- microcrystalline cellulose precursor that may be suitable for incorporation into a dispersion in order to form the modified microcrystalline cellulose excipient
- dispersible-grade microcrystalline celluloses typically comprise a mixture of microcrystalline cellulose and one or more hydrocolloids in which the microcrystalline cellulose and one or more hydrocolloids have been co-spray dried.
- the hydrocolloids used in dispersible-grade microcrystalline celluloses are typically carbohydrates. Examples of hydrocolloids that are suitable in this respect include carboxymethylcellulose (e.g. sodium carboxymethylcellulose), carrageenan, guar gum, sodium alginate (as well as mixtures thereof).
- the microcrystalline cellulose in the dispersion is a dispersible-grade microcrystalline cellulose comprising microcrystalline cellulose and a hydrocolloid selected from the group consisting of carboxymethylcellulose (e.g. sodium carboxymethylcellulose), carrageenan, guar gum, sodium alginate, and mixtures thereof.
- carboxymethylcellulose e.g. sodium carboxymethylcellulose
- carrageenan e.g. sodium carboxymethylcellulose
- guar gum e.g. sodium alginate
- sodium alginate e.g. sodium carboxymethylcellulose
- examples of commercially available dispersible grades of microcrystalline cellulose include Avicel RC or CL grades, Vivapur MCG, Neocel, and the like.
- a conventional low surface area microcrystalline cellulose is used as the starting material it can be re-hydrolysed with a strong mineral acid, such as hydrochloric acid, sulphuric acid, nitric acid or phosphoric acid prior to the step of forming a dispersion for spray-drying, as required by the second aspect of the invention.
- a strong mineral acid such as hydrochloric acid, sulphuric acid, nitric acid or phosphoric acid
- the process further comprises acid hydrolysis of the microcrystalline cellulose precursor prior to the formation of the dispersion (i.e.
- the product obtained by such an acid hydrolysis step may be further processed prior to its incorporation into the dispersion for the spray-drying step.
- Such further processing steps include neutralisation, sonication and/or high-shear homogenisation (e.g. using a fluidiser).
- Homogenisation is particularly useful as it enables the formation of a homogeneous dispersion, which in turn enables greater control of the relative porosity of the final modified microcrystalline cellulose excipient.
- dispersible-grade microcrystalline celluloses as discussed above, in the formation of the modified microcrystalline cellulose excipient may be particularly advantageous as it should not be necessary to subject such materials to an additional acid hydrolysis step prior to dispersion and generally require lower energy input for dispersion prior to spray-drying.
- Spray-drying may be performed by way of a variety of routine techniques, and using standard equipment, known to the skilled person.
- the spray-drying is typically performed in inert atmosphere, i.e. in the substantial absence of oxygen gas.
- the inlet temperature which is invariably higher than that of the outlet, can vary depending on the composition of the solvent mixture, the outlet temperature is set to that corresponding to the evaporation temperature of the solvent featured with the highest boiling point. For instance, if water is one of the components and has the highest boiling temperature among the solvents, the outlet temperature is set at about 95-105 °C.
- a suspension of the microcrystalline cellulose precursor in a solvent system (e.g. as defined above) is typically fed into a spray-dryer.
- the suspension is then released in the form of a spray at a suitable flow rate, e.g. from 0.1 to 1000 L/h, in a nitrogen atmosphere.
- a suitable flow rate e.g. from 0.1 to 1000 L/h
- the feed rate of the liquid is adjusted so that the outlet temperature range is maintained as discussed above, as known by someone skilled in the art.
- the compositions of the invention comprise a microcrystalline cellulose excipient having a surface area of from about 2 to about 60 m 2 /g, and such microcrystalline cellulose excipients may be obtained by the processed described herein. Such celluloses may be referred to as "modified microcrystalline cellulose excipients".
- modified microcrystalline cellulose excipients in the composition of the invention is obtainable by a process as defined in the second aspect of the invention.
- the ability of the modified microcrystalline cellulose excipients disclosed herein to form stable amorphous mixtures with an active pharmaceutical ingredient is an important property of these materials.
- the data in Example 6 show that test material exhibits excellent stability following storage for at least 2 months at room temperature and 75% relative humidity, as followed by X-ray diffraction analysis.
- compositions of the invention have good physical stability.
- Physical stability refers to stability to undesired solid-state transformations of the drug, the modified microcrystalline cellulose excipient, or both. Examples of solid-state transformations include collapse of the cellulose porous structure, amorphous-crystalline transformations, and the formation of polymorphs.
- a stable amorphous composition is one in which the drug in the composition remains present in a predominantly amorphous state, e.g. ⁇ 90% amorphous following long term storage (e.g. storage for at least 2 months under ambient conditions). Changes in amorphicity may be quantified using melting enthalpy data obtained in differential scanning calorimetry (DSC).
- DSC differential scanning calorimetry
- the WHO essential drugs list includes the following Type 2 and 4 BCS drug categories: antihelminthic, anticonvulsant, antiepileptic, antibacterial, antiulcerative, antihyperlipidemic, anticholelithogenic, antimanic, antimigraine, antiamebic, antiviral, diuretic, antihypertensive, antipsychotic, analgesic, antipyretic, anti-inflammatory, antihistaminic, gastroprokinetic, antidiabetic, antiasthmatic, antianginal, immunosuppressant, antiobesity, antiosteoporotic, antihyperlididemicm, antiandrogen, contraceptive, antidysmenoreic, antiparkinsonian, anticoagulant, anticancer, anti- cirrhosis (biliary), anxyolithic, sedative, antifungal.
- Active ingredients that may be employed in compositions of the invention preferably include Type 2 or 4 BCS active pharmaceutical ingredients, including among others non- steroidal anti-inflammatory drugs (NSAIDs) as normally described in pharmacological literature.
- NSAIDs are particularly suited to the compositions of the present invention due to the enhanced bioavailability, rapid release characteristics and reduced administered dose that can be achieved when used in these compositions.
- NSAIDs are useful in the treatment of dysmenorrhea and migraine, and in both cases rapid delivery of the drug aids in alleviating symptoms quickly.
- NSAIDs are also frequently suited for the compositions of the present invention given the presence of one or more aromatic rings or polycondensed cyclic structures (fully or partially saturated) and one or more hydrogen bond donors/acceptors in the drug molecule structures.
- the composition contains one or more additional active pharmaceutical ingredients (i.e. in addition to the active pharmaceutical ingredient that is required by the first aspect of the invention), thus providing a combination medication which is useful in treating any of the diseases mentioned herein.
- compositions of the invention which contain an NSAID (which is preferably present in a predominantly amorphous form) may further contain a non-NSAID active pharmaceutical ingredient, which latter ingredient may be present in any degree of crystallinity.
- non-NSAID active pharmaceutical ingredients which may be mentioned in this respect include non-NSAID analgesics, such as paracetamol and codeine, and stimulants, such as caffeine.
- One or more of said additional active pharmaceutical ingredients may be present in a predominantly amorphous form (i.e.
- each of said one or more additional active pharmaceutical ingredients includes a Type 2 or 4 BCS active ingredient.
- Amorphisation of each of said one or more additional active pharmaceutical ingredients may be achieved by way of any of the methods mentioned herein, including heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, and the like.
- Such compositions may enable rapid release of a plurality of active pharmaceutical ingredients thus facilitating accelerated treatment of a broader range of symptoms in the patient.
- one or more of said additional active pharmaceutical ingredients may be present in a predominantly crystalline form, provided that each of the active pharmaceutical ingredients that is present in a predominantly crystalline form is different from the active pharmaceutical ingredient that is required by the first aspect of the invention (which is preferably provided in a predominantly amorphous form).
- compositions of the invention in which the active pharmaceutical ingredient is an NSAID are described in the examples. However, the invention is not limited to such drug compounds.
- the compositions of the invention may contain any active pharmaceutical ingredient, and preferably contain at least one active pharmaceutical ingredient that is a Type 2 or 4 BCS active pharmaceutical ingredient, which has the molecular features discussed above.
- compositions of the invention include antibiotics, steroids and cholates.
- Steroids in particular corticosteroids, are mainly used to reduce inflammation and suppress the immune system. They are used to treat conditions such as asthma, allergic rhinitis and hay fever, urticarial (hives), atopic eczema, chronic obstructive pulmonary disease (COPD), painful and inflamed joints, muscles and tendons, lupus, inflammatory bowel disease (IBD) (including Crohn's disease and ulcerative colitis), giant cell arteritis, polymyalgia rheumatica, and multiple sclerosis (MS).
- IBD inflammatory bowel disease
- MS multiple sclerosis
- Progesterone a natural steroid sex hormone, and its analogues, such as estradiol (e.g. ethynyl estradiol), progestin or estrogen, are used in treatment of primary dysmenorrhea.
- Cholates in particular cholic acid, deoxycholic acic, ursodeoxycholic acid
- Ursodeoxycholic acid is used for treatment of primary biliary cirrhosis.
- NSAIDs include propionic acid derivatives (such as alminoprofenb, benoxaprofen, carprofen, dexibuprofen, dexketoprofen, fenbufen, fenoprofen, flunoxaprofen, flurbiprofen, ibuprofen, ibuproxam, indoprofen, ketoprofen, loxoprofen, miroprofen, naproxen, oxaprozin, pirprofen, suprofen, tarenflurbil, tepoxalin, tiaprofenic acid, vedaprofen, and naproxcinod), acetic acid derivatives (such as aceclofenac, acemetacin, alclofenac, amfenac, bendazac, bromfenac, bumadizone, bufexamac,
- propionic acid derivatives such as alminoprofenb, benox
- NSAIDS include anthranilic acid derivatives (so-called "fenamates”; such as azapropazone, etofenamate, flufenamic acid, flunixin, meclofenamic acid, mefenamic acid, morniflumate, niflumic acid, and tolfenamic acid).
- fuseamates such as azapropazone, etofenamate, flufenamic acid, flunixin, meclofenamic acid, mefenamic acid, morniflumate, niflumic acid, and tolfenamic acid.
- these active ingredients are capable of being incorporated into the modified microcrystalline cellulose excipient described herein (e.g. using the heat- assisted methods disclosed herein) in spite of the relatively high melting points and relatively low vapour pressures associated with these substances.
- the reference to heat-assisted methods above includes heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, and the
- compositions of the invention include antineoplastic agents.
- Antineoplastic agents are mainly used to treat or prevent cancer.
- Particularly preferred antineoplastic agents include those which contain at least one aromatic ring (preferably at least two aromatic rings), and at least one hydrogen bond donor or hydrogen bond acceptor.
- antineoplastic agents include protein-kinase inhibitor substances, for example Afatinib, Alectinib, Axitinib, Binimetinib, Bosutinib, Brigatinib, Cabozantinib, Cediranib, Ceritinib, Cobimetinib, Crizotinib, Dasatinib, Entrectinib, Erlotinib, Fostamatinib, Gefitinib, Ibrutinib, Imatinib, Lapatinib, Lenvatinib, Lestaurtinib, Masitinib, Momelotinib, Mubritinib, Neratinib, Nilotinib, Nintedanib, Olmutinib, Osimertinib, Pacritinib, Pazopanib, Ponatinib, Radotinib, Regorafenib, Rociletinib, Ruxoli
- protein kinase inhibitors are drug substances that inhibit the action of one or several kinases by phosphorylation of liable aminoacids, e.g. tyrosine, threonine, serine. Selective phosphorylation can be used to regulate biological hyperactivity of various mutant or overexpressed protein kinases in cancer.
- Protein kinase inhibitors which are considered to be BCS Type 2 substances, exhibit dramatic structure-activity relationships. Many non-specific kinase inhibitors have dyelike structures, i.e. flat, highly conjugated polyaromatic systems. (Knight and Shokat Features of selective kinase inhibitors, Chemistry & Biology, 2005; 12: 621-637.) Further, selective kinase inhibitors almost always contain heterocycles. They are also typically entropically constrained, with four or fewer freely rotatable bonds connecting any two-ring systems. These compounds tend to be hydrophobic, bind to proteins non- specifically, and aggregate at high concentrations, which suggests high propensity to ⁇ - ⁇ interactions.
- Table 2 summarises some of the typical solid-state properties for selected substances.
- NSAIDS include arylpropionic acid derivatives (e.g. ibuprofen, ketoprofen, flurbiprofen, and naproxen) anthranilic acid derivatives (e.g. flufenamic acid, mefenamic acid, and tolfenamic acid), acetic acid derivatives (e.g. indomethacin, and sulindac), and enolic acid derivatives (e.g. pyroxicam).
- arylpropionic acid derivatives e.g. ibuprofen, ketoprofen, flurbiprofen, and naproxen
- anthranilic acid derivatives e.g. flufenamic acid, mefenamic acid, and tolfenamic acid
- acetic acid derivatives e.g. indomethacin, and sulindac
- enolic acid derivatives e.g. pyroxicam
- active pharmaceutical ingredient in which the active pharmaceutical ingredient is a steroid or a cholate, particularly preferred active pharmaceutical ingredients include a contraceptive such as progesterone, estradiol (e.g. ethynyl estradiol), progestin, estrogen, cholic acid, deoxycholic acic or ursodeoxycholic acid.
- a contraceptive such as progesterone, estradiol (e.g. ethynyl estradiol), progestin, estrogen, cholic acid, deoxycholic acic or ursodeoxycholic acid.
- Active pharmaceutical ingredients that are particularly useful in the compositions of the invention include compounds, which contain at least one aromatic ring or a polycondensed (fully or partially saturated) cyclic structure, optionally wherein the molecule also contains at least one hydrogen bond donor or hydrogen bond acceptor.
- Hydrogen bond acceptors include oxygen and nitrogen atoms.
- Other active pharmaceutical ingredients that are particularly useful in the compositions of the invention include compounds which contain at least one aromatic ring or a polycondensed cyclic structure, and at least one hydrogen bond donor or hydrogen bond acceptor.
- compositions containing flufenamic acid in the Examples will similarly be observed for compositions of the invention containing other drug molecules which have at least one aromatic ring or a polycondensed (fully or partially saturated) cyclic structure, particularly where the drug molecule also contains at least one hydrogen bond donor or hydrogen bond acceptor.
- compositions of the invention containing at least one active pharmaceutical ingredient (e.g.
- an NSAID having such structural features (i.e. at least one (e.g. at least two) aromatic ring, and at least one hydrogen bond donor or hydrogen bond acceptor).
- Particular NSAIDs that may be mentioned in this respect include arylpropionic acid derivatives (e.g. ibuprofen, ketoprofen, flurbiprofen, naproxen), anthranilic acid derivatives (e.g. flufenamic acid, mefenamic acid), acetic acid derivatives (e.g. indomethacin, sulindac), and enolic acid derivatives e.g. pyroxicam.
- arylpropionic acid derivatives e.g. ibuprofen, ketoprofen, flurbiprofen, naproxen
- anthranilic acid derivatives e.g. flufenamic acid, mefenamic acid
- acetic acid derivatives e.g. indomethacin, sulindac
- NSAIDs having at least two aromatic rings include arylpropionic acid derivatives (e.g. ketoprofen, flurbiprofen, naproxen); anthranilic acid derivatives (e.g. flufenamic acid, mefenamic acid), acetic acid derivatives (e.g. indomethacin, sulindac), and enolic acid derivatives (e.g. pyroxicam).
- arylpropionic acid derivatives e.g. ketoprofen, flurbiprofen, naproxen
- anthranilic acid derivatives e.g. flufenamic acid, mefenamic acid
- acetic acid derivatives e.g. indomethacin, sulindac
- enolic acid derivatives e.g. pyroxicam
- BCS Type 2 and 4 APIs that may be mentioned in this respect include compounds which contain at least two aromatic rings, and at least one hydrogen bond donor or hydrogen bond acceptor.
- a further embodiment relates to compositions of the invention containing at least one active pharmaceutical ingredient (e.g. an NSAID or an antineoplastic agent) having at least two aromatic rings, and at least one hydrogen bond donor or hydrogen bond acceptor.
- active pharmaceutical ingredient e.g. an NSAID or an antineoplastic agent
- these active ingredients are capable of being incorporated into the modified microcrystalline cellulose excipient described herein (e.g. using the heat-assisted methods disclosed herein) in spite of the relatively high melting points and relatively low vapour pressures associated with these substances.
- the reference to heat-assisted methods above includes heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, and the like.
- compositions of the invention containing at least one active pharmaceutical ingredient (e.g. an NSAID or an antineoplastic agent) having at least two aromatic rings and at least one hydrogen bond donor or hydrogen bond acceptor
- said active pharmaceutical ingredient may have a molecular weight of at least 200 g/mol, preferably at least 210 g/mol.
- the molecular weight may be at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290 or at least 300 g/mol.
- Active pharmaceutical ingredients having a melting point (or glass transition temperature) not greater than the temperature at which cellulose degrades are also preferred as this ensures that incorporation of the active pharmaceutical ingredient into the cellulose excipient can be more easily achieved.
- the active pharmaceutical ingredient will have a melting point (or glass transition temperature) of up to about 350°C, preferably up to about 300°C.
- compositions of the present invention are particularly suited for use in the compositions of the present invention.
- Such compositions may stably contain the active pharmaceutical ingredient in an amorphous form such that the composition is capable of rapidly releasing the active pharmaceutical ingredient in vivo. This in turn provides a significant clinical benefit to the patient, particularly where relief from painful discomfort is desired.
- Other classes of compounds that may be particularly suited to be formulated with the modified microcrystalline cellulose excipients described herein include those which contain a free carboxylic acid group (typically together with the one (or preferably two) aromatic ring), or an amide.
- the compositions of the present invention are particularly suited to the delivery of active pharmaceutical ingredients that are poorly soluble.
- compositions containing NSAIDs are not limited to the compositions containing NSAIDs.
- the advantages may be realised for any drug which has a relatively low solubility, i.e. Type 2 and 4 BCS drugs, and which is typically administered orally to patients and has desirable structural features which enable ⁇ - ⁇ and ⁇ - ⁇ interactions.
- the compositions of the invention may be administered by other routes, in particular transmucosally (such as via sublingual or buccal administration), or via rectal or vaginal administration.
- Active ingredients may further be employed in salt form or any other suitable form, such as e.g. a complex, solvate or prodrug thereof, or, if relevant, in any stereoisomeric form including any enantiomeric, diastereomeric or racemic form, or a combination of any of the above.
- Pharmaceutically-acceptable salts of active ingredients include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of an active ingredient with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g.
- Salts may also be prepared by exchanging a counter-ion of active ingredient in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
- suitable ion exchange resin examples include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric and sulphuric acids; from organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, arylsulphonic acids; and from metals such as sodium, magnesium, or preferably, potassium and calcium.
- the active pharmaceutical ingredient is present in a predominantly amorphous form.
- at least 80% of the active pharmaceutical ingredient in the composition is in an amorphous form.
- the active pharmaceutical ingredient may be essentially amorphous (i.e. at least about 90% amorphous, that is at least about 90% by weight of the active pharmaceutical ingredient is present in an amorphous form).
- the active pharmaceutical ingredient is at least about 95% amorphous, at least about 98% amorphous or most preferably at least about 99% amorphous.
- the degree of amorphicity in the active pharmaceutical ingredient may be determined by e.g. DSC.
- the XRD profile for a product which is in a predominantly amorphous form would be one in which sharp peaks associated with the crystalline product (i.e. the drug or its polymorphs in the compositions of the invention) are substantially absent.
- the degree of crystallinity of the active pharmaceutical ingredient may be found to be less than about 10%, when determined using DSC.
- Compositions in which the active pharmaceutical ingredient is present in a predominantly amorphous form may be qualitatively identified via their XRD profiles by virtue of the absence of sharp characteristic diffraction peaks typical for crystalline substances. Infra-red absorption spectroscopy (e.g.
- FT-IR can also show shifts in absorption frequencies indicating molecular rearrangement and interactions between API and excipient.
- shifts of other characteristic peaks for H-bond donors/acceptor groups may also be indicative of molecular rearrangement.
- the active pharmaceutical ingredient may be provided in any form, including a form that is crystalline or substantially crystalline.
- the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient described herein in the composition of the invention is at most about 1 :3.
- the weight ratio may be at most about 1 :5.
- the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient in the composition is not more than about 1 :9.
- the pharmaceutical composition comprises the active pharmaceutical ingredient at an amount of between 5 and 30% by weight of the composition.
- compositions of the invention include those in which the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient in the composition is not more than about 1 :3 (e.g. not more than about 1 :5), and the surface area of the modified microcrystalline cellulose excipient (prior to the introduction of the active pharmaceutical ingredient) is from about 5 to about 50 m 2 /g (e.g. from about 10 to about 40 m 2 /g) as measured by N2 gas adsorption technique according to the Brunauer Emmett Teller (BET) method.
- Further preferred compositions of the invention include those in which the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient in the composition is not more than about 1 :3 (e.g.
- compositions of the invention include those in which the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient in the composition is not more than 1 :5 (e.g. not more than 1 :9), and the active pharmaceutical ingredient is at least about 95% amorphous.
- compositions of the invention include those in which the modified microcrystalline cellulose excipient has a degree of crystallinity of from about 70% to about 90%, the surface area of the modified microcrystalline cellulose excipient (prior to the introduction of the active pharmaceutical ingredient) is from about 5 to about 50 m 2 /g (e.g. from about 10 to about 40 m 2 /g), and the active pharmaceutical ingredient is at least about 90% amorphous.
- compositions of the invention may be prepared by way of a variety of routine techniques, and using standard equipment, known to the skilled person, including mixing together the active pharmaceutical ingredient and a modified microcrystalline cellulose excipient having the desired surface area.
- the active pharmaceutical ingredient will be initially provided in a crystalline form.
- Amorphisation of the active pharmaceutical ingredient may be achieved through any technique which is capable of reducing the level of crystallinity of the active pharmaceutical ingredient in the composition without substantially degrading the cellulose component or adversely altering its physical structure. Suitable techniques include heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, intensive mixing, reduced pressure mixing, moderate heating, combined grinding and heating, and co-spray drying with a solvent. Heat-assisted intensive mixing, for example melt extrusion, is particularly useful. During the heat-assisted mixing the components are heated while being mixed, while the heating temperature may be at or below the melting point of the active ingredient. An extruder equipped with heated compartments, e.g.
- the process involves the processing of a mixture containing the active pharmaceutical ingredient (optionally initially in a substantially crystalline form) and the modified microcrystalline cellulose excipient having the desired surface area.
- a classical melt extrusion process is a process during which at least one component in the blend is melted (or at least heated to close to its melting point or above its glass transition temperature) and the mixture is formed into products of different shapes and sizes by forcing the components and active substances through an orifice or die under controlled temperature, pressure, feeding rate, and screw speed.
- Suitable melt extrusion techniques that will be known to the skilled person include those described in Shah S., et ai, Int. J. Pharm., 453 (2013) 233.
- melt extrusion techniques are typically used with thermoplastic polymers, i.e. polymers that melt before they degrade.
- native cellulose such as MCC
- melt extruders and other heat-assisted intensive mixers are particularly useful in the context of the present invention.
- these techniques can also be used with the non-thermoplastic cellulose-based systems of the present invention, e.g. using single- or twin-screw extruders, preferably with a heating unit as long as the temperature inside the mixture does not exceed that at which the cellulose degrades by pyrolysis.
- the temperature inside the mixture should not exceed about 350°C, and preferably should not exceed about 300°C.
- compositions of the invention without extrusion by statically heating blends of pre-mixed active ingredient and the modified microcrystalline cellulose excipient described herein.
- the mixture is then heated to a temperature above the glass transition temperature of the active ingredient for a sufficient period of time but below the degradation temperature of the cellulose such that the active pharmaceutical ingredient is substantially present in an amorphous form.
- the blends are heated in the range between 50 and 300 °C.
- compositions of the invention may also be prepared by static heat sealing.
- mixtures of drug and cellulose are placed in a sealed vessel.
- the mixture is then heated to a temperature above the glass transition temperature of the active pharmaceutical ingredient for a sufficient period of time (the exact period to be determined empirically depending on the quantities of the used ingredients, e.g. between 5 min and 72 hours) such that the active pharmaceutical ingredient is substantially present in an amorphous form.
- a further aspect of the invention relates to a pharmaceutical composition
- a pharmaceutical composition comprising a modified microcrystalline cellulose excipient as defined herein and an active pharmaceutical ingredient, wherein the composition is obtained by a process involving heating a mixture of the active pharmaceutical ingredient and the modified microcrystalline cellulose excipient to a temperature close to or above the glass transition temperature of the active pharmaceutical ingredient.
- the mixture is heated to a temperature above about 50 °C.
- the temperature should not exceed that at which degradation of the cellulose component would occur.
- the temperature used should not exceed about 300 °C.
- Heat-assisted extrusion is a particularly preferred technique for use in the context of the present invention.
- the invention relates to a composition which is formed by a process involving heat-assisted extrusion of a mixture of the cellulose or cellulose derivative and the active pharmaceutical ingredient.
- the mixing time period is likely to vary according to the equipment used, and the skilled person will have no difficulty in determining by routine experimentation a suitable mixing time for a given combination of ingredient(s) empirically.
- Microcrystalline cellulose excipients which have a surface area in the region of from 2 to 60 m 2 /g (as measured by N2 gas adsorption technique according to the Brunauer Emmett Teller (BET) method) are particularly advantageous as these substances provide a sufficiently high area of interaction with the active pharmaceutical ingredient while also providing a formulation that has good stability (i.e. in which the active pharmaceutical ingredient is maintained in an amorphous form for longer periods). Strong interactions between the active pharmaceutical ingredient and the cellulose component also help to improve the dissolution characteristics of the overall composition. The strength of the interactions is governed, at least in part, by the affinity between API and cellulose and further amplified over a large surface area. Strong interactions between cellulose and the active pharmaceutical ingredient can be observed in many ways, including FTIR spectroscopy or sometimes through colour changes associated with the active pharmaceutical ingredient itself, as well as through analysing the solid state fluorescence spectra of the mixture.
- BET Brunauer Emmett Teller
- a preferred process for the formation of compositions of the invention involves the mixing together of an active pharmaceutical ingredient and a modified microcrystalline cellulose excipient having a surface area of from about 2 to about 60 m 2 /g.
- the modified microcrystalline cellulose excipient is obtained by any of the methods disclosed herein.
- the product obtained by the above-mentioned process may further be adapted by:
- heat-assisted extrusion e.g. melt-extrusion
- compositions of the invention may further comprise one or more further commonly- employed pharmaceutical excipients.
- Suitable excipients include inactive substances that are typically used as a carrier for the active ingredients in medications. Suitable excipients also include those that are employed in the pharmaceutical arts to bulk up pharmaceutical compositions that employ very potent active ingredients, to allow for convenient and accurate dosing. Alternatively, excipients may also be employed in manufacturing processes of the compositions of the invention to aid in the handling of the active ingredient concerned.
- additives apart from diluent may be employed such as taste masking agents, glidants, superdisintegrants, coating agents, etc.
- compositions of the invention are preferably administered orally to the gastrointestinal tract and may provide for rapid release of the active pharmaceutical ingredient in the stomach and/or, preferably, the intestinal system.
- the compositions of the invention may be incorporated into various kinds of pharmaceutical preparations intended for oral administration using standard techniques (see, for example, Lachman et al, "The Theory and Practice of Industrial Pharmacy', Lea & Febiger, 3 rd edition (1986) and “Remington: The Science and Practice of Pharmacy” , Gennaro (ed.), Philadelphia College of Pharmacy & Sciences, 19 th edition (1995)), for example to form a capsule, a powder or a tablet.
- compositions of the invention may also be administered to the patient through other routes, such as via transmucosal (e.g. sublingual or buccal), rectal or vaginal administration.
- transmucosal e.g. sublingual or buccal
- rectal or vaginal administration An appropriate route of administration is one which allows for rapid uptake of the active pharmaceutical ingredient into the bloodstream following administration.
- Sublingual, buccal, rectal and vaginal routes of administration are suitable in this respect as they allow the active pharmaceutical ingredient to rapidly enter into the bloodstream, thereby leading to a fast onset of action.
- compositions of the invention contain a pharmacologically effective amount of the active ingredient.
- pharmacologically effective amount we refer to an amount of active ingredient, which is capable of conferring a desired therapeutic effect on a treated patient, whether administered alone or in combination with another active ingredient. Such an effect may be objective (i.e. measurable by some test or marker) or subjective (i.e. the subject gives an indication of, or feels, an effect).
- compositions of the invention may be adapted (for example as described herein) to provide a sufficient dose of drug over the dosing interval (irrespective of the number of doses per unit time) to produce a desired therapeutic effect.
- compositions of the invention may thus be determined by the physician, or the skilled person, in relation to what will be most suitable for an individual patient. This is likely to vary with the route of administration, the type and severity of the condition that is to be treated, as well as the age, weight, sex, renal function, hepatic function and response of the particular patient to be treated.
- Suitable dosages of active ingredient in one oral delivery unit may be below 1 g, preferably below 100 mg and above 2 mg. Similar doses may also be appropriate for delivery by other routes, particularly via sublingual, buccal, rectal and vaginal administration.
- NSAIDs are typically used as analgesics, anti-inflammatory drugs and antipyretics, they can also be useful for treatment of primary dysmenorrhea and migraine.
- Dysmenorrhea is the pain associated with menstruation in women. It affects approximately 25% of women and in younger women (67-90%) it occurs without underlying problem. (Livshitz and Seidman (2010) Pharmaceuticals, 3, 2082-2089). It has been found that the over-production of uterine PGs is key factor to the painful cramps that are the major symptom of dysmenorrhea. NSAIDs decrease the menstrual pain by decreasing the intrauterine pressure and lowering PGF2a levels in menstrual fluid. (Dawood, M.Y. (1988) Am. J. Med., 20, 23-29).
- NSAIDs can be helpful for planning and timing in-vitro fertilization (IVF) treatments and in attenuating the progression of labour during pregnancy (Livshitz and Seidman (2010) Pharmaceuticals, 3, 2082-2089).
- Enhancement of NSAIDs bioavailability may implicate important clinical effects due to two reasons: reduced side effects and rapid onset of action.
- NSAIDs Due to the low solubility/bioavailability NSAIDs are typically administered in high doses to achieve a therapeutic effect. The latter is partly the reason for the side effects associated with NSAIDs, such as gastric ulcers. Thus, lowering the dose when the bioavailability is enhanced may reduce toxic effects.
- the relief in dysmenorrhea patients is associated with the rapid onset of action. It is the rapidity with which the NSAIDs are absorbed that determines how quickly the relief is obtained. Since the onset of menstrual flow is variable, it is practical to initiate the medication at the beginning of menstruation and to continue it for 3 days if necessary. With rapidly absorbed NSAIDs, pre-treatment before the onset of dysmenorrhea is unnecessary. This is of great importance since most of the women experiencing dysmenorrhea are young sexually active women.
- the fenamates and arylpropionic acid derivatives (profens) are the drugs of choice for treating dysmenorrhea.
- menstrual fluid PGs act by suppression of menstrual fluid PGs and by a direct analgesic effect. Reduction of menstrual fluid is the result of direct inhibition of PG biosynthesis and release in endometrial tissue - a phenomenon that occurs during the first 48 hours of menstruation.
- Arylpropionic acid derivatives such as ibuprofen, flurbiprofen, ketoprofen and naproxen
- fenamates such as mefenamic acid and flufenamic acid
- arylpropionic acid derivatives are preferred as they give rise to fewer side-effects, while fenamates are known to have stronger action (Marjoribanks J, et ai , 2009. Nonsteroidal anti-inflammatory drugs for primary Dysmenorrhea, Cochrane report, Wiley).
- the stronger mechanism of fenamate action is believed to be dual, i.e. inhibition of PG synthesis and antagonism to PG in certain tissues.
- There is a strong correlation between the solubility, dissolution rate and bioavailability of fenamates (Shinkuma, et ai, 1984, Int. J. Pharm., 21 , 187-200). Varying bioavailability due to poor solubility was the reason why some fenamates have been removed from the market, e.g. flufenamic acid.
- Another effective alternative for treatment of primary dysmenorrhea is administration of contraceptives, such as progesterone.
- contraceptives such as progesterone
- the pain associated with dysmenorrhea is postulated to result from progesterone withdrawal before the onset of menses, which causes an increase in PGs. Therefore, administration of progesterone can alleviate the symptoms of primary dysmenorrhea.
- migraine With a prevalence of 8% in males and 12-15% in females migraine is extremely common (Diener, H.C., et al. 2008, 22 (Suppl. 1), 51-58.). It is characterized by recurrent attacks of pulsatile, unilateral headache often accompanied by nausea and vomiting, photo- and phonophobia.
- compositions of the invention comprising NSAIDs are therefore useful in the treatment of migraine (e.g. acute migraine headache) and/or dysmenorrhea (e.g. primary dysmenorrhea).
- migraine e.g. acute migraine headache
- dysmenorrhea e.g. primary dysmenorrhea
- a method of treatment of migraine which method comprises administration of a composition of the invention to a person suffering from, or susceptible to, such a condition.
- dysmenorrhea e.g. primary dysmenorrhea
- Particular active pharmaceutical ingredients that are useful in such methods are non-steroidal anti-inflammatory drugs.
- an active pharmaceutical ingredient e.g. an NSAID
- migraine e.g. acute migraine headache
- an active pharmaceutical ingredient e.g. an NSAID or a contraceptive
- dysmenorrhea e.g. primary dysmenorrhea
- a pharmaceutical composition of the invention in the manufacture of a medicament for treating dysmenorrhea or migraine, wherein the active pharmaceutical ingredient is a non-steroidal anti-inflammatory drug or a contraceptive.
- treatment we include the therapeutic treatment, as well as the symptomatic treatment, the prophylaxis, or the diagnosis, of the condition.
- compositions of the invention comprise NSAIDs
- appropriate pharmacologically effective amounts of such compounds include those that are capable of producing (e.g. immediate) relief of pain or other symptoms when administered perorally.
- the amount of the NSAID active ingredient may be expressed as the amount in a unit dosage form. In such a case, the amount of NSAID active ingredient that may be present may be sufficient to provide a dose per unit dosage form that is in the range of between about 2 mg and about 1000 mg (e.g. about 200mg or 400 mg).
- the above-mentioned dosages are exemplary of the average case; there can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
- compositions of the invention possess the advantage of reducing the risks of producing side effects that are typically associated with high doses of drugs.
- the compositions may contain a lower overall quantity of the active pharmaceutical ingredient while still providing a rapid therapeutic benefit for the patient.
- the use of a lower overall quantity of the active pharmaceutical ingredient helps to reduce the occurrence of unwanted side effects that may occur.
- NSAIDs in particular may benefit from being used in the compositions of the invention as frequent usage of these drugs is associated with a substantially increased risk of gastrointestinal problems (e.g. gastrointestinal bleeding and ulcers) and kidney problems).
- Compositions of the invention may also have the advantage that they may be prepared using established pharmaceutical processing methods and may employ materials that are approved for use in foods or pharmaceuticals or of like regulatory status.
- compositions of the invention may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile than, and/or have other useful pharmacological, physical, or chemical properties over, pharmaceutical compositions known in the prior art, whether for use in the treatment of pain or otherwise.
- Figures 1A to 1 D show SEM micrographs showing MCC (Figs 1A. and 1 B) and P-MCC (Figs 1C and 1 D) powders at high magnification (Figs. 1A and 1 C) and low magnification (Figs 1 B and 1 D). Information of specific magnification and scaling is provided in each micrograph.
- Figure 2 shows an AMF height sensor image of P-MCC suspension adsorbed on gold plated surface.
- Figure 3 shows a pore size distribution curve derived from N2 adsorption and desorption isotherms at 77 K of MCC and P-MCC celluloses.
- Figure 4 shows FTIR spectra of (i) pure FFA, (ii) a MCC-FFA mixture stored at standard conditions ("MCC-FFA-N"), and (iii) a heated MCC-FFA mixture (120 °C, 2 h) ("MCC-FFA-H"). The data is normalized with respect to the C-H stretching vibration at 2897 cm “1 . Region presented in Fig 4A: 600 - 1000 cm "1 , and Fig 4B: 1400- 1800 cm “1 .
- Figure 5 shows FTIR spectra of (i) pure FFA, (ii) a MCC-FFA mixture stored at standard conditions ("P-MCC-FFA-N"), and (iii) a heated MCC-FFA mixture (120 °C, 2 h) ("P-MCC-FFA-H").
- the data is normalized with respect to the C-H stretching vibration at 2897 cm “1 .
- Fig 5B 1400 - 1800 cm "1 .
- Figure 6 shows a representative DSC thermogram of MCC-FFA mixtures and pure FFA. The heat flow of the mixtures is normalised with respect to the total weight of sample.
- Figure 7 shows a representative DSC thermogram of P-MCC-FFA mixtures heated to 120 °C for 2 hours and pure FFA. The heat flow of the mixtures is normalised with respect to the total weight of sample.
- Figure 8A shows an X-ray powder diffractogram for pure FFA and physical mixture of MCC with FFA.
- Figure 8B shows an X-ray powder diffractogram for pure FFA and heated mixture of MCC with FFA.
- Figure 9A shows an X-ray powder diffractogram for pure FFA and physical mixture of P-MCC with FFA.
- Figure 9B shows an X-ray powder diffractogram for pure FFA and heated mixture of P-MCC with FFA.
- Figure 10A shows dissolution profiles of FFA from MCC into SIF together with dissolution profile for pure FFA at 37.0 ⁇ 0.5 °C. Each data point is the average of two experiments.
- Figure 10B shows dissolution profiles of FFA from P-MCC into SIF together with dissolution profile for pure FFA at 37.0 ⁇ 0.5 °C. Each data point is the average of two experiments.
- Figure 11 shows X-ray powder diffractograms for (a) as-prepared normal mixture of MCC with FFA; (b) as-prepared heated mixture of MCC with FFA; and (c) heated mixture of MCC with FFA stored at RH 75% for 61 days.
- Figure 12 shows X-ray powder diffractograms for (a) as-prepared normal mixture of MCC with FFA; (b) as-prepared heated mixture of MCC with FFA; and (c) heated mixture of MCC with FFA stored at RH 75% for 61 days. Examples
- Flufenamic Acid [N-(a,a,a-trifluoro-m-tolyl) anthranilic acid]
- FFA Flufenamic Acid
- NSAID non-steroidal anti-inflammatory drug
- MCC Microcrystalline Cellulose
- MCC was the original cellulose used in the preparation of P-MCC (modified microcrystalline cellulose) as is detailed in Example 1.
- the AFM images were acquired using a Dimension Icon (Bruker, Germany) instrument.
- a gold surface was coated by submerging it in a 10 mM solution of 6-amino-1- hexanethiol hydrochloride for 2 hours and then dried by gently blowing compressed air over the surface.
- a drop (100 ⁇ _) of highly diluted dispersion containing cellulose crystallites (0.01 % wt) was placed on the precoated gold plate surface (5 mm in diameter) and dried.
- the gold plate was then mounted on the AFM instrument using a double adhesive tape.
- the height images were acquired in the peak force tapping mode, using manufacturer's Scan Asyst cantilever and ScanAsyst optimization algorithm.
- Nitrogen gas (N2 at 77 K) adsorption and desorption isotherms were obtained using an ASAP 2020 instrument (Micromeritics, USA). The gas absorption was conducted on pure, solid powders of P-MCC (>200 mg) and MCC (2 g). The specific surface area was calculated according to the BET method during N2 adsorption, while the pore size distribution was determined based on N2 adsorption data using DFT method. The total pore volume of the powders was obtained as the volume of adsorbed nitrogen at a relative pressure (p/pO) approximating unity. All calculations were determined using the ASAP 2020 instrument software. The samples were degassed at 100 °C under vacuum for at least 6 h prior to analysis. Filler rods were used for each sample during N2 sorption measurement.
- FTIR analysis was conducted on FFA, as a pure substrate, and FFA in blend with different celluloses.
- Cellulose-FFA blends from both heated and normal, i.e. unheated, samples were analysed.
- the FTIR spectra were obtained on a Bruker Tensor 27 (Germany) with KBr pellets. A background scan on air was subtracted from all spectra using the instrument software (Opus 7.0, Bruker, Germany).
- the approximate sample content in 200 mg KBr pellets was 10 wt% (i.e. 1 wt% drug).
- the collected data was normalized with respect to C-H stretching vibration at 2897 cm -1 .
- the FTIR spectra were collected with the following parameters: 64 scans at a spectrum resolution of 4 cm -1 over a range from 4000 to 400 cm -1 .
- Thermal analysis was performed with Model Q-2000 TA instrument (USA) on both cellulose-drug mixtures and pure substances. Samples were placed inside hermetically sealed aluminium crucibles with punctured lids, in order to avoid overpressure caused by water evaporation. An empty pan was used as a reference. The analysis was conducted in the temperature range from -40°C to 150°C with a heating rate of 10°C min -1 . N2 gas, at a flow of 50 ml_ min -1 , was applied during analysis. Initially the samples were cooled from room temperature to -40°C, then heated to 150°C and finally cooled to 25°C again. All of the samples were stored at ambient conditions for 24 hours prior to DSC measurements.
- the heating conditions were 120 °C for 2 hours.
- the measurements were performed in triplicate, and the estimated amount of drug in the mixtures was 10 wt%. Enthalpies and temperatures were estimated by Universal Analysis 2000 Software (TA Instruments).
- the characteristic X-ray diffraction patterns were generated using a D8 Twin-Twin, Bruker (Germany) instrument with Bragg- Brentano geometry for both FFA, as a pure drug, and FFA in a blend with different celluloses.
- Pure FFA (5 mg) was used as the reference and 50 mg of both normal and heated samples of cellulose-FFA blends (approx. 10 wt% FFA in each sample) were scanned once during the analysis.
- MCC a modified microcrystalline cellulose excipient
- the aqueous suspension of MCC was sonicated for 20 minutes with a high intensity ultrasound processor (Sonication Pulse Rate: 30 seconds On, 30 seconds Off) at 70% amplitude to yield a thick white gel which was then homogenized using a high-pressure fluidizer.
- Two sets of chambers with different sizes connected in series were used with internal diameters of 400/200 ⁇ and 200/100 ⁇ , respectively.
- the suspension was passed once through the larger chamber and six times through the small chamber at 1600-1700 bar.
- the dry matter content of 1.96 wt% for the dispersion was calculated gravi metrically by drying the sample to a constant weight at 150 °C.
- the sample was dispersed further with additional 150 mL deionized water until a 0.96 wt% dry matter content was achieved.
- the sample was sonicated one more time for 20 minutes (Sonication Pulse Rate: 30 seconds On, 30 seconds Off) at 70% amplitude.
- the produced dispersion was diluted with ethanol (95%) until a concentration of 50 vol% ethanol was achieved.
- the obtained dispersion was then spray-dried in nitrogen atmosphere at inlet temperature 195 ⁇ 5 °C, outlet temperature 103 ⁇ 3 °C, pump flow rate setting 15%, and spray flow rate 500 L/h (Buchner mini spray-dryer B-290).
- the collected product was stored at room temperature and 0% relative humidity in a desiccator until used.
- Example 2 Characterisation of microcrystalline cellulose excipient
- SEM images of the celluloses of Example 1 show roughly spherical particles of modified microcrystalline cellulose unlike rod-like particles of MCC.
- Figure 2 shows the cellulose crystallites, which were released upon hydrolytic cleavage in hydrochloric acid and high-shear homogenisation. The homogenization resulted in relatively well-dispersed individual or slightly aggregated cellulose fragments (whiskers). The length of the whiskers normally did not exceed 500 nm. The cellulose whiskers (in the form of a suspension) were spray-dried to produce the final P-MCC product, as shown in Figure 1. Nitrogen Gas Sorption
- Cellulose-FFA samples for characterization and release study were prepared the day before analysis with an approximate storage time of 24 hours at ambient conditions.
- a 50 mg blend of cellulose and drug was prepared by mixing 5 mg FFA with 45 mg of selected cellulose powder in 1 ml_ glass vials. The vials were sealed with plastic screw caps and vortexed for 30 seconds.
- Each cellulose-FFA blend was analysed in both heated and unheated (normal) form. Vials containing the cellulose-FFA blends were placed in a preheated oil bath at 120 °C for 2 hours in the heat-treatment procedure.
- Figures 4 and 5 display spectra collected on FFA in formulation with different celluloses in both normal and heated mixtures together with a reference band for pure crystalline FFA.
- Several distinctive functional groups of FFA are detectible by FTIR spectroscopy (Jabeen S., et ai, Spectrochim. Acta. Part A Mol. Biomol. Spectrosc. vol. 96, 972-985, 2012) most of which are normally absent in the chemical structure of cellulose.
- the FTIR spectra for normal blend of P-MCC-FFA ( Figure 5) display an apparent difference in band characteristics for both selected regions, i.e. the characteristic bands appeared to shift to higher wavenumbers, become broader and generally decrease in intensity. This is observed particularly for bands corresponding to CF3 vibration at 659 and 652 cm -1 , aromatic out-of-plane C-H deformations at 889 cm -1 and in-plane aromatic C-H deformation at 1422 cm -1 .
- the characteristic FFA bands in normal mixture with MCC demonstrated no particular deviation from relevant band positions.
- the FTIR spectra for heated samples revealed substantial deviations from the characteristic band positions of FFA in reference spectra.
- the shift in the wavenumber position at this band is particularly indicative of interaction involving carboxylic group of FFA and cellulose.
- the observed shift in the band position at 1655 cm -1 in the heated mixture of MCC-FFA was reduced when compared to FFA in formulation with P-MCC. While the positions of some peaks for heated P-MCC-FFA and MCC-FFA shifted, other peaks such as 760, 1519 and 1578 cm -1 were diffuse and of remarkably low intensity, which could be due to interference from water in these samples.
- the results from FTIR analysis suggest that a potential interaction is present between FFA and the different celluloses, particularly for FFA in formulation with P-MCC in the heated samples.
- FIGS 6 and 7 display representative thermograms from differential scanning calorimetry (DSC) analysis conducted on samples of pure FFA and physical mixtures of FFA with P-MCC and MCC.
- the profiles for physical mixtures are normalized with respect to the total weight of the sample (i.e. 10 wt% FFA).
- the DSC profile for pure FFA showed a distinct endotherm at 135°C corresponding to the melting temperature of crystalline FFA.
- the exothermic event at about 72 °C in the back-scan DSC profile of pure FFA is representative of recrystallization of FFA from the melt upon cooling.
- DSC profiles for FFA in mixtures with celluloses were generally characterized by two events: evaporation of water from cellulose in the region between 40 and 110 °C and a distinct endothermic peak at temperature around 133-139 °C, which corresponds to the melting of crystalline FFA.
- the exothermic event on the back-scan for physical mixtures, corresponding to recrystallization of FFA, could be distinguished only in the normal samples of P-MCC mixed with FFA and.
- the recrystallization exotherm in the physical mixture of P-MCC-FFA is slightly shifted to higher temperatures accompanied with significant decrease in size compared with recrystallization of pure FFA.
- DSC profiles for heated mixtures of FFA with celluloses were characterised by the lack of endothermic peaks in the region corresponding to melting of crystalline FFA.
- DSC profile of MCC in mixture with FFA displayed several distinguishable peaks close to the melting region of FFA, typically below the specific melting temperature.
- the absence of the melting peak of FFA (at 135 °C) in the DSC profiles is indicative of an amorphous structure of FFA in the heated samples, since fully amorphous materials do not exhibit a melting endotherm.
- the broad halo at 40 - 110 °C, characteristic of water evaporation is present in the DSC profiles for heated samples as well. This indicates that water is resorbed in the samples during storage after heat treatment to 120 °C prior to analysis.
- Tables 7 and 8 present enthalpies with corresponding temperatures for water evaporation, melting and recrystallization of FFA in mixtures together with values of standard deviation for each result.
- Table 7 - Melting enthalpies of FFA in pure form and in mixtures with different celluloses. Results are presented as averages with standard deviation in parentheses (n 3).
- FIG. 8 and 9 displays X-ray diffraction patterns for pure FFA and physical mixtures of FFA in both heated and normal samples with celluloses.
- the diffractogram of FFA is displayed in each graph as a reference.
- Several well-defined peaks of high intensity appear in the diffraction pattern of pure FFA. The peaks at 14°, 19° and 21 ° are the most prominent.
- the diffraction pattern for pure MCC is expected to display a diffused peak at 13-16°, a narrow peak at 22-23° and a peak with low intensity at 35° (Mihranyan A., et al., Int. J. Pharm., vol. 269, no. 2, pp. 433-442, 2004). Consequently, the sharp diffraction peaks over-imposed in the profile for normal mixture of MCC-FFA are clearly derived from the FFA content in the mixture. Moreover, it is observed that the peaks associated with FFA in the normal mixture with MCC have shifted from their original positions when compared with the fingerprint peaks for pure FFA. This may indicate substantial molecular rearrangement and formation of FFA polymorphs upon processing.
- the intensity of crystalline FFA peaks in the diffractograms for heated mixtures is generally reduced compared to the normal mixture of MCC-FFA, and only residual crystalline peaks of low intensity are visible in the diffractogram, in line with the DSC analysis on the MCC-FFA mixture.
- Figure 9 peaks with equivalent positions and appearance as the ones representative of the cellulosic content in MCC-FFA mixtures appeared and were therefore attributed to the cellulosic content of the P-MCC sample.
- the diffractogram for the normal mixture of P- MCC-FFA showed analogous results as the mixture of MCC with FFA, where the normal mixture displayed peaks corresponding to cellulosic content along with the characteristic peaks for crystalline FFA that continued to appear in the diffractograms. However, the peaks, corresponding to crystalline FFA, appear with a slightly different shift in the peak positions and decrease in the relative intensity. In view of the DSC results for normal mixtures of P-MCC-FFA, in which only 48 % of FFA was found to be crystalline, it is plausible that partial amorphisation is responsible for the overall decrease in relative intensity, while the shift in peak positions is likely to be related to the presence of polymorphism in FFA.
- the X-ray diffraction patterns for the heated sample of P-MCC-FFA revealed characteristic peaks for cellulose, identical to the peaks in normal mixture. However, the heated mixture of P-MCC-FFA showed no peaks indicating on absence of crystalline FFA. Since amorphous materials do not diffract X-rays in a coherent manner, it is regarded that FFA is present explicitly in its amorphous state in the heated mixture of P- MCC-FFA. Consequently, the results of X-ray diffraction confirmed the previous DSC results suggesting that the drug is present in its crystalline form in the normal mixtures and transformed to fully amorphous state during heat treatment.
- Dissolution measurements were made on normal and heated physical mixtures of FFA with either MCC or P-MCC to evaluate the release kinetics of FFA.
- a reference measurement was made using pure FFA.
- Dissolution kinetics of FFA in formulation with cellulose were determined by the standardized USP paddle method. Release profiles for the drug were created by spectrofluorimetric analysis on samples at various time points.
- FFA flufenamic acid
- SIF simulated intestinal fluid
- Various amounts of stock solution between 0.1 mL and 1 mL were transferred to plastic vials and frozen at -27 °C.
- the vials containing stock solution were freeze-dried overnight using a Scanvac CoolSafe 55-4 (LaboGene ApS, Lynge, Denmark).
- Seven working standard solutions with FFA concentration range between 0.1 and 10 ⁇ g/mL were prepared by dissolving the contents of the vials containing freeze-dried stock solution with 1 mL of a polar solvent, Acetonitrile-DMSO (4: 1 , vol/vol).
- Dissolution measurements were performed with the rotating paddle technique at 37.0 ⁇ 0.5 °C and 50 rpm with SOT AX (AT7 Smart, Switzerland) dissolution apparatus.
- SIF was selected as the dissolution medium and prepared by diluting 20 mL of concentrated SIF with 480 mL deionized water. Normal and heated cellulose mixtures were poured into dissolution vessels with 500 mL dissolution medium and samples of 1 ml were extracted at various time points (15 min, 30 min, 1 h, 2 h, 3h, 4h and 5h). A total of seven samples with a volume of 1 mL were collected for each cellulose-FFA formulation and passed through a syringe filter into 2 ml plastic vials.
- the vials were frozen at -27 °C and freeze-dried further until all of the water had sublimated from the sample and the dried FFA filtrate remained.
- the vials containing dried FFA were filled with 1 ml_ Acetonitrile-DMSO (4: 1 , vol/vol) solvent and manually shaken until the collected FFA was dissolved. Two parallel measurements were performed for each formulation. Spectrofluorometric analysis together with previously described regression analysis was used to estimate the concentration of released FFA at different time points.
- the dissolution profile of FFA in formulation with MCC shows a pattern similar to the one of FFA alone. Generally, the release behaviour of FFA from both celluloses in normal formulations was similar to that of FFA alone. The normal FFA-cellulose mixtures and the sample with pure FFA released over 50 % of the drug after 2 hours and between 82-86 % was released after 5 hours of experiment.
- the dissolution profiles of FFA in the heated formulation with P-MCC ( Figure 10B) displayed a considerable acceleration of the dissolution rate of the drug. An initial burst release was apparent and a rapid increase in the concentration of released drug from the heated mixture with P-MCC was observed during the first 50 minutes of the experiment.
- the concentration of released drug was observed to be three times as high for the heated mixture with P-MCC compared to FFA after the initial 15 minutes where the amount of FFA released from heated P-MCC-FFA mixture was 51 %, compared to 15% for pure crystalline FFA.
- the cumulative concentration of FFA released from P-MCC after 2 h was 95 % whereas FFA alone did not reach that value within the experimental time frame and displayed a maximum of 87 % after 5 h. It should also be noted that the heated mixture of P-MCC-FFA is the only sample that released the entire amount of preloaded FFA within 5 hours.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Polymers & Plastics (AREA)
- Organic Chemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Inorganic Chemistry (AREA)
- Medicinal Preparation (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
There is provided a pharmaceutical composition comprising a modified microcrystalline cellulose excipient and an active pharmaceutical ingredient (e.g. from the Type 2 or 4 BCS class), wherein the modified microcrystalline cellulose excipient has a relatively high5 surface area. Compositions of the invention find particularly utility as formulations comprising BCS Type 2 and 4 drugs, including NSAIDs or other drugs, that may be employed in the treatment of migraine and dysmenorrhea, as well as formulations comprising other poorly soluble active ingredients where rapid release in vivo is advantageous.
Description
New Compositions
Field of the Invention This invention relates to new cellulose-based pharmaceutical compositions that provide for enhanced solubility of active ingredients, such as Type 2 and 4 BCS drugs, featured with low solubility in the gastrointestinal tract. The invention also relates to methods of manufacturing such pharmaceutical compositions. Background
The drug needs to dissolve first before it can be absorbed. According to Biopharmaceutics Classification System (BCS) all drugs are divided in 4 general groups based on their solubility and permeability (Amidon, G.L. et al. 1995. Pharm. Res. 12(3):413-420).
According to FDA BCS guidelines, the following boundaries are currently valid:
• A drug substance is considered HIGHLY SOLUBLE when the highest dose strength is soluble in < 250 ml water over a pH range of 1 to 7.5.
• A drug substance is considered HIGHLY PERMEABLE when the extent of absorption in humans is determined to be > 90% of an administered dose, based on mass-balance or in comparison to an intravenous reference dose.
• A drug product is considered to be RAPIDLY DISSOLVING when > 85% of the labeled amount of drug substance dissolves within 30 minutes using USP apparatus I or II in a volume of < 900 ml buffer solutions.
Further, a dose number Do may also be useful as a solubility indicator:
M
D = o.
V 0 xS app
(1)
wherein Mo is the maximum dose, Vo is the volume available for dissolution, normally 250 ml, and Sapp is the apparent solubility of the drug in the medium. Thus, Do<1 in a range of pH between 1 and 7.5 indicates good solubility, whereas if Do>1 at any pH the solubility of a drug is poor.
The present invention pertains to enhancing the solubility and bioavailability of Type 2 and 4 BCS drugs. Various APIs which are classified as Type 2 or 4 BCS drugs are disclosed in Takagi et al. Mol. Pharm. 2006; 3(6): 631-643; Lindenberg et al., Eur. J. Pharm. Biopharm. 2004; 58: 265-278; and Pham T. et al., Mol. Pharm. 2013; 10: 2445-2461. These references mainly deal with APIs on the WHO essential drugs list. However, the invention is useful also for developmental substances since it is known that at least more than half and as much as 90% of drug candidates are poorly soluble substances. In the context of the drug substances useful in the present invention, the molecular descriptors for typical BCS Type 2 and 4 substances can be used as summarized by Pham T. et al. Mol. Pharm. 2013; 10: 2445-2461 , provided that they contain at least one aromatic ring or polycondensed cyclic structure.
Table 1 - Typical molecular descriptors of various classes BCS drugs. Table adapted from Pham T. et al. Mol. Pharm. 2013; 10: 2445-2461.
Type 1 BCS Type 2 BCS Type 3 BCS Type 4 BCS logD pH6 0.71/0.65 1.81/1.9 -0.88/- 1.11 1.37/1.28 mean/median
logD pH7.5 -1.72 to 5.18 -8.24 to 5.07 -7.69 to 5.18 -8.11 to 6.6 range
logD pH7.5 1.34/1.18 1.63/1.67 -0.56/-0.73 1.40/1.00 mean/median
nHA+B range 1 -15 1-40 2-27 1 -28 nHA+B 4.97/4 6.24/6 8.81/8 10.81/10 mean/median
RBN, range 0-13 0-19 0-16 1 -20
RBN, mean 4.12/4 4.34/4 5.07/4 7.56/7 /median
RBN, rotating bonds number; nHA+B total number of hydrogen bond acceptors and donors; Papp apparent permeability; logD pH dependent distribution coefficient in octanol-water; logP partition coefficient in octanol-water; Mw molecular weight Most of NSAIDs are normally Type 2 or 4 BCS substances with low solubility although some of them, such as salicylic acid derivatives e.g. ASA, salicylic acid, ethenzamide, etc., or acetaminophen, are classed as Type 1 (or Type 3) substances. NSAIDs are a practical model of poorly soluble drugs because they are affordable and have relatively low toxicity as compared to other pharmacological classes of drugs and therefore are less hazardous for research personnel and environment.
NSAIDs mimic arachidonic acid (AA) to be able to inhibit COX, i.e. (i) they contain a centre of acidity or a free carboxylic group and (ii) are largely lipophilic and feature at least one aromatic ring. Normally, NSAIDs have a centre of acidity, typically represented by a carboxylic acid group or alternatively by enolic group, hydroxamic acid, sulfonamide, or a tetrazole ring. The acidic group of NSAIDs is important for H-bonding with the polar region at Arg120 and oppositely located Tyr355 of COX (Hawkey 1999. Lancet 353:307- 314). All NSAIDs possess at least one aromatic ring, which renders NSAIDs lipophilic, which is essential for inhibiting COX enzymes and ensures unhindered passive diffusion across biological membranes. The overall lipophilic character of NSAIDs results in poor solubility-limited bioavailability, i.e. Type 2 and 4 BCS drugs.
Another important parameter is rate of absorption. Because most NSAIDs have poor solubility, their rate of absorption is limited and this results in poor bioavailability. To enhance the bioavailability and achieve a therapeutic effect, NSAIDs are typically administered in high doses and the peak plasma concentration is reached usually in 2 hours.
The administration of high doses of NSAIDs is partly the reason for the side effects, such as peptic ulcers and ultimately bleeding or perforation of the stomach, associated with these drugs. Some 10% to 50% of patients are unable to tolerate treatment with NSAIDs because of side effects, including abdominal pain, diarrhoea, and upset stomach. There is therefore a need to provide formulations containing NSAIDs, which are able to produce a therapeutic effect as rapidly as possible while minimising the risk of side effects for the patient. Oral route of drug administration is the most frequently used and convenient route for administration of many drugs. Microcrystalline cellulose is an important tableting excipient for oral administration. European Pharmacopoiea defines microcrystalline cellulose (MCC) as purified, partially depolymerized cellulose, prepared by treating a- cellulose, obtained as a pulp from fibrous plant material, with mineral acids. Although the primary aim of treating a-cellulose with mineral acid to a so-called level-off degree of polymerization (DP 100-350) is to remove amorphous regions of cellulose, the overall degree of crystallinity of MCC does not exceed 80% as measured by XRD (Mihranyan et al. Int. J. Pharm. 2004; 269 (2), 433-442). Normally MCC is an essentially non-porous powder featuring a surface area around 0.5-1 m2/g as measured by nitrogen gas adsorption (ibid). MCC is normally produced by spray-drying of the cellulose slurry that is obtained following the acidic hydrolysis.
Oguchi T. et al., Yakuzaigaku, Vol. 57, No. 3 (1997) 168 discusses the use of a porous form of microcrystalline cellulose, referred to as "porous crystalline cellulose" PCC, as a drug carrier in a composition with naproxen. However, the manner in which the drug carrier is prepared is not disclosed. Here and throughout the text the term PCC refers solely to an undisclosed material supplied by Asahi Kasei, Japan. In the disclosed document, PCC is a material featured with pores, which are predominantly 4 nm (40 A) in size.
Matsumoto K. et al. , Chem. Pharm. Bull. 46(2) 314-318 (1998) and Matsumoto K. et ai, Int. J. Pharm. 1994: 108: 167 discloses the use of PCC in mixtures with ethenzamide, i.e.
a type 1 (3) BCS substance. No details regarding the preparation of the excipient are provided and PCC is used having pores, which are predominantly below 10 nm and centred around 4 nm in size. The material described therein displays poor stability in the presence of moisture as it progressively loses its specific surface area when exposed to various relative humidities.
Tozuka Y., et al. Bull. Chem. Soc. Jpn., 73, 1567-1572 (2000) discloses a composition of PCC with 2-naphthoic acid, a non-pharmaceutical ingredient. However, very little information concerning the origin of the PCC is provided.
Nakai Y. et al., Chem. Pharm. Bull. 26(10) 2983-2989 (1978) discloses compositions of MCC with naphthalene (non-pharmaceutical ingredient), camphor and o-cresol, non- pharmaceutical ingredient. Nakai Y., Drug Dev. Ind. Pharm. 1986; 12(7): 1017-1039. discloses formulations containing ground MCC mixtures having enhanced solubility. It is clear from Nakai (1986) that the drugs are not stable in the formulations disclosed therein. Furthermore, it is evident from the XRD data that the structure of the MCC gradually changes resulting in the MCC becoming progressively more amorphous over time. The literature also discusses the effect of pore size on amorphization inside the pores of various adsorbent materials. Oguchi T. et al., Yakuzaigaku, Vol. 57, No. 3 (1997) 168; Matsumoto et al. Int. J. Pharm. 1994: 108: 167; Yonemochi and Yamamoto in Encyclopedia of Surface and Colloid Science, Ed. P. Somasundaran, 2nd ed., 2006, vol. 6, pp. 5068-5077 discuss in great detail the importance of pore size in controlled pore glasses on amorphisation behaviour in porous matrices. In particular, amorphisation behaviour is strongest when the size of the pores is less than 30 nm, and more preferably less than 10 nm. It is believed that in sufficiently small pores the reorientation and crystallisation is effectively hindered. Further, Yonemochi et al. Chem Pharm Bull 1991 ; 39: 1027 report higher chemical instability of aspirin was associated mainly with pores below 30 nm because in these pores aspirin was essentially amorphous, while above 30 nm more of the aspirin was crystalline and therefore more stable.
The listing or discussion of an apparently prior-published document in this specification should not necessarily be taken as an acknowledgement that the document is part of the state of the art or is common general knowledge.
Disclosure of the Invention
According to a first aspect of the invention, there is provided a pharmaceutical composition comprising a modified microcrystalline cellulose excipient and an active pharmaceutical ingredient, wherein the modified microcrystalline cellulose excipient has a surface area of from about 2 to about 60 m2/g. Compositions comprising such features are hereinafter referred to together as "the compositions of the invention".
The inventor has advantageously found that compositions of the invention provide for rapid release of active pharmaceutical ingredients, particularly Type 2 and 4 BCS active ingredients, and also provide for an enhancement of their bioavailability in the gastrointestinal tract (GIT) following peroral administration of the composition. In one embodiment, substantially all (e.g. at least about 85%) of the drug is released from the formulation in the first hour following administration of the formulation. The release may also be such that amount of drug released from the composition reaches a plateau within a short time (e.g. within about 1 hour, such as within about 15 minutes). That is, substantially all (e.g. at least about 90%) of the total amount of drug that is released during the first 24 hours may be released during the first two hours (e.g. during the first 60 minutes). In the present application, the terms "rapid release" and "immediate release" are used interchangeably. The compositions of the invention are not limited to oral administration, and can be delivered by other routes, as is discussed elsewhere herein.
The term "rapid release" will be understood by the skilled person to refer to compositions that provide for, and/or are adapted to provide for, a "quick" and/or an "immediate" release of drug (in which drug is released at a sufficiently high rate for a sufficient period of time to produce a therapeutic response in a very short time) from the formulation. Release from the formulation refers to release of the drug from the network structure of the modified microcrystalline cellulose excipient.
The compositions of the invention are also advantageous as they exhibit an increased physical stability of the active pharmaceutical ingredient (e.g. an increased stability of its physical form) within the formulation. In the compositions of the invention, the active pharmaceutical ingredient, which is typically a Type 2 or 4 BCS substance, is preferably present in a predominantly amorphous form. The compositions allow the active pharmaceutical ingredient to remain in such a predominantly amorphous form for an extended period of time (e.g. up to at least 1 month, such as up to at least six months).
Upon release from the cellulose carrier, the active pharmaceutical ingredient is provided in a form having a high degree of amorphicity compared to other formulations. This in turn enhances the rate of dissolution of the active pharmaceutical ingredient in surrounding media, and accelerates uptake by the body.
As is mentioned elsewhere herein, the compositions of the invention may contain a plurality of active pharmaceutical ingredients, provided that at least one of said active pharmaceutical ingredients, which is typically a Type 2 or 4 BCS substance, is present in a predominantly amorphous form.
Enhanced dissolution is particularly important for Type 2 and 4 BCS active ingredients as these substances have a low solubility profile. By enhancing the dissolution rate of the active pharmaceutical ingredient following administration, the overall dose of drug that is provided to the subject may be reduced while still achieving the intended therapeutic benefits.
Microcrystalline cellulose (MCC) is a well-defined and thoroughly studied natural polymer that is profoundly important in the pharmaceutical industry, especially in oral tablet formulations. MCC is particularly useful as a pharmaceutical tableting aid. A complete description of MCC is provided in European Pharmacopoeia.
Microcrystalline cellulose is produced by mineral acid hydrolysis of refined cellulose to a level-off degree of polymerisation (DP 100-350). Under controlled conditions, the acid induces hydrolytic cleavage of glycosidic bonds in the amorphous regions of cellulose in the fibres leaving crystalline regions intact. The free crystallites flocculate and form compact structures of the cellulose fibre aggregates in the dispersion. The produced slurry is then washed and spray-dried to finally produce dry solid powder. The surface area of such conventional MCC is typically about 0.5-1 m2/g, measured by N2 gas adsorption.
The compositions of the invention comprise a modified microcrystalline cellulose excipient, which is obtainable from microcrystalline cellulose. The modified microcrystalline cellulose excipient has a surface area of from about 2 to about 60 m2/g which is significantly higher than that associated with conventional microcrystalline cellulose (which typically has a surface area in the region of 0.5-1 m2/g, measured by N2 gas adsorption), and significantly lower than that associated with "porous crystalline cellulose", PCC (which typically has a surface area in excess of 90 m2/g).
It is surprisingly observed that the modified microcrystalline cellulose excipient disclosed herein (also referred to herein as P-MCC) can inhibit recrystallization of amorphous drug even if it features pores predominantly of about 50 nm in size (diameter), while previously it was suggested that pores smaller than 30 nm, and more preferably smaller than 10 nm, are needed to observe such effect. For instance, in PCC the active pores are predominantly in the region around 4 nm in size, see Oguchi et al. Yakuzaigaku, Vol. 57, No. 3 (1997) 168. Thus in one embodiment, the modified microcrystalline cellulose excipient comprises pores having a median pore size of greater than or about 40 nm (e.g. from about 40 nm to about 60 nm), or particularly greater than or about 50 nm.
It is believed that the surface area is also an important feature in this respect. In one embodiment, the modified microcrystalline cellulose excipient has a surface area of from about 2.5 to about 50 m2/g, for example from about 5 to about 50 m2/g. In a further embodiment, the modified microcrystalline cellulose excipient has a surface area of from about 5 to about 40 m2/g, e.g. from about 5 to about 30 m2/g, such from about 5 to about 20 m2/g. References herein to the surface area of the modified microcrystalline cellulose excipient are to the surface area of that excipient in the absence of any active pharmaceutical ingredient, i.e. prior to the introduction of an active pharmaceutical ingredient. When an active pharmaceutical ingredient is brought into association with a modified microcrystalline cellulose excipient as defined herein and subjected to processing conditions that are suitable for allowing these two ingredients to become thoroughly mixed, a portion the active pharmaceutical ingredient is expected to become incorporated into the pores in the modified microcrystalline cellulose excipient with the result that the apparent surface area of the modified microcrystalline cellulose excipient will be measurably lower than the surface area prior to introduction of the API.
Surface area values mentioned herein refer to values determined through measuring adsorption and desorption of a gas (typically nitrogen gas, at 77 K), and calculating the specific surface area according to the BET method during adsorption. One skilled in the art will attest that different gases for measuring the surface area may yield different values. Further, different non-gas adsorption methods may also yield different values due to the different accessibility of the measuring analyte to the pores. The modified microcrystalline cellulose excipients used in the compositions of the invention may also have total pore volumes that are particularly suited to the uses disclosed herein. Suitable pore volumes that may be mentioned in this respect include
pore volumes of at least about 0.005 cm3/g (e.g. ranging from about 0.005 to about 0.2 cm3/g). The pore volumes are obtained from condensation of gas at relative pressures approaching 1. Other methods may yield different volumes. Thus, in one embodiment, the modified microcrystalline cellulose excipient has a total pore volume of at least about 0.005 cm3/g. In a further embodiment, the modified microcrystalline cellulose excipient has a total pore volume of from about 0.005 to about 0.2 cm3/g, such as from about 0.01 to about 0.1 cm3/g, preferably from about 0.01 to about 0.05 cm3/g. The pore size distribution may be determined using any standard techniques known to those skilled in the art. One such suitable method includes calculation via the DFT method based on N2 adsorption data.
The pore volume of the modified microcrystalline cellulose excipients disclosed herein may also vary in a characteristic manner with the size of said pores. Compared to ordinary MCC, which is essentially non-porous, the modified microcrystalline cellulose excipient disclosed here shows specific surface area and total pore volume values which are about an order of magnitude larger. Furthermore, the pore size distribution is broad and predominantly centred at around 50 nm. It has been unexpectedly found that an excipient having such larges pores is capable of being used to enhance the solubility of the BCS Class 2 and 4 drugs mentioned herein. PCC on the other hand typically has very much smaller pore sizes (with a median value in the region around 4 to 5 nm) and so there would be a greatly reduced contribution to the total pore volume from pores having a diameter in the region of 50 nm.
The chemical structure of said cellulose may be provided either in the form in which it is naturally produced, or as a derivative of such a form. Derivatives that may be mentioned in this respect are produced by surface limited modification of cellulose without its dissolution. The chemical routes of modification may include any of the known surface limited reactions involving primary and secondary alcohols such as but not limited to TEMPO-mediated oxidation, acylation, esterification, etherification, epoxylation, sulfonation, phosphorylation, and halogenation, e.g. chlorination, bromination, or iodination. Such derivatives are typically only present at the 2nd, 3rd and 6th carbon atoms of the D-glucose units. It is also preferred that, when the cellulose is provided in the form of a derivative, it is a surface modified cellulose without dissolution. By this we mean that the cellulose has been modified primarily at only the exposed surfaces of the network structure rather than at every modifiable position on every D-glucose unit within the cellulose network. By limiting the modifications to only the surface regions, the large scale structure of the cellulose that is present prior to chemical modification can be
preserved during and after chemical modification. For the avoidance of doubt, the surface- and bulk-modified celluloses can be discerned by their degree of crystallinity, i.e. the degree of crystallinity of surface modified cellulose will be essentially unaltered following chemical modification, whereas that of bulk modified cellulose will be significantly reduced, compared to the unmodified cellulose.
The modified microcrystalline cellulose excipients that are useful in the compositions of the invention may be prepared by way of a variety of techniques, using standard equipment, such as by spray-drying a low surface area microcrystalline cellulose (i.e. MCC having a surface area of from about 0.5 to about 1 m2/g).
Processes for the formation of compositions of the invention typically involve spray- drying a dispersion comprising microcrystalline cellulose precursor (e.g. a low surface area microcrystalline cellulose) in a suitable solvent. Such a process requires the formation of a dispersion comprising microcrystalline cellulose precursor and a suitable solvent system. Solvent systems that may be used in this respect include aqueous solvents, e.g. mixtures of water with one or more additional water-miscible solvents (preferably one or more additional volatile water-miscible solvents). Thus, according to a second aspect of the invention, there is provided a process for preparing a modified microcrystalline cellulose excipient as hereinbefore defined, which process comprises:
(i) forming a dispersion comprising a microcrystalline cellulose precursor and a solvent system, wherein the solvent system comprises a mixture of water and one or more volatile water-miscible solvents; and
(ii) spray-drying said dispersion.
Particular solvent systems that are useful in the formation of modified microcrystalline cellulose excipients having the desired surface area include mixtures of water with one or more additional water-miscible solvents. Water-miscible solvents that may be used in this context are non-aqueous solvents with a relatively high volatility. Without wishing to be bound by theory, it is believed that the evaporation of the water-miscible solvent during the spray drying process accelerates the removal of the water from the dispersion, which in turn reduces the extent to which the porous structure of the spray-dried microcrystalline cellulose excipient collapses during the drying process. This allows the relatively high porosity in the final modified microcrystalline cellulose excipient to be attained. Suitable solvents for use in this respect include water-miscible solvents having a vapour pressure of at least about 1 kPa, e.g. at least about 5 kPa, at 25°C. Examples of such solvents include alcohols (such as methanol, ethanol and propanol (e.g. n-
propanol and isopropanol)), ketones (such as acetone and methyl ethyl ketone), ethers (such as tetrahydrofuran), acetonitrile and mixtures thereof.
Further particular solvent systems that may be mentioned include solvent systems in which the one or more volatile water-miscible solvents are present in an amount ranging from about 10 to about 70% by volume of the solvent system. In one embodiment, the one or more volatile water-miscible solvents are present in an amount ranging from about 30 to about 60% by volume of the solvent system. The precursor of microcrystalline cellulose that may be used in such dispersions may be a particulate material, preferably one which has a D50 value (prior to the spray drying step) of up to 1 μηι, e.g. up to 500 nm.
Other forms of microcrystalline cellulose precursor that may be suitable for incorporation into a dispersion in order to form the modified microcrystalline cellulose excipient include so-called dispersible-grade microcrystalline celluloses. Dispersible-grade microcrystalline celluloses typically comprise a mixture of microcrystalline cellulose and one or more hydrocolloids in which the microcrystalline cellulose and one or more hydrocolloids have been co-spray dried. The hydrocolloids used in dispersible-grade microcrystalline celluloses are typically carbohydrates. Examples of hydrocolloids that are suitable in this respect include carboxymethylcellulose (e.g. sodium carboxymethylcellulose), carrageenan, guar gum, sodium alginate (as well as mixtures thereof). Thus, in one embodiment of the second aspect of the invention, the microcrystalline cellulose in the dispersion is a dispersible-grade microcrystalline cellulose comprising microcrystalline cellulose and a hydrocolloid selected from the group consisting of carboxymethylcellulose (e.g. sodium carboxymethylcellulose), carrageenan, guar gum, sodium alginate, and mixtures thereof. Examples of commercially available dispersible grades of microcrystalline cellulose include Avicel RC or CL grades, Vivapur MCG, Neocel, and the like.
If a conventional low surface area microcrystalline cellulose is used as the starting material it can be re-hydrolysed with a strong mineral acid, such as hydrochloric acid, sulphuric acid, nitric acid or phosphoric acid prior to the step of forming a dispersion for spray-drying, as required by the second aspect of the invention. The use of the phrase "re-hydrolysed with a strong mineral acid" in this context, refers to an acid hydrolysis step that is additional to any acid hydrolysis step that was conducted during the production of the microcrystalline cellulose from refined cellulose. Thus, in one embodiment of the
second aspect of the invention, the process further comprises acid hydrolysis of the microcrystalline cellulose precursor prior to the formation of the dispersion (i.e. the dispersion with a solvent system comprising a mixture of water and one or more water- miscible solvents). The product obtained by such an acid hydrolysis step may be further processed prior to its incorporation into the dispersion for the spray-drying step. Such further processing steps include neutralisation, sonication and/or high-shear homogenisation (e.g. using a fluidiser). Homogenisation is particularly useful as it enables the formation of a homogeneous dispersion, which in turn enables greater control of the relative porosity of the final modified microcrystalline cellulose excipient.
The use of dispersible-grade microcrystalline celluloses, as discussed above, in the formation of the modified microcrystalline cellulose excipient may be particularly advantageous as it should not be necessary to subject such materials to an additional acid hydrolysis step prior to dispersion and generally require lower energy input for dispersion prior to spray-drying.
Spray-drying may be performed by way of a variety of routine techniques, and using standard equipment, known to the skilled person. The spray-drying is typically performed in inert atmosphere, i.e. in the substantial absence of oxygen gas. While the inlet temperature, which is invariably higher than that of the outlet, can vary depending on the composition of the solvent mixture, the outlet temperature is set to that corresponding to the evaporation temperature of the solvent featured with the highest boiling point. For instance, if water is one of the components and has the highest boiling temperature among the solvents, the outlet temperature is set at about 95-105 °C. A suspension of the microcrystalline cellulose precursor in a solvent system (e.g. as defined above) is typically fed into a spray-dryer. The suspension is then released in the form of a spray at a suitable flow rate, e.g. from 0.1 to 1000 L/h, in a nitrogen atmosphere. The feed rate of the liquid is adjusted so that the outlet temperature range is maintained as discussed above, as known by someone skilled in the art.
As is stated above, the compositions of the invention comprise a microcrystalline cellulose excipient having a surface area of from about 2 to about 60 m2/g, and such microcrystalline cellulose excipients may be obtained by the processed described herein. Such celluloses may be referred to as "modified microcrystalline cellulose excipients". Thus, in one embodiment, the modified microcrystalline cellulose excipient in the composition of the invention is obtainable by a process as defined in the second aspect of the invention.
The ability of the modified microcrystalline cellulose excipients disclosed herein to form stable amorphous mixtures with an active pharmaceutical ingredient is an important property of these materials. The data in Example 6 show that test material exhibits excellent stability following storage for at least 2 months at room temperature and 75% relative humidity, as followed by X-ray diffraction analysis.
The compositions of the invention have good physical stability. Physical stability refers to stability to undesired solid-state transformations of the drug, the modified microcrystalline cellulose excipient, or both. Examples of solid-state transformations include collapse of the cellulose porous structure, amorphous-crystalline transformations, and the formation of polymorphs. A stable amorphous composition is one in which the drug in the composition remains present in a predominantly amorphous state, e.g.≥90% amorphous following long term storage (e.g. storage for at least 2 months under ambient conditions). Changes in amorphicity may be quantified using melting enthalpy data obtained in differential scanning calorimetry (DSC). Alternatively, this may be qualitatively verified by the absence of sharp peak in XRD which would otherwise be indicative of crystalline drug and/or its polymorph. The WHO essential drugs list includes the following Type 2 and 4 BCS drug categories: antihelminthic, anticonvulsant, antiepileptic, antibacterial, antiulcerative, antihyperlipidemic, anticholelithogenic, antimanic, antimigraine, antiamebic, antiviral, diuretic, antihypertensive, antipsychotic, analgesic, antipyretic, anti-inflammatory, antihistaminic, gastroprokinetic, antidiabetic, antiasthmatic, antianginal, immunosuppressant, antiobesity, antiosteoporotic, antihyperlididemicm, antiandrogen, contraceptive, antidysmenoreic, antiparkinsonian, anticoagulant, anticancer, anti- cirrhosis (biliary), anxyolithic, sedative, antifungal. The compositions of the invention are particularly suited for use with these drugs. In one embodiment, the composition of the invention contains an active pharmaceutical ingredient that is considered to have low solubility according to FDA BCS guidelines.
Active ingredients that may be employed in compositions of the invention preferably include Type 2 or 4 BCS active pharmaceutical ingredients, including among others non- steroidal anti-inflammatory drugs (NSAIDs) as normally described in pharmacological literature. NSAIDs are particularly suited to the compositions of the present invention due to the enhanced bioavailability, rapid release characteristics and reduced
administered dose that can be achieved when used in these compositions. Apart from their several pharmacological effects, NSAIDs are useful in the treatment of dysmenorrhea and migraine, and in both cases rapid delivery of the drug aids in alleviating symptoms quickly. NSAIDs are also frequently suited for the compositions of the present invention given the presence of one or more aromatic rings or polycondensed cyclic structures (fully or partially saturated) and one or more hydrogen bond donors/acceptors in the drug molecule structures.
In one embodiment of the invention, the composition contains one or more additional active pharmaceutical ingredients (i.e. in addition to the active pharmaceutical ingredient that is required by the first aspect of the invention), thus providing a combination medication which is useful in treating any of the diseases mentioned herein. For example, compositions of the invention which contain an NSAID (which is preferably present in a predominantly amorphous form) may further contain a non-NSAID active pharmaceutical ingredient, which latter ingredient may be present in any degree of crystallinity. Examples of non-NSAID active pharmaceutical ingredients which may be mentioned in this respect include non-NSAID analgesics, such as paracetamol and codeine, and stimulants, such as caffeine. One or more of said additional active pharmaceutical ingredients may be present in a predominantly amorphous form (i.e. similar to the active pharmaceutical ingredient required by the first aspect of the invention), particularly where said one or more additional active pharmaceutical ingredients includes a Type 2 or 4 BCS active ingredient. Amorphisation of each of said one or more additional active pharmaceutical ingredients may be achieved by way of any of the methods mentioned herein, including heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, and the like. Such compositions may enable rapid release of a plurality of active pharmaceutical ingredients thus facilitating accelerated treatment of a broader range of symptoms in the patient.
In a further such embodiment, one or more of said additional active pharmaceutical ingredients may be present in a predominantly crystalline form, provided that each of the active pharmaceutical ingredients that is present in a predominantly crystalline form is different from the active pharmaceutical ingredient that is required by the first aspect of the invention (which is preferably provided in a predominantly amorphous form).
To illustrate the inventive concept, compositions of the invention in which the active pharmaceutical ingredient is an NSAID are described in the examples. However, the invention is not limited to such drug compounds. For the avoidance of doubt, the compositions of the invention may contain any active pharmaceutical ingredient, and preferably contain at least one active pharmaceutical ingredient that is a Type 2 or 4 BCS active pharmaceutical ingredient, which has the molecular features discussed above.
Other particular active ingredients that may be employed in compositions of the invention include antibiotics, steroids and cholates. Steroids, in particular corticosteroids, are mainly used to reduce inflammation and suppress the immune system. They are used to treat conditions such as asthma, allergic rhinitis and hay fever, urticarial (hives), atopic eczema, chronic obstructive pulmonary disease (COPD), painful and inflamed joints, muscles and tendons, lupus, inflammatory bowel disease (IBD) (including Crohn's disease and ulcerative colitis), giant cell arteritis, polymyalgia rheumatica, and multiple sclerosis (MS). Progesterone, a natural steroid sex hormone, and its analogues, such as estradiol (e.g. ethynyl estradiol), progestin or estrogen, are used in treatment of primary dysmenorrhea. Cholates (in particular cholic acid, deoxycholic acic, ursodeoxycholic acid) may be used in the treatment of bile acid synthesis disorders due to single enzyme defects and peroxisomal disorders (such as Zellweger syndrome). Ursodeoxycholic acid is used for treatment of primary biliary cirrhosis.
In embodiments of the invention in which the active pharmaceutical ingredient may be an NSAID, particularly preferred NSAIDs include propionic acid derivatives (such as alminoprofenb, benoxaprofen, carprofen, dexibuprofen, dexketoprofen, fenbufen, fenoprofen, flunoxaprofen, flurbiprofen, ibuprofen, ibuproxam, indoprofen, ketoprofen, loxoprofen, miroprofen, naproxen, oxaprozin, pirprofen, suprofen, tarenflurbil, tepoxalin, tiaprofenic acid, vedaprofen, and naproxcinod), acetic acid derivatives (such as aceclofenac, acemetacin, alclofenac, amfenac, bendazac, bromfenac, bumadizone, bufexamac, diclofenac, difenpiramide, etodolac, felbinac, fentiazac, indomethacin, indomethacin farnesil, ketorolac, lonazolac, oxametacin, proglumetacin, sulindac, tolmetin, zomepirac, and nabumetone), oxicams (also referred to as "enolic acid derivatives"; such as ampiroxicam, droxicam, isoxicam , lornoxicam, meloxicam, piroxicam, tenoxicam, and phenylbutazone (bute)), anthranilic acid derivatives (so-called "fenamates"; such as azapropazone, etofenamate, flufenamic acid, flunixin, meclofenamic acid, mefenamic acid, morniflumate, niflumic acid, and tolfenamic acid), selective COX-2 inhibitors (such as apricoxib, celecoxib, cimicoxib, deracoxib, etoricoxib,
firocoxib, lumiracoxib, mavacoxib, parecoxib, robenacoxib, rofecoxib, and valdecoxib), pyrazolones and pyrazolidines (such as aminophenazone, ampyrone, azapropazone, clofezone, famprofazone, feprazone, kebuzone, metamizole, mofebutazone, morazone, nifenazone, oxyphenbutazone, phenazone, phenylbutazone, propyphenazone, sulfinpyrazone and suxibuzone), and sulfonamides (such as nimesulide).
Particularly preferred NSAIDS include anthranilic acid derivatives (so-called "fenamates"; such as azapropazone, etofenamate, flufenamic acid, flunixin, meclofenamic acid, mefenamic acid, morniflumate, niflumic acid, and tolfenamic acid). It has been surprisingly found that these active ingredients are capable of being incorporated into the modified microcrystalline cellulose excipient described herein (e.g. using the heat- assisted methods disclosed herein) in spite of the relatively high melting points and relatively low vapour pressures associated with these substances. The reference to heat-assisted methods above includes heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, and the like.
Still further active ingredients that may be employed in compositions of the invention include antineoplastic agents. Antineoplastic agents are mainly used to treat or prevent cancer. Particularly preferred antineoplastic agents include those which contain at least one aromatic ring (preferably at least two aromatic rings), and at least one hydrogen bond donor or hydrogen bond acceptor. Examples of suitable antineoplastic agents include protein-kinase inhibitor substances, for example Afatinib, Alectinib, Axitinib, Binimetinib, Bosutinib, Brigatinib, Cabozantinib, Cediranib, Ceritinib, Cobimetinib, Crizotinib, Dasatinib, Entrectinib, Erlotinib, Fostamatinib, Gefitinib, Ibrutinib, Imatinib, Lapatinib, Lenvatinib, Lestaurtinib, Masitinib, Momelotinib, Mubritinib, Neratinib, Nilotinib, Nintedanib, Olmutinib, Osimertinib, Pacritinib, Pazopanib, Ponatinib, Radotinib, Regorafenib, Rociletinib, Ruxolitinib, Selumetinib, Semaxanib, Sorafenib, Sunitinib, Tivozanib, Toceranib, Trametinib, Vandetanib and Vemurafenib. Pharmacologically, protein kinase inhibitors are drug substances that inhibit the action of one or several kinases by phosphorylation of liable aminoacids, e.g. tyrosine, threonine, serine. Selective phosphorylation can be used to regulate biological hyperactivity of various mutant or overexpressed protein kinases in cancer.
Protein kinase inhibitors, which are considered to be BCS Type 2 substances, exhibit dramatic structure-activity relationships. Many non-specific kinase inhibitors have dyelike structures, i.e. flat, highly conjugated polyaromatic systems. (Knight and Shokat Features of selective kinase inhibitors, Chemistry & Biology, 2005; 12: 621-637.)
Further, selective kinase inhibitors almost always contain heterocycles. They are also typically entropically constrained, with four or fewer freely rotatable bonds connecting any two-ring systems. These compounds tend to be hydrophobic, bind to proteins non- specifically, and aggregate at high concentrations, which suggests high propensity to ττ- π interactions.
Table 2 summarises some of the typical solid-state properties for selected substances.
Table 2 - solid-state properties
Substance Mw Tm, C pK logP solubility, mg/ml dasatinib 488 280 8.5 2.8 0.0128 sorafenib 464 205 11.5 4.1 0.0017 nilotinib 529 230 11.9 4.5 0.0020 erlotinib 393 230 16.1 3.1 0.0089 imatinib 493 226 12.4 3.5 0.0146
Particularly preferred NSAIDS include arylpropionic acid derivatives (e.g. ibuprofen, ketoprofen, flurbiprofen, and naproxen) anthranilic acid derivatives (e.g. flufenamic acid, mefenamic acid, and tolfenamic acid), acetic acid derivatives (e.g. indomethacin, and sulindac), and enolic acid derivatives (e.g. pyroxicam).
In embodiments of the invention in which the active pharmaceutical ingredient is a steroid or a cholate, particularly preferred active pharmaceutical ingredients include a contraceptive such as progesterone, estradiol (e.g. ethynyl estradiol), progestin, estrogen, cholic acid, deoxycholic acic or ursodeoxycholic acid.
Active pharmaceutical ingredients that are particularly useful in the compositions of the invention include compounds, which contain at least one aromatic ring or a polycondensed (fully or partially saturated) cyclic structure, optionally wherein the molecule also contains at least one hydrogen bond donor or hydrogen bond acceptor. Hydrogen bond donors include at least one XH moiety wherein X = C, N, O, or S (preferably wherein X = N or O). Hydrogen bond acceptors include oxygen and nitrogen atoms. Other active pharmaceutical ingredients that are particularly useful in the compositions of the invention include compounds which contain at least one aromatic ring or a polycondensed cyclic structure, and at least one hydrogen bond donor or hydrogen bond acceptor. Without wishing to be bound by theory, it is believed that such compounds are able to participate in hydrophobic interactions (ττ-ΧΗ interactions
wherein X = C, N, O, S; and ττ-π interactions) with large surface area hydroxyl-rich portions of the cellulose.
Results obtained from molecular modelling studies suggest that the rapid-release characteristics observed for the compositions containing flufenamic acid in the Examples will similarly be observed for compositions of the invention containing other drug molecules which have at least one aromatic ring or a polycondensed (fully or partially saturated) cyclic structure, particularly where the drug molecule also contains at least one hydrogen bond donor or hydrogen bond acceptor.
The molecular modelling studies are summarised here. Interaction energies between aromatic organic molecules (AOM) and cellulose were probed using density functional theory (DFT) with respect to weak intermolecular interactions such as H-bonding, ττ-π stacking, and ττ-ΟΗ bonds.
Three model AOMs, viz. benzene, benzamide and benzoic acid, were used for ab initio simulations with cellulose using cellobiose as the structural monomer. No consideration to different cellulose allomorphs or cellulose crystal planes was taken. All pre-relaxation molecular coordinates were acquired from the PubChem Compounds Database. The Van der Waals exchange-correlation functional vdW-DF by Dion et al (Dion M. et al. (2004), Phys. Rev. Lett. 92, 246401) as implemented by Roman-Perez and Soler (Soler JM et al (2002) J. Phys. Condens. Matter 14, 2745-2779; Roman-Perez et al (2009) Phys. Rev. Lett. 103, 096102) was used. The basis set was DZP and the energy cut-off was set at 300 Ry. The molecular systems were structurally relaxed using the iterative Conjugate Gradient (CG) method.
Results
Table 3 - interaction energies of AOM dimers
It was found that the studied aromatic organic molecules are in general more likely to interact with cellulose than to form a dimer. Although H-bonding was particularly strong, the contribution of ττ-ΟΗ interactions was still significant. The results predict that there is a high affinity between cellulose and AOM and this affinity will be further amplified over the large surface area that cellulose may provide, wherein the interactions at a monolayer or close to a monolayer will be the strongest.
Particularly preferred BCS Type 2 and 4 APIs that may be mentioned in this respect include compounds which contain at least one aromatic ring (preferably at least two aromatic rings), and at least one hydrogen bond donor or hydrogen bond acceptor. Thus, a further embodiment relates to compositions of the invention containing at least one active pharmaceutical ingredient (e.g. an NSAID) having such structural features (i.e. at least one (e.g. at least two) aromatic ring, and at least one hydrogen bond donor or hydrogen bond acceptor). Particular NSAIDs that may be mentioned in this respect include arylpropionic acid derivatives (e.g. ibuprofen, ketoprofen, flurbiprofen, naproxen), anthranilic acid derivatives (e.g. flufenamic acid, mefenamic acid), acetic acid derivatives (e.g. indomethacin, sulindac), and enolic acid derivatives e.g. pyroxicam. Particular NSAIDs having at least two aromatic rings that may be mentioned in this respect include arylpropionic acid derivatives (e.g. ketoprofen, flurbiprofen, naproxen); anthranilic acid derivatives (e.g. flufenamic acid, mefenamic acid), acetic acid derivatives (e.g. indomethacin, sulindac), and enolic acid derivatives (e.g. pyroxicam).
Further particularly preferred BCS Type 2 and 4 APIs that may be mentioned in this respect include compounds which contain at least two aromatic rings, and at least one hydrogen bond donor or hydrogen bond acceptor. Thus, a further embodiment relates to compositions of the invention containing at least one active pharmaceutical ingredient (e.g. an NSAID or an antineoplastic agent) having at least two aromatic rings, and at least one hydrogen bond donor or hydrogen bond acceptor. It has been surprisingly found that these active ingredients are capable of being incorporated into the modified microcrystalline cellulose excipient described herein (e.g. using the heat-assisted methods disclosed herein) in spite of the relatively high melting points and relatively low vapour pressures associated with these substances. The reference to heat-assisted methods above includes heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, and the like.
In a further embodiment, including embodiments in which the compositions of the invention containing at least one active pharmaceutical ingredient (e.g. an NSAID or an antineoplastic agent) having at least two aromatic rings and at least one hydrogen bond donor or hydrogen bond acceptor, said active pharmaceutical ingredient may have a molecular weight of at least 200 g/mol, preferably at least 210 g/mol. In still further embodiments, the molecular weight may be at least 220, at least 230, at least 240, at least 250, at least 260, at least 270, at least 280, at least 290 or at least 300 g/mol. Active pharmaceutical ingredients having a melting point (or glass transition temperature) not greater than the temperature at which cellulose degrades are also preferred as this
ensures that incorporation of the active pharmaceutical ingredient into the cellulose excipient can be more easily achieved. Typically, therefore, the active pharmaceutical ingredient will have a melting point (or glass transition temperature) of up to about 350°C, preferably up to about 300°C.
The active pharmaceutical ingredients mentioned herein, in particular the NSAIDs and antineoplastic agents mentioned above as well as compounds having at least two aromatic rings and at least one hydrogen bond donor or hydrogen bond acceptor, are particularly suited for use in the compositions of the present invention. Such compositions may stably contain the active pharmaceutical ingredient in an amorphous form such that the composition is capable of rapidly releasing the active pharmaceutical ingredient in vivo. This in turn provides a significant clinical benefit to the patient, particularly where relief from painful discomfort is desired. Other classes of compounds that may be particularly suited to be formulated with the modified microcrystalline cellulose excipients described herein include those which contain a free carboxylic acid group (typically together with the one (or preferably two) aromatic ring), or an amide. The compositions of the present invention are particularly suited to the delivery of active pharmaceutical ingredients that are poorly soluble.
It should be noted that the advantages that have been identified for the compounds of the present invention are not limited to the compositions containing NSAIDs. The advantages may be realised for any drug which has a relatively low solubility, i.e. Type 2 and 4 BCS drugs, and which is typically administered orally to patients and has desirable structural features which enable ττ-π and ττ-ΟΗ interactions. For the avoidance of doubt, the compositions of the invention may be administered by other routes, in particular transmucosally (such as via sublingual or buccal administration), or via rectal or vaginal administration.
Active ingredients may further be employed in salt form or any other suitable form, such as e.g. a complex, solvate or prodrug thereof, or, if relevant, in any stereoisomeric form including any enantiomeric, diastereomeric or racemic form, or a combination of any of the above.
Pharmaceutically-acceptable salts of active ingredients that may be mentioned include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of an active ingredient with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of active ingredient in the form of a salt with another counter-ion, for example using a suitable ion exchange resin. Examples of pharmaceutically acceptable addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric and sulphuric acids; from organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, arylsulphonic acids; and from metals such as sodium, magnesium, or preferably, potassium and calcium.
In the compositions of the invention, the active pharmaceutical ingredient is present in a predominantly amorphous form. In one embodiment, at least 80% of the active pharmaceutical ingredient in the composition is in an amorphous form. For example, the active pharmaceutical ingredient may be essentially amorphous (i.e. at least about 90% amorphous, that is at least about 90% by weight of the active pharmaceutical ingredient is present in an amorphous form). Preferably, the active pharmaceutical ingredient is at least about 95% amorphous, at least about 98% amorphous or most preferably at least about 99% amorphous. The degree of amorphicity in the active pharmaceutical ingredient may be determined by e.g. DSC. Additionally, the XRD profile for a product which is in a predominantly amorphous form would be one in which sharp peaks associated with the crystalline product (i.e. the drug or its polymorphs in the compositions of the invention) are substantially absent. For example, for a composition in which the active pharmaceutical ingredient is present in a predominantly amorphous form, the degree of crystallinity of the active pharmaceutical ingredient may be found to be less than about 10%, when determined using DSC. Compositions in which the active pharmaceutical ingredient is present in a predominantly amorphous form may be qualitatively identified via their XRD profiles by virtue of the absence of sharp characteristic diffraction peaks typical for crystalline substances. Infra-red absorption spectroscopy (e.g. FT-IR) can also show shifts in absorption frequencies indicating molecular rearrangement and interactions between API and excipient. Where the drug is a substance that contains an aromatic group and a carbonyl
group (such groups are typically present in NSAIDs) the changes in the frequencies of the aromatic vibrations (between around 1000 and 800 cm-1) and C=0 vibrations (between around 1500 and 1800 cm-1) will be indicative of significant molecular rearrangement. One skilled in the art will understand that shifts of other characteristic peaks for H-bond donors/acceptor groups may also be indicative of molecular rearrangement.
Prior to introduction to the composition, the active pharmaceutical ingredient may be provided in any form, including a form that is crystalline or substantially crystalline.
In a further embodiment, the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient described herein in the composition of the invention is at most about 1 :3. For example, the weight ratio may be at most about 1 :5. In a preferred embodiment, the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient in the composition is not more than about 1 :9.
In further preferred embodiment, the pharmaceutical composition comprises the active pharmaceutical ingredient at an amount of between 5 and 30% by weight of the composition.
Particularly preferred compositions of the invention include those in which the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient in the composition is not more than about 1 :3 (e.g. not more than about 1 :5), and the surface area of the modified microcrystalline cellulose excipient (prior to the introduction of the active pharmaceutical ingredient) is from about 5 to about 50 m2/g (e.g. from about 10 to about 40 m2/g) as measured by N2 gas adsorption technique according to the Brunauer Emmett Teller (BET) method. Further preferred compositions of the invention include those in which the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose excipient in the composition is not more than about 1 :3 (e.g. not more than about 1 :5), and the active pharmaceutical ingredient is at least about 90% amorphous. Still further preferred compositions of the invention include those in which the weight ratio of the active pharmaceutical ingredient to the modified microcrystalline cellulose
excipient in the composition is not more than 1 :5 (e.g. not more than 1 :9), and the active pharmaceutical ingredient is at least about 95% amorphous.
Still further preferred compositions of the invention include those in which the modified microcrystalline cellulose excipient has a degree of crystallinity of from about 70% to about 90%, the surface area of the modified microcrystalline cellulose excipient (prior to the introduction of the active pharmaceutical ingredient) is from about 5 to about 50 m2/g (e.g. from about 10 to about 40 m2/g), and the active pharmaceutical ingredient is at least about 90% amorphous.
Compositions of the invention may be prepared by way of a variety of routine techniques, and using standard equipment, known to the skilled person, including mixing together the active pharmaceutical ingredient and a modified microcrystalline cellulose excipient having the desired surface area.
Typically the active pharmaceutical ingredient will be initially provided in a crystalline form. Amorphisation of the active pharmaceutical ingredient may be achieved through any technique which is capable of reducing the level of crystallinity of the active pharmaceutical ingredient in the composition without substantially degrading the cellulose component or adversely altering its physical structure. Suitable techniques include heat-assisted intensive mixing (e.g. melt extrusion), static heat sealing, intensive mixing, reduced pressure mixing, moderate heating, combined grinding and heating, and co-spray drying with a solvent. Heat-assisted intensive mixing, for example melt extrusion, is particularly useful. During the heat-assisted mixing the components are heated while being mixed, while the heating temperature may be at or below the melting point of the active ingredient. An extruder equipped with heated compartments, e.g. similar to those used for melt extrusion, is particularly useful. For the compositions of the invention, the process involves the processing of a mixture containing the active pharmaceutical ingredient (optionally initially in a substantially crystalline form) and the modified microcrystalline cellulose excipient having the desired surface area. A classical melt extrusion process is a process during which at least one component in the blend is melted (or at least heated to close to its melting point or above its glass transition temperature) and the mixture is formed into products of different shapes and sizes by forcing the components and active substances through an orifice or die under controlled temperature, pressure, feeding rate, and screw speed. Suitable melt extrusion techniques that will be known to the
skilled person include those described in Shah S., et ai, Int. J. Pharm., 453 (2013) 233. Conventional melt extrusion techniques are typically used with thermoplastic polymers, i.e. polymers that melt before they degrade. In contrast, native cellulose, such as MCC, is a non-thermoplastic polymer, which degrades by pyrolysis before it melts. Nonetheless, melt extruders and other heat-assisted intensive mixers are particularly useful in the context of the present invention. Thus, these techniques can also be used with the non-thermoplastic cellulose-based systems of the present invention, e.g. using single- or twin-screw extruders, preferably with a heating unit as long as the temperature inside the mixture does not exceed that at which the cellulose degrades by pyrolysis. Thus, the temperature inside the mixture should not exceed about 350°C, and preferably should not exceed about 300°C.
It may also be possible to form compositions of the invention without extrusion by statically heating blends of pre-mixed active ingredient and the modified microcrystalline cellulose excipient described herein. The mixture is then heated to a temperature above the glass transition temperature of the active ingredient for a sufficient period of time but below the degradation temperature of the cellulose such that the active pharmaceutical ingredient is substantially present in an amorphous form. In practice, the blends are heated in the range between 50 and 300 °C.
Compositions of the invention may also be prepared by static heat sealing. In this process, mixtures of drug and cellulose are placed in a sealed vessel. The mixture is then heated to a temperature above the glass transition temperature of the active pharmaceutical ingredient for a sufficient period of time (the exact period to be determined empirically depending on the quantities of the used ingredients, e.g. between 5 min and 72 hours) such that the active pharmaceutical ingredient is substantially present in an amorphous form.
Thus a further aspect of the invention relates to a pharmaceutical composition comprising a modified microcrystalline cellulose excipient as defined herein and an active pharmaceutical ingredient, wherein the composition is obtained by a process involving heating a mixture of the active pharmaceutical ingredient and the modified microcrystalline cellulose excipient to a temperature close to or above the glass transition temperature of the active pharmaceutical ingredient. In practice, we mean that the mixture is heated to a temperature above about 50 °C. The temperature should not exceed that at which degradation of the cellulose component would occur. Thus,
typically in processes in which the cellulose component is heated in the presence of the active pharmaceutical ingredient, the temperature used should not exceed about 300 °C.
Heat-assisted extrusion, including melt extrusion, is a particularly preferred technique for use in the context of the present invention. Thus, in one embodiment, the invention relates to a composition which is formed by a process involving heat-assisted extrusion of a mixture of the cellulose or cellulose derivative and the active pharmaceutical ingredient. The mixing time period is likely to vary according to the equipment used, and the skilled person will have no difficulty in determining by routine experimentation a suitable mixing time for a given combination of ingredient(s) empirically.
Microcrystalline cellulose excipients which have a surface area in the region of from 2 to 60 m2/g (as measured by N2 gas adsorption technique according to the Brunauer Emmett Teller (BET) method) are particularly advantageous as these substances provide a sufficiently high area of interaction with the active pharmaceutical ingredient while also providing a formulation that has good stability (i.e. in which the active pharmaceutical ingredient is maintained in an amorphous form for longer periods). Strong interactions between the active pharmaceutical ingredient and the cellulose component also help to improve the dissolution characteristics of the overall composition. The strength of the interactions is governed, at least in part, by the affinity between API and cellulose and further amplified over a large surface area. Strong interactions between cellulose and the active pharmaceutical ingredient can be observed in many ways, including FTIR spectroscopy or sometimes through colour changes associated with the active pharmaceutical ingredient itself, as well as through analysing the solid state fluorescence spectra of the mixture.
A preferred process for the formation of compositions of the invention involves the mixing together of an active pharmaceutical ingredient and a modified microcrystalline cellulose excipient having a surface area of from about 2 to about 60 m2/g. In an embodiment of such a process, the modified microcrystalline cellulose excipient is obtained by any of the methods disclosed herein. The product obtained by the above-mentioned process may further be adapted by:
heat-assisted extrusion, e.g. melt-extrusion;
static heat sealing;
heat-assisted intensive mixing,
mixing under reduced pressure,
heating;
mild grinding (i.e. grinding which does not adversely affect the pore structure or the physical-chemical properties of the excipient); and/or
co-spray drying, or rotary evaporation at reduced pressure, with a solvent, preferably wherein the solvent is a mixture of water and lower alkyl alcohol;
using routine techniques in all cases. Compositions of the invention may further comprise one or more further commonly- employed pharmaceutical excipients. Suitable excipients include inactive substances that are typically used as a carrier for the active ingredients in medications. Suitable excipients also include those that are employed in the pharmaceutical arts to bulk up pharmaceutical compositions that employ very potent active ingredients, to allow for convenient and accurate dosing. Alternatively, excipients may also be employed in manufacturing processes of the compositions of the invention to aid in the handling of the active ingredient concerned. One skilled in the art will understand that in pharmaceutical formulation other additives apart from diluent may be employed such as taste masking agents, glidants, superdisintegrants, coating agents, etc.
The compositions of the invention are preferably administered orally to the gastrointestinal tract and may provide for rapid release of the active pharmaceutical ingredient in the stomach and/or, preferably, the intestinal system. In this respect, the compositions of the invention may be incorporated into various kinds of pharmaceutical preparations intended for oral administration using standard techniques (see, for example, Lachman et al, "The Theory and Practice of Industrial Pharmacy', Lea & Febiger, 3rd edition (1986) and "Remington: The Science and Practice of Pharmacy" , Gennaro (ed.), Philadelphia College of Pharmacy & Sciences, 19th edition (1995)), for example to form a capsule, a powder or a tablet.
The compositions of the invention may also be administered to the patient through other routes, such as via transmucosal (e.g. sublingual or buccal), rectal or vaginal administration. An appropriate route of administration is one which allows for rapid uptake of the active pharmaceutical ingredient into the bloodstream following administration. Sublingual, buccal, rectal and vaginal routes of administration are
suitable in this respect as they allow the active pharmaceutical ingredient to rapidly enter into the bloodstream, thereby leading to a fast onset of action.
Pharmaceutical preparations comprising compositions of the invention contain a pharmacologically effective amount of the active ingredient. By "pharmacologically effective amount", we refer to an amount of active ingredient, which is capable of conferring a desired therapeutic effect on a treated patient, whether administered alone or in combination with another active ingredient. Such an effect may be objective (i.e. measurable by some test or marker) or subjective (i.e. the subject gives an indication of, or feels, an effect).
More preferred compositions of the invention may be adapted (for example as described herein) to provide a sufficient dose of drug over the dosing interval (irrespective of the number of doses per unit time) to produce a desired therapeutic effect.
The amounts of active ingredients that may be employed in compositions of the invention may thus be determined by the physician, or the skilled person, in relation to what will be most suitable for an individual patient. This is likely to vary with the route of administration, the type and severity of the condition that is to be treated, as well as the age, weight, sex, renal function, hepatic function and response of the particular patient to be treated.
Suitable dosages of active ingredient in one oral delivery unit (e.g. one tablet) may be below 1 g, preferably below 100 mg and above 2 mg. Similar doses may also be appropriate for delivery by other routes, particularly via sublingual, buccal, rectal and vaginal administration.
While NSAIDs are typically used as analgesics, anti-inflammatory drugs and antipyretics, they can also be useful for treatment of primary dysmenorrhea and migraine.
Dysmenorrhea is the pain associated with menstruation in women. It affects approximately 25% of women and in younger women (67-90%) it occurs without underlying problem. (Livshitz and Seidman (2010) Pharmaceuticals, 3, 2082-2089). It has been found that the over-production of uterine PGs is key factor to the painful cramps that are the major symptom of dysmenorrhea. NSAIDs decrease the menstrual pain by decreasing the intrauterine pressure and lowering PGF2a levels in menstrual fluid. (Dawood, M.Y. (1988) Am. J. Med., 20, 23-29). They relieve pain in 80-85% of
patients and reduce menorrhagia (bleeding), which has also been correlated with excessive PG synthesis (Ylikorkala, O. (1994) Pharmacol. Toxicol., 75, 86-88.), in 30- 40%. Further, NSAIDs can be helpful for planning and timing in-vitro fertilization (IVF) treatments and in attenuating the progression of labour during pregnancy (Livshitz and Seidman (2010) Pharmaceuticals, 3, 2082-2089).
Enhancement of NSAIDs bioavailability may implicate important clinical effects due to two reasons: reduced side effects and rapid onset of action.
Side effects: Due to the low solubility/bioavailability NSAIDs are typically administered in high doses to achieve a therapeutic effect. The latter is partly the reason for the side effects associated with NSAIDs, such as gastric ulcers. Thus, lowering the dose when the bioavailability is enhanced may reduce toxic effects.
Onset of action: The relief in dysmenorrhea patients is associated with the rapid onset of action. It is the rapidity with which the NSAIDs are absorbed that determines how quickly the relief is obtained. Since the onset of menstrual flow is variable, it is practical to initiate the medication at the beginning of menstruation and to continue it for 3 days if necessary. With rapidly absorbed NSAIDs, pre-treatment before the onset of dysmenorrhea is unnecessary. This is of great importance since most of the women experiencing dysmenorrhea are young sexually active women. The fenamates and arylpropionic acid derivatives (profens) are the drugs of choice for treating dysmenorrhea. They act by suppression of menstrual fluid PGs and by a direct analgesic effect. Reduction of menstrual fluid is the result of direct inhibition of PG biosynthesis and release in endometrial tissue - a phenomenon that occurs during the first 48 hours of menstruation. Arylpropionic acid derivatives (such as ibuprofen, flurbiprofen, ketoprofen and naproxen) and fenamates (such as mefenamic acid and flufenamic acid) are particularly useful for treatment of dysmenorrhea. As a rule of thumb, arylpropionic acid derivatives are preferred as they give rise to fewer side-effects, while fenamates are known to have stronger action (Marjoribanks J, et ai , 2009. Nonsteroidal anti-inflammatory drugs for primary Dysmenorrhea, Cochrane report, Wiley). The stronger mechanism of fenamate action is believed to be dual, i.e. inhibition of PG synthesis and antagonism to PG in certain tissues. There is a strong correlation between the solubility, dissolution rate and bioavailability of fenamates (Shinkuma, et ai,
1984, Int. J. Pharm., 21 , 187-200). Varying bioavailability due to poor solubility was the reason why some fenamates have been removed from the market, e.g. flufenamic acid.
Another effective alternative for treatment of primary dysmenorrhea is administration of contraceptives, such as progesterone. The pain associated with dysmenorrhea is postulated to result from progesterone withdrawal before the onset of menses, which causes an increase in PGs. Therefore, administration of progesterone can alleviate the symptoms of primary dysmenorrhea. With a prevalence of 8% in males and 12-15% in females migraine is extremely common (Diener, H.C., et al. 2008, 22 (Suppl. 1), 51-58.). It is characterized by recurrent attacks of pulsatile, unilateral headache often accompanied by nausea and vomiting, photo- and phonophobia. In about 20% of patients the headache is preceded by an aura consisting of transient neurological symptoms, most frequently a scintillating scotoma. While the "triptans" -e.g., sumatriptan (Imitrex), rizatriptan (Maxalt), eletriptan (Relpax)-have been promoted over the last 2 decades, NSAIDs still remain a treatment option for acute migraine headache.
Compositions of the invention comprising NSAIDs are therefore useful in the treatment of migraine (e.g. acute migraine headache) and/or dysmenorrhea (e.g. primary dysmenorrhea). According to a further aspect of the invention there is provided a method of treatment of migraine (e.g. acute migraine headache) which method comprises administration of a composition of the invention to a person suffering from, or susceptible to, such a condition. According to a still further aspect of the invention there is provided a method of treatment of dysmenorrhea (e.g. primary dysmenorrhea) which method comprises administration of a composition of the invention to a person suffering from, or susceptible to, such a condition. Particular active pharmaceutical ingredients that are useful in such methods are non-steroidal anti-inflammatory drugs. According to a further aspect of the invention there is provided the use of an active pharmaceutical ingredient (e.g. an NSAID) in the manufacture of a composition of the invention for treating migraine (e.g. acute migraine headache). Similarly, there is provided the use of an active pharmaceutical ingredient (e.g. an NSAID or a contraceptive) in the manufacture of a composition of the invention for treating dysmenorrhea (e.g. primary dysmenorrhea). Still further, there is provided the use of a pharmaceutical composition of the invention in the manufacture of a medicament for
treating dysmenorrhea or migraine, wherein the active pharmaceutical ingredient is a non-steroidal anti-inflammatory drug or a contraceptive.
For the avoidance of doubt, by "treatment" we include the therapeutic treatment, as well as the symptomatic treatment, the prophylaxis, or the diagnosis, of the condition.
When compositions of the invention comprise NSAIDs, appropriate pharmacologically effective amounts of such compounds include those that are capable of producing (e.g. immediate) relief of pain or other symptoms when administered perorally. The amount of the NSAID active ingredient may be expressed as the amount in a unit dosage form. In such a case, the amount of NSAID active ingredient that may be present may be sufficient to provide a dose per unit dosage form that is in the range of between about 2 mg and about 1000 mg (e.g. about 200mg or 400 mg). The above-mentioned dosages are exemplary of the average case; there can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
Compositions of the invention possess the advantage of reducing the risks of producing side effects that are typically associated with high doses of drugs. By delivering the active pharmaceutical ingredient in such a way that the higher rates of dissolution than normal can be achieved, the compositions may contain a lower overall quantity of the active pharmaceutical ingredient while still providing a rapid therapeutic benefit for the patient. The use of a lower overall quantity of the active pharmaceutical ingredient helps to reduce the occurrence of unwanted side effects that may occur. NSAIDs in particular may benefit from being used in the compositions of the invention as frequent usage of these drugs is associated with a substantially increased risk of gastrointestinal problems (e.g. gastrointestinal bleeding and ulcers) and kidney problems). Compositions of the invention may also have the advantage that they may be prepared using established pharmaceutical processing methods and may employ materials that are approved for use in foods or pharmaceuticals or of like regulatory status.
Compositions of the invention may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile than, and/or have other useful pharmacological, physical, or
chemical properties over, pharmaceutical compositions known in the prior art, whether for use in the treatment of pain or otherwise.
Wherever the word "about" is employed herein in the context of dimensions (e.g. values, temperatures, pressures (exerted forces), relative humidities, sizes and weights, crystallinities, surface area, particle or grain sizes, etc.), amounts (e.g. relative amounts (e.g. numbers or percentages) of particles, individual constituents in a composition or a component of a composition and absolute amounts, such as doses of active ingredients, numbers of particles, etc.), deviations (from constants, degrees of degradation, etc.) it will be appreciated that such variables are approximate and as such may vary by ± 10%, for example ± 5% and preferably ± 2% (e.g. ± 1 %) from the numbers specified herein.
Preferences and options for a given aspect, feature or parameter of the invention should, unless the context indicates otherwise, be regarded as having been disclosed in combination with any and all preferences and options for all other aspects, features and parameters of the invention. For example, combinations of descriptions of the porosity of the modified microcrystalline cellulose excipient, e.g. the preferred surface area and the preferred pore volume, should be regarded as having been disclosed. The invention is best demonstrated by a comparison of a modified microcrystalline cellulose (hereinafter referred to as "P-MCC") with MCC. For each cellulose, two types of mixtures have been prepared, i.e. normal, physical mixtures and staticly heat sealed mixtures with flufenamic acid ("FFA") as a model drug substance. The invention is illustrated by the following examples in which:
Figures 1A to 1 D show SEM micrographs showing MCC (Figs 1A. and 1 B) and P-MCC (Figs 1C and 1 D) powders at high magnification (Figs. 1A and 1 C) and low magnification (Figs 1 B and 1 D). Information of specific magnification and scaling is provided in each micrograph.
Figure 2 shows an AMF height sensor image of P-MCC suspension adsorbed on gold plated surface.
Figure 3 shows a pore size distribution curve derived from N2 adsorption and desorption isotherms at 77 K of MCC and P-MCC celluloses.
Figure 4 shows FTIR spectra of (i) pure FFA, (ii) a MCC-FFA mixture stored at standard conditions ("MCC-FFA-N"), and (iii) a heated MCC-FFA mixture (120 °C, 2 h) ("MCC-FFA-H"). The data is normalized with respect to the C-H stretching vibration at 2897 cm"1. Region presented in Fig 4A: 600 - 1000 cm"1 , and Fig 4B: 1400- 1800 cm"1.
Figure 5 shows FTIR spectra of (i) pure FFA, (ii) a MCC-FFA mixture stored at standard conditions ("P-MCC-FFA-N"), and (iii) a heated MCC-FFA mixture (120 °C, 2 h) ("P-MCC-FFA-H"). The data is normalized with respect to the C-H stretching vibration at 2897 cm"1. Region presented in Fig 5A: 600 - 1000 cm"1 , and Fig 5B: 1400 - 1800 cm"1.
Figure 6 shows a representative DSC thermogram of MCC-FFA mixtures and pure FFA. The heat flow of the mixtures is normalised with respect to the total weight of sample.
Figure 7 shows a representative DSC thermogram of P-MCC-FFA mixtures heated to 120 °C for 2 hours and pure FFA. The heat flow of the mixtures is normalised with respect to the total weight of sample.
Figure 8A shows an X-ray powder diffractogram for pure FFA and physical mixture of MCC with FFA. Figure 8B shows an X-ray powder diffractogram for pure FFA and heated mixture of MCC with FFA.
Figure 9A shows an X-ray powder diffractogram for pure FFA and physical mixture of P-MCC with FFA. Figure 9B shows an X-ray powder diffractogram for pure FFA and heated mixture of P-MCC with FFA.
Figure 10A shows dissolution profiles of FFA from MCC into SIF together with dissolution profile for pure FFA at 37.0 ± 0.5 °C. Each data point is the average of two experiments.
Figure 10B shows dissolution profiles of FFA from P-MCC into SIF together with dissolution profile for pure FFA at 37.0 ± 0.5 °C. Each data point is the average of two experiments.
Figure 11 shows X-ray powder diffractograms for (a) as-prepared normal mixture of MCC with FFA; (b) as-prepared heated mixture of MCC with FFA; and (c) heated mixture of MCC with FFA stored at RH 75% for 61 days.
Figure 12 shows X-ray powder diffractograms for (a) as-prepared normal mixture of MCC with FFA; (b) as-prepared heated mixture of MCC with FFA; and (c) heated mixture of MCC with FFA stored at RH 75% for 61 days. Examples
Experimental
Flufenamic Acid ([N-(a,a,a-trifluoro-m-tolyl) anthranilic acid]), hereinafter "FFA", was used as the model drug in the following studies. FFA is a non-steroidal anti-inflammatory drug (NSAID) that was obtained from Sigma-Aldrich in the form of analytical standard for drug analysis.
Microcrystalline Cellulose ("MCC"). The specific powder for this study is provided by Sigma-Aldrich: Avicel® PH-101 , ~50 μηι particle size.
MCC was the original cellulose used in the preparation of P-MCC (modified microcrystalline cellulose) as is detailed in Example 1.
Scanning Electron Microscopy - SEM
SEM images of cellulose powder samples were acquired with a scanning electron microscope (Merlin FEG-SEM, Zeiss, Germany). The membranes were sputtered with Au/Pd prior to analysis to minimize charging effects of the samples.
Atomic force microscopy - AFM
The AFM images were acquired using a Dimension Icon (Bruker, Germany) instrument. A gold surface was coated by submerging it in a 10 mM solution of 6-amino-1- hexanethiol hydrochloride for 2 hours and then dried by gently blowing compressed air over the surface. A drop (100 μΙ_) of highly diluted dispersion containing cellulose crystallites (0.01 % wt) was placed on the precoated gold plate surface (5 mm in diameter) and dried. The gold plate was then mounted on the AFM instrument using a double adhesive tape. The height images were acquired in the peak force tapping mode, using manufacturer's Scan Asyst cantilever and ScanAsyst optimization algorithm.
Nitrogen Gas Sorption
Nitrogen gas (N2 at 77 K) adsorption and desorption isotherms were obtained using an ASAP 2020 instrument (Micromeritics, USA). The gas absorption was conducted on pure, solid powders of P-MCC (>200 mg) and MCC (2 g). The specific surface area was
calculated according to the BET method during N2 adsorption, while the pore size distribution was determined based on N2 adsorption data using DFT method. The total pore volume of the powders was obtained as the volume of adsorbed nitrogen at a relative pressure (p/pO) approximating unity. All calculations were determined using the ASAP 2020 instrument software. The samples were degassed at 100 °C under vacuum for at least 6 h prior to analysis. Filler rods were used for each sample during N2 sorption measurement.
Fourier Transform Infrared Spectroscopy - FTIR
FTIR analysis was conducted on FFA, as a pure substrate, and FFA in blend with different celluloses. Cellulose-FFA blends from both heated and normal, i.e. unheated, samples were analysed. The FTIR spectra were obtained on a Bruker Tensor 27 (Germany) with KBr pellets. A background scan on air was subtracted from all spectra using the instrument software (Opus 7.0, Bruker, Germany). The approximate sample content in 200 mg KBr pellets was 10 wt% (i.e. 1 wt% drug). The collected data was normalized with respect to C-H stretching vibration at 2897 cm-1. The FTIR spectra were collected with the following parameters: 64 scans at a spectrum resolution of 4 cm-1 over a range from 4000 to 400 cm-1. Differential Scanning Calorimetry - DSC
Thermal analysis was performed with Model Q-2000 TA instrument (USA) on both cellulose-drug mixtures and pure substances. Samples were placed inside hermetically sealed aluminium crucibles with punctured lids, in order to avoid overpressure caused by water evaporation. An empty pan was used as a reference. The analysis was conducted in the temperature range from -40°C to 150°C with a heating rate of 10°C min-1. N2 gas, at a flow of 50 ml_ min-1 , was applied during analysis. Initially the samples were cooled from room temperature to -40°C, then heated to 150°C and finally cooled to 25°C again. All of the samples were stored at ambient conditions for 24 hours prior to DSC measurements. For the heated cellulose-FFA mixtures the heating conditions were 120 °C for 2 hours. The measurements were performed in triplicate, and the estimated amount of drug in the mixtures was 10 wt%. Enthalpies and temperatures were estimated by Universal Analysis 2000 Software (TA Instruments).
X-ray diffraction -XRD
The characteristic X-ray diffraction patterns were generated using a D8 Twin-Twin, Bruker (Germany) instrument with Bragg- Brentano geometry for both FFA, as a pure drug, and FFA in a blend with different celluloses. The samples were scanned at room
temperature (25 °C), CuKa radiation was utilized (λ = 1.54 A) with 2Θ angle set between 10 and 60°. Pure FFA (5 mg) was used as the reference and 50 mg of both normal and heated samples of cellulose-FFA blends (approx. 10 wt% FFA in each sample) were scanned once during the analysis.
Example 1 - Formation of microcrystalline cellulose excipient
MCC was used to prepare P-MCC (a modified microcrystalline cellulose excipient) particles in a hydrolysis reaction with hydrochloric acid followed by high-shear homogenisation to release nanocellulose whiskers, and spray-drying.
Pure MCC powder (20 g) was soaked in water 364 mL of water for 24 h at room temperature and then stirred in a round-bottomed flask at 105°C for 60 min under reflux. Concentrated HCI (236 mL, 37%) was added to the dispersion until a final acid concentration of 4 M was obtained, and the reaction mixture was maintained in a preheated oil bath at 105 °C for an additional 60 min. Later, pH-adjustment and neutralization of the solution to pH 5.7 was made by addition of 4M sodium hydroxide solution under stirring. The prepared dispersion was washed by centrifugation at 500 rpm for 10 min 5 times until a final pH value of 6.9 was obtained. The aqueous suspension of MCC was sonicated for 20 minutes with a high intensity ultrasound processor (Sonication Pulse Rate: 30 seconds On, 30 seconds Off) at 70% amplitude to yield a thick white gel which was then homogenized using a high-pressure fluidizer. Two sets of chambers with different sizes connected in series were used with internal diameters of 400/200 μηι and 200/100 μηι, respectively. The suspension was passed once through the larger chamber and six times through the small chamber at 1600-1700 bar. The dry matter content of 1.96 wt% for the dispersion was calculated gravi metrically by drying the sample to a constant weight at 150 °C. The sample was dispersed further with additional 150 mL deionized water until a 0.96 wt% dry matter content was achieved. The sample was sonicated one more time for 20 minutes (Sonication Pulse Rate: 30 seconds On, 30 seconds Off) at 70% amplitude.
The produced dispersion was diluted with ethanol (95%) until a concentration of 50 vol% ethanol was achieved. The obtained dispersion was then spray-dried in nitrogen atmosphere at inlet temperature 195±5 °C, outlet temperature 103±3 °C, pump flow rate setting 15%, and spray flow rate 500 L/h (Buchner mini spray-dryer B-290). The collected product was stored at room temperature and 0% relative humidity in a desiccator until used.
Example 2 - Characterisation of microcrystalline cellulose excipient
Scanning Electron Microscopy
At lower magnification, SEM images of the celluloses of Example 1 show roughly spherical particles of modified microcrystalline cellulose unlike rod-like particles of MCC.
Atomic force microscopy (AFM)
Figure 2 shows the cellulose crystallites, which were released upon hydrolytic cleavage in hydrochloric acid and high-shear homogenisation. The homogenization resulted in relatively well-dispersed individual or slightly aggregated cellulose fragments (whiskers). The length of the whiskers normally did not exceed 500 nm. The cellulose whiskers (in the form of a suspension) were spray-dried to produce the final P-MCC product, as shown in Figure 1. Nitrogen Gas Sorption
Nitrogen sorption isotherms were used to determine the BET specific surface areas, pore sizes and total pore volumes. The pore size characteristics and BET specific surface areas are tabulated in Table 5. Table 5: Specific surface area and pore volume
a The values are the average with standard deviation presented in parentheses (n = 2).
b Single point desorption total pore volume of pores less than 1 17 nm width at p/po = 0.9832 c Single point desorption total pore volume of pores less than 139±4 nm width at p/po = 0.9860 The obtained specific surface area and pore volume of P-MCC were about 9 m2/g and 0.022 cm3/g, respectively, which is much higher than that for ordinary MCC. Figure 3 shows a pore size distribution curve of MCC and P-MCC celluloses. It is clear that the pores having a size of from 10 to 80 nm contribute a substantially greater pore volume for P-MCC than the same sized pores do for MCC. This is consistent with the data in Table 5.
Example 3 - Formation of cellulose-API mixtures
Modified and ordinary MCC were blended with FFA in 1 :9 ratio as shown in Table 6).
Table 6: Summary of cellulose-FFA blends and preparation/storage conditions
Cellulose-FFA samples for characterization and release study were prepared the day before analysis with an approximate storage time of 24 hours at ambient conditions. Typically, a 50 mg blend of cellulose and drug was prepared by mixing 5 mg FFA with 45 mg of selected cellulose powder in 1 ml_ glass vials. The vials were sealed with plastic screw caps and vortexed for 30 seconds. Each cellulose-FFA blend was analysed in both heated and unheated (normal) form. Vials containing the cellulose-FFA blends were placed in a preheated oil bath at 120 °C for 2 hours in the heat-treatment procedure.
Example 4 - Characterisation of cellulose-API mixtures
The potential interactions between the drug and cellulose in the mixtures of Example 3 were analysed and evaluated both for normal and heated samples.
Fourier Transform Infrared Spectroscopy
Figures 4 and 5 display spectra collected on FFA in formulation with different celluloses in both normal and heated mixtures together with a reference band for pure crystalline FFA. Several distinctive functional groups of FFA are detectible by FTIR spectroscopy (Jabeen S., et ai, Spectrochim. Acta. Part A Mol. Biomol. Spectrosc. vol. 96, 972-985, 2012) most of which are normally absent in the chemical structure of cellulose.
The FTIR spectra for normal blend of P-MCC-FFA (Figure 5) display an apparent difference in band characteristics for both selected regions, i.e. the characteristic bands appeared to shift to higher wavenumbers, become broader and generally decrease in intensity. This is observed particularly for bands corresponding to CF3 vibration at 659 and 652 cm-1 , aromatic out-of-plane C-H deformations at 889 cm-1 and in-plane aromatic C-H deformation at 1422 cm-1. The characteristic FFA bands in normal mixture with MCC demonstrated no particular deviation from relevant band positions.
The FTIR spectra for heated samples revealed substantial deviations from the characteristic band positions of FFA in reference spectra. In the selected spectral regions, a shift in band positions was observed at nearly all bands of pure FFA for each heated cellulose-FFA mixture (Figs. 4 and 5). The characteristic bands of FFA shifted in their wavenumber position and were further notable in their broader appearance and lower intensity in spectra for heated P-MCC-FFA and MCC-FFA. The shift in the band position at 1655 cm-1 was prominent in spectra for the heated sample of P-MCC-FFA in which the band position shifted to lower wavenumber in the heated sample of P-MCC- FFA. The band associated with carbonyl stretching at 1655 cm-1 is known to be especially sensitive to changes in the electrostatic environment of the molecule. Thus, the shift in the wavenumber position at this band is particularly indicative of interaction involving carboxylic group of FFA and cellulose. The observed shift in the band position at 1655 cm-1 in the heated mixture of MCC-FFA was reduced when compared to FFA in formulation with P-MCC. While the positions of some peaks for heated P-MCC-FFA and MCC-FFA shifted, other peaks such as 760, 1519 and 1578 cm-1 were diffuse and of remarkably low intensity, which could be due to interference from water in these samples. Overall, the results from FTIR analysis suggest that a potential interaction is present between FFA and the different celluloses, particularly for FFA in formulation with P-MCC in the heated samples.
Differential Scanning Calorimetry
Figures 6 and 7 display representative thermograms from differential scanning calorimetry (DSC) analysis conducted on samples of pure FFA and physical mixtures of FFA with P-MCC and MCC. The profiles for physical mixtures are normalized with respect to the total weight of the sample (i.e. 10 wt% FFA). The DSC profile for pure FFA showed a distinct endotherm at 135°C corresponding to the melting temperature of crystalline FFA. The exothermic event at about 72 °C in the back-scan DSC profile of pure FFA is representative of recrystallization of FFA from the melt upon cooling. DSC profiles for FFA in mixtures with celluloses were generally characterized by two events: evaporation of water from cellulose in the region between 40 and 110 °C and a distinct endothermic peak at temperature around 133-139 °C, which corresponds to the melting of crystalline FFA. The exothermic event on the back-scan for physical mixtures, corresponding to recrystallization of FFA, could be distinguished only in the normal samples of P-MCC mixed with FFA and. The recrystallization exotherm in the physical mixture of P-MCC-FFA is slightly shifted to higher temperatures accompanied with significant decrease in size compared with recrystallization of pure FFA.
DSC profiles for heated mixtures of FFA with celluloses were characterised by the lack of endothermic peaks in the region corresponding to melting of crystalline FFA. DSC profile of MCC in mixture with FFA displayed several distinguishable peaks close to the melting region of FFA, typically below the specific melting temperature. The absence of the melting peak of FFA (at 135 °C) in the DSC profiles is indicative of an amorphous structure of FFA in the heated samples, since fully amorphous materials do not exhibit a melting endotherm. The broad halo at 40 - 110 °C, characteristic of water evaporation is present in the DSC profiles for heated samples as well. This indicates that water is resorbed in the samples during storage after heat treatment to 120 °C prior to analysis. The results from DSC analysis suggest that FFA is present predominantly in an amorphous state in the heated samples with the exception of MCC-FFA mixture. The peaks present in the region of 120 - 133 °C in the heated MCC-FFA sample suggests the presence of FFA polymorphs with different melting temperatures in the sample.
Tables 7 and 8 present enthalpies with corresponding temperatures for water evaporation, melting and recrystallization of FFA in mixtures together with values of standard deviation for each result. Table 7 - Melting enthalpies of FFA in pure form and in mixtures with different celluloses. Results are presented as averages with standard deviation in parentheses (n=3).
a Two peaks could be detected at near the melting temperature of FFA in each sample, b A peak for melting of FFA was detected only in one sample out of three.
Table 8 - crystallization enthalpies of FFA in pure form and in mixtures with different celluloses. Results are presented as averages with standard deviation in parentheses (n=3).
The degree of crystallinity for FFA in each mixture is presented in Table 7 where a decreasing trend in FFA crystallinity from normal to heated samples is apparent. The highest content of crystalline FFA at 58% is observed for the mixture of FFA with MCC in normal blends. Corresponding values for crystallinity index of FFA in the normal mixture with P-MCC show that the drug is only partially crystalline.
In the heated samples, the FFA crystallinity is very low suggesting that FFA is present predominantly in the amorphous state in heated P-MCC-FFA samples. These results suggest that heated mixtures with celluloses exhibit strong tendency to stabilize the amorphous structure of FFA, while celluloses in normal mixtures have only a limited ability to conserve the amorphicity of the drug.
The recrystallization of FFA occurred explicitly in the normal sample of P-MCC-FFA and the reduced value of the enthalpy (see Table 8) is associated with the lower content of FFA in the mixture (the amount for FFA is 10 times higher in the pure sample of FFA compared to P-MCC-FFA). A repeated measurement for both pure FFA and normal mixture of P-MCC-FFA showed that the peak for recrystallization could arise at several different temperatures indicating that FFA polymorphs with different crystallization behaviour were present. X-ray diffraction
In order to further confirm the physical state of FFA in the heated and normal samples, X-ray diffraction is conducted on samples of pure FFA and FFA in physical mixtures with the different celluloses.
Figures 8 and 9 displays X-ray diffraction patterns for pure FFA and physical mixtures of FFA in both heated and normal samples with celluloses. The diffractogram of FFA is displayed in each graph as a reference. Several well-defined peaks of high intensity appear in the diffraction pattern of pure FFA. The peaks at 14°, 19° and 21 ° are the most prominent. The diffraction pattern for pure MCC is expected to display a diffused peak at 13-16°, a narrow peak at 22-23° and a peak with low intensity at 35° (Mihranyan A., et al., Int. J. Pharm., vol. 269, no. 2, pp. 433-442, 2004). Consequently, the sharp diffraction peaks over-imposed in the profile for normal mixture of MCC-FFA are clearly derived from the FFA content in the mixture. Moreover, it is observed that the peaks associated with FFA in the normal mixture with MCC have shifted from their original positions when compared with the fingerprint peaks for pure FFA. This may indicate substantial molecular rearrangement and formation of FFA polymorphs upon processing. The intensity of crystalline FFA peaks in the diffractograms for heated mixtures is generally reduced compared to the normal mixture of MCC-FFA, and only residual crystalline peaks of low intensity are visible in the diffractogram, in line with the DSC analysis on the MCC-FFA mixture. In the diffractograms for mixtures of P-MCC with FFA (Figure 9) peaks with equivalent positions and appearance as the ones representative of the cellulosic content in MCC-FFA mixtures appeared and were therefore attributed to the cellulosic content of the P-MCC sample. The diffractogram for the normal mixture of P- MCC-FFA showed analogous results as the mixture of MCC with FFA, where the normal mixture displayed peaks corresponding to cellulosic content along with the characteristic peaks for crystalline FFA that continued to appear in the diffractograms. However, the peaks, corresponding to crystalline FFA, appear with a slightly different shift in the peak positions and decrease in the relative intensity. In view of the DSC results for normal mixtures of P-MCC-FFA, in which only 48 % of FFA was found to be crystalline, it is plausible that partial amorphisation is responsible for the overall decrease in relative intensity, while the shift in peak positions is likely to be related to the presence of polymorphism in FFA. The X-ray diffraction patterns for the heated sample of P-MCC-FFA revealed characteristic peaks for cellulose, identical to the peaks in normal mixture. However, the heated mixture of P-MCC-FFA showed no peaks indicating on absence of crystalline FFA. Since amorphous materials do not diffract X-rays in a coherent manner, it is regarded that FFA is present explicitly in its amorphous state in the heated mixture of P- MCC-FFA. Consequently, the results of X-ray diffraction confirmed the previous DSC results suggesting that the drug is present in its crystalline form in the normal mixtures and transformed to fully amorphous state during heat treatment.
Example 5 - Dissolution studies
Dissolution measurements were made on normal and heated physical mixtures of FFA with either MCC or P-MCC to evaluate the release kinetics of FFA. A reference measurement was made using pure FFA. Dissolution kinetics of FFA in formulation with cellulose were determined by the standardized USP paddle method. Release profiles for the drug were created by spectrofluorimetric analysis on samples at various time points.
Method - Standard drug solutions and fluorimetric calibration
Stock solution containing 10 μg/mL flufenamic acid (FFA) was prepared by dissolving FFA in simulated intestinal fluid (SIF). Various amounts of stock solution between 0.1 mL and 1 mL were transferred to plastic vials and frozen at -27 °C. The vials containing stock solution were freeze-dried overnight using a Scanvac CoolSafe 55-4 (LaboGene ApS, Lynge, Denmark). Seven working standard solutions with FFA concentration range between 0.1 and 10 μg/mL were prepared by dissolving the contents of the vials containing freeze-dried stock solution with 1 mL of a polar solvent, Acetonitrile-DMSO (4: 1 , vol/vol). Fluorescence spectral measurements were performed on an Infinite M200 Tecan (Austria) microplate reader equipped with two monochromators (excitation and emission) and UV Xenon light source. Black 96-well round-bottom (Corning 96 Round Bottom, Polystyrol) microplates were used. Excitation and emission spectra were collected using the instrument software and the maximum excitation wavelengths of FFA in the solvent was estimated to Aex = 289 with a full band scan from 200 nm to 400 nm. The maximum emission intensity of the drug in the working standards was measured spectrophotometrically at Aex = 289 with a full band scan from 400 nm to 500 nm. A linear regression analysis was made on the collected data set.
Method - Drug Release
Dissolution measurements were performed with the rotating paddle technique at 37.0±0.5 °C and 50 rpm with SOT AX (AT7 Smart, Switzerland) dissolution apparatus. SIF was selected as the dissolution medium and prepared by diluting 20 mL of concentrated SIF with 480 mL deionized water. Normal and heated cellulose mixtures were poured into dissolution vessels with 500 mL dissolution medium and samples of 1 ml were extracted at various time points (15 min, 30 min, 1 h, 2 h, 3h, 4h and 5h). A total of seven samples with a volume of 1 mL were collected for each cellulose-FFA formulation and passed through a syringe filter into 2 ml plastic vials. The vials were frozen at -27 °C and freeze-dried further until all of the water had sublimated from the sample and the dried FFA filtrate remained. The vials containing dried FFA were filled
with 1 ml_ Acetonitrile-DMSO (4: 1 , vol/vol) solvent and manually shaken until the collected FFA was dissolved. Two parallel measurements were performed for each formulation. Spectrofluorometric analysis together with previously described regression analysis was used to estimate the concentration of released FFA at different time points.
Results
The results from in vitro release experiments, conducted on heated and normal mixtures of FFA with either MCC or P-MCC are presented in Figures 10A and 10B respectively. The dissolution profiles from each physical mixture are displayed together with the profile of the drug alone in the dissolution medium (SIF).
The dissolution profile of FFA in formulation with MCC (Figure 10A) shows a pattern similar to the one of FFA alone. Generally, the release behaviour of FFA from both celluloses in normal formulations was similar to that of FFA alone. The normal FFA-cellulose mixtures and the sample with pure FFA released over 50 % of the drug after 2 hours and between 82-86 % was released after 5 hours of experiment. The dissolution profiles of FFA in the heated formulation with P-MCC (Figure 10B) displayed a considerable acceleration of the dissolution rate of the drug. An initial burst release was apparent and a rapid increase in the concentration of released drug from the heated mixture with P-MCC was observed during the first 50 minutes of the experiment. The concentration of released drug was observed to be three times as high for the heated mixture with P-MCC compared to FFA after the initial 15 minutes where the amount of FFA released from heated P-MCC-FFA mixture was 51 %, compared to 15% for pure crystalline FFA. The cumulative concentration of FFA released from P-MCC after 2 h was 95 % whereas FFA alone did not reach that value within the experimental time frame and displayed a maximum of 87 % after 5 h. It should also be noted that the heated mixture of P-MCC-FFA is the only sample that released the entire amount of preloaded FFA within 5 hours.
Example 6 - Stability studies
Glass vials containing samples were stored at constant relative humidity (75 %) over saturated NaCI solution at room temperature for 61 days inside a desiccator. X-ray diffraction was conducted on samples after 61 days according to the principle specified above.
The results from the stability study showed that recrystallization and transformation of the physical state of FFA was facilitated in normal mixtures during storage where new peaks of high intensity for crystalline FFA were observed in the X-ray profiles. For the normal mixtures, MCC-FFA (Fig. 1 1a) and P-MCC-FFA (Fig. 12a), new peaks of high intensity appeared at 18° and 22° (for MCC-FFA) and 14°, 21 ° and 43° (for P-MCC-FFA). Moreover, the recurring peaks in the profiles for normal samples after storage exhibited much lower intensity than the original ones. Two of the new peaks, at 14° and 21 °, in the profile for normal mixture of P-MCC-FFA (75% RH) appeared with a high intensity at a position identical to the ones shown in the profile for pure crystalline FFA. The appearance of new peaks in the profiles for normal mixtures was accompanied by the complete disappearance of other characteristic FFA peaks present in the original X-ray profiles. This effect was most apparent in the results for the normal mixture of MCC-FFA, where the characteristic peaks at 13°, 24°, 26° and 27° had disappeared in the new profile (Fig. 1 1a). The peak positions and overall profiles differed between the two physical mixtures of selected celluloses in normal samples. The appearance of new peaks of high intensity and disappearance of some characteristic peaks of FFA in the profiles for normal samples suggest that the physical state of FFA undergoes a substantial structural transformation during storage.
X-ray profiles obtained from the stability study on heated mixtures of FFA in formulation with MCC (Figs. 11 b and 11 c) and P-MCC (Figs. 12b and 12c) showed no significant differences from the original samples. In contrast to results for normal mixtures, FFA in the heated mixtures does not undergo a phase transition and long-term exposure to moisture does not induce recrystallization of FFA when in formulation with MCC and P- MCC. The absence of new crystalline peaks in the profiles of MCC-FFA and P-MCC-FFA revealed that the physical state of FFA is preserved during storage and the drug is present in the amorphous state even when high levels of moisture were introduced in the system over time.
Claims
1. A pharmaceutical composition comprising a modified microcrystalline cellulose excipient and an active pharmaceutical ingredient, wherein the modified microcrystalline cellulose excipient has a surface area of from about 2 to about 60 m2/g.
2. The pharmaceutical composition according to Claim 1 , wherein the modified microcrystalline cellulose excipient has a surface area of from about 5 to about 50 m2/g.
3. The pharmaceutical composition according to Claim 1 or Claim 2, wherein the modified microcrystalline cellulose excipient has a total pore volume of at least about 0.005 cm3/g.
4. The pharmaceutical composition according to any one of the preceding claims, wherein the modified microcrystalline cellulose excipient has a total pore volume of from about 0.01 to about 0.1 cm3/g, preferably from about 0.01 to about 0.05 cm3/g.
5. The pharmaceutical composition according to any one of the preceding claims, wherein the active pharmaceutical ingredient is in a predominantly amorphous form.
6. The pharmaceutical composition according to Claim 5, wherein at least 80% of the active pharmaceutical ingredient in the composition is in an amorphous form.
7. The pharmaceutical composition according to any one of the preceding claims, wherein the pharmaceutical composition comprises the active pharmaceutical ingredient at an amount of between 5 and 30% by weight of the composition.
8. The pharmaceutical composition according to any one of the preceding claims, wherein the active pharmaceutical ingredient is a molecule which contains at least one aromatic ring or a polycondensed cyclic structure, optionally wherein the molecule also contains at least one hydrogen bond donor or hydrogen bond acceptor.
9. The pharmaceutical composition according to any one of the preceding claims, wherein the active pharmaceutical ingredient is a non-steroidal anti-inflammatory drug, a steroid and a cholate, optionally selected from the group consisting of ibuprofen, ketoprofen, flurbiprofen, naproxen, aspirin, ethenzamide, mefenamic acid, flufenamic
acid, tolfenamic acid, indomethacin, sulindac, pyroxicam, progesterone, estradiol, progestin, estrogen, cholic acid, deoxycholic acic and ursodeoxycholic acid.
10. The pharmaceutical composition according to any one of the preceding claims, wherein the active pharmaceutical ingredient:
(a) contains at least two aromatic rings, and at least one hydrogen bond donor or hydrogen bond acceptor; and/or
(b) is an NSAID or an antineoplastic agent.
1 1. The pharmaceutical composition according to any one of the preceding claims, wherein the active pharmaceutical ingredient is selected from the group consisting of: anthranilic acid derivatives (such as azapropazone, etofenamate, flufenamic acid, flunixin, meclofenamic acid, mefenamic acid, morniflumate, niflumic acid, and tolfenamic acid); and protein-kinase inhibitor substances (such as Afatinib, Alectinib, Axitinib, Binimetinib, Bosutinib, Brigatinib, Cabozantinib, Cediranib, Ceritinib, Cobimetinib, Crizotinib, Dasatinib, Entrectinib, Erlotinib, Fostamatinib, Gefitinib, Ibrutinib, Imatinib, Lapatinib, Lenvatinib, Lestaurtinib, Masitinib, Momelotinib, Mubritinib, Neratinib, Nilotinib, Nintedanib, Olmutinib, Osimertinib, Pacritinib, Pazopanib, Ponatinib, Radotinib, Regorafenib, Rociletinib, Ruxolitinib, Selumetinib, Semaxanib, Sorafenib, Sunitinib, Tivozanib, Toceranib, Trametinib, Vandetanib and Vemurafenib).
12. A pharmaceutical composition according to any one of Claims 1 to 1 1 , wherein the composition is obtained by a process involving heating a mixture of the modified microcrystalline cellulose excipient and the active pharmaceutical ingredient to a temperature close to or above the glass transition temperature of the active pharmaceutical ingredient.
13. A process for preparing a modified microcrystalline cellulose excipient as defined in any one of Claims 1 to 4, which process comprises:
(i) forming a dispersion comprising a microcrystalline cellulose precursor and a solvent system, wherein the solvent system comprises a mixture of water and one or more volatile water-miscible solvents; and
(ii) spray drying said dispersion.
14. The process according to Claim 13, wherein the microcrystalline cellulose precursor in the dispersion has a D50 value prior to the spray drying step of up to 1 μηι.
15. The process according to Claim 13, wherein the microcrystalline cellulose precursor in the dispersion is a dispersible-grade microcrystalline cellulose.
16. The process according to Claim 15, wherein the dispersible-grade microcrystalline cellulose is a mixture comprising microcrystalline cellulose and a hydrocolloid selected from the group consisting of carboxymethylcellulose, carrageenan, guar gum, sodium alginate, and mixtures thereof.
17. The process according to Claim 13 or Claim 14, further comprising acid hydrolysis of the microcrystalline cellulose precursor prior to the formation of the dispersion.
18. The process according to any one of Claims 13 to 17, wherein the one or more volatile water-miscible solvents is selected from the group consisting of methanol, ethanol, isopropanol, n-propanol, acetone, methyl ethyl ketone, tetrahydrofuran, acetonitrile, and mixtures thereof.
19. The process according to any one of Claims 13 to 18, wherein the one or more volatile water-miscible solvents are present in an amount ranging from about 10 to about 70 % by volume of the solvent system.
20. The pharmaceutical composition according to any one of Claims 1 to 12, wherein the modified microcrystalline cellulose excipient is obtainable by a process as defined in any one of Claims 13 to 19.
21. Method of treating dysmenorrhea or migraine, said method comprising administering a pharmaceutical composition as defined in any one of Claims 1 to 12 or 20 to a subject suffering from dysmenorrhea or migraine, wherein the active pharmaceutical ingredient is a non-steroidal anti-inflammatory drug.
22. A pharmaceutical composition as defined in any one of Claims 1 to 12 or 20 for use in treating dysmenorrhea or migraine, wherein the active pharmaceutical ingredient is a non-steroidal anti-inflammatory drug.
23. Use of a pharmaceutical composition as defined in any one of Claims 1 to 12 or 20 in the manufacture of a medicament for treating dysmenorrhea or migraine, wherein the active pharmaceutical ingredient is a non-steroidal anti-inflammatory drug.
24. A method of preparing a pharmaceutical composition as defined in any one of Claims 1 to 11 or 20, comprising mixing together an active pharmaceutical ingredient and a modified microcrystalline cellulose excipient having a surface area of from about 2 to about 60 m2/g, optionally wherein the modified microcrystalline cellulose excipient is obtained by a process according to any one of Claims 13 to 19.
25. The method of Claim 24, further comprising processing the mixture by:
heat-assisted-extrusion;
static heat sealing;
heat-assisted intensive mixing,
mixing under reduced pressure,
heating;
mild grinding which does not adversely affect the pore structure of excipient; and/or co-spray drying, or rotary evaporation at reduced pressure, with a solvent, preferably wherein the solvent is a mixture of water and lower alkyl alcohol.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
GBGB1610628.8A GB201610628D0 (en) | 2016-06-17 | 2016-06-17 | New compositions |
GB1610628.8 | 2016-06-17 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2017216584A1 true WO2017216584A1 (en) | 2017-12-21 |
Family
ID=56895328
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/GB2017/051782 WO2017216584A1 (en) | 2016-06-17 | 2017-06-19 | New compositions |
Country Status (2)
Country | Link |
---|---|
GB (1) | GB201610628D0 (en) |
WO (1) | WO2017216584A1 (en) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108938584A (en) * | 2018-08-17 | 2018-12-07 | 湖北欣瑞康医药科技有限公司 | A kind of tablet and preparation method thereof of the Tivozanib of inhibitor containing vegf receptor salt |
US11040027B2 (en) | 2017-01-17 | 2021-06-22 | Heparegenix Gmbh | Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JPH03264537A (en) * | 1990-03-15 | 1991-11-25 | Asahi Chem Ind Co Ltd | Method for improving solubility of scarcely soluble drug |
EP1712583A1 (en) * | 2004-01-30 | 2006-10-18 | Asahi Kasei Chemicals Corporation | Porous cellulose aggregate and formed product composition comprising the same |
EP1873196A1 (en) * | 2005-04-22 | 2008-01-02 | Asahi Kasei Chemicals Corporation | Porous cellulose aggregate and molding composition thereof |
EP2689785A1 (en) * | 2010-04-15 | 2014-01-29 | Shin Nippon Biomedical Laboratories, Ltd. | Method for generating dry vaccine powder formulation for intranasal delivery |
-
2016
- 2016-06-17 GB GBGB1610628.8A patent/GB201610628D0/en not_active Ceased
-
2017
- 2017-06-19 WO PCT/GB2017/051782 patent/WO2017216584A1/en active Application Filing
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JPH03264537A (en) * | 1990-03-15 | 1991-11-25 | Asahi Chem Ind Co Ltd | Method for improving solubility of scarcely soluble drug |
EP1712583A1 (en) * | 2004-01-30 | 2006-10-18 | Asahi Kasei Chemicals Corporation | Porous cellulose aggregate and formed product composition comprising the same |
EP1873196A1 (en) * | 2005-04-22 | 2008-01-02 | Asahi Kasei Chemicals Corporation | Porous cellulose aggregate and molding composition thereof |
EP2689785A1 (en) * | 2010-04-15 | 2014-01-29 | Shin Nippon Biomedical Laboratories, Ltd. | Method for generating dry vaccine powder formulation for intranasal delivery |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US11040027B2 (en) | 2017-01-17 | 2021-06-22 | Heparegenix Gmbh | Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death |
CN108938584A (en) * | 2018-08-17 | 2018-12-07 | 湖北欣瑞康医药科技有限公司 | A kind of tablet and preparation method thereof of the Tivozanib of inhibitor containing vegf receptor salt |
CN108938584B (en) * | 2018-08-17 | 2021-01-26 | 湖北欣瑞康医药科技有限公司 | Tablet containing VEGF receptor inhibitor Tivozanib salt and preparation method thereof |
Also Published As
Publication number | Publication date |
---|---|
GB201610628D0 (en) | 2016-08-03 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN102985072A (en) | Pharmaceutical compositions | |
AU2015295846A1 (en) | Directly compressible composition containing micro-crystalline cellulose | |
EA014443B1 (en) | Pharmaceutical composition (variants) and method for manufacturing thereof | |
WO2018199282A1 (en) | Orally administrable enzalutamide-containing pharmaceutical composition | |
TW201526921A (en) | Ticagrelor solid dispersion and preparation method thereof | |
JP2022037240A (en) | New compositions | |
WO2017216584A1 (en) | New compositions | |
WO2016029496A1 (en) | Methosulide tablet and preparation method therefor | |
TWI658841B (en) | The novel formulation comprising a benzimidazole derivative | |
BR112020015217A2 (en) | crystalline form of sodium bictegravir, composition, use, pharmaceutical composition and sodium bictegravir solvate | |
Akhlaq et al. | Physicochemical characterization and in-vitro evaluation of flubiprofen oral controlled release matrix tablets: Role of ether derivative polymer ethocel | |
WO2016175230A1 (en) | Pharmaceutical composition for oral administration | |
WO2019080830A1 (en) | Pharmaceutical composition containing quinoline derivative | |
JP2023036924A (en) | Pharmaceutical composition containing lenalidomide | |
TW201840320A (en) | Pharmaceutical formulations comprising 5-chloro-n4-[2-(dimethylphosphoryl)phenyl]-n2-{2-methoxy-4-[4-(4-methylpiperazin-1-yl)piperidin-1-yl]phenyl}pyrimidine-2,4-diamine | |
EP2672959A1 (en) | Granulated composition comprising tadalafil and a disintegrant | |
WO2009034409A2 (en) | Pharmaceutical compositions of rhein or diacerein | |
Thampi et al. | A NOVEL VAGINAL TABLETS LOADED WITH FLUCONAZOLE NANOSPONGES FOR VULVOVAGINAL CANDIDIASIS TO OVERCOME ANTIFUNGAL DRUG RESISTANCE | |
EP3052086A1 (en) | Pharmaceutical composition comprising low dose active pharmaceutical ingredient and preparation thereof | |
Tong et al. | Enhancement of dissolution and oral bioavailability by adjusting microenvironment pH in crocetin ternary solid dispersions: Optimization, characterization, in vitro evaluation, and pharmacokinetics | |
Jaiswar et al. | Hot melt extrusion: continuous process of preparation of sustained released matrix tablet by using hydroxypropylcellulose | |
WO2022138717A1 (en) | Oral solid preparation | |
JP2019526641A (en) | Tesofensin composition | |
Jagtap et al. | Acrylic co-polymer and organic acid-based press coated pulsatile tablet of nifedipine using 32 factorial design: use of novel solubilizer for solubility enhancement | |
WO2022264004A1 (en) | Pharmaceutical composition comprising itraconazole |
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: 17743377 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: 17743377 Country of ref document: EP Kind code of ref document: A1 |