US20230277566A1 - Broad spectrum antiviral and methods of use - Google Patents
Broad spectrum antiviral and methods of use Download PDFInfo
- Publication number
- US20230277566A1 US20230277566A1 US17/959,512 US202217959512A US2023277566A1 US 20230277566 A1 US20230277566 A1 US 20230277566A1 US 202217959512 A US202217959512 A US 202217959512A US 2023277566 A1 US2023277566 A1 US 2023277566A1
- Authority
- US
- United States
- Prior art keywords
- virus
- hydrogen
- dna
- administering
- amifostine
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 238000000034 method Methods 0.000 title claims abstract description 34
- 230000000840 anti-viral effect Effects 0.000 title claims description 49
- 150000001875 compounds Chemical class 0.000 claims abstract description 103
- 230000009385 viral infection Effects 0.000 claims abstract description 32
- 241000700605 Viruses Species 0.000 claims description 89
- JKOQGQFVAUAYPM-UHFFFAOYSA-N amifostine Chemical compound NCCCNCCSP(O)(O)=O JKOQGQFVAUAYPM-UHFFFAOYSA-N 0.000 claims description 68
- 229960001097 amifostine Drugs 0.000 claims description 62
- 229910052739 hydrogen Inorganic materials 0.000 claims description 41
- 239000001257 hydrogen Substances 0.000 claims description 41
- 230000002441 reversible effect Effects 0.000 claims description 36
- -1 isobutyryl Chemical group 0.000 claims description 33
- ZGSGBCBFJJASJA-UHFFFAOYSA-N 2-[3-(methylamino)propylamino]ethylsulfanylphosphonic acid Chemical compound CNCCCNCCSP(O)(O)=O ZGSGBCBFJJASJA-UHFFFAOYSA-N 0.000 claims description 28
- 241001493065 dsRNA viruses Species 0.000 claims description 28
- 150000003839 salts Chemical class 0.000 claims description 26
- 125000004435 hydrogen atom Chemical group [H]* 0.000 claims description 25
- 239000008194 pharmaceutical composition Substances 0.000 claims description 17
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 claims description 12
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 10
- 229910018828 PO3H2 Inorganic materials 0.000 claims description 7
- 239000003937 drug carrier Substances 0.000 claims description 6
- 239000012453 solvate Substances 0.000 claims description 6
- YQTCQNIPQMJNTI-UHFFFAOYSA-N 2,2-dimethylpropan-1-one Chemical group CC(C)(C)[C]=O YQTCQNIPQMJNTI-UHFFFAOYSA-N 0.000 claims description 5
- UFHFLCQGNIYNRP-UHFFFAOYSA-N Hydrogen Chemical compound [H][H] UFHFLCQGNIYNRP-UHFFFAOYSA-N 0.000 claims description 5
- 241000712907 Retroviridae Species 0.000 claims description 5
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 claims description 5
- 125000003236 benzoyl group Chemical group [H]C1=C([H])C([H])=C(C([H])=C1[H])C(*)=O 0.000 claims description 5
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 claims description 4
- 241000701242 Adenoviridae Species 0.000 claims description 2
- 241000712464 Orthomyxoviridae Species 0.000 claims description 2
- 150000002431 hydrogen Chemical class 0.000 claims 12
- 150000003573 thiols Chemical group 0.000 claims 4
- 108700010877 adenoviridae proteins Proteins 0.000 claims 1
- 238000011282 treatment Methods 0.000 abstract description 20
- 230000002265 prevention Effects 0.000 abstract description 8
- 241000712461 unidentified influenza virus Species 0.000 abstract description 2
- 208000010370 Adenoviridae Infections Diseases 0.000 abstract 1
- 206010060931 Adenovirus infection Diseases 0.000 abstract 1
- 208000011589 adenoviridae infectious disease Diseases 0.000 abstract 1
- YHPLKWQJMAYFCN-UHFFFAOYSA-N WR-1065 Chemical compound NCCCNCCS YHPLKWQJMAYFCN-UHFFFAOYSA-N 0.000 description 84
- 108020004414 DNA Proteins 0.000 description 51
- 239000003814 drug Substances 0.000 description 39
- 102000039446 nucleic acids Human genes 0.000 description 39
- 108020004707 nucleic acids Proteins 0.000 description 39
- 230000000694 effects Effects 0.000 description 38
- 150000007523 nucleic acids Chemical class 0.000 description 38
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 35
- 241000725303 Human immunodeficiency virus Species 0.000 description 33
- 235000002639 sodium chloride Nutrition 0.000 description 31
- 239000003443 antiviral agent Substances 0.000 description 29
- 229940079593 drug Drugs 0.000 description 29
- 230000003612 virological effect Effects 0.000 description 29
- 210000004027 cell Anatomy 0.000 description 28
- 102000004169 proteins and genes Human genes 0.000 description 28
- 230000010076 replication Effects 0.000 description 27
- 108010047620 Phytohemagglutinins Proteins 0.000 description 25
- 230000001885 phytohemagglutinin Effects 0.000 description 25
- 208000036142 Viral infection Diseases 0.000 description 23
- 108090000623 proteins and genes Proteins 0.000 description 22
- 239000003419 rna directed dna polymerase inhibitor Substances 0.000 description 22
- 239000000203 mixture Substances 0.000 description 20
- 125000003396 thiol group Chemical class [H]S* 0.000 description 20
- 238000002474 experimental method Methods 0.000 description 18
- 210000001519 tissue Anatomy 0.000 description 17
- 230000027455 binding Effects 0.000 description 15
- 230000037396 body weight Effects 0.000 description 15
- UFULAYFCSOUIOV-UHFFFAOYSA-N cysteamine Chemical compound NCCS UFULAYFCSOUIOV-UHFFFAOYSA-N 0.000 description 15
- 239000002777 nucleoside Substances 0.000 description 15
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 15
- 239000000126 substance Substances 0.000 description 15
- 102100034349 Integrase Human genes 0.000 description 14
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 14
- 229960003151 mercaptamine Drugs 0.000 description 14
- 239000000243 solution Substances 0.000 description 14
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 13
- 208000037797 influenza A Diseases 0.000 description 13
- 241000450599 DNA viruses Species 0.000 description 12
- 239000003112 inhibitor Substances 0.000 description 12
- 150000003833 nucleoside derivatives Chemical class 0.000 description 12
- 108090000765 processed proteins & peptides Proteins 0.000 description 12
- PFNFFQXMRSDOHW-UHFFFAOYSA-N spermine Chemical compound NCCCNCCCCNCCCN PFNFFQXMRSDOHW-UHFFFAOYSA-N 0.000 description 12
- 229960002555 zidovudine Drugs 0.000 description 12
- HBOMLICNUCNMMY-XLPZGREQSA-N zidovudine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](N=[N+]=[N-])C1 HBOMLICNUCNMMY-XLPZGREQSA-N 0.000 description 12
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 11
- 239000003795 chemical substances by application Substances 0.000 description 11
- 230000006870 function Effects 0.000 description 11
- 208000015181 infectious disease Diseases 0.000 description 11
- 125000000217 alkyl group Chemical group 0.000 description 10
- 230000000798 anti-retroviral effect Effects 0.000 description 10
- 239000000463 material Substances 0.000 description 10
- 229940002612 prodrug Drugs 0.000 description 10
- 239000000651 prodrug Substances 0.000 description 10
- 108091005461 Nucleic proteins Proteins 0.000 description 9
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 9
- 230000015572 biosynthetic process Effects 0.000 description 9
- 238000009472 formulation Methods 0.000 description 9
- 230000003993 interaction Effects 0.000 description 9
- 230000007246 mechanism Effects 0.000 description 9
- 239000003826 tablet Substances 0.000 description 9
- 229940124597 therapeutic agent Drugs 0.000 description 9
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 8
- 239000003638 chemical reducing agent Substances 0.000 description 8
- 239000000306 component Substances 0.000 description 8
- OOTFVKOQINZBBF-UHFFFAOYSA-N cystamine Chemical compound CCSSCCN OOTFVKOQINZBBF-UHFFFAOYSA-N 0.000 description 8
- 229940099500 cystamine Drugs 0.000 description 8
- 239000002609 medium Substances 0.000 description 8
- 230000029812 viral genome replication Effects 0.000 description 8
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 7
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 7
- 206010028980 Neoplasm Diseases 0.000 description 7
- 230000009471 action Effects 0.000 description 7
- 239000004480 active ingredient Substances 0.000 description 7
- 125000005365 aminothiol group Chemical group 0.000 description 7
- 230000036436 anti-hiv Effects 0.000 description 7
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 7
- 239000002775 capsule Substances 0.000 description 7
- 230000014509 gene expression Effects 0.000 description 7
- 230000005764 inhibitory process Effects 0.000 description 7
- 230000000977 initiatory effect Effects 0.000 description 7
- 239000007788 liquid Substances 0.000 description 7
- 239000002207 metabolite Substances 0.000 description 7
- 229920001223 polyethylene glycol Polymers 0.000 description 7
- 239000003755 preservative agent Substances 0.000 description 7
- 230000004224 protection Effects 0.000 description 7
- 230000004223 radioprotective effect Effects 0.000 description 7
- 230000002829 reductive effect Effects 0.000 description 7
- TZYVRXZQAWPIAB-FCLHUMLKSA-N 5-amino-3-[(2r,3r,4s,5r)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-4h-[1,3]thiazolo[4,5-d]pyrimidine-2,7-dione Chemical compound O=C1SC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O TZYVRXZQAWPIAB-FCLHUMLKSA-N 0.000 description 6
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 6
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- 239000012980 RPMI-1640 medium Substances 0.000 description 6
- 239000001768 carboxy methyl cellulose Substances 0.000 description 6
- 239000013553 cell monolayer Substances 0.000 description 6
- 238000006243 chemical reaction Methods 0.000 description 6
- 238000002360 preparation method Methods 0.000 description 6
- 229940063675 spermine Drugs 0.000 description 6
- 238000002560 therapeutic procedure Methods 0.000 description 6
- 239000002562 thickening agent Substances 0.000 description 6
- RSPCKAHMRANGJZ-UHFFFAOYSA-N thiohydroxylamine Chemical compound SN RSPCKAHMRANGJZ-UHFFFAOYSA-N 0.000 description 6
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 6
- 229920002472 Starch Polymers 0.000 description 5
- 150000001413 amino acids Chemical class 0.000 description 5
- 125000003277 amino group Chemical group 0.000 description 5
- 239000011230 binding agent Substances 0.000 description 5
- 238000002648 combination therapy Methods 0.000 description 5
- 230000001419 dependent effect Effects 0.000 description 5
- 239000008121 dextrose Substances 0.000 description 5
- 201000010099 disease Diseases 0.000 description 5
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 5
- 208000002672 hepatitis B Diseases 0.000 description 5
- 206010022000 influenza Diseases 0.000 description 5
- 208000037798 influenza B Diseases 0.000 description 5
- 229940079322 interferon Drugs 0.000 description 5
- 238000001990 intravenous administration Methods 0.000 description 5
- 238000004519 manufacturing process Methods 0.000 description 5
- 239000002105 nanoparticle Substances 0.000 description 5
- 229920000768 polyamine Polymers 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 235000019698 starch Nutrition 0.000 description 5
- NZVYCXVTEHPMHE-ZSUJOUNUSA-N thymalfasin Chemical compound CC(=O)N[C@@H](CO)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O NZVYCXVTEHPMHE-ZSUJOUNUSA-N 0.000 description 5
- 241001430294 unidentified retrovirus Species 0.000 description 5
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 5
- GVJHHUAWPYXKBD-UHFFFAOYSA-N (±)-α-Tocopherol Chemical compound OC1=C(C)C(C)=C2OC(CCCC(C)CCCC(C)CCCC(C)C)(C)CCC2=C1C GVJHHUAWPYXKBD-UHFFFAOYSA-N 0.000 description 4
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 4
- 208000030507 AIDS Diseases 0.000 description 4
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 4
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 4
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 4
- 102000053602 DNA Human genes 0.000 description 4
- 102000004190 Enzymes Human genes 0.000 description 4
- 108090000790 Enzymes Proteins 0.000 description 4
- 108010010803 Gelatin Proteins 0.000 description 4
- 208000031886 HIV Infections Diseases 0.000 description 4
- 208000037357 HIV infectious disease Diseases 0.000 description 4
- 108010050904 Interferons Proteins 0.000 description 4
- 102000014150 Interferons Human genes 0.000 description 4
- 239000002202 Polyethylene glycol Substances 0.000 description 4
- 229940123066 Polymerase inhibitor Drugs 0.000 description 4
- SMWDFEZZVXVKRB-UHFFFAOYSA-N Quinoline Chemical compound N1=CC=CC2=CC=CC=C21 SMWDFEZZVXVKRB-UHFFFAOYSA-N 0.000 description 4
- 108010078233 Thymalfasin Proteins 0.000 description 4
- 239000002253 acid Substances 0.000 description 4
- DZBUGLKDJFMEHC-UHFFFAOYSA-N acridine Chemical compound C1=CC=CC2=CC3=CC=CC=C3N=C21 DZBUGLKDJFMEHC-UHFFFAOYSA-N 0.000 description 4
- YMARZQAQMVYCKC-OEMFJLHTSA-N amprenavir Chemical compound C([C@@H]([C@H](O)CN(CC(C)C)S(=O)(=O)C=1C=CC(N)=CC=1)NC(=O)O[C@@H]1COCC1)C1=CC=CC=C1 YMARZQAQMVYCKC-OEMFJLHTSA-N 0.000 description 4
- 238000013459 approach Methods 0.000 description 4
- 125000003118 aryl group Chemical group 0.000 description 4
- 210000004369 blood Anatomy 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- 201000011510 cancer Diseases 0.000 description 4
- 125000004432 carbon atom Chemical group C* 0.000 description 4
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 4
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 4
- 229940105329 carboxymethylcellulose Drugs 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 230000001120 cytoprotective effect Effects 0.000 description 4
- 231100000135 cytotoxicity Toxicity 0.000 description 4
- 230000003013 cytotoxicity Effects 0.000 description 4
- 230000007123 defense Effects 0.000 description 4
- 238000000326 densiometry Methods 0.000 description 4
- 239000003085 diluting agent Substances 0.000 description 4
- 150000002148 esters Chemical class 0.000 description 4
- 239000008273 gelatin Substances 0.000 description 4
- 229920000159 gelatin Polymers 0.000 description 4
- 229940014259 gelatin Drugs 0.000 description 4
- 235000019322 gelatine Nutrition 0.000 description 4
- 235000011852 gelatine desserts Nutrition 0.000 description 4
- 208000033519 human immunodeficiency virus infectious disease Diseases 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 238000001727 in vivo Methods 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- 229950003954 isatoribine Drugs 0.000 description 4
- 230000014759 maintenance of location Effects 0.000 description 4
- 229920000609 methyl cellulose Polymers 0.000 description 4
- 235000010981 methylcellulose Nutrition 0.000 description 4
- 239000001923 methylcellulose Substances 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 239000002726 nonnucleoside reverse transcriptase inhibitor Substances 0.000 description 4
- 239000003921 oil Substances 0.000 description 4
- 235000019198 oils Nutrition 0.000 description 4
- 230000036961 partial effect Effects 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 238000002962 plaque-reduction assay Methods 0.000 description 4
- 241000114864 ssRNA viruses Species 0.000 description 4
- 239000003381 stabilizer Substances 0.000 description 4
- 229960004231 thymalfasin Drugs 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- YKPQUSLRUFLVDA-UHFFFAOYSA-N $l^{2}-azanylmethane Chemical compound [NH]C YKPQUSLRUFLVDA-UHFFFAOYSA-N 0.000 description 3
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2r,3r,4s,5r,6s)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2s,3r,4s,5r,6r)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2r,3r,4s,5r,6r)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 3
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 229920000936 Agarose Polymers 0.000 description 3
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 3
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 3
- 241000416162 Astragalus gummifer Species 0.000 description 3
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- VWFCHDSQECPREK-LURJTMIESA-N Cidofovir Chemical compound NC=1C=CN(C[C@@H](CO)OCP(O)(O)=O)C(=O)N=1 VWFCHDSQECPREK-LURJTMIESA-N 0.000 description 3
- 230000004568 DNA-binding Effects 0.000 description 3
- 241000709661 Enterovirus Species 0.000 description 3
- 239000001856 Ethyl cellulose Substances 0.000 description 3
- 108010078049 Interferon alpha-2 Proteins 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 108020005196 Mitochondrial DNA Proteins 0.000 description 3
- 108091093105 Nuclear DNA Proteins 0.000 description 3
- 229940122313 Nucleoside reverse transcriptase inhibitor Drugs 0.000 description 3
- 108091034117 Oligonucleotide Proteins 0.000 description 3
- 241000709664 Picornaviridae Species 0.000 description 3
- 229930006000 Sucrose Natural products 0.000 description 3
- 210000001744 T-lymphocyte Anatomy 0.000 description 3
- 229920001615 Tragacanth Polymers 0.000 description 3
- WREGKURFCTUGRC-POYBYMJQSA-N Zalcitabine Chemical compound O=C1N=C(N)C=CN1[C@@H]1O[C@H](CO)CC1 WREGKURFCTUGRC-POYBYMJQSA-N 0.000 description 3
- MKUXAQIIEYXACX-UHFFFAOYSA-N aciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCO)C=N2 MKUXAQIIEYXACX-UHFFFAOYSA-N 0.000 description 3
- 239000000654 additive Substances 0.000 description 3
- 229940008201 allegra Drugs 0.000 description 3
- 230000003510 anti-fibrotic effect Effects 0.000 description 3
- 230000002790 anti-mutagenic effect Effects 0.000 description 3
- 239000003963 antioxidant agent Substances 0.000 description 3
- 235000006708 antioxidants Nutrition 0.000 description 3
- 239000007900 aqueous suspension Substances 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 230000003833 cell viability Effects 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 238000002512 chemotherapy Methods 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 230000002939 deleterious effect Effects 0.000 description 3
- 238000011161 development Methods 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 235000014113 dietary fatty acids Nutrition 0.000 description 3
- 125000002228 disulfide group Chemical group 0.000 description 3
- 239000002552 dosage form Substances 0.000 description 3
- 238000012377 drug delivery Methods 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- PEASPLKKXBYDKL-FXEVSJAOSA-N enfuvirtide Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(N)=O)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(C)=O)[C@@H](C)O)[C@@H](C)CC)C1=CN=CN1 PEASPLKKXBYDKL-FXEVSJAOSA-N 0.000 description 3
- 229920001249 ethyl cellulose Polymers 0.000 description 3
- 210000003527 eukaryotic cell Anatomy 0.000 description 3
- 239000000194 fatty acid Substances 0.000 description 3
- 229930195729 fatty acid Natural products 0.000 description 3
- 239000000796 flavoring agent Substances 0.000 description 3
- 235000013355 food flavoring agent Nutrition 0.000 description 3
- 230000006872 improvement Effects 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 230000002458 infectious effect Effects 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 229940057995 liquid paraffin Drugs 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 239000000314 lubricant Substances 0.000 description 3
- 210000004072 lung Anatomy 0.000 description 3
- 238000012423 maintenance Methods 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 239000000178 monomer Substances 0.000 description 3
- 231100000350 mutagenesis Toxicity 0.000 description 3
- 230000035772 mutation Effects 0.000 description 3
- 231100000252 nontoxic Toxicity 0.000 description 3
- 230000003000 nontoxic effect Effects 0.000 description 3
- 239000000346 nonvolatile oil Substances 0.000 description 3
- 210000004789 organ system Anatomy 0.000 description 3
- 230000003647 oxidation Effects 0.000 description 3
- 238000007254 oxidation reaction Methods 0.000 description 3
- 244000052769 pathogen Species 0.000 description 3
- 230000001717 pathogenic effect Effects 0.000 description 3
- XRQDFNLINLXZLB-CKIKVBCHSA-N peramivir Chemical compound CCC(CC)[C@H](NC(C)=O)[C@@H]1[C@H](O)[C@@H](C(O)=O)C[C@H]1NC(N)=N XRQDFNLINLXZLB-CKIKVBCHSA-N 0.000 description 3
- 229960001084 peramivir Drugs 0.000 description 3
- 230000005855 radiation Effects 0.000 description 3
- 230000009467 reduction Effects 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 229920006395 saturated elastomer Polymers 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 238000001179 sorption measurement Methods 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 239000008107 starch Substances 0.000 description 3
- 229940032147 starch Drugs 0.000 description 3
- 239000005720 sucrose Substances 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000014616 translation Effects 0.000 description 3
- PUPZLCDOIYMWBV-UHFFFAOYSA-N (+/-)-1,3-Butanediol Chemical compound CC(O)CCO PUPZLCDOIYMWBV-UHFFFAOYSA-N 0.000 description 2
- AOFUBOWZWQFQJU-SNOJBQEQSA-N (2r,3s,4s,5r)-2,5-bis(hydroxymethyl)oxolane-2,3,4-triol;(2s,3r,4s,5s,6r)-6-(hydroxymethyl)oxane-2,3,4,5-tetrol Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O.OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@@H]1O AOFUBOWZWQFQJU-SNOJBQEQSA-N 0.000 description 2
- CWVMWSZEMZOUPC-JUAXIXHSSA-N (3s,5s,8r,9s,10s,13s,14s,16r)-16-bromo-3-hydroxy-10,13-dimethyl-1,2,3,4,5,6,7,8,9,11,12,14,15,16-tetradecahydrocyclopenta[a]phenanthren-17-one Chemical compound C1[C@@H](O)CC[C@]2(C)[C@H]3CC[C@](C)(C([C@H](Br)C4)=O)[C@@H]4[C@@H]3CC[C@H]21 CWVMWSZEMZOUPC-JUAXIXHSSA-N 0.000 description 2
- IWUCXVSUMQZMFG-RGDLXGNYSA-N 1-[(2s,3s,4r,5s)-3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]-1,2,4-triazole-3-carboxamide Chemical compound N1=C(C(=O)N)N=CN1[C@@H]1[C@@H](O)[C@@H](O)[C@H](CO)O1 IWUCXVSUMQZMFG-RGDLXGNYSA-N 0.000 description 2
- ABEXEQSGABRUHS-UHFFFAOYSA-N 16-methylheptadecyl 16-methylheptadecanoate Chemical compound CC(C)CCCCCCCCCCCCCCCOC(=O)CCCCCCCCCCCCCCC(C)C ABEXEQSGABRUHS-UHFFFAOYSA-N 0.000 description 2
- XYLUJZDXASMNBC-UHFFFAOYSA-N 2-[3-(methylamino)propylamino]ethanethiol Chemical compound CNCCCNCCS XYLUJZDXASMNBC-UHFFFAOYSA-N 0.000 description 2
- CERZMXAJYMMUDR-QBTAGHCHSA-N 5-amino-3,5-dideoxy-D-glycero-D-galacto-non-2-ulopyranosonic acid Chemical compound N[C@@H]1[C@@H](O)CC(O)(C(O)=O)O[C@H]1[C@H](O)[C@H](O)CO CERZMXAJYMMUDR-QBTAGHCHSA-N 0.000 description 2
- GWNHAOBXDGOXRR-HJFSHJIFSA-N 9-[2-[[(2r,4s)-4-(3-chlorophenyl)-2-oxo-1,3,2$l^{5}-dioxaphosphinan-2-yl]methoxy]ethyl]purin-6-amine Chemical compound C1([C@@H]2CCO[P@](O2)(=O)COCCN2C=3N=CN=C(C=3N=C2)N)=CC=CC(Cl)=C1 GWNHAOBXDGOXRR-HJFSHJIFSA-N 0.000 description 2
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 2
- 108010019625 Atazanavir Sulfate Proteins 0.000 description 2
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- 102100025064 Cellular tumor antigen p53 Human genes 0.000 description 2
- 241000711573 Coronaviridae Species 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 2
- ZZZCUOFIHGPKAK-UHFFFAOYSA-N D-erythro-ascorbic acid Natural products OCC1OC(=O)C(O)=C1O ZZZCUOFIHGPKAK-UHFFFAOYSA-N 0.000 description 2
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 2
- SRBFZHDQGSBBOR-IOVATXLUSA-N D-xylopyranose Chemical compound O[C@@H]1COC(O)[C@H](O)[C@H]1O SRBFZHDQGSBBOR-IOVATXLUSA-N 0.000 description 2
- 102000003915 DNA Topoisomerases Human genes 0.000 description 2
- 108090000323 DNA Topoisomerases Proteins 0.000 description 2
- 230000005778 DNA damage Effects 0.000 description 2
- 231100000277 DNA damage Toxicity 0.000 description 2
- 229920002307 Dextran Polymers 0.000 description 2
- BXZVVICBKDXVGW-NKWVEPMBSA-N Didanosine Chemical compound O1[C@H](CO)CC[C@@H]1N1C(NC=NC2=O)=C2N=C1 BXZVVICBKDXVGW-NKWVEPMBSA-N 0.000 description 2
- XQSPYNMVSIKCOC-NTSWFWBYSA-N Emtricitabine Chemical compound C1=C(F)C(N)=NC(=O)N1[C@H]1O[C@@H](CO)SC1 XQSPYNMVSIKCOC-NTSWFWBYSA-N 0.000 description 2
- 108010032976 Enfuvirtide Proteins 0.000 description 2
- 241000991587 Enterovirus C Species 0.000 description 2
- ZZSNKZQZMQGXPY-UHFFFAOYSA-N Ethyl cellulose Chemical compound CCOCC1OC(OC)C(OCC)C(OCC)C1OC1C(O)C(O)C(OC)C(CO)O1 ZZSNKZQZMQGXPY-UHFFFAOYSA-N 0.000 description 2
- 108060003393 Granulin Proteins 0.000 description 2
- 241000700721 Hepatitis B virus Species 0.000 description 2
- NTYJJOPFIAHURM-UHFFFAOYSA-N Histamine Chemical compound NCCC1=CN=CN1 NTYJJOPFIAHURM-UHFFFAOYSA-N 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 2
- 208000001953 Hypotension Diseases 0.000 description 2
- XQFRJNBWHJMXHO-RRKCRQDMSA-N IDUR Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(I)=C1 XQFRJNBWHJMXHO-RRKCRQDMSA-N 0.000 description 2
- 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 2
- 208000002979 Influenza in Birds Diseases 0.000 description 2
- 101710200424 Inosine-5'-monophosphate dehydrogenase Proteins 0.000 description 2
- 102000000588 Interleukin-2 Human genes 0.000 description 2
- 108010002350 Interleukin-2 Proteins 0.000 description 2
- 241000764238 Isis Species 0.000 description 2
- KJHKTHWMRKYKJE-SUGCFTRWSA-N Kaletra Chemical compound N1([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=2C=CC=CC=2)NC(=O)COC=2C(=CC=CC=2C)C)CC=2C=CC=CC=2)CCCNC1=O KJHKTHWMRKYKJE-SUGCFTRWSA-N 0.000 description 2
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 2
- 206010053961 Mitochondrial toxicity Diseases 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- 201000003793 Myelodysplastic syndrome Diseases 0.000 description 2
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 2
- HSHXDCVZWHOWCS-UHFFFAOYSA-N N'-hexadecylthiophene-2-carbohydrazide Chemical compound CCCCCCCCCCCCCCCCNNC(=O)c1cccs1 HSHXDCVZWHOWCS-UHFFFAOYSA-N 0.000 description 2
- MBBZMMPHUWSWHV-BDVNFPICSA-N N-methylglucamine Chemical compound CNC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO MBBZMMPHUWSWHV-BDVNFPICSA-N 0.000 description 2
- 206010028813 Nausea Diseases 0.000 description 2
- 229940123424 Neuraminidase inhibitor Drugs 0.000 description 2
- 235000019483 Peanut oil Nutrition 0.000 description 2
- JNTOCHDNEULJHD-UHFFFAOYSA-N Penciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(CCC(CO)CO)C=N2 JNTOCHDNEULJHD-UHFFFAOYSA-N 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- 206010057190 Respiratory tract infections Diseases 0.000 description 2
- 108091027981 Response element Proteins 0.000 description 2
- IWUCXVSUMQZMFG-AFCXAGJDSA-N Ribavirin Chemical compound N1=C(C(=O)N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 IWUCXVSUMQZMFG-AFCXAGJDSA-N 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 2
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 2
- XNKLLVCARDGLGL-JGVFFNPUSA-N Stavudine Chemical compound O=C1NC(=O)C(C)=CN1[C@H]1C=C[C@@H](CO)O1 XNKLLVCARDGLGL-JGVFFNPUSA-N 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 102400000800 Thymosin alpha-1 Human genes 0.000 description 2
- HDOVUKNUBWVHOX-QMMMGPOBSA-N Valacyclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCOC(=O)[C@@H](N)C(C)C)C=N2 HDOVUKNUBWVHOX-QMMMGPOBSA-N 0.000 description 2
- WPVFJKSGQUFQAP-GKAPJAKFSA-N Valcyte Chemical compound N1C(N)=NC(=O)C2=C1N(COC(CO)COC(=O)[C@@H](N)C(C)C)C=N2 WPVFJKSGQUFQAP-GKAPJAKFSA-N 0.000 description 2
- 108010067390 Viral Proteins Proteins 0.000 description 2
- 229930003268 Vitamin C Natural products 0.000 description 2
- 229930003427 Vitamin E Natural products 0.000 description 2
- 206010047700 Vomiting Diseases 0.000 description 2
- HTJGLYIJVSDQAE-VWNXEWBOSA-N [(1s,6s,7s,8r,8ar)-1,7,8-trihydroxy-1,2,3,5,6,7,8,8a-octahydroindolizin-6-yl] butanoate Chemical compound O[C@H]1[C@H](O)[C@@H](OC(=O)CCC)CN2CC[C@H](O)[C@@H]21 HTJGLYIJVSDQAE-VWNXEWBOSA-N 0.000 description 2
- TVRCRTJYMVTEFS-ICGCPXGVSA-N [(2r,3r,4r,5r)-5-(4-amino-2-oxopyrimidin-1-yl)-4-hydroxy-2-(hydroxymethyl)-4-methyloxolan-3-yl] (2s)-2-amino-3-methylbutanoate Chemical compound C[C@@]1(O)[C@H](OC(=O)[C@@H](N)C(C)C)[C@@H](CO)O[C@H]1N1C(=O)N=C(N)C=C1 TVRCRTJYMVTEFS-ICGCPXGVSA-N 0.000 description 2
- 229960004748 abacavir Drugs 0.000 description 2
- MCGSCOLBFJQGHM-SCZZXKLOSA-N abacavir Chemical compound C=12N=CN([C@H]3C=C[C@@H](CO)C3)C2=NC(N)=NC=1NC1CC1 MCGSCOLBFJQGHM-SCZZXKLOSA-N 0.000 description 2
- YQNQNVDNTFHQSW-UHFFFAOYSA-N acetic acid [2-[[(5-nitro-2-thiazolyl)amino]-oxomethyl]phenyl] ester Chemical compound CC(=O)OC1=CC=CC=C1C(=O)NC1=NC=C([N+]([O-])=O)S1 YQNQNVDNTFHQSW-UHFFFAOYSA-N 0.000 description 2
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 2
- 229960004150 aciclovir Drugs 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- ORILYTVJVMAKLC-UHFFFAOYSA-N adamantane Chemical compound C1C(C2)CC3CC1CC2C3 ORILYTVJVMAKLC-UHFFFAOYSA-N 0.000 description 2
- WOZSCQDILHKSGG-UHFFFAOYSA-N adefovir depivoxil Chemical compound N1=CN=C2N(CCOCP(=O)(OCOC(=O)C(C)(C)C)OCOC(=O)C(C)(C)C)C=NC2=C1N WOZSCQDILHKSGG-UHFFFAOYSA-N 0.000 description 2
- 229960003205 adefovir dipivoxil Drugs 0.000 description 2
- 150000001299 aldehydes Chemical class 0.000 description 2
- 235000010443 alginic acid Nutrition 0.000 description 2
- 229920000615 alginic acid Polymers 0.000 description 2
- 239000000783 alginic acid Substances 0.000 description 2
- 229960001126 alginic acid Drugs 0.000 description 2
- 150000004781 alginic acids Chemical class 0.000 description 2
- 229940100198 alkylating agent Drugs 0.000 description 2
- 239000002168 alkylating agent Substances 0.000 description 2
- DKNWSYNQZKUICI-UHFFFAOYSA-N amantadine Chemical compound C1C(C2)CC3CC2CC1(N)C3 DKNWSYNQZKUICI-UHFFFAOYSA-N 0.000 description 2
- 150000001412 amines Chemical class 0.000 description 2
- 229960001830 amprenavir Drugs 0.000 description 2
- 230000003466 anti-cipated effect Effects 0.000 description 2
- 230000003474 anti-emetic effect Effects 0.000 description 2
- 230000000692 anti-sense effect Effects 0.000 description 2
- 239000002111 antiemetic agent Substances 0.000 description 2
- 239000004599 antimicrobial Substances 0.000 description 2
- 239000002246 antineoplastic agent Substances 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- PYMYPHUHKUWMLA-UHFFFAOYSA-N arabinose Natural products OCC(O)C(O)C(O)C=O PYMYPHUHKUWMLA-UHFFFAOYSA-N 0.000 description 2
- AXRYRYVKAWYZBR-GASGPIRDSA-N atazanavir Chemical compound C([C@H](NC(=O)[C@@H](NC(=O)OC)C(C)(C)C)[C@@H](O)CN(CC=1C=CC(=CC=1)C=1N=CC=CC=1)NC(=O)[C@@H](NC(=O)OC)C(C)(C)C)C1=CC=CC=C1 AXRYRYVKAWYZBR-GASGPIRDSA-N 0.000 description 2
- 206010064097 avian influenza Diseases 0.000 description 2
- 239000002585 base Substances 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 229960000686 benzalkonium chloride Drugs 0.000 description 2
- CADWTSSKOVRVJC-UHFFFAOYSA-N benzyl(dimethyl)azanium;chloride Chemical compound [Cl-].C[NH+](C)CC1=CC=CC=C1 CADWTSSKOVRVJC-UHFFFAOYSA-N 0.000 description 2
- SRBFZHDQGSBBOR-UHFFFAOYSA-N beta-D-Pyranose-Lyxose Natural products OC1COC(O)C(O)C1O SRBFZHDQGSBBOR-UHFFFAOYSA-N 0.000 description 2
- 229910000019 calcium carbonate Inorganic materials 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 229910000389 calcium phosphate Inorganic materials 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- BPKIGYQJPYCAOW-FFJTTWKXSA-I calcium;potassium;disodium;(2s)-2-hydroxypropanoate;dichloride;dihydroxide;hydrate Chemical compound O.[OH-].[OH-].[Na+].[Na+].[Cl-].[Cl-].[K+].[Ca+2].C[C@H](O)C([O-])=O BPKIGYQJPYCAOW-FFJTTWKXSA-I 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 125000002091 cationic group Chemical group 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 210000000349 chromosome Anatomy 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- GBBJCSTXCAQSSJ-XQXXSGGOSA-N clevudine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1[C@H](F)[C@@H](O)[C@H](CO)O1 GBBJCSTXCAQSSJ-XQXXSGGOSA-N 0.000 description 2
- 239000003086 colorant Substances 0.000 description 2
- 239000013078 crystal Substances 0.000 description 2
- 230000000120 cytopathologic effect Effects 0.000 description 2
- 229940127089 cytotoxic agent Drugs 0.000 description 2
- WHBIGIKBNXZKFE-UHFFFAOYSA-N delavirdine Chemical compound CC(C)NC1=CC=CN=C1N1CCN(C(=O)C=2NC3=CC=C(NS(C)(=O)=O)C=C3C=2)CC1 WHBIGIKBNXZKFE-UHFFFAOYSA-N 0.000 description 2
- 239000003599 detergent Substances 0.000 description 2
- 238000009792 diffusion process Methods 0.000 description 2
- 230000003292 diminished effect Effects 0.000 description 2
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 2
- 239000002270 dispersing agent Substances 0.000 description 2
- 231100000673 dose–response relationship Toxicity 0.000 description 2
- 241001492478 dsDNA viruses, no RNA stage Species 0.000 description 2
- XPOQHMRABVBWPR-ZDUSSCGKSA-N efavirenz Chemical compound C([C@]1(C2=CC(Cl)=CC=C2NC(=O)O1)C(F)(F)F)#CC1CC1 XPOQHMRABVBWPR-ZDUSSCGKSA-N 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- QDGZDCVAUDNJFG-FXQIFTODSA-N entecavir (anhydrous) Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@H]1C[C@H](O)[C@@H](CO)C1=C QDGZDCVAUDNJFG-FXQIFTODSA-N 0.000 description 2
- CAYJBRBGZBCZKO-BHGBQCOSSA-N ethyl (e,4s)-4-[[(2r,5s)-2-[(4-fluorophenyl)methyl]-6-methyl-5-[(5-methyl-1,2-oxazole-3-carbonyl)amino]-4-oxoheptanoyl]amino]-5-[(3s)-2-oxopyrrolidin-3-yl]pent-2-enoate Chemical compound C([C@@H](/C=C/C(=O)OCC)NC(=O)[C@@H](CC(=O)[C@@H](NC(=O)C1=NOC(C)=C1)C(C)C)CC=1C=CC(F)=CC=1)[C@@H]1CCNC1=O CAYJBRBGZBCZKO-BHGBQCOSSA-N 0.000 description 2
- 235000019325 ethyl cellulose Nutrition 0.000 description 2
- 229940098617 ethyol Drugs 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- GGXKWVWZWMLJEH-UHFFFAOYSA-N famcyclovir Chemical compound N1=C(N)N=C2N(CCC(COC(=O)C)COC(C)=O)C=NC2=C1 GGXKWVWZWMLJEH-UHFFFAOYSA-N 0.000 description 2
- 150000004665 fatty acids Chemical class 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 235000003599 food sweetener Nutrition 0.000 description 2
- WIGCFUFOHFEKBI-UHFFFAOYSA-N gamma-tocopherol Natural products CC(C)CCCC(C)CCCC(C)CCCC1CCC2C(C)C(O)C(C)C(C)C2O1 WIGCFUFOHFEKBI-UHFFFAOYSA-N 0.000 description 2
- IRSCQMHQWWYFCW-UHFFFAOYSA-N ganciclovir Chemical compound O=C1NC(N)=NC2=C1N=CN2COC(CO)CO IRSCQMHQWWYFCW-UHFFFAOYSA-N 0.000 description 2
- 239000007903 gelatin capsule Substances 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 150000002334 glycols Chemical class 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 208000005252 hepatitis A Diseases 0.000 description 2
- 125000000623 heterocyclic group Chemical group 0.000 description 2
- 229960001340 histamine Drugs 0.000 description 2
- PPZMYIBUHIPZOS-UHFFFAOYSA-N histamine dihydrochloride Chemical compound Cl.Cl.NCCC1=CN=CN1 PPZMYIBUHIPZOS-UHFFFAOYSA-N 0.000 description 2
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 2
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 2
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 2
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 2
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 2
- 230000036543 hypotension Effects 0.000 description 2
- 238000005417 image-selected in vivo spectroscopy Methods 0.000 description 2
- DOUYETYNHWVLEO-UHFFFAOYSA-N imiquimod Chemical compound C1=CC=CC2=C3N(CC(C)C)C=NC3=C(N)N=C21 DOUYETYNHWVLEO-UHFFFAOYSA-N 0.000 description 2
- 239000002955 immunomodulating agent Substances 0.000 description 2
- 229940121354 immunomodulator Drugs 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 238000000099 in vitro assay Methods 0.000 description 2
- 230000002779 inactivation Effects 0.000 description 2
- CBVCZFGXHXORBI-PXQQMZJSSA-N indinavir Chemical compound C([C@H](N(CC1)C[C@@H](O)C[C@@H](CC=2C=CC=CC=2)C(=O)N[C@H]2C3=CC=CC=C3C[C@H]2O)C(=O)NC(C)(C)C)N1CC1=CC=CN=C1 CBVCZFGXHXORBI-PXQQMZJSSA-N 0.000 description 2
- 229940090438 infergen Drugs 0.000 description 2
- 239000004615 ingredient Substances 0.000 description 2
- 238000012739 integrated shape imaging system Methods 0.000 description 2
- 108010010648 interferon alfacon-1 Proteins 0.000 description 2
- 108010042414 interferon gamma-1b Proteins 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 230000005865 ionizing radiation Effects 0.000 description 2
- 125000000468 ketone group Chemical group 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 239000008101 lactose Substances 0.000 description 2
- 229960001375 lactose Drugs 0.000 description 2
- JTEGQNOMFQHVDC-NKWVEPMBSA-N lamivudine Chemical compound O=C1N=C(N)C=CN1[C@H]1O[C@@H](CO)SC1 JTEGQNOMFQHVDC-NKWVEPMBSA-N 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 239000012533 medium component Substances 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- 239000002480 mineral oil Substances 0.000 description 2
- 235000010446 mineral oil Nutrition 0.000 description 2
- 231100000296 mitochondrial toxicity Toxicity 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 238000000465 moulding Methods 0.000 description 2
- 230000008693 nausea Effects 0.000 description 2
- SYJXFKPQNSDJLI-HKEUSBCWSA-N neamine Chemical compound N[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@H](N)C[C@@H]1N SYJXFKPQNSDJLI-HKEUSBCWSA-N 0.000 description 2
- QAGYKUNXZHXKMR-HKWSIXNMSA-N nelfinavir Chemical compound CC1=C(O)C=CC=C1C(=O)N[C@H]([C@H](O)CN1[C@@H](C[C@@H]2CCCC[C@@H]2C1)C(=O)NC(C)(C)C)CSC1=CC=CC=C1 QAGYKUNXZHXKMR-HKWSIXNMSA-N 0.000 description 2
- CERZMXAJYMMUDR-UHFFFAOYSA-N neuraminic acid Natural products NC1C(O)CC(O)(C(O)=O)OC1C(O)C(O)CO CERZMXAJYMMUDR-UHFFFAOYSA-N 0.000 description 2
- 239000002547 new drug Substances 0.000 description 2
- VSZGPKBBMSAYNT-RRFJBIMHSA-N oseltamivir Chemical compound CCOC(=O)C1=C[C@@H](OC(CC)CC)[C@H](NC(C)=O)[C@@H](N)C1 VSZGPKBBMSAYNT-RRFJBIMHSA-N 0.000 description 2
- 239000000312 peanut oil Substances 0.000 description 2
- 108010092853 peginterferon alfa-2a Proteins 0.000 description 2
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 150000003904 phospholipids Chemical class 0.000 description 2
- ZJAOAACCNHFJAH-UHFFFAOYSA-N phosphonoformic acid Chemical compound OC(=O)P(O)(O)=O ZJAOAACCNHFJAH-UHFFFAOYSA-N 0.000 description 2
- 239000002504 physiological saline solution Substances 0.000 description 2
- 230000007505 plaque formation Effects 0.000 description 2
- 230000036470 plasma concentration Effects 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- 230000001124 posttranscriptional effect Effects 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 230000002335 preservative effect Effects 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- NCDNCNXCDXHOMX-XGKFQTDJSA-N ritonavir Chemical compound N([C@@H](C(C)C)C(=O)N[C@H](C[C@H](O)[C@H](CC=1C=CC=CC=1)NC(=O)OCC=1SC=NC=1)CC=1C=CC=CC=1)C(=O)N(C)CC1=CSC(C(C)C)=N1 NCDNCNXCDXHOMX-XGKFQTDJSA-N 0.000 description 2
- 229960004641 rituximab Drugs 0.000 description 2
- 210000003079 salivary gland Anatomy 0.000 description 2
- QWAXKHKRTORLEM-UGJKXSETSA-N saquinavir Chemical compound C([C@@H]([C@H](O)CN1C[C@H]2CCCC[C@H]2C[C@H]1C(=O)NC(C)(C)C)NC(=O)[C@H](CC(N)=O)NC(=O)C=1N=C2C=CC=CC2=CC=1)C1=CC=CC=C1 QWAXKHKRTORLEM-UGJKXSETSA-N 0.000 description 2
- 239000002911 sialidase inhibitor Substances 0.000 description 2
- 235000015424 sodium Nutrition 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 2
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 2
- 239000007901 soft capsule Substances 0.000 description 2
- 239000000600 sorbitol Substances 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 235000000346 sugar Nutrition 0.000 description 2
- 150000008163 sugars Chemical class 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 239000003765 sweetening agent Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 239000006188 syrup Substances 0.000 description 2
- 235000020357 syrup Nutrition 0.000 description 2
- 239000000454 talc Substances 0.000 description 2
- 229910052623 talc Inorganic materials 0.000 description 2
- IQFYYKKMVGJFEH-CSMHCCOUSA-N telbivudine Chemical compound O=C1NC(=O)C(C)=CN1[C@H]1O[C@@H](CO)[C@H](O)C1 IQFYYKKMVGJFEH-CSMHCCOUSA-N 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 238000011287 therapeutic dose Methods 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 229940021747 therapeutic vaccine Drugs 0.000 description 2
- SUJUHGSWHZTSEU-FYBSXPHGSA-N tipranavir Chemical compound C([C@@]1(CCC)OC(=O)C([C@H](CC)C=2C=C(NS(=O)(=O)C=3N=CC(=CC=3)C(F)(F)F)C=CC=2)=C(O)C1)CC1=CC=CC=C1 SUJUHGSWHZTSEU-FYBSXPHGSA-N 0.000 description 2
- 235000010487 tragacanth Nutrition 0.000 description 2
- 239000000196 tragacanth Substances 0.000 description 2
- 229940116362 tragacanth Drugs 0.000 description 2
- 230000032258 transport Effects 0.000 description 2
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 2
- 241001515965 unidentified phage Species 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 238000012800 visualization Methods 0.000 description 2
- 235000019154 vitamin C Nutrition 0.000 description 2
- 239000011718 vitamin C Substances 0.000 description 2
- 235000019165 vitamin E Nutrition 0.000 description 2
- 229940046009 vitamin E Drugs 0.000 description 2
- 239000011709 vitamin E Substances 0.000 description 2
- 230000008673 vomiting Effects 0.000 description 2
- 239000000080 wetting agent Substances 0.000 description 2
- 229960000523 zalcitabine Drugs 0.000 description 2
- IVWWFWFVSWOTLP-YVZVNANGSA-N (3'as,4r,7'as)-2,2,2',2'-tetramethylspiro[1,3-dioxolane-4,6'-4,7a-dihydro-3ah-[1,3]dioxolo[4,5-c]pyran]-7'-one Chemical compound C([C@@H]1OC(O[C@@H]1C1=O)(C)C)O[C@]21COC(C)(C)O2 IVWWFWFVSWOTLP-YVZVNANGSA-N 0.000 description 1
- ASWBNKHCZGQVJV-UHFFFAOYSA-N (3-hexadecanoyloxy-2-hydroxypropyl) 2-(trimethylazaniumyl)ethyl phosphate Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(O)COP([O-])(=O)OCC[N+](C)(C)C ASWBNKHCZGQVJV-UHFFFAOYSA-N 0.000 description 1
- FELGMEQIXOGIFQ-CYBMUJFWSA-N (3r)-9-methyl-3-[(2-methylimidazol-1-yl)methyl]-2,3-dihydro-1h-carbazol-4-one Chemical compound CC1=NC=CN1C[C@@H]1C(=O)C(C=2C(=CC=CC=2)N2C)=C2CC1 FELGMEQIXOGIFQ-CYBMUJFWSA-N 0.000 description 1
- SCVHJVCATBPIHN-SJCJKPOMSA-N (3s)-3-[[(2s)-2-[[2-(2-tert-butylanilino)-2-oxoacetyl]amino]propanoyl]amino]-4-oxo-5-(2,3,5,6-tetrafluorophenoxy)pentanoic acid Chemical compound N([C@@H](C)C(=O)N[C@@H](CC(O)=O)C(=O)COC=1C(=C(F)C=C(F)C=1F)F)C(=O)C(=O)NC1=CC=CC=C1C(C)(C)C SCVHJVCATBPIHN-SJCJKPOMSA-N 0.000 description 1
- WRIDQFICGBMAFQ-UHFFFAOYSA-N (E)-8-Octadecenoic acid Natural products CCCCCCCCCC=CCCCCCCC(O)=O WRIDQFICGBMAFQ-UHFFFAOYSA-N 0.000 description 1
- ZORQXIQZAOLNGE-UHFFFAOYSA-N 1,1-difluorocyclohexane Chemical compound FC1(F)CCCCC1 ZORQXIQZAOLNGE-UHFFFAOYSA-N 0.000 description 1
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 1
- XQFRJNBWHJMXHO-UHFFFAOYSA-N 1-[4-hydroxy-5-(hydroxymethyl)oxolan-2-yl]-5-iodopyrimidine-2,4-dione Chemical compound C1C(O)C(CO)OC1N1C(=O)NC(=O)C(I)=C1 XQFRJNBWHJMXHO-UHFFFAOYSA-N 0.000 description 1
- JLPULHDHAOZNQI-ZTIMHPMXSA-N 1-hexadecanoyl-2-(9Z,12Z-octadecadienoyl)-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCC\C=C/C\C=C/CCCCC JLPULHDHAOZNQI-ZTIMHPMXSA-N 0.000 description 1
- 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 description 1
- OWEGMIWEEQEYGQ-UHFFFAOYSA-N 100676-05-9 Natural products OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(OC2C(OC(O)C(O)C2O)CO)O1 OWEGMIWEEQEYGQ-UHFFFAOYSA-N 0.000 description 1
- VBRUONUESYTIDA-UHFFFAOYSA-N 2-(4-fluorophenyl)-6-(methanesulfonamido)-n-methyl-5-propan-2-yloxy-1-benzofuran-3-carboxamide Chemical compound O1C2=CC(NS(C)(=O)=O)=C(OC(C)C)C=C2C(C(=O)NC)=C1C1=CC=C(F)C=C1 VBRUONUESYTIDA-UHFFFAOYSA-N 0.000 description 1
- SPCKHVPPRJWQRZ-UHFFFAOYSA-N 2-benzhydryloxy-n,n-dimethylethanamine;2-hydroxypropane-1,2,3-tricarboxylic acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O.C=1C=CC=CC=1C(OCCN(C)C)C1=CC=CC=C1 SPCKHVPPRJWQRZ-UHFFFAOYSA-N 0.000 description 1
- RQCBPOPQTLHDFC-UHFFFAOYSA-N 2-phenyl-1,3-oxazole Chemical compound C1=COC(C=2C=CC=CC=2)=N1 RQCBPOPQTLHDFC-UHFFFAOYSA-N 0.000 description 1
- LQJBNNIYVWPHFW-UHFFFAOYSA-N 20:1omega9c fatty acid Natural products CCCCCCCCCCC=CCCCCCCCC(O)=O LQJBNNIYVWPHFW-UHFFFAOYSA-N 0.000 description 1
- CDOUZKKFHVEKRI-UHFFFAOYSA-N 3-bromo-n-[(prop-2-enoylamino)methyl]propanamide Chemical compound BrCCC(=O)NCNC(=O)C=C CDOUZKKFHVEKRI-UHFFFAOYSA-N 0.000 description 1
- HSBKFSPNDWWPSL-VDTYLAMSSA-N 4-amino-5-fluoro-1-[(2s,5r)-5-(hydroxymethyl)-2,5-dihydrofuran-2-yl]pyrimidin-2-one Chemical compound C1=C(F)C(N)=NC(=O)N1[C@@H]1C=C[C@H](CO)O1 HSBKFSPNDWWPSL-VDTYLAMSSA-N 0.000 description 1
- WTDWVLJJJOTABN-UHFFFAOYSA-N 5-cyclopropyl-2-(4-fluorophenyl)-6-[(2-hydroxyethyl)(methylsulfonyl)amino]-n-methyl-1-benzofuran-3-carboxamide Chemical compound C1=C2C(C(=O)NC)=C(C=3C=CC(F)=CC=3)OC2=CC(N(CCO)S(C)(=O)=O)=C1C1CC1 WTDWVLJJJOTABN-UHFFFAOYSA-N 0.000 description 1
- VHRSUDSXCMQTMA-PJHHCJLFSA-N 6alpha-methylprednisolone Chemical compound C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)CO)CC[C@H]21 VHRSUDSXCMQTMA-PJHHCJLFSA-N 0.000 description 1
- HCAJQHYUCKICQH-VPENINKCSA-N 8-Oxo-7,8-dihydro-2'-deoxyguanosine Chemical compound C1=2NC(N)=NC(=O)C=2NC(=O)N1[C@H]1C[C@H](O)[C@@H](CO)O1 HCAJQHYUCKICQH-VPENINKCSA-N 0.000 description 1
- QSBYPNXLFMSGKH-UHFFFAOYSA-N 9-Heptadecensaeure Natural products CCCCCCCC=CCCCCCCCC(O)=O QSBYPNXLFMSGKH-UHFFFAOYSA-N 0.000 description 1
- 108091092742 A-DNA Proteins 0.000 description 1
- 229930000680 A04AD01 - Scopolamine Natural products 0.000 description 1
- 244000215068 Acacia senegal Species 0.000 description 1
- 235000006491 Acacia senegal Nutrition 0.000 description 1
- NIXOWILDQLNWCW-UHFFFAOYSA-N Acrylic acid Chemical compound OC(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-N 0.000 description 1
- 241000321096 Adenoides Species 0.000 description 1
- 206010001258 Adenoviral infections Diseases 0.000 description 1
- 102100036774 Afamin Human genes 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 239000005995 Aluminium silicate Substances 0.000 description 1
- 235000003911 Arachis Nutrition 0.000 description 1
- 244000105624 Arachis hypogaea Species 0.000 description 1
- AXRYRYVKAWYZBR-UHFFFAOYSA-N Atazanavir Natural products C=1C=C(C=2N=CC=CC=2)C=CC=1CN(NC(=O)C(NC(=O)OC)C(C)(C)C)CC(O)C(NC(=O)C(NC(=O)OC)C(C)(C)C)CC1=CC=CC=C1 AXRYRYVKAWYZBR-UHFFFAOYSA-N 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- GUBGYTABKSRVRQ-DCSYEGIMSA-N Beta-Lactose Chemical compound OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-DCSYEGIMSA-N 0.000 description 1
- WVJXFHQMGZSQDL-UHFFFAOYSA-N CNCCCNCC[S+]=P(O)(O)O Chemical compound CNCCCNCC[S+]=P(O)(O)O WVJXFHQMGZSQDL-UHFFFAOYSA-N 0.000 description 1
- QAGYKUNXZHXKMR-UHFFFAOYSA-N CPD000469186 Natural products CC1=C(O)C=CC=C1C(=O)NC(C(O)CN1C(CC2CCCCC2C1)C(=O)NC(C)(C)C)CSC1=CC=CC=C1 QAGYKUNXZHXKMR-UHFFFAOYSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- JDVVGAQPNNXQDW-WCMLQCRESA-N Castanospermine Natural products O[C@H]1[C@@H](O)[C@H]2[C@@H](O)CCN2C[C@H]1O JDVVGAQPNNXQDW-WCMLQCRESA-N 0.000 description 1
- JDVVGAQPNNXQDW-TVNFTVLESA-N Castinospermine Chemical compound C1[C@H](O)[C@@H](O)[C@H](O)[C@H]2[C@@H](O)CCN21 JDVVGAQPNNXQDW-TVNFTVLESA-N 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 108091060290 Chromatid Proteins 0.000 description 1
- FKLJPTJMIBLJAV-UHFFFAOYSA-N Compound IV Chemical compound O1N=C(C)C=C1CCCCCCCOC1=CC=C(C=2OCCN=2)C=C1 FKLJPTJMIBLJAV-UHFFFAOYSA-N 0.000 description 1
- 206010010741 Conjunctivitis Diseases 0.000 description 1
- 229920002261 Corn starch Polymers 0.000 description 1
- 241000700626 Cowpox virus Species 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 1
- 102000016559 DNA Primase Human genes 0.000 description 1
- 108010092681 DNA Primase Proteins 0.000 description 1
- 230000004543 DNA replication Effects 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 241000199914 Dinophyceae Species 0.000 description 1
- 206010059866 Drug resistance Diseases 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 238000008157 ELISA kit Methods 0.000 description 1
- XPOQHMRABVBWPR-UHFFFAOYSA-N Efavirenz Natural products O1C(=O)NC2=CC=C(Cl)C=C2C1(C(F)(F)F)C#CC1CC1 XPOQHMRABVBWPR-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 102100031780 Endonuclease Human genes 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- 101710121417 Envelope glycoprotein Proteins 0.000 description 1
- 241000701533 Escherichia virus T4 Species 0.000 description 1
- 208000000461 Esophageal Neoplasms Diseases 0.000 description 1
- 108010008165 Etanercept Proteins 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- IAYPIBMASNFSPL-UHFFFAOYSA-N Ethylene oxide Chemical compound C1CO1 IAYPIBMASNFSPL-UHFFFAOYSA-N 0.000 description 1
- PIICEJLVQHRZGT-UHFFFAOYSA-N Ethylenediamine Chemical class NCCN PIICEJLVQHRZGT-UHFFFAOYSA-N 0.000 description 1
- 241000710781 Flaviviridae Species 0.000 description 1
- 229930091371 Fructose Natural products 0.000 description 1
- 239000005715 Fructose Substances 0.000 description 1
- RFSUNEUAIZKAJO-ARQDHWQXSA-N Fructose Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O RFSUNEUAIZKAJO-ARQDHWQXSA-N 0.000 description 1
- 108010044091 Globulins Proteins 0.000 description 1
- 102000006395 Globulins Human genes 0.000 description 1
- 108010081687 Glutamate-cysteine ligase Proteins 0.000 description 1
- 102100039696 Glutamate-cysteine ligase catalytic subunit Human genes 0.000 description 1
- 229920000084 Gum arabic Polymers 0.000 description 1
- 241000606768 Haemophilus influenzae Species 0.000 description 1
- 206010061192 Haemorrhagic fever Diseases 0.000 description 1
- 241000711549 Hepacivirus C Species 0.000 description 1
- 241000700739 Hepadnaviridae Species 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 241000709721 Hepatovirus A Species 0.000 description 1
- 241000700586 Herpesviridae Species 0.000 description 1
- 102100032742 Histone-lysine N-methyltransferase SETD2 Human genes 0.000 description 1
- 101000654725 Homo sapiens Histone-lysine N-methyltransferase SETD2 Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N Hydrogen bromide Chemical compound Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- 239000004354 Hydroxyethyl cellulose Substances 0.000 description 1
- 229920000663 Hydroxyethyl cellulose Polymers 0.000 description 1
- STECJAGHUSJQJN-GAUPFVANSA-N Hyoscine Natural products C1([C@H](CO)C(=O)OC2C[C@@H]3N([C@H](C2)[C@@H]2[C@H]3O2)C)=CC=CC=C1 STECJAGHUSJQJN-GAUPFVANSA-N 0.000 description 1
- 108010091358 Hypoxanthine Phosphoribosyltransferase Proteins 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 108010054698 Interferon Alfa-n3 Proteins 0.000 description 1
- 108010005716 Interferon beta-1a Proteins 0.000 description 1
- 108010047761 Interferon-alpha Proteins 0.000 description 1
- 102000006992 Interferon-alpha Human genes 0.000 description 1
- 102000003814 Interleukin-10 Human genes 0.000 description 1
- 108090000174 Interleukin-10 Proteins 0.000 description 1
- PWWVAXIEGOYWEE-UHFFFAOYSA-N Isophenergan Chemical compound C1=CC=C2N(CC(C)N(C)C)C3=CC=CC=C3SC2=C1 PWWVAXIEGOYWEE-UHFFFAOYSA-N 0.000 description 1
- 102100023408 KH domain-containing, RNA-binding, signal transduction-associated protein 1 Human genes 0.000 description 1
- 101710094958 KH domain-containing, RNA-binding, signal transduction-associated protein 1 Proteins 0.000 description 1
- 229930194542 Keto Natural products 0.000 description 1
- LKDRXBCSQODPBY-AMVSKUEXSA-N L-(-)-Sorbose Chemical compound OCC1(O)OC[C@H](O)[C@@H](O)[C@@H]1O LKDRXBCSQODPBY-AMVSKUEXSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical class NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 1
- 240000007472 Leucaena leucocephala Species 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 241000282560 Macaca mulatta Species 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- GUBGYTABKSRVRQ-PICCSMPSSA-N Maltose Natural products 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-PICCSMPSSA-N 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 201000005505 Measles Diseases 0.000 description 1
- OCJYIGYOJCODJL-UHFFFAOYSA-N Meclizine Chemical compound CC1=CC=CC(CN2CCN(CC2)C(C=2C=CC=CC=2)C=2C=CC(Cl)=CC=2)=C1 OCJYIGYOJCODJL-UHFFFAOYSA-N 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 102100037510 Metallothionein-1E Human genes 0.000 description 1
- 241000711386 Mumps virus Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 229940121948 Muscarinic receptor antagonist Drugs 0.000 description 1
- STECJAGHUSJQJN-UHFFFAOYSA-N N-Methyl-scopolamin Natural products C1C(C2C3O2)N(C)C3CC1OC(=O)C(CO)C1=CC=CC=C1 STECJAGHUSJQJN-UHFFFAOYSA-N 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- 229930193140 Neomycin Natural products 0.000 description 1
- 241001292005 Nidovirales Species 0.000 description 1
- 206010030155 Oesophageal carcinoma Diseases 0.000 description 1
- 240000007817 Olea europaea Species 0.000 description 1
- 239000005642 Oleic acid Substances 0.000 description 1
- ZQPPMHVWECSIRJ-UHFFFAOYSA-N Oleic acid Natural products CCCCCCCCC=CCCCCCCCC(O)=O ZQPPMHVWECSIRJ-UHFFFAOYSA-N 0.000 description 1
- 102000016304 Origin Recognition Complex Human genes 0.000 description 1
- 108010067244 Origin Recognition Complex Proteins 0.000 description 1
- 101710111048 Para-Rep C9 Proteins 0.000 description 1
- 241000711504 Paramyxoviridae Species 0.000 description 1
- 241000701945 Parvoviridae Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 102000006335 Phosphate-Binding Proteins Human genes 0.000 description 1
- 108010058514 Phosphate-Binding Proteins Proteins 0.000 description 1
- 244000038594 Phyllanthus urinaria Species 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 229920000954 Polyglycolide Polymers 0.000 description 1
- 241000700625 Poxviridae Species 0.000 description 1
- 101710150344 Protein Rev Proteins 0.000 description 1
- 101800001554 RNA-directed RNA polymerase Proteins 0.000 description 1
- 101710118046 RNA-directed RNA polymerase Proteins 0.000 description 1
- 241000711798 Rabies lyssavirus Species 0.000 description 1
- 108010012770 Rebetron Proteins 0.000 description 1
- 241000702247 Reoviridae Species 0.000 description 1
- 108020005091 Replication Origin Proteins 0.000 description 1
- 102000018780 Replication Protein A Human genes 0.000 description 1
- 108010027643 Replication Protein A Proteins 0.000 description 1
- 108010022187 Rev peptide Proteins 0.000 description 1
- 241000711931 Rhabdoviridae Species 0.000 description 1
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 1
- OZBDFBJXRJWNAV-UHFFFAOYSA-N Rimantadine hydrochloride Chemical compound Cl.C1C(C2)CC3CC2CC1(C(N)C)C3 OZBDFBJXRJWNAV-UHFFFAOYSA-N 0.000 description 1
- NCDNCNXCDXHOMX-UHFFFAOYSA-N Ritonavir Natural products C=1C=CC=CC=1CC(NC(=O)OCC=1SC=NC=1)C(O)CC(CC=1C=CC=CC=1)NC(=O)C(C(C)C)NC(=O)N(C)CC1=CSC(C(C)C)=N1 NCDNCNXCDXHOMX-UHFFFAOYSA-N 0.000 description 1
- 241000710799 Rubella virus Species 0.000 description 1
- 241000315672 SARS coronavirus Species 0.000 description 1
- 102000012479 Serine Proteases Human genes 0.000 description 1
- 108010022999 Serine Proteases Proteins 0.000 description 1
- 229940122055 Serine protease inhibitor Drugs 0.000 description 1
- 101710102218 Serine protease inhibitor Proteins 0.000 description 1
- 208000001203 Smallpox Diseases 0.000 description 1
- 229920002125 Sokalan® Polymers 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 235000015125 Sterculia urens Nutrition 0.000 description 1
- 240000001058 Sterculia urens Species 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- 108010022394 Threonine synthase Proteins 0.000 description 1
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Natural products O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 1
- 102000005497 Thymidylate Synthase Human genes 0.000 description 1
- SUJUHGSWHZTSEU-UHFFFAOYSA-N Tipranavir Natural products C1C(O)=C(C(CC)C=2C=C(NS(=O)(=O)C=3N=CC(=CC=3)C(F)(F)F)C=CC=2)C(=O)OC1(CCC)CCC1=CC=CC=C1 SUJUHGSWHZTSEU-UHFFFAOYSA-N 0.000 description 1
- 241000710924 Togaviridae Species 0.000 description 1
- 101710183280 Topoisomerase Proteins 0.000 description 1
- 101800001690 Transmembrane protein gp41 Proteins 0.000 description 1
- 239000007983 Tris buffer Chemical class 0.000 description 1
- 241000223105 Trypanosoma brucei Species 0.000 description 1
- 206010054094 Tumour necrosis Diseases 0.000 description 1
- 102000007537 Type II DNA Topoisomerases Human genes 0.000 description 1
- 108010046308 Type II DNA Topoisomerases Proteins 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 241000870995 Variola Species 0.000 description 1
- 241000700647 Variola virus Species 0.000 description 1
- OIRDTQYFTABQOQ-UHTZMRCNSA-N Vidarabine Chemical compound C1=NC=2C(N)=NC=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@@H]1O OIRDTQYFTABQOQ-UHTZMRCNSA-N 0.000 description 1
- 108020000999 Viral RNA Proteins 0.000 description 1
- 241000710886 West Nile virus Species 0.000 description 1
- 241000120645 Yellow fever virus group Species 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- ZWELIJXAKMASLK-UGKPPGOTSA-N [(2r,3r,4r,5r)-4-acetyloxy-5-(5-amino-2-oxo-[1,3]thiazolo[4,5-d]pyrimidin-3-yl)-2-(hydroxymethyl)oxolan-3-yl] acetate Chemical compound CC(=O)O[C@@H]1[C@H](OC(=O)C)[C@@H](CO)O[C@H]1N1C(=O)SC2=CN=C(N)N=C21 ZWELIJXAKMASLK-UGKPPGOTSA-N 0.000 description 1
- VFCYZPOEGWLYRM-QCZKYFFMSA-N [(2s,3r,5s)-5-(4-amino-2-oxopyrimidin-1-yl)-2-(hydroxymethyl)oxolan-3-yl] (2s)-2-amino-3-methylbutanoate Chemical compound O1[C@@H](CO)[C@H](OC(=O)[C@@H](N)C(C)C)C[C@H]1N1C(=O)N=C(N)C=C1 VFCYZPOEGWLYRM-QCZKYFFMSA-N 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 235000010489 acacia gum Nutrition 0.000 description 1
- 229940099550 actimmune Drugs 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 239000011149 active material Substances 0.000 description 1
- 230000009056 active transport Effects 0.000 description 1
- 125000002015 acyclic group Chemical group 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 210000002534 adenoid Anatomy 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 235000010419 agar Nutrition 0.000 description 1
- 108010080374 albuferon Proteins 0.000 description 1
- 150000001298 alcohols Chemical class 0.000 description 1
- 229940060265 aldara Drugs 0.000 description 1
- 229940054685 alinia Drugs 0.000 description 1
- 150000001338 aliphatic hydrocarbons Chemical class 0.000 description 1
- 229910052783 alkali metal Inorganic materials 0.000 description 1
- 229910052784 alkaline earth metal Inorganic materials 0.000 description 1
- 125000005907 alkyl ester group Chemical group 0.000 description 1
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-PHYPRBDBSA-N alpha-D-galactose Chemical compound OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@H]1O WQZGKKKJIJFFOK-PHYPRBDBSA-N 0.000 description 1
- 108010090535 alpha-albumin Proteins 0.000 description 1
- 230000004075 alteration Effects 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
- 235000012211 aluminium silicate Nutrition 0.000 description 1
- 229960003805 amantadine Drugs 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 150000001409 amidines Chemical class 0.000 description 1
- 239000002647 aminoglycoside antibiotic agent Substances 0.000 description 1
- 229960004977 anhydrous lactose Drugs 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 239000002260 anti-inflammatory agent Substances 0.000 description 1
- 229940121363 anti-inflammatory agent Drugs 0.000 description 1
- 230000001139 anti-pruritic effect Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 229940125715 antihistaminic agent Drugs 0.000 description 1
- 239000000739 antihistaminic agent Substances 0.000 description 1
- 229940082988 antihypertensives serotonin antagonists Drugs 0.000 description 1
- 229960005475 antiinfective agent Drugs 0.000 description 1
- 230000003078 antioxidant effect Effects 0.000 description 1
- 239000003908 antipruritic agent Substances 0.000 description 1
- 239000003903 antiretrovirus agent Substances 0.000 description 1
- 239000003420 antiserotonin agent Substances 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 229940030139 aptivus Drugs 0.000 description 1
- PYMYPHUHKUWMLA-WDCZJNDASA-N arabinose Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)C=O PYMYPHUHKUWMLA-WDCZJNDASA-N 0.000 description 1
- 125000006615 aromatic heterocyclic group Chemical group 0.000 description 1
- 239000003212 astringent agent Substances 0.000 description 1
- 229960003277 atazanavir Drugs 0.000 description 1
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 229940002637 baraclude Drugs 0.000 description 1
- OGBUMNBNEWYMNJ-UHFFFAOYSA-N batilol Chemical class CCCCCCCCCCCCCCCCCCOCC(O)CO OGBUMNBNEWYMNJ-UHFFFAOYSA-N 0.000 description 1
- 239000000440 bentonite Substances 0.000 description 1
- 229910000278 bentonite Inorganic materials 0.000 description 1
- SVPXDRXYRYOSEX-UHFFFAOYSA-N bentoquatam Chemical compound O.O=[Si]=O.O=[Al]O[Al]=O SVPXDRXYRYOSEX-UHFFFAOYSA-N 0.000 description 1
- JUHORIMYRDESRB-UHFFFAOYSA-N benzathine Chemical class C=1C=CC=CC=1CNCCNCC1=CC=CC=C1 JUHORIMYRDESRB-UHFFFAOYSA-N 0.000 description 1
- 229960001950 benzethonium chloride Drugs 0.000 description 1
- UREZNYTWGJKWBI-UHFFFAOYSA-M benzethonium chloride Chemical compound [Cl-].C1=CC(C(C)(C)CC(C)(C)C)=CC=C1OCCOCC[N+](C)(C)CC1=CC=CC=C1 UREZNYTWGJKWBI-UHFFFAOYSA-M 0.000 description 1
- SDQJTWBNWQABLE-UHFFFAOYSA-N benzoylene urea Natural products C1=CC=C2C(=O)NC(=O)NC2=C1 SDQJTWBNWQABLE-UHFFFAOYSA-N 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QUYVBRFLSA-N beta-maltose Chemical compound OC[C@H]1O[C@H](O[C@H]2[C@H](O)[C@@H](O)[C@H](O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@@H]1O GUBGYTABKSRVRQ-QUYVBRFLSA-N 0.000 description 1
- 239000003613 bile acid Substances 0.000 description 1
- 238000012925 biological evaluation Methods 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 230000003592 biomimetic effect Effects 0.000 description 1
- 229920001222 biopolymer Polymers 0.000 description 1
- 229960000517 boceprevir Drugs 0.000 description 1
- LHHCSNFAOIFYRV-DOVBMPENSA-N boceprevir Chemical compound O=C([C@@H]1[C@@H]2[C@@H](C2(C)C)CN1C(=O)[C@@H](NC(=O)NC(C)(C)C)C(C)(C)C)NC(C(=O)C(N)=O)CC1CCC1 LHHCSNFAOIFYRV-DOVBMPENSA-N 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 239000010839 body fluid Substances 0.000 description 1
- KGBXLFKZBHKPEV-UHFFFAOYSA-N boric acid Chemical compound OB(O)O KGBXLFKZBHKPEV-UHFFFAOYSA-N 0.000 description 1
- 239000004327 boric acid Substances 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 229960001705 buclizine Drugs 0.000 description 1
- MOYGZHXDRJNJEP-UHFFFAOYSA-N buclizine Chemical compound C1=CC(C(C)(C)C)=CC=C1CN1CCN(C(C=2C=CC=CC=2)C=2C=CC(Cl)=CC=2)CC1 MOYGZHXDRJNJEP-UHFFFAOYSA-N 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 235000010216 calcium carbonate Nutrition 0.000 description 1
- 159000000007 calcium salts Chemical class 0.000 description 1
- 229910000394 calcium triphosphate Inorganic materials 0.000 description 1
- PMDQGYMGQKTCSX-HQROKSDRSA-L calcium;[(2r,3s)-1-[(4-aminophenyl)sulfonyl-(2-methylpropyl)amino]-3-[[(3s)-oxolan-3-yl]oxycarbonylamino]-4-phenylbutan-2-yl] phosphate Chemical compound [Ca+2].C([C@@H]([C@H](OP([O-])([O-])=O)CN(CC(C)C)S(=O)(=O)C=1C=CC(N)=CC=1)NC(=O)O[C@@H]1COCC1)C1=CC=CC=C1 PMDQGYMGQKTCSX-HQROKSDRSA-L 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 210000000234 capsid Anatomy 0.000 description 1
- 229940077731 carbohydrate nutrients Drugs 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 229960001631 carbomer Drugs 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 239000003183 carcinogenic agent Substances 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 239000004359 castor oil Substances 0.000 description 1
- 235000019438 castor oil Nutrition 0.000 description 1
- 229950003414 celgosivir Drugs 0.000 description 1
- 230000022131 cell cycle Effects 0.000 description 1
- 230000006369 cell cycle progression Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 229940107810 cellcept Drugs 0.000 description 1
- 241000902900 cellular organisms Species 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 229920001429 chelating resin Polymers 0.000 description 1
- UKTAZPQNNNJVKR-KJGYPYNMSA-N chembl2368925 Chemical compound C1=CC=C2C(C(O[C@@H]3C[C@@H]4C[C@H]5C[C@@H](N4CC5=O)C3)=O)=CNC2=C1 UKTAZPQNNNJVKR-KJGYPYNMSA-N 0.000 description 1
- 230000002113 chemopreventative effect Effects 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- VDANGULDQQJODZ-UHFFFAOYSA-N chloroprocaine Chemical class CCN(CC)CCOC(=O)C1=CC=C(N)C=C1Cl VDANGULDQQJODZ-UHFFFAOYSA-N 0.000 description 1
- 229960002023 chloroprocaine Drugs 0.000 description 1
- ZPEIMTDSQAKGNT-UHFFFAOYSA-N chlorpromazine Chemical compound C1=C(Cl)C=C2N(CCCN(C)C)C3=CC=CC=C3SC2=C1 ZPEIMTDSQAKGNT-UHFFFAOYSA-N 0.000 description 1
- 229960001076 chlorpromazine Drugs 0.000 description 1
- OEYIOHPDSNJKLS-UHFFFAOYSA-N choline Chemical class C[N+](C)(C)CCO OEYIOHPDSNJKLS-UHFFFAOYSA-N 0.000 description 1
- 229960001231 choline Drugs 0.000 description 1
- 239000000812 cholinergic antagonist Substances 0.000 description 1
- 210000004756 chromatid Anatomy 0.000 description 1
- 229960000724 cidofovir Drugs 0.000 description 1
- PJZPDFUUXKKDNB-KNINVFKUSA-N ciluprevir Chemical compound N([C@@H]1C(=O)N2[C@H](C(N[C@@]3(C[C@H]3\C=C/CCCCC1)C(O)=O)=O)C[C@H](C2)OC=1C2=CC=C(C=C2N=C(C=1)C=1N=C(NC(C)C)SC=1)OC)C(=O)OC1CCCC1 PJZPDFUUXKKDNB-KNINVFKUSA-N 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 229960005338 clevudine Drugs 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 238000004891 communication Methods 0.000 description 1
- 230000009918 complex formation Effects 0.000 description 1
- 239000008139 complexing agent Substances 0.000 description 1
- 239000007891 compressed tablet Substances 0.000 description 1
- 238000007906 compression Methods 0.000 description 1
- 230000006835 compression Effects 0.000 description 1
- 230000001010 compromised effect Effects 0.000 description 1
- 239000007859 condensation product Substances 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 230000008094 contradictory effect Effects 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 239000008120 corn starch Substances 0.000 description 1
- 229940099112 cornstarch Drugs 0.000 description 1
- 229940088900 crixivan Drugs 0.000 description 1
- 210000001100 crypt cell Anatomy 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 229960003564 cyclizine Drugs 0.000 description 1
- UVKZSORBKUEBAZ-UHFFFAOYSA-N cyclizine Chemical compound C1CN(C)CCN1C(C=1C=CC=CC=1)C1=CC=CC=C1 UVKZSORBKUEBAZ-UHFFFAOYSA-N 0.000 description 1
- 229960004397 cyclophosphamide Drugs 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 229940124569 cytoprotecting agent Drugs 0.000 description 1
- 229940087451 cytovene Drugs 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 229960005319 delavirdine Drugs 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 229940075882 denavir Drugs 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 229960003957 dexamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- ANCLJVISBRWUTR-UHFFFAOYSA-N diaminophosphinic acid Chemical compound NP(N)(O)=O ANCLJVISBRWUTR-UHFFFAOYSA-N 0.000 description 1
- 229960002656 didanosine Drugs 0.000 description 1
- ZBCBWPMODOFKDW-UHFFFAOYSA-N diethanolamine Chemical class OCCNCCO ZBCBWPMODOFKDW-UHFFFAOYSA-N 0.000 description 1
- 229940043237 diethanolamine Drugs 0.000 description 1
- 230000009699 differential effect Effects 0.000 description 1
- MZDOIJOUFRQXHC-UHFFFAOYSA-N dimenhydrinate Chemical compound O=C1N(C)C(=O)N(C)C2=NC(Cl)=N[C]21.C=1C=CC=CC=1C(OCCN(C)C)C1=CC=CC=C1 MZDOIJOUFRQXHC-UHFFFAOYSA-N 0.000 description 1
- 229960004993 dimenhydrinate Drugs 0.000 description 1
- 235000019329 dioctyl sodium sulphosuccinate Nutrition 0.000 description 1
- LRCFXGAMWKDGLA-UHFFFAOYSA-N dioxosilane;hydrate Chemical compound O.O=[Si]=O LRCFXGAMWKDGLA-UHFFFAOYSA-N 0.000 description 1
- 229960000520 diphenhydramine Drugs 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- NOPFSRXAKWQILS-UHFFFAOYSA-N docosan-1-ol Chemical compound CCCCCCCCCCCCCCCCCCCCCCO NOPFSRXAKWQILS-UHFFFAOYSA-N 0.000 description 1
- 229960003413 dolasetron Drugs 0.000 description 1
- 239000003210 dopamine receptor blocking agent Substances 0.000 description 1
- RMEDXOLNCUSCGS-UHFFFAOYSA-N droperidol Chemical compound C1=CC(F)=CC=C1C(=O)CCCN1CC=C(N2C(NC3=CC=CC=C32)=O)CC1 RMEDXOLNCUSCGS-UHFFFAOYSA-N 0.000 description 1
- 229960000394 droperidol Drugs 0.000 description 1
- 238000001647 drug administration Methods 0.000 description 1
- 238000009513 drug distribution Methods 0.000 description 1
- 230000002183 duodenal effect Effects 0.000 description 1
- 210000001198 duodenum Anatomy 0.000 description 1
- 239000000975 dye Substances 0.000 description 1
- 229960003804 efavirenz Drugs 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 229950006528 elvucitabine Drugs 0.000 description 1
- 229950000234 emricasan Drugs 0.000 description 1
- 229960000366 emtricitabine Drugs 0.000 description 1
- 229940001018 emtriva Drugs 0.000 description 1
- 229940073621 enbrel Drugs 0.000 description 1
- 229960002062 enfuvirtide Drugs 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 229960000980 entecavir Drugs 0.000 description 1
- 108700004025 env Genes Proteins 0.000 description 1
- 230000009088 enzymatic function Effects 0.000 description 1
- 108700020302 erbB-2 Genes Proteins 0.000 description 1
- 210000003617 erythrocyte membrane Anatomy 0.000 description 1
- 201000004101 esophageal cancer Diseases 0.000 description 1
- 229960004667 ethyl cellulose Drugs 0.000 description 1
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 description 1
- 229940012017 ethylenediamine Drugs 0.000 description 1
- 229960004396 famciclovir Drugs 0.000 description 1
- 239000010685 fatty oil Substances 0.000 description 1
- LLYJISDUHFXOHK-GOCONZMPSA-N ferroptocide Chemical compound C[C@@H]1CC[C@@]23C[C@@H](C(=O)[C@]2([C@@]1([C@@H](C[C@H]([C@@H]3C)C4=CCN5C(=O)N(C(=O)N5C4)C6=CC=CC=C6)OC(=O)CCl)C)O)O LLYJISDUHFXOHK-GOCONZMPSA-N 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 229940028864 flumadine Drugs 0.000 description 1
- 238000001506 fluorescence spectroscopy Methods 0.000 description 1
- 229960001447 fomivirsen Drugs 0.000 description 1
- XCWFZHPEARLXJI-UHFFFAOYSA-N fomivirsen Chemical compound C1C(N2C3=C(C(NC(N)=N3)=O)N=C2)OC(CO)C1OP(O)(=S)OCC1OC(N(C)C(=O)\N=C(\N)C=C)CC1OP(O)(=S)OCC1OC(N2C3=C(C(NC(N)=N3)=O)N=C2)CC1OP(O)(=S)OCC1OC(N2C(NC(=O)C(C)=C2)=O)CC1OP(O)(=S)OCC1OC(N2C(NC(=O)C(C)=C2)=O)CC1OP(O)(=S)OCC1OC(N2C(NC(=O)C(C)=C2)=O)CC1OP(O)(=S)OCC1OC(N2C3=C(C(NC(N)=N3)=O)N=C2)CC1OP(O)(=S)OCC1OC(N2C(N=C(N)C=C2)=O)CC1OP(O)(=S)OCC(C(C1)OP(S)(=O)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)O)OC1N1C=C(C)C(=O)NC1=O XCWFZHPEARLXJI-UHFFFAOYSA-N 0.000 description 1
- DLKYYJFLRUUGHJ-SSJCJZGYSA-A fomivirsen sodium Chemical compound [Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].[Na+].O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C3=C(C(NC(N)=N3)=O)N=C2)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C(NC(=O)C(C)=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C3=C(C(NC(N)=N3)=O)N=C2)COP([S-])(=O)O[C@@H]2[C@H](O[C@H](C2)N2C(N=C(N)C=C2)=O)COP([O-])(=S)O[C@@H]2[C@H](O[C@H](C2)N2C3=C(C(NC(N)=N3)=O)N=C2)CO)[C@@H](OP([O-])(=S)OC[C@@H]2[C@H](C[C@@H](O2)N2C3=C(C(NC(N)=N3)=O)N=C2)OP([O-])(=S)OC[C@@H]2[C@H](C[C@@H](O2)N2C(N=C(N)C=C2)=O)OP([O-])(=S)OC[C@@H]2[C@H](C[C@@H](O2)N2C3=C(C(NC(N)=N3)=O)N=C2)O)C1 DLKYYJFLRUUGHJ-SSJCJZGYSA-A 0.000 description 1
- 229960003142 fosamprenavir Drugs 0.000 description 1
- MLBVMOWEQCZNCC-OEMFJLHTSA-N fosamprenavir Chemical compound C([C@@H]([C@H](OP(O)(O)=O)CN(CC(C)C)S(=O)(=O)C=1C=CC(N)=CC=1)NC(=O)O[C@@H]1COCC1)C1=CC=CC=C1 MLBVMOWEQCZNCC-OEMFJLHTSA-N 0.000 description 1
- 229960005102 foscarnet Drugs 0.000 description 1
- 229940108452 foscavir Drugs 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- PGBHMTALBVVCIT-VCIWKGPPSA-N framycetin Chemical compound N[C@@H]1[C@@H](O)[C@H](O)[C@H](CN)O[C@@H]1O[C@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](N)C[C@@H](N)[C@@H]2O)O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CN)O2)N)O[C@@H]1CO PGBHMTALBVVCIT-VCIWKGPPSA-N 0.000 description 1
- 229960003704 framycetin Drugs 0.000 description 1
- 229910021485 fumed silica Inorganic materials 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 229940099052 fuzeon Drugs 0.000 description 1
- 229930182830 galactose Natural products 0.000 description 1
- 229960002963 ganciclovir Drugs 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000003304 gavage Methods 0.000 description 1
- 231100000025 genetic toxicology Toxicity 0.000 description 1
- 230000001738 genotoxic effect Effects 0.000 description 1
- 108010050669 glucosidase I Proteins 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- YQEMORVAKMFKLG-UHFFFAOYSA-N glycerine monostearate Natural products CCCCCCCCCCCCCCCCCC(=O)OC(CO)CO YQEMORVAKMFKLG-UHFFFAOYSA-N 0.000 description 1
- SVUQHVRAGMNPLW-UHFFFAOYSA-N glycerol monostearate Natural products CCCCCCCCCCCCCCCCC(=O)OCC(O)CO SVUQHVRAGMNPLW-UHFFFAOYSA-N 0.000 description 1
- ZEMPKEQAKRGZGQ-XOQCFJPHSA-N glycerol triricinoleate Natural products CCCCCC[C@@H](O)CC=CCCCCCCCC(=O)OC[C@@H](COC(=O)CCCCCCCC=CC[C@@H](O)CCCCCC)OC(=O)CCCCCCCC=CC[C@H](O)CCCCCC ZEMPKEQAKRGZGQ-XOQCFJPHSA-N 0.000 description 1
- 229940075507 glyceryl monostearate Drugs 0.000 description 1
- MFWNKCLOYSRHCJ-BTTYYORXSA-N granisetron Chemical compound C1=CC=C2C(C(=O)N[C@H]3C[C@H]4CCC[C@@H](C3)N4C)=NN(C)C2=C1 MFWNKCLOYSRHCJ-BTTYYORXSA-N 0.000 description 1
- 229960003727 granisetron Drugs 0.000 description 1
- 239000003979 granulating agent Substances 0.000 description 1
- 239000007902 hard capsule Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 150000002373 hemiacetals Chemical class 0.000 description 1
- 230000011132 hemopoiesis Effects 0.000 description 1
- 208000006454 hepatitis Diseases 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 229940060415 hepatitis b immune globulin Drugs 0.000 description 1
- 125000005842 heteroatom Chemical group 0.000 description 1
- 229960004931 histamine dihydrochloride Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 102000011749 human hepatitis C immune globulin Human genes 0.000 description 1
- 108010062138 human hepatitis C immune globulin Proteins 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 235000019447 hydroxyethyl cellulose Nutrition 0.000 description 1
- 229940071676 hydroxypropylcellulose Drugs 0.000 description 1
- 229960004716 idoxuridine Drugs 0.000 description 1
- 210000003405 ileum Anatomy 0.000 description 1
- 229960002751 imiquimod Drugs 0.000 description 1
- 230000007124 immune defense Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000002584 immunomodulator Effects 0.000 description 1
- 239000003018 immunosuppressive agent Substances 0.000 description 1
- 229940124589 immunosuppressive drug Drugs 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 229960001936 indinavir Drugs 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 239000003999 initiator Substances 0.000 description 1
- 229960003521 interferon alfa-2a Drugs 0.000 description 1
- 229960003507 interferon alfa-2b Drugs 0.000 description 1
- 229940076144 interleukin-10 Drugs 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 229940088976 invirase Drugs 0.000 description 1
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 description 1
- QXJSBBXBKPUZAA-UHFFFAOYSA-N isooleic acid Natural products CCCCCCCC=CCCCCCCCCC(O)=O QXJSBBXBKPUZAA-UHFFFAOYSA-N 0.000 description 1
- 125000001972 isopentyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 210000001630 jejunum Anatomy 0.000 description 1
- 229940112586 kaletra Drugs 0.000 description 1
- NLYAJNPCOHFWQQ-UHFFFAOYSA-N kaolin Chemical compound O.O.O=[Al]O[Si](=O)O[Si](=O)O[Al]=O NLYAJNPCOHFWQQ-UHFFFAOYSA-N 0.000 description 1
- 229960001627 lamivudine Drugs 0.000 description 1
- 238000002386 leaching Methods 0.000 description 1
- VHOGYURTWQBHIL-UHFFFAOYSA-N leflunomide Chemical class O1N=CC(C(=O)NC=2C=CC(=CC=2)C(F)(F)F)=C1C VHOGYURTWQBHIL-UHFFFAOYSA-N 0.000 description 1
- 229940113354 lexiva Drugs 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 239000003589 local anesthetic agent Substances 0.000 description 1
- 229960005015 local anesthetics Drugs 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 229960004525 lopinavir Drugs 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- ZLNQQNXFFQJAID-UHFFFAOYSA-L magnesium carbonate Chemical compound [Mg+2].[O-]C([O-])=O ZLNQQNXFFQJAID-UHFFFAOYSA-L 0.000 description 1
- 239000001095 magnesium carbonate Substances 0.000 description 1
- 229910000021 magnesium carbonate Inorganic materials 0.000 description 1
- 229940037627 magnesium lauryl sulfate Drugs 0.000 description 1
- 159000000003 magnesium salts Chemical class 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- HBNDBUATLJAUQM-UHFFFAOYSA-L magnesium;dodecyl sulfate Chemical compound [Mg+2].CCCCCCCCCCCCOS([O-])(=O)=O.CCCCCCCCCCCCOS([O-])(=O)=O HBNDBUATLJAUQM-UHFFFAOYSA-L 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 229960001855 mannitol Drugs 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 229960001474 meclozine Drugs 0.000 description 1
- 238000002483 medication Methods 0.000 description 1
- 229960003194 meglumine Drugs 0.000 description 1
- 229910021645 metal ion Inorganic materials 0.000 description 1
- 229960002900 methylcellulose Drugs 0.000 description 1
- 229960004584 methylprednisolone Drugs 0.000 description 1
- TTWJBBZEZQICBI-UHFFFAOYSA-N metoclopramide Chemical compound CCN(CC)CCNC(=O)C1=CC(Cl)=C(N)C=C1OC TTWJBBZEZQICBI-UHFFFAOYSA-N 0.000 description 1
- 229960004503 metoclopramide Drugs 0.000 description 1
- 239000000693 micelle Substances 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 239000004530 micro-emulsion Substances 0.000 description 1
- 239000004005 microsphere Substances 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 239000007932 molded tablet Substances 0.000 description 1
- 239000001788 mono and diglycerides of fatty acids Substances 0.000 description 1
- 210000004980 monocyte derived macrophage Anatomy 0.000 description 1
- 238000011294 monotherapeutic Methods 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 239000003471 mutagenic agent Substances 0.000 description 1
- 231100000707 mutagenic chemical Toxicity 0.000 description 1
- 230000003505 mutagenic effect Effects 0.000 description 1
- RTGDFNSFWBGLEC-SYZQJQIISA-N mycophenolate mofetil Chemical compound COC1=C(C)C=2COC(=O)C=2C(O)=C1C\C=C(/C)CCC(=O)OCCN1CCOCC1 RTGDFNSFWBGLEC-SYZQJQIISA-N 0.000 description 1
- 229960004866 mycophenolate mofetil Drugs 0.000 description 1
- 125000004108 n-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001280 n-hexyl group Chemical group C(CCCCC)* 0.000 description 1
- 125000000740 n-pentyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000004123 n-propyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])* 0.000 description 1
- 125000001624 naphthyl group Chemical group 0.000 description 1
- 229930014626 natural product Natural products 0.000 description 1
- 229960000884 nelfinavir Drugs 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- NQDJXKOVJZTUJA-UHFFFAOYSA-N nevirapine Chemical compound C12=NC=CC=C2C(=O)NC=2C(C)=CC=NC=2N1C1CC1 NQDJXKOVJZTUJA-UHFFFAOYSA-N 0.000 description 1
- KUZYDHCVYKUFKF-LPLPFJSBSA-N niruriside Chemical compound CC(=O)O[C@@H]1[C@@H](O)[C@H](OC(C)=O)[C@@H](COC(=O)C)O[C@H]1O[C@@]1(COC(C)=O)[C@@H](OC(=O)\C=C\C=2C=CC=CC=2)[C@H](O)[C@@H](COC(=O)\C=C\C=2C=CC=CC=2)O1 KUZYDHCVYKUFKF-LPLPFJSBSA-N 0.000 description 1
- 229940042402 non-nucleoside reverse transcriptase inhibitor Drugs 0.000 description 1
- 231100001221 nontumorigenic Toxicity 0.000 description 1
- 229940072250 norvir Drugs 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- 210000004940 nucleus Anatomy 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- 239000012053 oil suspension Substances 0.000 description 1
- ZQPPMHVWECSIRJ-KTKRTIGZSA-N oleic acid Chemical compound CCCCCCCC\C=C/CCCCCCCC(O)=O ZQPPMHVWECSIRJ-KTKRTIGZSA-N 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- 231100000590 oncogenic Toxicity 0.000 description 1
- 230000002246 oncogenic effect Effects 0.000 description 1
- 229960005343 ondansetron Drugs 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 229960003752 oseltamivir Drugs 0.000 description 1
- PGZUMBJQJWIWGJ-ONAKXNSWSA-N oseltamivir phosphate Chemical compound OP(O)(O)=O.CCOC(=O)C1=C[C@@H](OC(CC)CC)[C@H](NC(C)=O)[C@@H](N)C1 PGZUMBJQJWIWGJ-ONAKXNSWSA-N 0.000 description 1
- 229940045681 other alkylating agent in atc Drugs 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- 108700025694 p53 Genes Proteins 0.000 description 1
- 239000006179 pH buffering agent Substances 0.000 description 1
- 230000009057 passive transport Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229940002988 pegasys Drugs 0.000 description 1
- 108010092851 peginterferon alfa-2b Proteins 0.000 description 1
- 229960003931 peginterferon alfa-2b Drugs 0.000 description 1
- 229960001179 penciclovir Drugs 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- RFWLACFDYFIVMC-UHFFFAOYSA-D pentacalcium;[oxido(phosphonatooxy)phosphoryl] phosphate Chemical compound [Ca+2].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[O-]P([O-])(=O)OP([O-])(=O)OP([O-])([O-])=O.[O-]P([O-])(=O)OP([O-])(=O)OP([O-])([O-])=O RFWLACFDYFIVMC-UHFFFAOYSA-D 0.000 description 1
- 210000005259 peripheral blood Anatomy 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- 150000003021 phthalic acid derivatives Chemical class 0.000 description 1
- 239000000049 pigment Substances 0.000 description 1
- 239000004014 plasticizer Substances 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- 229960000471 pleconaril Drugs 0.000 description 1
- KQOXLKOJHVFTRN-UHFFFAOYSA-N pleconaril Chemical compound O1N=C(C)C=C1CCCOC1=C(C)C=C(C=2N=C(ON=2)C(F)(F)F)C=C1C KQOXLKOJHVFTRN-UHFFFAOYSA-N 0.000 description 1
- 229920001606 poly(lactic acid-co-glycolic acid) Polymers 0.000 description 1
- 239000010695 polyglycol Substances 0.000 description 1
- 229920000151 polyglycol Polymers 0.000 description 1
- 239000004633 polyglycolic acid Substances 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 239000000244 polyoxyethylene sorbitan monooleate Substances 0.000 description 1
- 229920001451 polypropylene glycol Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 150000004804 polysaccharides Chemical class 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 229940068965 polysorbates Drugs 0.000 description 1
- 239000004810 polytetrafluoroethylene Substances 0.000 description 1
- 229920001343 polytetrafluoroethylene Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 159000000001 potassium salts Chemical class 0.000 description 1
- 229950001452 pradefovir Drugs 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical class CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- WIKYUJGCLQQFNW-UHFFFAOYSA-N prochlorperazine Chemical compound C1CN(C)CCN1CCCN1C2=CC(Cl)=CC=C2SC2=CC=CC=C21 WIKYUJGCLQQFNW-UHFFFAOYSA-N 0.000 description 1
- 229960003111 prochlorperazine Drugs 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 229960003910 promethazine Drugs 0.000 description 1
- 230000004845 protein aggregation Effects 0.000 description 1
- 230000006916 protein interaction Effects 0.000 description 1
- 150000003230 pyrimidines Chemical class 0.000 description 1
- 239000001397 quillaja saponaria molina bark Substances 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- 238000011127 radiochemotherapy Methods 0.000 description 1
- 230000003537 radioprotector Effects 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 229940053146 rebetol Drugs 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 229940107904 reyataz Drugs 0.000 description 1
- 230000000630 rising effect Effects 0.000 description 1
- 229960000311 ritonavir Drugs 0.000 description 1
- 229950007656 rupintrivir Drugs 0.000 description 1
- PCFWUYJHCYCQKP-QMMMGPOBSA-N s-[2-[[(2r)-2-acetamido-3-sulfanylpropanoyl]amino]ethyl] ethanethioate Chemical class CC(=O)N[C@@H](CS)C(=O)NCCSC(C)=O PCFWUYJHCYCQKP-QMMMGPOBSA-N 0.000 description 1
- 229930182490 saponin Natural products 0.000 description 1
- 150000007949 saponins Chemical class 0.000 description 1
- 229960001852 saquinavir Drugs 0.000 description 1
- 230000002000 scavenging effect Effects 0.000 description 1
- STECJAGHUSJQJN-FWXGHANASA-N scopolamine Chemical compound C1([C@@H](CO)C(=O)O[C@H]2C[C@@H]3N([C@H](C2)[C@@H]2[C@H]3O2)C)=CC=CC=C1 STECJAGHUSJQJN-FWXGHANASA-N 0.000 description 1
- 229960002646 scopolamine Drugs 0.000 description 1
- 125000002914 sec-butyl group Chemical group [H]C([H])([H])C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 239000003001 serine protease inhibitor Substances 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 238000009097 single-agent therapy Methods 0.000 description 1
- HELHAJAZNSDZJO-OLXYHTOASA-L sodium L-tartrate Chemical compound [Na+].[Na+].[O-]C(=O)[C@H](O)[C@@H](O)C([O-])=O HELHAJAZNSDZJO-OLXYHTOASA-L 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- APSBXTVYXVQYAB-UHFFFAOYSA-M sodium docusate Chemical group [Na+].CCCCC(CC)COC(=O)CC(S([O-])(=O)=O)C(=O)OCC(CC)CCCC APSBXTVYXVQYAB-UHFFFAOYSA-M 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 239000008109 sodium starch glycolate Substances 0.000 description 1
- 229920003109 sodium starch glycolate Polymers 0.000 description 1
- 229940079832 sodium starch glycolate Drugs 0.000 description 1
- 239000001433 sodium tartrate Substances 0.000 description 1
- 229960002167 sodium tartrate Drugs 0.000 description 1
- 235000011004 sodium tartrates Nutrition 0.000 description 1
- MIXCUJKCXRNYFM-UHFFFAOYSA-M sodium;diiodomethanesulfonate;n-propyl-n-[2-(2,4,6-trichlorophenoxy)ethyl]imidazole-1-carboxamide Chemical compound [Na+].[O-]S(=O)(=O)C(I)I.C1=CN=CN1C(=O)N(CCC)CCOC1=C(Cl)C=C(Cl)C=C1Cl MIXCUJKCXRNYFM-UHFFFAOYSA-M 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 235000011069 sorbitan monooleate Nutrition 0.000 description 1
- 239000001593 sorbitan monooleate Substances 0.000 description 1
- 229940035049 sorbitan monooleate Drugs 0.000 description 1
- 229940083466 soybean lecithin Drugs 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 229960001203 stavudine Drugs 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 150000005846 sugar alcohols Polymers 0.000 description 1
- BDHFUVZGWQCTTF-UHFFFAOYSA-M sulfonate Chemical compound [O-]S(=O)=O BDHFUVZGWQCTTF-UHFFFAOYSA-M 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 229940054565 sustiva Drugs 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 230000035488 systolic blood pressure Effects 0.000 description 1
- 239000007916 tablet composition Substances 0.000 description 1
- 229940061367 tamiflu Drugs 0.000 description 1
- NHKZSTHOYNWEEZ-AFCXAGJDSA-N taribavirin Chemical compound N1=C(C(=N)N)N=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](CO)O1 NHKZSTHOYNWEEZ-AFCXAGJDSA-N 0.000 description 1
- 229950006081 taribavirin Drugs 0.000 description 1
- 229960002935 telaprevir Drugs 0.000 description 1
- 229960005311 telbivudine Drugs 0.000 description 1
- SGOIRFVFHAKUTI-ZCFIWIBFSA-N tenofovir (anhydrous) Chemical compound N1=CN=C2N(C[C@@H](C)OCP(O)(O)=O)C=NC2=C1N SGOIRFVFHAKUTI-ZCFIWIBFSA-N 0.000 description 1
- 125000000999 tert-butyl group Chemical group [H]C([H])([H])C(*)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 229940126585 therapeutic drug Drugs 0.000 description 1
- RTKIYNMVFMVABJ-UHFFFAOYSA-L thimerosal Chemical compound [Na+].CC[Hg]SC1=CC=CC=C1C([O-])=O RTKIYNMVFMVABJ-UHFFFAOYSA-L 0.000 description 1
- 229940033663 thimerosal Drugs 0.000 description 1
- 229960000838 tipranavir Drugs 0.000 description 1
- 229940044655 toll-like receptor 9 agonist Drugs 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- VSQQQLOSPVPRAZ-RRKCRQDMSA-N trifluridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(C(F)(F)F)=C1 VSQQQLOSPVPRAZ-RRKCRQDMSA-N 0.000 description 1
- FEZBIKUBAYAZIU-UHFFFAOYSA-N trimethobenzamide Chemical compound COC1=C(OC)C(OC)=CC(C(=O)NCC=2C=CC(OCCN(C)C)=CC=2)=C1 FEZBIKUBAYAZIU-UHFFFAOYSA-N 0.000 description 1
- 229960004161 trimethobenzamide Drugs 0.000 description 1
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 1
- DFHAXXVZCFXGOQ-UHFFFAOYSA-K trisodium phosphonoformate Chemical compound [Na+].[Na+].[Na+].[O-]C(=O)P([O-])([O-])=O DFHAXXVZCFXGOQ-UHFFFAOYSA-K 0.000 description 1
- UXQDWARBDDDTKG-UHFFFAOYSA-N tromantadine Chemical compound C1C(C2)CC3CC2CC1(NC(=O)COCCN(C)C)C3 UXQDWARBDDDTKG-UHFFFAOYSA-N 0.000 description 1
- 229960000832 tromantadine Drugs 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 229940093257 valacyclovir Drugs 0.000 description 1
- 229940010343 valcyte Drugs 0.000 description 1
- 229960002149 valganciclovir Drugs 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 229950002810 valopicitabine Drugs 0.000 description 1
- 229950002819 valtorcitabine Drugs 0.000 description 1
- 229940108442 valtrex Drugs 0.000 description 1
- 239000012178 vegetable wax Substances 0.000 description 1
- 229940023080 viracept Drugs 0.000 description 1
- 230000017613 viral reproduction Effects 0.000 description 1
- 210000000605 viral structure Anatomy 0.000 description 1
- 210000002845 virion Anatomy 0.000 description 1
- 229940032699 vistide Drugs 0.000 description 1
- 229940124024 weight reducing agent Drugs 0.000 description 1
- 238000009736 wetting Methods 0.000 description 1
- 229920001285 xanthan gum Polymers 0.000 description 1
- 239000000230 xanthan gum Substances 0.000 description 1
- 235000010493 xanthan gum Nutrition 0.000 description 1
- 229940082509 xanthan gum Drugs 0.000 description 1
- ARAIBEBZBOPLMB-UFGQHTETSA-N zanamivir Chemical compound CC(=O)N[C@@H]1[C@@H](N=C(N)N)C=C(C(O)=O)O[C@H]1[C@H](O)[C@H](O)CO ARAIBEBZBOPLMB-UFGQHTETSA-N 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229940107931 zovirax Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/66—Phosphorus compounds
- A61K31/661—Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/13—Amines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/66—Phosphorus compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
Definitions
- a method for the prevention or treatment of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses infection by administering an effective amount of a compound of Formula (I), Formula (II) or a derivative thereof to an individual in need is provided.
- Viruses can be divided into several (arbitrary) classifications based upon the type of nucleic acid that they carry and the mode of expression.
- Table 1 illustrates one classification scheme based upon “The International Code of Virus Classification and Nomenclature”, and the classification system developed by Dr. David Baltimore (incorporated herein by reference)
- DNA viruses Double stranded Order Ex. Enterobacteria phage T4 DNA viruses Caudovirales Unassigned Ex. Family Adenoviridae viruses Ex. Family Herpesviridae Ex. Family Polymaviridae (simiam virus 40) Ex. Family Poxviridae (Cowpox virus, Variola virus-smallpox) Single stranded Unassigned DNA viruses bacteriophages Unassigned Ex. Family Parvoviridae viruses RNA viruses Double stranded Unassigned Ex. Family Reoviridae RNA viruses viruses (+) single-stranded Order: Ex.
- Family Coronaviridae coronavirus, RNA or mRNA-like Nidovirales SARS viruses Unassigned Ex.
- Family Flaviviridae Yellow fever viruses virus, West Nile virus, Hepatitis C virus
- Family Picornaviridae Poliovirus, rhino virus, hepatitis A virus
- Togaviridae Rubella virus
- ⁇ single-stranded Order: Ex. Family Paramyxoviridae (measles RNA viruses Mononegavirales virus, mumps virus) (non-segmented Ex. Family Rhabdoviridae (rabies virus) negative stranded viruses) Segmented Ex.
- Family Orthomyxoviridae Influenza negative stranded viruses
- Family Retroviridae Retroviruses, reverse RNA reverse viruses HIV
- transcribing transcribing viruses viruses
- Family Hepadnaviridae Hepatitis B DNA reverse viruses virus
- a limited number of chronic viral diseases which affect millions of people world-wide (e.g., 40 million individuals are HIV-infected), can be controlled to some degree, but no cures are currently available.
- infected individuals provide a reservoir for the respective virus through which na ⁇ ve individuals become infected, thereby, perpetuating the problem of viral infection of significant human populations with these pathogens.
- antiviral drugs such as the nucleoside reverse transcriptase inhibitors (NRTIs) used to inhibit viral replication of specific pathogenic viruses, have resulted in recognizable improvements in the ability to control infections with these pathogens and to improve the quality and length of life of infected individuals.
- NRTIs nucleoside reverse transcriptase inhibitors
- a partial list of currently available antiviral drugs used to inhibit viral replication of specific pathogenic viruses is provided in Appendix 2 (for eg. the NRTIs).
- the therapies listed have resulted in recognizable improvements in the ability to control infections from these pathogens and to improve the quality and length of life of infected individuals.
- currently available classes of antiviral agents have limited utilities due to their narrow scope of activities against different viruses and/or problems with significant drug-induced toxicities.
- the modes of action of the NRTIs and other drugs in current clinical use predispose to the development of drug resistance through viral mutations.
- many currently available drugs have considerable side effects that prevent their wide spread use to achieve treatment or prevention goals.
- compositions and methods of the preferred embodiments provide such agents and associated methods of treatment.
- Amifostine is a pro-drug that is metabolized by alkaline phosphatase to the reduced free thiol (WR-1065); oxidation of WR-1065 leads to formation of the disulfide (WR-33278).
- spermine chemical formula H 2 N(CH 2 ) 3 NH(CH 2 ) 2 (CH 2 ) 2 NH(CH 2 ) 3 NH 2
- WR-1065 appears to function as an analog of the polyamine spermine, and to compete with spermine for sites on DNA, and probably also on other nucleic acids and proteins.
- analogs of WR-1065 and the other compounds of preferred embodiments, as discussed below may function as spermine or other polyamine analogs, and may mimic the antiviral and polyamine-like activity of WR-1065.
- Thiol metabolites of amifostine are believed to be responsible for most of the cytoprotective and radioprotective properties of amifostine.
- Amifostine is taken up by cells through a combination of passive and active transport processes where it is metabolized to its active forms; these metabolites participate in a number of functions.
- These functions include, but are not limited to, (i) protection against radiation induced cytotoxicity and cell killing, (ii) protection against radiation-induced mutagenesis/carcinogenesis, (iii) modulation of topoisomerase I and topoisomerase II alpha activities, (iv) modulation of conformational changes in chromatid structure, (v) inhibition of cell cycle progression, (vi) inhibition of endonuclease activity, (vii) competition with spermine in polyamine transport systems, (viii) induction and repression of gene expression (effect dependent upon the particular gene).
- Other activities include detoxifying cisplatin and other alkylating agents, scavenging free radicals, modulating apoptosis, and modifying the activity of specific enzymes/proteins.
- tissues are especially effective at uptake of the drugs (and/or their metabolites) and retention of those metabolites; these tissues include: kidney, liver, salivary gland, and lung (Rasey et al., (1984), Radiat. Res. 97(3): 598-607). It is possible that other tissues that have not been tested also perform like the above mentioned tissues. These studies establish the ability to obtain therapeutic levels of these drugs and/or their metabolites in the cells of these tissues/organ systems. Therapeutic drug levels are also expected to be obtained in tissues or organs systems where drug pharmacokinetics resemble that of the above mentioned tissues.
- the cytoprotective effects of amifostine appear to be dependent upon a number of factors including, but not limited to, oxygen tension, pH, gene status (including the presence of a functional p53 gene), and enzyme status (including the expression of alkaline phosphatase in the cell membrane). Differences in these factors appear to be responsible for the differential cytoprotective effect mediated by amifostine between tumor cells and normal, nontumorigenic tissue.
- amifostine, phosphonol and their derivatives and analogs are particularly effective in inhibiting replication of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses.
- a method of treating or preventing a DNA virus, a RNA virus and/or a DNA or RNA reverse transcribing virus infection in an individual in need thereof comprising the step of administering to the individual an effective antiviral amount of a compound or a pharmaceutically acceptable salt or solvate thereof, wherein the compound comprises Formula (I) or Formula (II)
- X is selected from the group consisting of —PO 3 H 2 , hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl, wherein each of R 1 , R 2 , and R 3 is independently selected from hydrogen and C 1-6 alkyl, and wherein n is an integer of from 1 to 10.
- the compound is of Formula (I), R 1 is methyl, R 2 is hydrogen, R 3 is hydrogen, and n is 3.
- the compound is of Formula (I), R 1 is methyl, R 2 is hydrogen, R 3 is hydrogen, and X is hydrogen.
- the compound is of Formula (I), R 1 is hydrogen, R 2 is hydrogen, R 3 is hydrogen, and n is 3.
- the compound is of Formula (I), R 1 is hydrogen, R 2 is hydrogen, R 3 is hydrogen, and X is hydrogen.
- the compound is of Formula (II), R 1 is methyl, R 2 is hydrogen, R 3 is hydrogen, and n is 3.
- the compound is of Formula (II), R 1 is hydrogen, R 2 is hydrogen, R 3 is hydrogen, and n is 3.
- the compound is administered to the individual at a daily dosage of from about 200 mg/m 2 to about 3000 mg/m 2 .
- the step of administering is selected from the group consisting of orally administering, subcutaneously administering, intravenously administering, parenterally administering, and administering by inhalation.
- the method further comprises the step of administering to the individual an effective antiviral amount of an antiviral drug, for example (see the list of antiviral drugs in Appendix 2) to treat or prevent an viral infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus.
- an antiviral drug for example (see the list of antiviral drugs in Appendix 2) to treat or prevent an viral infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus.
- a method of treating or preventing a human or animal viral infection caused by a DNA virus, a RNA virus, a DNA or a RNA reverse transcribing virus in an individual in need thereof comprising the step of administering to the individual an effective antiviral amount of amifostine, the free thiol form of amifostine, the disulfide of amifostine (WR-33278), a combination of both of the free thiol and the disulfide of amifostine, or other structurally and functionally related compounds as described in this document.
- a method of treating or preventing an infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus, or combination thereof in an individual in need thereof comprising the step of administering to the individual an effective antiviral amount of phosphonol, the free thiol form of phosphonol, the disulfide of phosphonol, a combination of both the free thiol and the disulfide of phosphonol, or other structurally and functionally related compounds as described in this document.
- the viral infection is caused by a dsDNA virus.
- the viral infection is caused by a dsDNA virus.
- the viral infection is caused by a dsRNA virus.
- the viral infection is caused by a dsRNA virus.
- the viral infection is caused by a (+)ssRNA virus.
- the viral infection is caused by a (+)ssRNA virus.
- the viral infection is caused by a ( ⁇ )ssRNA virus.
- the viral infection is caused by a ( ⁇ )ssRNA virus.
- the viral infection is caused by a non Retroviridae ssRNA reverse transcribing virus.
- the viral infection is caused by a non Retroviradae ssRNA reverse transcribing virus.
- the viral infection is caused by a dsDNA reverse transcribing virus.
- the viral infection is caused by a dsDNA reverse transcribing virus.
- the viral infection is caused by a ssDNA reverse transcribing virus.
- the viral infection is caused by a ssDNA reverse transcribing virus.
- a pharmaceutical kit comprising a pharmaceutical composition comprising a compound or pharmaceutically acceptable salt or solvate thereof in a pharmaceutically acceptable carrier, the compound comprising Formula (I), Formula (II), or a combination thereof:
- X is selected from the group consisting of —PO 3 H 2 , hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl, wherein each of R 1 , R 2 , and R 3 is independently selected from hydrogen and C 1-6 alkyl, and wherein n is an integer of from 1 to 10; and directions for administering the pharmaceutical composition to a patient infected with a DNA virus, an RNA virus, or a DNA or RNA reverse transcribing virus.
- the kit further comprises an antiviral drug, selected on the basis of its effectiveness for treating the given viral agent, in a pharmaceutically acceptable carrier.
- the kit further comprises an antiviral drug in a pharmaceutically acceptable carrier and directions for administering the antiviral drug in a pharmaceutically acceptable carrier to the patient.
- Amifostine is an organic thiophosphate which selectively protects normal tissues but not tumors against cytotoxicity of ionizing radiations, DNA-binding chemotherapeutic agents (e.g., classical alkylating agents such as cyclophosphamide and non-classical alkylating agents such as mitomycin-C and platinum analogs).
- DNA-binding chemotherapeutic agents e.g., classical alkylating agents such as cyclophosphamide and non-classical alkylating agents such as mitomycin-C and platinum analogs.
- Amifostine is a prodrug that is dephosphorylated to the active metabolite, the free thiol form, by alkaline phosphatase and exits the bloodstream rapidly.
- the compounds of preferred embodiments including amifostine and its derivatives and analogs, are particularly effective antiviral compounds for use in inhibiting replication of diverse species of DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses.
- Amifostine (referred to as “WR-2721”) is marketed under the name Ethyol® by Schering-Plough Pty Ltd. It has the chemical name S-2-(3-aminopropyl)aminoethyl phosphorothioic acid and the structure:
- a particularly preferred antiviral compound for use in inhibiting replication of DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses includes the free thiol form of amifostine.
- the free thiol form (referred to as “WR-1065”) has the chemical name 2-(3-aminopropylamino)ethanethiol and the following structure:
- the disulfide form of amifostine (referred to as WR-33278) has the chemical name N 1 ,N 3 ,(dithiodiethane-2,1-diyl)dipropane-1,3-diamine
- Phosphonol (referred to as “WR-3689”) is structurally similar to amifostine, the only difference being the presence of a terminal methyl group.
- Phosphonol has the chemical name S-2-(3-(methylamino)propylamino)ethyl phosphorothioic acid and the following structure:
- WR-255591 The free thiol form of phosphonol (referred to as WR-255591) has the chemical name 2-(3-(methylamino)propylamino) ethanethiol and the following structure:
- the disulfide form of phosphonol (referred to as WR-33278) has the following structure:
- the compounds of preferred embodiments include prodrug forms of the above-described free thiol forms of amifostine, phosphonol, and analogs thereof.
- prodrugs include compounds of the structure:
- each of R 1 , R 2 , and R 3 is independently selected from hydrogen and lower alkyl, wherein —(C n H 2n )— is lower alkyl and n is 1, 2, 3, 4, 5, or (up to 10), and wherein X is a suitable leaving group.
- Suitable leaving groups include —PO 3 H 2 , hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl); however, any other suitable leaving group can be employed that yields the active free thiol form of the compound when metabolized in vivo.
- alkyl groups e.g., —(C 1-6 alkyl)
- keto groups e.g., —C( ⁇ O)—(C 1-6 alkyl) or —C( ⁇ O)—(C 6-18 aryl).
- lower alkyl as used herein is a broad term, and is to be given its ordinary and customary meaning to a person of ordinary skill in the art (and is not to be limited to a special or customized meaning), and refers without limitation to a straight chain or branched chain, acyclic or cyclic, saturated aliphatic hydrocarbon containing 1, 2, 3, 4, 5, or 6 carbon atoms.
- Saturated straight chain lower alkyls include methyl, ethyl, n-propyl, n-butyl, n-pentyl, and n-hexyl; while representative saturated branched chain alkyls include isopropyl, sec-butyl, isobutyl, tert-butyl, and isopentyl.
- aryl as used herein is a broad term and is used in its ordinary sense, including, without limitation, to refer to an aromatic carbocyclic moiety such as phenyl or naphthyl, including mono-, di-, and poly-homocyclic aromatic ring systems (e.g., C 6-18 aryl).
- prodrug forms described above are metabolized into active thiols of the formula:
- each of R 1 , R 2 , R 3 , —(C n H 2n )—, and n is as defined above.
- —(C n H 2n )— is a straight alkyl chain having three carbon atoms. It is also particularly preferred that R 2 and R 3 are both hydrogen, and that R 1 is hydrogen or methyl.
- disulfide forms also exhibit antiviral activity, for example, a disulfide of the following structure:
- antiviral agents of preferred embodiments incorporate a cysteamine-like group (i.e., a group containing the moiety >N—(CH 2 ) 2 —S—), or a moiety with the structure of WR 2721 or WR-1065, tethered to a DNA or nucleic acid binding agent, or other agents with some affinity for nucleic acids or proteins.
- a cysteamine-like group i.e., a group containing the moiety >N—(CH 2 ) 2 —S—
- a moiety with the structure of WR 2721 or WR-1065 tethered to a DNA or nucleic acid binding agent, or other agents with some affinity for nucleic acids or proteins.
- the compounds of preferred embodiments are particularly effective antiviral agents for monotherapeutic or combined-therapeutic use in treating DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses.
- the compounds of preferred embodiments are generally administered at dosages equal to or less than the oral radioprotective dosage of amifostine (e.g., 1456 mg total dose, or 910 mg/m 2 for a 60 kg body weight (BW) adult human) or phosphonol (e.g., 725 mg/m 2 ).
- the recommended starting dose of amifostine is 910 mg/m 2 for a 60 kg BW adult human administered once daily as a 15-minute i.v. infusion, starting 30 minutes prior to chemotherapy.
- Dosages can be converted from mg/m 2 to total mg or mg/kg BW (see, e.g., Freireich et al. (1966), Cancer Chemother. Reports, 50 (4) 219-244) as in Table 2.
- amifostine or other compounds of preferred embodiments
- dosages of 910 mg/m 2 or less This dosage is equivalent to 24.3 mg/kg BW for a 60 kg BW adult human being (or a total dose of 1456 mg for a 60 kg BW adult, as described above); however, in certain embodiments it can be desirable to administer the compounds of preferred embodiments at higher dosage levels.
- children have been given up to a 2700 mg/m 2 total dose of amifostine prior to administration of a chemotherapeutic agent. In some individuals such high doses are associated with side effects.
- a dose of 740 mg/m 2 amifostine (1148 mg total, for a 60 kg BW human adult) is associated with fewer side effects (List et al.
- the maximally tolerated dose (MTD) for WR-2721 in mice was 432 mg/kg (BW) administered i.p. and 720 mg/kg BW administered p.o., and the 100% effective radioprotective dose was about one half of the MTD.
- the MTD was 893 mg/kg BW administered i.p. and 1488 mg/kg BW administered p.o., and the 100% effective radioprotective dose was about one half of the MTD.
- All of the aminothiols have MTDs in rodents of greater than 400 mg/kg BW.
- Amifostine and WR-1065 can be efficacious at very low concentrations, for example, down to 0.4 micromolar concentrations in some in vitro studies.
- a drug delivery system that obtains relatively constant intracellular concentrations over a period of time that could extend from several days to up to one year or more is desired.
- drug delivery systems include subcutaneous administration, formulation in nanoparticles, were formulation in other slow release systems.
- Table 3 provides plasma concentrations of amifostine metabolites (see Geary et al., Res. Comm. Chem. Path. Pharmacol., 65(2), 147-159 (1989)) and phosphonol metabolites (see Buckpitt et al., Contract #DAMD 17-86-C-6177, reference obtained from Dr. D. Grdina, personal communication) after duodenal administration of 150 mg/kg BW of each drug in rhesus macaques. This data was collected as part of work performed to evaluate the radioprotective activity of the compounds. These data may be useful in estimating plasma concentrations in humans.
- amifostine While it is generally preferred to formulate amifostine or the other compounds of preferred embodiments for oral administration, the compounds of preferred embodiments can be formulated so as to allow them to be administered by other routes, as discussed herein. It can be desirable in certain embodiments to formulate amifostine for intravenous administration in order to maximize efficacy. Because of the structural similarities between amifostine and phosphonol, especially the similarities in the sulfhydryl ends of the molecules, phosphonol is expected to behave in a manner similar to amifostine rather than WR-151327 (chemical formula CH 3 NH(CH 2 ) 3 NH(CH 2 ) 3 SPO 3 H 2 ). WR-151327 has been previously shown to have anti-HIV activity; this activity was attributed to the ability of the compound to modulate cytokine levels in a manner that inhibited HIV replication and/or proliferation (Kalebic et al., 1994).
- Phosphonol may not be quite as efficacious as amifostine, based upon the work of Gutschow et al., who found lower activity when a methyl group was substituted for a hydrogen atom in a position similar to that of the methyl group of phosphonol that distinguishes its structure from that of amifostine (Gutschow et al. (1995), Pharmazie 50(10): 672-5).
- phosphonol is expected to be significantly more efficacious than WR-151327. This consideration should be viewed in light of the fact that the overall structures of the compounds tested by Gutschow et al. were significantly different from the structures of the preferred embodiments. The overall structures of the compounds tested by Gutschow et al. are significantly different from the structures of the preferred embodiments, and it is expected that phosphonol will be significantly more efficacious than WR-151327.
- Amifostine, its analogs, and its derivatives can be administered in combination with other antiviral agents employed to treat HIV infection.
- One of the benefits of such combination therapies is that lower doses of the other antiviral agents can be administered while still achieving a similar level of antiviral efficacy.
- Such lower dosages can be particularly advantageous for drugs known to have genotoxicity and mitochondrial toxicity (for example, some nucleoside analogs).
- greater efficacy might be achieved using therapeutic doses of two drugs than could be achieved using only a single drug.
- nucleoside analogs members of the class of drugs known as nucleoside analogs.
- Other agents that could be used potentially are included in the list of antiviral drugs included in Appendix 2.
- nucleoside analogs has been associated with a variety of side effects due to the fact that these drugs are analogs of naturally occurring nucleosides, are incorporated into host cell DNA, and function as obligate DNA chain terminators. As such, these drugs are associated with the induction of mutations in host cell DNA, increased risks for certain types of cancer, as well as increased risks for a variety of diseases associated with mutation induction in nuclear and mitochondrial DNA.
- Nucleoside analogs are also associated with mitochondrial toxicity to a degree that varies depending upon the specific nucleoside analog in question. Thus, it is desirable to be able to minimize the doses of nucleoside analogs used, and/or to use these drugs in combination with other drugs with demonstrated antiviral efficacy that does not compromise the effectiveness of the other treatment, and that also has demonstrated antimutagenic efficacy.
- compositions Comprising Amifostine and Analogs Thereof:
- the compounds of preferred embodiments can be incorporated into a pharmaceutically acceptable carrier, including incorporation into nanoparticles for administration to an individual having a viral infection (for example, an adenoviral infection as an example of a DNA virus, an influenza infection as an example of a RNA virus, or an HIV infection as an example of a DNA or RNA reverse transcribing virus) or can be administered prophylactically to prevent viral infection, or postinfection to decrease symptoms upon viral infection.
- a viral infection for example, an adenoviral infection as an example of a DNA virus, an influenza infection as an example of a RNA virus, or an HIV infection as an example of a DNA or RNA reverse transcribing virus
- the compounds of preferred embodiments can be employed as the sole agent in the prevention or treatment of DNA viruses, RNA viruses, or DNA or RNA reverse transcribing viruses, and/or combinations thereof two or more such compounds can be employed, optionally in combination with other therapeutic agents, e.g., conventional or newly developed drugs employed in the treatment of AIDS or HIV, or other viral infections.
- reducing agents including, but not limited to, vitamin C and vitamin E.
- Other reducing agents include organic aldehydes, hydroxyl-containing aldehydes, and reducing sugars such as glucose, mannose, galactose, xylose, ribose, and arabinose.
- Other reducing sugars containing hemiacetal or keto groupings can be employed, for example, maltose, sucrose, lactose, fructose, and sorbose.
- reducing agents include alcohols, preferably polyhydric alcohols, such as glycerol, sorbitol, glycols, especially ethylene glycol and propylene glycol, and polyglycols such as polyethylene and polypropylene glycols.
- polyhydric alcohols such as glycerol, sorbitol, glycols, especially ethylene glycol and propylene glycol, and polyglycols such as polyethylene and polypropylene glycols.
- pharmaceutically acceptable salts and “a pharmaceutically acceptable salt thereof” as used herein are broad terms, and are to be given their ordinary and customary meaning to a person of ordinary skill in the art (and are not to be limited to a special or customized meaning), and refer without limitation to salts prepared from pharmaceutically acceptable, non-toxic acids or bases.
- Suitable pharmaceutically acceptable salts include metallic salts, e.g., salts of aluminum, zinc, alkali metal salts such as lithium, sodium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts; organic salts, e.g., salts of lysine, N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), procaine, and tris; salts of free acids and bases; inorganic salts, e.g., sulfate, hydrochloride, and hydrobromide; and other salts which are currently in widespread pharmaceutical use and are listed in sources well known to those of skill in the art, such as, for example, The Merck Index.
- metallic salts e.g., salts of aluminum, zinc, alkali metal salts such as lithium, sodium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts
- Any suitable constituent can be selected to make a salt of the therapeutic agents discussed herein, provided that it is non-toxic and does not substantially interfere with the desired activity.
- pharmaceutically acceptable precursors and derivatives of the compounds can be employed.
- Pharmaceutically acceptable amides, lower alkyl esters, and protected derivatives can also be suitable for use in compositions and methods of preferred embodiments. While it may be possible to administer the compounds of the preferred embodiments in the form of pharmaceutically acceptable salts, it is generally preferred to administer the compounds in neutral form.
- Contemplated routes of administration include but are not limited to oral, sublingual, parenteral, transcutaneous, subcutaneous, intravenous, and by inhalation.
- Compounds of preferred embodiments can be formulated into liquid preparations for, e.g., oral administration. Suitable forms for such administration include suspensions, syrups, and elixirs.
- compositions of preferred embodiments are preferably isotonic with the blood or other body fluid of the recipient.
- the isotonicity of the compositions can be attained using sodium tartrate, propylene glycol or other inorganic or organic solutes.
- Sodium chloride is particularly preferred.
- Buffering agents can be employed, such as acetic acid and salts, citric acid and salts, boric acid and salts, and phosphoric acid and salts.
- Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils.
- Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like.
- a reducing agent such as vitamin C, vitamin E, or other reducing agents as are known in the pharmaceutical arts, in the formulation.
- Viscosity of the pharmaceutical compositions can be maintained at the selected level using a pharmaceutically acceptable thickening agent.
- Methylcellulose is preferred because it is readily and economically available and is easy to work with.
- Other suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like.
- the preferred concentration of the thickener will depend upon the thickening agent selected. An amount is preferably used that will achieve the selected viscosity. Viscous compositions are normally prepared from solutions by the addition of such thickening agents.
- a pharmaceutically acceptable preservative can be employed to increase the shelf life of the pharmaceutical compositions.
- Benzyl alcohol can be suitable, although a variety of preservatives including, for example, parabens, thimerosal, chlorobutanol, or benzalkonium chloride can also be employed.
- a suitable concentration of the preservative is typically from about 0.02% to about 2% based on the total weight of the composition, although larger or smaller amounts can be desirable depending upon the agent selected. Reducing agents, as described above, can be advantageously used to maintain good shelf life of the formulation.
- the compounds of preferred embodiments can be in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, or the like, and can contain auxiliary substances such as wetting or emulsifying agents, pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
- a suitable carrier diluent, or excipient
- auxiliary substances such as wetting or emulsifying agents, pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
- Such preparations can include complexing agents, metal ions, polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, dextran, and the like, liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts.
- Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. The presence of such additional components can influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance, and are thus chosen according to the intended application, such that the characteristics of the carrier are tailored to the selected route of administration.
- compositions intended for oral use can be prepared according to any method known in the art for the manufacture of pharmaceutical compositions and can include one or more of the following agents: sweeteners, flavoring agents, coloring agents and preservatives.
- Aqueous suspensions can contain the active ingredient in admixture with excipients suitable for the manufacture of aqueous suspensions.
- Formulations for oral use can also be provided as hard gelatin capsules, wherein the active ingredient(s) are mixed with an inert solid diluent, such as calcium carbonate, calcium phosphate, or kaolin, or as soft gelatin capsules.
- an inert solid diluent such as calcium carbonate, calcium phosphate, or kaolin
- the active compounds can be dissolved or suspended in suitable liquids, such as water or an oil medium, such as peanut oil, olive oil, fatty oils, liquid paraffin, or liquid polyethylene glycols.
- Stabilizers and microspheres formulated for oral administration can also be used.
- Capsules can include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
- the push-fit capsules can contain the active ingredient in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
- fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
- binders such as starches
- lubricants such as talc or magnesium stearate
- stabilizers optionally, stabilizers.
- a reducing agent in the capsule.
- Tablets can be uncoated or coated by known methods to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period of time.
- a time delay material such as glyceryl monostearate can be used.
- the solid form When administered in solid form, such as tablet form, the solid form typically comprises from about 0.001 wt. % or less to about 50 wt. % or more of active ingredient(s), preferably from about 0.005, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1 wt. % to about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, or 45 wt. %.
- Tablets can contain the active ingredients in admixture with non-toxic pharmaceutically acceptable excipients including inert materials.
- a tablet can be prepared by compression or molding, optionally, with one or more additional ingredients.
- Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding, in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
- each tablet or capsule contains from about 10 mg or less to about 1,000 mg or more of a compound of the preferred embodiments, more preferably from about 20, 30, 40, 50, 60, 70, 80, 90, or 100 mg to about 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, or 900 mg.
- tablets or capsules are provided in a range of dosages to permit divided dosages to be administered. A dosage appropriate to the patient and the number of doses to be administered daily can thus be conveniently selected.
- Suitable inert materials include diluents, such as carbohydrates, mannitol, lactose, anhydrous lactose, cellulose, sucrose, modified dextrans, starch, and the like, or inorganic salts such as calcium triphosphate, calcium phosphate, sodium phosphate, calcium carbonate, sodium carbonate, magnesium carbonate, and sodium chloride.
- diluents such as carbohydrates, mannitol, lactose, anhydrous lactose, cellulose, sucrose, modified dextrans, starch, and the like
- inorganic salts such as calcium triphosphate, calcium phosphate, sodium phosphate, calcium carbonate, sodium carbonate, magnesium carbonate, and sodium chloride.
- Disintegrants or granulating agents can be included in the formulation, for example, starches such as corn starch, alginic acid, sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite, insoluble cationic exchange resins, powdered gums such as agar, karaya or tragacanth, or alginic acid or salts thereof.
- starches such as corn starch, alginic acid, sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite, insoluble cationic exchange resins, powdered gums such as agar, karaya or tragacanth, or alginic acid or salts thereof.
- Binders can be used to form a hard tablet. Binders include materials from natural products such as acacia, tragacanth, starch and gelatin, methyl cellulose, ethyl cellulose, carboxymethyl cellulose, polyvinyl pyrrolidone, hydroxypropylmethyl cellulose, and the like.
- Lubricants such as stearic acid or magnesium or calcium salts thereof, polytetrafluoroethylene, liquid paraffin, vegetable oils and waxes, sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol, starch, talc, pyrogenic silica, hydrated silicoaluminate, and the like, can be included in tablet formulations.
- Surfactants can also be employed, for example, anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate, cationic such as benzalkonium chloride or benzethonium chloride, or nonionic detergents such as polyoxyethylene hydrogenated castor oil, glycerol monostearate, polysorbates, sucrose fatty acid ester, methyl cellulose, or carboxymethyl cellulose.
- anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate
- cationic such as benzalkonium chloride or benzethonium chloride
- nonionic detergents such as polyoxyethylene hydrogenated castor oil, glycerol monostearate, polysorbates, sucrose fatty acid ester, methyl cellulose, or carboxymethyl cellulose.
- Controlled release formulations can be employed wherein the amifostine or analog(s) thereof is incorporated into an inert matrix that permits release by either diffusion or leaching mechanisms. Slowly degenerating matrices can also be incorporated into the formulation.
- Other delivery systems can include timed release, delayed release, or sustained release delivery systems.
- Coatings can be used, for example, nonenteric materials such as methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methylhydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl-methyl cellulose, sodium carboxy-methyl cellulose, providone and the polyethylene glycols, or enteric materials such as phthalic acid esters.
- Dyestuffs or pigments can be added for identification or to characterize different combinations of active compound doses.
- a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils can be added to the active ingredient(s).
- Physiological saline solution, dextrose, or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol are also suitable liquid carriers.
- the pharmaceutical compositions can also be in the form of oil-in-water emulsions.
- the oily phase can be a vegetable oil, such as olive or arachis oil, a mineral oil such as liquid paraffin, or a mixture thereof.
- Suitable emulsifying agents include naturally-occurring gums such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan mono-oleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan mono-oleate.
- the emulsions can also contain sweetening and flavoring agents.
- a compound of the preferred embodiments When a compound of the preferred embodiments is administered by intravenous, parenteral, or other injection, it is preferably in the form of a pyrogen-free, parenterally acceptable aqueous solution or oleaginous suspension.
- Suspensions can be formulated according to methods well known in the art using suitable dispersing or wetting agents and suspending agents. The preparation of acceptable aqueous solutions with suitable pH, isotonicity, stability, and the like, is within the skill in the art.
- a preferred pharmaceutical composition for injection preferably contains an isotonic vehicle such as 1,3-butanediol, water, isotonic sodium chloride solution, Ringer's solution, dextrose solution, dextrose and sodium chloride solution, lactated Ringer's solution, or other vehicles as are known in the art.
- an isotonic vehicle such as 1,3-butanediol, water, isotonic sodium chloride solution, Ringer's solution, dextrose solution, dextrose and sodium chloride solution, lactated Ringer's solution, or other vehicles as are known in the art.
- sterile fixed oils can be employed conventionally as a solvent or suspending medium.
- any bland fixed oil can be employed including synthetic mono or diglycerides.
- fatty acids such as oleic acid can likewise be used in the formation of injectable preparations.
- the pharmaceutical compositions can also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art
- the duration of the injection can be adjusted depending upon various factors, and can comprise a single injection administered over the course of a few seconds or less, to 0.5, 0.1, 0.25, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours or more of continuous intravenous administration.
- compositions of the preferred embodiments can additionally employ adjunct components conventionally found in pharmaceutical compositions in their art-established fashion and at their art-established levels.
- the compositions can contain additional compatible pharmaceutically active materials for combination therapy (such as supplementary antimicrobials, antipruritics, astringents, local anesthetics, anti-inflammatory agents, reducing agents, and the like), or can contain materials useful in physically formulating various dosage forms of the preferred embodiments, such as excipients, dyes, thickening agents, stabilizers, preservatives or antioxidants.
- the compounds of the preferred embodiments can be provided to an administering physician or other health care professional in the form of a kit.
- the kit is a package which houses a container which contains the compound(s) in a suitable pharmaceutical composition, and instructions for administering the pharmaceutical composition to a subject.
- the kit can optionally also contain one or more additional therapeutic agents, e.g., antiviral nucleoside analog, peptide analog, protease inhibitor, monoclonal antibody, or any other antiviral used for the treatment or prevention of virus disease (see for example Appendix 2).
- kits containing one or more compositions comprising compound(s) of the preferred embodiments in combination with one or more additional antiviral, antibacterial, and/or anti-infective agents can be provided, or separate pharmaceutical compositions containing a compound of the preferred embodiments and additional therapeutic agents can be provided.
- the kit can also contain separate doses of a compound of the preferred embodiments for serial or sequential administration.
- the kit can optionally contain one or more diagnostic tools and instructions for use.
- the kit can contain suitable delivery devices, e.g., syringes, and the like, along with instructions for administering the compound(s) and any other therapeutic agent.
- the kit can optionally contain instructions for storage, reconstitution (if applicable), and administration of any or all therapeutic agents included.
- kits can include a plurality of containers reflecting the number of administrations to be given to a subject.
- a kit for the treatment of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses, and combinations thereof is provided that includes amifostine or another compound of a preferred embodiment and one or more antiviral agents currently employed to treat the virus.
- Antiviral agents include for example, nucleoside analogs such as acyclovir, reverse transcriptase inhibitors such as zidovudine, didanosine, zalcitabine, stavudine, 3TC; non-nucleoside reverse transcriptase inhibitors; protease inhibitors; cytokines; immunomodulators, and antibodies but are not limited thereto.
- the compounds of the preferred embodiments can be administered prophylactically for the prevention of DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection.
- therapy is preferably initiated as early as possible following the onset of signs and symptoms of a viral infection, or following exposure to a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus infection.
- the administration route, amount administered, and frequency of administration will vary depending on the age of the patient, the severity of the infection, and any associated conditions.
- Contemplated amounts, dosages, and routes of administration for the compounds of preferred embodiments for treatment of a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus infection are similar to those established for conventional antiviral agents.
- anti-viral therapies combining an aminothiol with another antiviral therapy will have an additive or synergistic activity, making it possible to reduce the doses of the drugs currently used to treat DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection, or making it possible to achieve higher efficacy by using therapeutic doses of two or more agents together.
- the combination of an aminothiol with a nucleoside analog is expected to simultaneously provide improved antiviral therapy and protection against nucleoside analog-induced side effects, especially those associated with nuclear DNA or mitochondrial DNA damage. It is further anticipated that an aminothiol could protect against deleterious side effects of another antiviral drugs, especially those that induce nuclear DNA or mitochondrial DNA damage.
- contemplated amounts of the compounds of the preferred embodiments for oral administration to treat DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection are from about 10 mg or less to about 2000 mg or more administered from about every 4 hours or less to about every 6 hours or more (or from about 4 times daily to about 6 times daily) for about 5 days or less to about 10 days or more (40 mg/day or less to about 15,000 mg/day or more) or until there is a significant improvement in the condition.
- doses of from about 10 mg or less to about 1000 mg or more are orally administered once, twice, or multiple times a day, typically for up to about 12 months, or, in certain circumstances, indefinitely (from about 10 mg/day to about 1,000 mg/day).
- doses of from about 10 mg or less to about 1000 mg or more are orally administered once, twice, or multiple times a day, typically for up to about 12 months, or, in certain circumstances, indefinitely (from about 10 mg/day to about 1,000 mg/day).
- it can be desirable to vary the dosage employing a higher dosage early in the treatment, and a lower dosage later in the treatment.
- the single highest dose of amifostine administered to an adult human as documented in the literature was 1330 mg/m 2 .
- Children have been administered single doses of amifostine of up to 2700 mg/m 2 with no untoward effects.
- the literature indicates that multiple doses (up to three times the recommended single dose of 740 to 910 mg/m 2 ) have been safely administered within a 24-hour period.
- Repeated administration of amifostine at two and four hours after the initial dose does not appear to result in an increase in side effects, especially nausea, vomiting, or hypotension. It appears that the most significant deleterious side effect from the administration of amifostine is hypotension.
- contemplated amounts of the compounds of the preferred embodiments, methods of administration, and treatment schedules for individuals with infections caused by DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses are generally similar to those described above for the prevention of and/or treatment of radiation- or chemotherapy-induced cytotoxicity; they may also be similar to those used for the treatment of myelodysplastic syndrome.
- amifostine Known side effects of amifostine include decrease in systolic blood pressure, nausea, and vomiting. If such side effects are observed for the particular thiophosphate administered, it is generally preferred to administer an antiemetic medication prior to, or in conjunction with the thiophosphate.
- Suitable antiemetic medications include antihistamines (e.g., buclizine, cyclizine, dimenhydrinate, diphenhydramine, meclizine), anticholinergic agents (e.g., scopolamine), dopamine antagonists (e.g., chlorpromazine, droperidol, metoclopramide, prochlorperazine, promethazine), serotonin antagonists (e.g., dolasetron, granisetron, ondansetron), or other agents (e.g., dexamethasone, methylprednisolone, trimethobenzamide).
- antihistamines e.g., buclizine, cyclizine, dimenhydrinate, diphenhydramine, meclizine
- anticholinergic agents e.g., scopolamine
- dopamine antagonists e.g., chlorpromazine, droperidol, metoclopramide, prochlorperazine
- WR-1065 The purity of the WR-1065 used in the experiments described below is unknown. WR-1065 is sensitive to oxidation (and possibly other reactions) and can undergo reaction to forms that appear to lack antiviral activity. Accordingly, in the tables below and in the preferred dosages provided above, the indicated concentrations represent the maximum amount of active compound that could be present; the true concentration of active compound is less than that indicated but cannot be determined definitively. In addition, for concentrations of WR-1065 of less than 100 ⁇ M, estimates of the true efficacy of the compound are limited by the fact that the compound has been found to be inactivated by a variety of medium components (Grdina et al. (2000), Drug Metabol Drug Interact 16(4): 237-79.). Below approximately 50 ⁇ M, this problem becomes especially severe.
- A549 or MDCK cells were used for the plaque reduction assays.
- Cells were grown to near confluence in Minimal Essential Medium (MEM) in six-well plates.
- MEM Minimal Essential Medium
- PFUs plaque forming units
- Cell monolayers were subsequently overlaid with 0.6% agarose in MEM.
- the overlay medium was prepared to contain a final concentration of 10 ⁇ M, 33 ⁇ M and 100 ⁇ M amount of WR 1065. Each concentration was assayed in triplicate.
- MDCK cells growing in 6-well plates were infected with 100 plaque forming units (PFUs) of virus in 100 ⁇ l and adsorption was allowed for one hour at 37° C.
- Cell monolayers were subsequently overlaid with 0.6% agarose in MEM.
- the overlay medium was prepared to contain a final concentration of 10 ⁇ M, 33 ⁇ M and 100 ⁇ M amount of WR 1065. Each concentration was assayed in triplicate.
- Wells overlaid with media without WR-1065 were set up as 100% infectious virus yield controls.
- Cultures were maintained at 37° C. in 5% CO 2 for 3 days. At the end of this time period, cell cultures were fixed with 1% formaldehyde and stained with Crystal Violet for visualization of plaques and evaluation of plaque size and number. Reduction of plaque formation was expressed as a percentage of the untreated controls.
- Table 5 includes data from an experiment to measure WR-1065 protection against cytopathic effects in MDCK cell monolayers infected with influenza A/Puerto Rico/8/34 (H1N1), using densitometry readings of the stained cell monolayers to estimate relative cell survival. Similar results were obtained in a second independent experiment using influenza A/Puerto Rico/8/34 (H1N1), and in an experiment using the same approach with influenza B/Lee/40. In a third experiment with influenza A, WR 1065 was added to an agarose overlay following a one-hour adsorption with a different virus strain, A/HKx31 (H3N2), and a plaque reduction assay was performed (Table 5). The results illustrate protection of MDCK cells from Influenza A by WR 1065, based upon densitometry readings (experiment 1) or a plaque reduction assay (experiment 2)
- the reduced form of amifostine (WR 2721 or ethyol), was the most active moiety in the observed antiviral effects.
- WR-1065 was the most active moiety in the observed antiviral effects.
- a common medium component (phenylalanine) blocks the activity of WR 1065 to some undetermined degree.
- PBMCs Peripheral blood mononuclear cells obtained from the NIH Blood Bank were cultured with phytohemagglutinin (PHA) for 48 hours to create PHA blasts.
- PHA phytohemagglutinin
- the cells were cultured in RPMI-1640 medium with 10% fetal bovine serum (FBS), 1% penicillin/streptomycin and glutamine, and 10% Interleukin-2 (IL-2).
- FBS fetal bovine serum
- IL-2 Interleukin-2
- WR-1065 from the NCI Chemical Carcinogen Repository; see Hoffman et al. (2001), Env. Mol. Mut. 37:117) was weighed (Mol. Wt. 134) and stored frozen in RPMI-1640 medium as a 10 mg/ml solution (13.4 ⁇ L of the solution added to 1 ml of RPMI-1640 yields 1000 ⁇ M solution).
- AZT SIGMA, Mol. Wt.
- HIV infection status was monitored in five experimental groups by measuring p24 using an ELISA kit (RETRO-TEK HIV-1, p24 Extended Range ELISA, ZMC catalog #0801137).
- the HIV infection status for the five experimental groups is provided in Table 3 (see Experiment #1 data).
- Table 7 illustrates the antiviral efficacy of AZT and WR-1065 on PHA blasts. Subsequent experiments confirmed this antiretroviral effect, and demonstrated that the ability of WR 1065 to inhibit viral replication was dose-dependent and comparable in concentration and effect to that seen with AZT alone using this in vitro assay system.
- WR-1065 was prepared from a 10 mg/ml solution in RPMI-1640 medium and diluted to the desired concentrations. Cell viability was measured 72 hours after exposure to WR-1065. Percentage of viable cells (compared to unexposed controls) is provided in Table 8.)
- Cysteamine (chemical formula H 2 NCH 2 CH 2 SH) has been demonstrated to have anti-HIV activity (Ho et al. (1995), AIDS Res Hum Retroviruses 11(4): 451-9; Bergamini et al. (1996), J Infect Dis 174(1): 214-8).
- 200 ⁇ M cysteamine effectively suppressed ( ⁇ 100%) HIV replication.
- WR-1065 effectively suppressed (>99.9%) HIV replication at a concentration of less than 100 ⁇ M.
- the in vitro anti-HIV activity of WR-1065 is over 2-fold higher than that of cysteamine.
- cysteamine's duration of action was determined to be very short; to achieve an anti-HIV effect, fresh cysteamine had to be added to the cell culture system every 12 hours.
- WR-1065 had a long duration of action—it only had to be added to the culture system once in a 72 hour period.
- Cystamine (chemical formula H 2 N(CH 2 ) 2 SS(CH 2 ) 2 NH 2 ), the oxidized form of cysteamine, has been demonstrated to have anti-HIV activity, DNA binding capacity, radioprotective capacity, and the ability to shift the equilibrium of DNA from the A-form towards the B-form (Allegra et al. (2002), Amino Acids 22(2): 155-66).
- WR-1065 the active form of WR-2721, has also been shown to bind to DNA in the minor groove, and also to shift the B/A-DNA equilibrium towards the B-form.
- cystamine and its reduced form cysteamine are capable of binding to other nucleic acids in addition to DNA, and have some ability to bind to and/or to interact with proteins. It should be noted that the anti-HIV activity of cystamine is considered to be due, at least in large part, to its rapid in vivo conversion to cysteamine.
- WR-1065 is a larger molecule than cysteamine because it contains an additional moiety: —(CH 2 ) 3 NH 2 .
- WR-1065 can bind to and thereby block a larger fragment of a nucleic acid than cysteamine. It can therefore be hypothesized that the differential binding characteristic of WR-1065 versus cysteamine may be responsible for WR-1065's improved antiviral efficacy.
- WR-1065's antiviral activity is due in part to its ability to bind to and to block critical sites on nucleic acids and/or proteins.
- Blockage of nucleic acid sites and/or proteins is considered to be a possible mechanism for the aminothiols' modulation of enzyme function (Brekken et al. (1986), J Biol Chem 273(41): 26317-22). The mechanism by which an aminothiol could bind to and/or block nucleic acids and/or proteins without inducing significant cytotoxicity to eukaryotic cells is unknown at this time.
- WR-1065 has antiviral efficacy against three different species of adenovirus and two different strains of influenza. These are dramatically different types of viruses with significantly different modes of replication.
- the demonstration of broad-spectrum antiviral activity supports the hypothesis that the ability of the antiviral agent WR-1065 to bind to RNA plays a role in the observed antiviral effect.
- one or more of the compounds of the present invention function as an antiviral agent because its structure renders it bi-functional, combining two distinct properties.
- the —(CH 2 ) 3 NH 2 portion of WR-1065 is believed to be responsible for binding to nucleic acids (DNA, RNA), and possibly also to some proteins.
- a second portion of WR-1065 —NH(CH 2 ) 2 SH is believed to be largely responsible for the antiretroviral/antiviral effects that have been observed.
- the sulfhydryl group needs to be in the reduced state in order for antiretroviral/antiviral effects to be observed for the compound; however, it is possible that the sulfhydryl group may remain functional if oxidized to the disulfide (as in WR-33278). Both of these components contribute to maximizing antiretroviral/antiviral effects of the compounds of preferred embodiments (e.g., both the —(CH 2 ) 3 NH 2 and the —NH(CH 2 ) 2 SH moieties of amifostine).
- the first portion of the molecule functions to align the molecule in close proximity to critical binding sites on nucleic acids and/or proteins, and the second portion of the molecule functions in a reaction that contributes to antiviral efficacy.
- Variations to the chemical structure of the moiety —(CH 2 ) 3 NH 2 may be tolerated without losing or altering the antiretroviral/antiviral efficacy of the compound, so long as the resulting structure retains a binding capacity/affinity that is similar to that observed with —(CH 2 ) 3 NH 2 itself.
- Such variations may include more or less than three carbon atoms in the alkyl group, and a branched alkyl chain, lower alkyl substituents on the amino group.
- Gutschow et al. describe two compounds that are demonstrated to display significant antiretroviral/antiviral activity (Gutschow et al. (1995), Pharmazie 50(10): 672-5.). These compounds differ in structure considerably from the compounds of preferred embodiments, but the two most efficacious compounds have, as a portion of their structure, a cysteamine-like group. Gutschow et al.
- WR-151327 and WR-151326 In contrast to WR-151327 and WR-151326, other compounds of preferred embodiments have only two —CH 2 — groups separating the sulfhydryl group from the first amino group, and thus possess superior antiretroviral/antiviral properties.
- the antiretroviral/activity of WR-1065 is significantly greater than that reported for WR-151326, and thus WR-1065 has functional capacities that are different from those of WR-151327 and WR-151326.
- WR-1065 and its analogs interfere with critical components of the viral life cycle of many/all viruses through interactions with viral nucleic acids, nucleic acid-associated proteins, and/or molecular machines involved in nucleic acid and/or protein production and maintenance.
- nucleic acids of higher organisms have a protein-to-DNA ratio of approximately 1:10.
- nucleic acids of many viruses, vegetative bacteriophage, and dinoflagellates have a protein-to-DNA ratio of approximately 1:1 (Cremisi, 1979; Kellenberger, 1988).
- the significantly higher ratio of proteins to nucleic acids of viruses would allow for enhanced interaction between proteins associated with nucleic acids, the nucleic acids themselves, and a molecule with nucleic acid and protein binding affinity.
- the enhanced opportunity for binding between WR-1065 and viral nucleic acids and proteins could result in deleterious effects upon viral nucleic acid production, maintenance, or functionality.
- WR-1065 could affect the functionality of molecular machines.
- molecular machines known as origin recognition complexes will be discussed, but the considerations presented below are applicable to many/all molecular machines of viruses versus higher organisms. Because the components and structure of these machines differ between simple life forms and higher life forms, WR-1065-associated effects could result in different consequences. The basis for this hypothesis comes from two separate areas of consideration.
- the replicon model as proposed by Jacob et al. in 1964 has been shown to be applicable to all bacterial replication systems as well as to several viral families, including simian virus-40 (SV-40). Bergsma et al.
- WR-1065 and similar compounds could block replication initiation through interactions with a defined replicator sequence on viral nucleic acids, while not exerting a similar effect upon the nucleic acids of higher organisms because of the degenerate nature of their initiation-site selection. Binding of WR-1065 to replication initiation sites of eukaryotic cells would be hypothesized to slow down nucleic acid processing and, hence, cell division, but not block it completely. This hypothesis is supported by the fact that WR-1065 has been shown to delay the progression of cells through the cell cycle, but the exact mechanism remains unclear.
- replication origin recognition complexes of higher animals share many similarities with analogous complexes in simple life forms, including viruses and archaeal cells (Gai et al., 2004). However, critical differences have been noted in the components and in the mode of action of these molecular machines (Gai et al., 2004).
- the molecular machine known as the origin recognition complex consists of the oncogenic large tumor antigen that is formed as a monomer; six monomers then form a hexamer, and two hexamers assemble together, along with replication protein A, topoisomerase I, and polymerase-alpha/primase. These molecules form the replication initiation complex for SV-40.
- This complex is highly dependent upon the conformation of individual monomers, as well as the conformation of the hexamer (Gai et al. 2004).
- the functionality of the entire replication initiation complex is also dependent upon the conformation of the target nucleic acids and the target initiation site (Gai et al., 2004).
- WR-1065 is known to have the capacity to alter the conformation of both nucleic acids and proteins, a complex such as that described for SV-40 would be especially vulnerable to induced conformational changes. Partial support for this hypothesis comes from studies demonstrating interactions between WR-1065 and topoisomerase enzymes. Analogous molecular machines that operate in cells of higher organisms would not be expected to share identical conformational requirements.
- origin recognition complexes of eukaryotic cells do not appear to require an origin of replication site, but rather are capable of using a variety of sites, it is reasonable to hypothesize that these latter origin recognition complexes could potentially tolerate a degree of conformational change that SV-40 and other virally-associated origin recognition complexes cannot tolerate.
- Herpes virus Vidarabine (Vira-A) NRTI Viral polymerase Acyclovir (Zovirax) NRTI Viral polymerase Famcylovir (Famvir) Idoxuridine (Herplex liquifilm, Viral polymerase stoxil) Trifluridine (Viroptic) Inhibitor of thymidylate synthetase Tromantadine Derivate of Inhibits viral replication adamantane Penciclovir (Denavir) Interferes with enzymes for viral replication Docosanol (Abreva) Long chain alcohol Changes cell membrane of healthy cells Valacyclovir (Valtrex) Imiquimod (Aldara) Cytomegalo Gancyclovir (Cytovene) NRTI Viral polymerase virus Valganciclovir (Valcyte) NRTI Viral polymerase Fomivirsen (Vi)
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Virology (AREA)
- Oncology (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Communicable Diseases (AREA)
- Molecular Biology (AREA)
- Engineering & Computer Science (AREA)
- Biotechnology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
A method for the prevention or treatment of Influenza virus infection or Adenovirus infection by administering an effective amount of a compound of Formula (I), Formula (II), or similar compound to an individual in need is provided.
Description
- This application claims priority to and the benefit of the filing of U.S. Provisional Patent Application Ser. No. 60/792,738, entitled “Broad Spectrum Antiviral and Methods of Use”, filed on Apr. 17, 2006, and the specification and claims thereof are incorporated herein by reference.
- A method for the prevention or treatment of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses infection by administering an effective amount of a compound of Formula (I), Formula (II) or a derivative thereof to an individual in need is provided.
- Viruses can be divided into several (arbitrary) classifications based upon the type of nucleic acid that they carry and the mode of expression. Table 1 illustrates one classification scheme based upon “The International Code of Virus Classification and Nomenclature”, and the classification system developed by Dr. David Baltimore (incorporated herein by reference)
-
TABLE 1 Group Name Nucleic Acid Type Order Examples DNA viruses Double stranded Order: Ex. Enterobacteria phage T4 DNA viruses Caudovirales Unassigned Ex. Family Adenoviridae viruses Ex. Family Herpesviridae Ex. Family Polymaviridae (simiam virus 40) Ex. Family Poxviridae (Cowpox virus, Variola virus-smallpox) Single stranded Unassigned DNA viruses bacteriophages Unassigned Ex. Family Parvoviridae viruses RNA viruses Double stranded Unassigned Ex. Family Reoviridae RNA viruses viruses (+) single-stranded Order: Ex. Family Coronaviridae (coronavirus, RNA or mRNA-like Nidovirales SARS) viruses Unassigned Ex. Family Flaviviridae (Yellow fever viruses virus, West Nile virus, Hepatitis C virus) Ex. Family Picornaviridae (poliovirus, rhino virus, hepatitis A virus) Ex. Togaviridae (Rubella virus) (−) single-stranded Order: Ex. Family Paramyxoviridae (measles RNA viruses Mononegavirales virus, mumps virus) (non-segmented Ex. Family Rhabdoviridae (rabies virus) negative stranded viruses) Segmented Ex. Family Orthomyxoviridae (Influenza negative stranded viruses) viruses DNA and RNA Single-stranded Unassigned Ex. Family Retroviridae (Retroviruses, reverse RNA reverse viruses HIV) transcribing transcribing viruses viruses Double-stranded Unassigned Ex. Family Hepadnaviridae (Hepatitis B DNA reverse viruses virus) transcribing virus - The design and discovery of new antiviral drugs can be directed against either viral or cellular targets. Drugs that inhibit viral proteins are more likely to be virus-specific and are more prone to the development of resistance. Thus, there is an urgent need for broad spectrum antiviral drugs that can be used in mono- or combined therapy to treat the numerous viral diseases caused by DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses for which limited or no therapeutic options are currently available. The demand for new effective and safe antiviral strategies has become an increasingly important problem to solve in recent years due to the rising prevalence of chronic viral infections such as HIV and hepatitis B and C, the emergence of new viruses or viral strains such as the SARS coronavirus and pathogenic Avian Influenza strains, the ever-present threat of viral pandemics from agents like virulent strains of influenza A, and the potential danger of hemorrhagic fever viruses and eradicated viruses such as variola virus being exploited as bioterrorist weapons.
- A limited number of chronic viral diseases, which affect millions of people world-wide (e.g., 40 million individuals are HIV-infected), can be controlled to some degree, but no cures are currently available. As a result, infected individuals provide a reservoir for the respective virus through which naïve individuals become infected, thereby, perpetuating the problem of viral infection of significant human populations with these pathogens. Currently available antiviral drugs, such as the nucleoside reverse transcriptase inhibitors (NRTIs) used to inhibit viral replication of specific pathogenic viruses, have resulted in recognizable improvements in the ability to control infections with these pathogens and to improve the quality and length of life of infected individuals. A partial list of currently available antiviral drugs used to inhibit viral replication of specific pathogenic viruses is provided in Appendix 2 (for eg. the NRTIs). The therapies listed have resulted in recognizable improvements in the ability to control infections from these pathogens and to improve the quality and length of life of infected individuals. However, currently available classes of antiviral agents have limited utilities due to their narrow scope of activities against different viruses and/or problems with significant drug-induced toxicities. In addition, the modes of action of the NRTIs and other drugs in current clinical use predispose to the development of drug resistance through viral mutations. Finally, many currently available drugs have considerable side effects that prevent their wide spread use to achieve treatment or prevention goals.
- There is a clear need for new antiviral agents and combination therapies to suppress diseases caused by DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses. The compositions and methods of the preferred embodiments provide such agents and associated methods of treatment.
- Amifostine is a pro-drug that is metabolized by alkaline phosphatase to the reduced free thiol (WR-1065); oxidation of WR-1065 leads to formation of the disulfide (WR-33278). These reactions can be depicted schematically as follows:
- Structural similarities can be observed between spermine (chemical formula H2N(CH2)3NH(CH2)2(CH2)2NH(CH2)3NH2) and WR-1065 and WR-33278. Spermine is associated with nucleic acids and is thought to stabilize helical structure, particularly in viruses. WR-1065 appears to function as an analog of the polyamine spermine, and to compete with spermine for sites on DNA, and probably also on other nucleic acids and proteins. Thus, analogs of WR-1065 and the other compounds of preferred embodiments, as discussed below, may function as spermine or other polyamine analogs, and may mimic the antiviral and polyamine-like activity of WR-1065.
- Thiol metabolites of amifostine are believed to be responsible for most of the cytoprotective and radioprotective properties of amifostine. Amifostine is taken up by cells through a combination of passive and active transport processes where it is metabolized to its active forms; these metabolites participate in a number of functions. These functions include, but are not limited to, (i) protection against radiation induced cytotoxicity and cell killing, (ii) protection against radiation-induced mutagenesis/carcinogenesis, (iii) modulation of topoisomerase I and topoisomerase II alpha activities, (iv) modulation of conformational changes in chromatid structure, (v) inhibition of cell cycle progression, (vi) inhibition of endonuclease activity, (vii) competition with spermine in polyamine transport systems, (viii) induction and repression of gene expression (effect dependent upon the particular gene). Other activities include detoxifying cisplatin and other alkylating agents, scavenging free radicals, modulating apoptosis, and modifying the activity of specific enzymes/proteins.
- To better understand their radioprotective activity, pharmacokinetic studies of amifostine and phosphonol have been performed; because of the stated goal of the studies, tissues known to be sensitive to radiation-induced damage were evaluated most extensively. The results have shown that the drugs are distributed to most normal tissues and to a lower degree to tumor tissue. The highest levels of drug distribution occurred in the following tissues/organ systems (in order from highest to lowest): kidney, liver, salivary gland, heart, spleen, lung, muscle, and brain (bone marrow was also referred to as having high levels, but levels relative to other tissues are not given) (Rasey et al. 1988, Pharmac Ther. 39: 33-43). It has further been demonstrated that certain normal tissues are especially effective at uptake of the drugs (and/or their metabolites) and retention of those metabolites; these tissues include: kidney, liver, salivary gland, and lung (Rasey et al., (1984), Radiat. Res. 97(3): 598-607). It is possible that other tissues that have not been tested also perform like the above mentioned tissues. These studies establish the ability to obtain therapeutic levels of these drugs and/or their metabolites in the cells of these tissues/organ systems. Therapeutic drug levels are also expected to be obtained in tissues or organs systems where drug pharmacokinetics resemble that of the above mentioned tissues.
- Studies with amifostine incorporated into nanoparticles have shown that this preparation technique constitutes an effective mechanism for delivering amifostine to multiple organ systems over a prolonged time period. There was no significant difference in survival of bone marrow progenitor cells in mice treated with 500 mg/kilogram amifostine by either IP injection or by nanoparticles (administered by gavage, equivalent to oral dosing in humans) one hour prior to a dose of 8 to 9 Gy whole-body gamma irradiation. These studies also showed significant protection of jejunal crypt cells following the same exposures. Pharmacokinetic studies of amifostine distribution and retention in a variety of tissues following oral administration of PLGA/amifostine nanoparticles showed high levels of retention in all evaluated tissues 30 minutes after administration. Retention of drug four hours after administration was demonstrated in the following tissues (in order from highest to lowest): liver, jejunum, stomach, ileum, duodenum, bone marrow, and spleen.
- The cytoprotective effects of amifostine appear to be dependent upon a number of factors including, but not limited to, oxygen tension, pH, gene status (including the presence of a functional p53 gene), and enzyme status (including the expression of alkaline phosphatase in the cell membrane). Differences in these factors appear to be responsible for the differential cytoprotective effect mediated by amifostine between tumor cells and normal, nontumorigenic tissue.
- According to one aspect amifostine, phosphonol and their derivatives and analogs are particularly effective in inhibiting replication of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses.
- Accordingly, in a first aspect a method of treating or preventing a DNA virus, a RNA virus and/or a DNA or RNA reverse transcribing virus infection in an individual in need thereof is provided, comprising the step of administering to the individual an effective antiviral amount of a compound or a pharmaceutically acceptable salt or solvate thereof, wherein the compound comprises Formula (I) or Formula (II)
- or a pharmaceutically acceptable salt or solvate thereof, wherein X is selected from the group consisting of —PO3H2, hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl, wherein each of R1, R2, and R3 is independently selected from hydrogen and C1-6 alkyl, and wherein n is an integer of from 1 to 10.
- In an embodiment of the first aspect, the compound is of Formula (I), R1 is methyl, R2 is hydrogen, R3 is hydrogen, and n is 3.
- In an embodiment of the first aspect, the compound is of Formula (I), R1 is methyl, R2 is hydrogen, R3 is hydrogen, and X is hydrogen.
- In an embodiment of the first aspect, the compound is of Formula (I), R1 is hydrogen, R2 is hydrogen, R3 is hydrogen, and n is 3.
- In an embodiment of the first aspect, the compound is of Formula (I), R1 is hydrogen, R2 is hydrogen, R3 is hydrogen, and X is hydrogen.
- In an embodiment of the first aspect, the compound is of Formula (II), R1 is methyl, R2 is hydrogen, R3 is hydrogen, and n is 3.
- In an embodiment of the first aspect, the compound is of Formula (II), R1 is hydrogen, R2 is hydrogen, R3 is hydrogen, and n is 3.
- In an embodiment of the first aspect, the compound is administered to the individual at a daily dosage of from about 200 mg/m2 to about 3000 mg/m2.
- In an embodiment of the first aspect, the step of administering is selected from the group consisting of orally administering, subcutaneously administering, intravenously administering, parenterally administering, and administering by inhalation.
- In an embodiment of the first aspect, the method further comprises the step of administering to the individual an effective antiviral amount of an antiviral drug, for example (see the list of antiviral drugs in Appendix 2) to treat or prevent an viral infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus.
- In a second aspect, a method of treating or preventing a human or animal viral infection caused by a DNA virus, a RNA virus, a DNA or a RNA reverse transcribing virus in an individual in need thereof is provided, comprising the step of administering to the individual an effective antiviral amount of amifostine, the free thiol form of amifostine, the disulfide of amifostine (WR-33278), a combination of both of the free thiol and the disulfide of amifostine, or other structurally and functionally related compounds as described in this document.
- In a third aspect, a method of treating or preventing an infection caused by a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus, or combination thereof in an individual in need thereof is provided, comprising the step of administering to the individual an effective antiviral amount of phosphonol, the free thiol form of phosphonol, the disulfide of phosphonol, a combination of both the free thiol and the disulfide of phosphonol, or other structurally and functionally related compounds as described in this document.
- In an embodiment of the second aspect, the viral infection is caused by a dsDNA virus.
- In an embodiment of the third aspect, the viral infection is caused by a dsDNA virus.
- In an embodiment of the second aspect, the viral infection is caused by a dsRNA virus.
- In an embodiment of the third aspect, the viral infection is caused by a dsRNA virus.
- In an embodiment of the second aspect, the viral infection is caused by a (+)ssRNA virus.
- In an embodiment of the third aspect, the viral infection is caused by a (+)ssRNA virus.
- In an embodiment of the second aspect, the viral infection is caused by a (−)ssRNA virus.
- In an embodiment of the third aspect, the viral infection is caused by a (−)ssRNA virus.
- In an embodiment of the second aspect, the viral infection is caused by a non Retroviridae ssRNA reverse transcribing virus.
- In an embodiment of the third aspect, the viral infection is caused by a non Retroviradae ssRNA reverse transcribing virus.
- In an embodiment of the second aspect, the viral infection is caused by a dsDNA reverse transcribing virus.
- In an embodiment of the third aspect, the viral infection is caused by a dsDNA reverse transcribing virus.
- In an embodiment of the second aspect, the viral infection is caused by a ssDNA reverse transcribing virus.
- In an embodiment of the third aspect, the viral infection is caused by a ssDNA reverse transcribing virus.
- In a fourth aspect, a pharmaceutical kit is provided comprising a pharmaceutical composition comprising a compound or pharmaceutically acceptable salt or solvate thereof in a pharmaceutically acceptable carrier, the compound comprising Formula (I), Formula (II), or a combination thereof:
- wherein X is selected from the group consisting of —PO3H2, hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl, wherein each of R1, R2, and R3 is independently selected from hydrogen and C1-6 alkyl, and wherein n is an integer of from 1 to 10; and
directions for administering the pharmaceutical composition to a patient infected with a DNA virus, an RNA virus, or a DNA or RNA reverse transcribing virus. - In an embodiment of the fourth aspect, the kit further comprises an antiviral drug, selected on the basis of its effectiveness for treating the given viral agent, in a pharmaceutically acceptable carrier.
- In an embodiment of the fourth aspect, the kit further comprises an antiviral drug in a pharmaceutically acceptable carrier and directions for administering the antiviral drug in a pharmaceutically acceptable carrier to the patient.
- The following description and examples illustrate some exemplary embodiments of the disclosed invention in detail. Those of skill in the art will recognize that there are numerous variations and modifications of this invention that are encompassed by its scope. Accordingly, the description of a certain exemplary embodiment should not be deemed to limit the scope of the present invention.
- Amifostine is an organic thiophosphate which selectively protects normal tissues but not tumors against cytotoxicity of ionizing radiations, DNA-binding chemotherapeutic agents (e.g., classical alkylating agents such as cyclophosphamide and non-classical alkylating agents such as mitomycin-C and platinum analogs). Amifostine is a prodrug that is dephosphorylated to the active metabolite, the free thiol form, by alkaline phosphatase and exits the bloodstream rapidly.
- It has surprisingly been discovered that the compounds of preferred embodiments, including amifostine and its derivatives and analogs, are particularly effective antiviral compounds for use in inhibiting replication of diverse species of DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses.
- Amifostine (referred to as “WR-2721”) is marketed under the name Ethyol® by Schering-Plough Pty Ltd. It has the chemical name S-2-(3-aminopropyl)aminoethyl phosphorothioic acid and the structure:
- A particularly preferred antiviral compound for use in inhibiting replication of DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses includes the free thiol form of amifostine. The free thiol form (referred to as “WR-1065”) has the chemical name 2-(3-aminopropylamino)ethanethiol and the following structure:
- The disulfide form of amifostine (referred to as WR-33278) has the chemical name N1,N3,(dithiodiethane-2,1-diyl)dipropane-1,3-diamine
- Phosphonol (referred to as “WR-3689”) is structurally similar to amifostine, the only difference being the presence of a terminal methyl group. Phosphonol has the chemical name S-2-(3-(methylamino)propylamino)ethyl phosphorothioic acid and the following structure:
- The free thiol form of phosphonol (referred to as WR-255591) has the chemical name 2-(3-(methylamino)propylamino) ethanethiol and the following structure:
- The disulfide form of phosphonol (referred to as WR-33278) has the following structure:
- The compounds of preferred embodiments include prodrug forms of the above-described free thiol forms of amifostine, phosphonol, and analogs thereof. Such prodrugs include compounds of the structure:
- wherein each of R1, R2, and R3 is independently selected from hydrogen and lower alkyl,
wherein —(CnH2n)— is lower alkyl and n is 1, 2, 3, 4, 5, or (up to 10), and
wherein X is a suitable leaving group. Suitable leaving groups include —PO3H2, hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl); however, any other suitable leaving group can be employed that yields the active free thiol form of the compound when metabolized in vivo. Other leaving groups can include alkyl groups, e.g., —(C1-6 alkyl), and keto groups, e.g., —C(═O)—(C1-6 alkyl) or —C(═O)—(C6-18 aryl). - The term “lower alkyl” as used herein is a broad term, and is to be given its ordinary and customary meaning to a person of ordinary skill in the art (and is not to be limited to a special or customized meaning), and refers without limitation to a straight chain or branched chain, acyclic or cyclic, saturated aliphatic hydrocarbon containing 1, 2, 3, 4, 5, or 6 carbon atoms. Saturated straight chain lower alkyls include methyl, ethyl, n-propyl, n-butyl, n-pentyl, and n-hexyl; while representative saturated branched chain alkyls include isopropyl, sec-butyl, isobutyl, tert-butyl, and isopentyl. “aryl,” as used herein is a broad term and is used in its ordinary sense, including, without limitation, to refer to an aromatic carbocyclic moiety such as phenyl or naphthyl, including mono-, di-, and poly-homocyclic aromatic ring systems (e.g., C6-18 aryl).
- The prodrug forms described above are metabolized into active thiols of the formula:
- wherein each of R1, R2, R3, —(CnH2n)—, and n is as defined above. In a particularly preferred embodiment, —(CnH2n)— is a straight alkyl chain having three carbon atoms. It is also particularly preferred that R2 and R3 are both hydrogen, and that R1 is hydrogen or methyl.
- In addition to the thiols, certain disulfide forms also exhibit antiviral activity, for example, a disulfide of the following structure:
- can also be employed as an antiviral agent.
- Other antiviral agents of preferred embodiments incorporate a cysteamine-like group (i.e., a group containing the moiety >N—(CH2)2—S—), or a moiety with the structure of WR 2721 or WR-1065, tethered to a DNA or nucleic acid binding agent, or other agents with some affinity for nucleic acids or proteins.
- The compounds of preferred embodiments are particularly effective antiviral agents for monotherapeutic or combined-therapeutic use in treating DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses. The compounds of preferred embodiments are generally administered at dosages equal to or less than the oral radioprotective dosage of amifostine (e.g., 1456 mg total dose, or 910 mg/m2 for a 60 kg body weight (BW) adult human) or phosphonol (e.g., 725 mg/m2). In adults undergoing chemotherapy, the recommended starting dose of amifostine is 910 mg/m2 for a 60 kg BW adult human administered once daily as a 15-minute i.v. infusion, starting 30 minutes prior to chemotherapy. Similar dosing regimens can be employed for use of the compounds of preferred embodiments when used as antiviral agents. Dosages can be converted from mg/m2 to total mg or mg/kg BW (see, e.g., Freireich et al. (1966), Cancer Chemother. Reports, 50 (4) 219-244) as in Table 2.
-
TABLE 2 Body Wt. Body Surface area Species (Kg) (m2) Km factor Mouse 0.2 0.0066 3.0 Rat 0.15 0.025 5.9 Monkey 3.0 0.24 12 Dog 8.0 0.40 20 Human child 20.0 0.80 25 Human adult 60.0 1.60 37 - It is generally preferred to administer amifostine (or other compounds of preferred embodiments) at dosages of 910 mg/m2 or less. This dosage is equivalent to 24.3 mg/kg BW for a 60 kg BW adult human being (or a total dose of 1456 mg for a 60 kg BW adult, as described above); however, in certain embodiments it can be desirable to administer the compounds of preferred embodiments at higher dosage levels. For example, children have been given up to a 2700 mg/m2 total dose of amifostine prior to administration of a chemotherapeutic agent. In some individuals such high doses are associated with side effects. A dose of 740 mg/m2 amifostine (1148 mg total, for a 60 kg BW human adult) is associated with fewer side effects (List et al. (1997), Blood 90(9): 3364-9), and is thus generally preferred. For daily dosing, 200-340 mg/m2 of amifostine (544 mg total dose for a 60 kg BW adult) is generally preferred (Schuchter (1996), Semin Oncol 23(4 Suppl 8): 40-3; Santini et al. (1999), Haematologica 84(11): 1035-42).
- Rodent studies suggest the use of higher dosages. For example, the maximally tolerated dose (MTD) for WR-2721 in mice was 432 mg/kg (BW) administered i.p. and 720 mg/kg BW administered p.o., and the 100% effective radioprotective dose was about one half of the MTD. For phosphonol, the MTD was 893 mg/kg BW administered i.p. and 1488 mg/kg BW administered p.o., and the 100% effective radioprotective dose was about one half of the MTD. All of the aminothiols have MTDs in rodents of greater than 400 mg/kg BW.
- Amifostine and WR-1065 can be efficacious at very low concentrations, for example, down to 0.4 micromolar concentrations in some in vitro studies. In a preferred embodiment, a drug delivery system that obtains relatively constant intracellular concentrations over a period of time that could extend from several days to up to one year or more is desired. To achieve this goal, we anticipate using drug delivery systems demonstrated to achieve relatively constant intracellular drug levels over extended time periods. Examples of such drug delivery systems include subcutaneous administration, formulation in nanoparticles, were formulation in other slow release systems.
- Table 3 provides plasma concentrations of amifostine metabolites (see Geary et al., Res. Comm. Chem. Path. Pharmacol., 65(2), 147-159 (1989)) and phosphonol metabolites (see Buckpitt et al., Contract #DAMD 17-86-C-6177, reference obtained from Dr. D. Grdina, personal communication) after duodenal administration of 150 mg/kg BW of each drug in rhesus macaques. This data was collected as part of work performed to evaluate the radioprotective activity of the compounds. These data may be useful in estimating plasma concentrations in humans.
-
TABLE 3 Time after Concentration Drug Administration (h) (μg/ml) WR-10651 1 6.21 WR-10651 2 4.14 WR-10651 6 2.07 Phosphonol2 1 11.75 Phosphonol2 2 17.08 Phosphonol2 6 15.50 Note: 40 μM WR-1065 = 8.28 μg/ml - While it is generally preferred to formulate amifostine or the other compounds of preferred embodiments for oral administration, the compounds of preferred embodiments can be formulated so as to allow them to be administered by other routes, as discussed herein. It can be desirable in certain embodiments to formulate amifostine for intravenous administration in order to maximize efficacy. Because of the structural similarities between amifostine and phosphonol, especially the similarities in the sulfhydryl ends of the molecules, phosphonol is expected to behave in a manner similar to amifostine rather than WR-151327 (chemical formula CH3NH(CH2)3NH(CH2)3SPO3H2). WR-151327 has been previously shown to have anti-HIV activity; this activity was attributed to the ability of the compound to modulate cytokine levels in a manner that inhibited HIV replication and/or proliferation (Kalebic et al., 1994).
- Phosphonol may not be quite as efficacious as amifostine, based upon the work of Gutschow et al., who found lower activity when a methyl group was substituted for a hydrogen atom in a position similar to that of the methyl group of phosphonol that distinguishes its structure from that of amifostine (Gutschow et al. (1995), Pharmazie 50(10): 672-5). However, phosphonol is expected to be significantly more efficacious than WR-151327. This consideration should be viewed in light of the fact that the overall structures of the compounds tested by Gutschow et al. were significantly different from the structures of the preferred embodiments. The overall structures of the compounds tested by Gutschow et al. are significantly different from the structures of the preferred embodiments, and it is expected that phosphonol will be significantly more efficacious than WR-151327.
- Amifostine, its analogs, and its derivatives can be administered in combination with other antiviral agents employed to treat HIV infection. One of the benefits of such combination therapies is that lower doses of the other antiviral agents can be administered while still achieving a similar level of antiviral efficacy. Such lower dosages can be particularly advantageous for drugs known to have genotoxicity and mitochondrial toxicity (for example, some nucleoside analogs). Conversely, greater efficacy might be achieved using therapeutic doses of two drugs than could be achieved using only a single drug.
- The most common antiviral drugs that can be used in junction with the compounds of preferred embodiments include, but are not limited, to members of the class of drugs known as nucleoside analogs. Other agents that could be used potentially are included in the list of antiviral drugs included in Appendix 2. The use of nucleoside analogs has been associated with a variety of side effects due to the fact that these drugs are analogs of naturally occurring nucleosides, are incorporated into host cell DNA, and function as obligate DNA chain terminators. As such, these drugs are associated with the induction of mutations in host cell DNA, increased risks for certain types of cancer, as well as increased risks for a variety of diseases associated with mutation induction in nuclear and mitochondrial DNA. Nucleoside analogs are also associated with mitochondrial toxicity to a degree that varies depending upon the specific nucleoside analog in question. Thus, it is desirable to be able to minimize the doses of nucleoside analogs used, and/or to use these drugs in combination with other drugs with demonstrated antiviral efficacy that does not compromise the effectiveness of the other treatment, and that also has demonstrated antimutagenic efficacy.
- The use of the compounds of preferred embodiments have been tested in cell culture with one nucleoside analog (zidovudine), and it is been demonstrated that the efficacy of each compound is not diminished when used in combination with the other. It is anticipated that the compounds of preferred embodiments can also be effectively employed with other nucleoside analogs and with other antiviral agents (see Appendix 2).
- Pharmaceutical Compositions Comprising Amifostine and Analogs Thereof: The compounds of preferred embodiments (including derivatives, isomers, metabolites, prodrugs, or pharmaceutically acceptable esters, salts, and solvates thereof) can be incorporated into a pharmaceutically acceptable carrier, including incorporation into nanoparticles for administration to an individual having a viral infection (for example, an adenoviral infection as an example of a DNA virus, an influenza infection as an example of a RNA virus, or an HIV infection as an example of a DNA or RNA reverse transcribing virus) or can be administered prophylactically to prevent viral infection, or postinfection to decrease symptoms upon viral infection. The compounds of preferred embodiments can be employed as the sole agent in the prevention or treatment of DNA viruses, RNA viruses, or DNA or RNA reverse transcribing viruses, and/or combinations thereof two or more such compounds can be employed, optionally in combination with other therapeutic agents, e.g., conventional or newly developed drugs employed in the treatment of AIDS or HIV, or other viral infections.
- Because certain of the compounds of preferred embodiments can be sensitive to oxidation, it can be desirable to administer the compounds in combination with reducing agents including, but not limited to, vitamin C and vitamin E. Other reducing agents include organic aldehydes, hydroxyl-containing aldehydes, and reducing sugars such as glucose, mannose, galactose, xylose, ribose, and arabinose. Other reducing sugars containing hemiacetal or keto groupings can be employed, for example, maltose, sucrose, lactose, fructose, and sorbose. Other reducing agents include alcohols, preferably polyhydric alcohols, such as glycerol, sorbitol, glycols, especially ethylene glycol and propylene glycol, and polyglycols such as polyethylene and polypropylene glycols.
- The terms “pharmaceutically acceptable salts” and “a pharmaceutically acceptable salt thereof” as used herein are broad terms, and are to be given their ordinary and customary meaning to a person of ordinary skill in the art (and are not to be limited to a special or customized meaning), and refer without limitation to salts prepared from pharmaceutically acceptable, non-toxic acids or bases. Suitable pharmaceutically acceptable salts include metallic salts, e.g., salts of aluminum, zinc, alkali metal salts such as lithium, sodium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts; organic salts, e.g., salts of lysine, N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), procaine, and tris; salts of free acids and bases; inorganic salts, e.g., sulfate, hydrochloride, and hydrobromide; and other salts which are currently in widespread pharmaceutical use and are listed in sources well known to those of skill in the art, such as, for example, The Merck Index. Any suitable constituent can be selected to make a salt of the therapeutic agents discussed herein, provided that it is non-toxic and does not substantially interfere with the desired activity. In addition to salts, pharmaceutically acceptable precursors and derivatives of the compounds can be employed. Pharmaceutically acceptable amides, lower alkyl esters, and protected derivatives can also be suitable for use in compositions and methods of preferred embodiments. While it may be possible to administer the compounds of the preferred embodiments in the form of pharmaceutically acceptable salts, it is generally preferred to administer the compounds in neutral form.
- It is generally preferred to administer the compounds of preferred embodiments orally; however, other routes of administration are contemplated. Contemplated routes of administration include but are not limited to oral, sublingual, parenteral, transcutaneous, subcutaneous, intravenous, and by inhalation. Compounds of preferred embodiments can be formulated into liquid preparations for, e.g., oral administration. Suitable forms for such administration include suspensions, syrups, and elixirs.
- The pharmaceutical compositions of preferred embodiments are preferably isotonic with the blood or other body fluid of the recipient. The isotonicity of the compositions can be attained using sodium tartrate, propylene glycol or other inorganic or organic solutes. Sodium chloride is particularly preferred. Buffering agents can be employed, such as acetic acid and salts, citric acid and salts, boric acid and salts, and phosphoric acid and salts. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like. In certain embodiments it can be desirable to maintain the active compound in the reduced state. Accordingly, it can be desirable to include a reducing agent, such as vitamin C, vitamin E, or other reducing agents as are known in the pharmaceutical arts, in the formulation.
- Viscosity of the pharmaceutical compositions can be maintained at the selected level using a pharmaceutically acceptable thickening agent. Methylcellulose is preferred because it is readily and economically available and is easy to work with. Other suitable thickening agents include, for example, xanthan gum, carboxymethyl cellulose, hydroxypropyl cellulose, carbomer, and the like. The preferred concentration of the thickener will depend upon the thickening agent selected. An amount is preferably used that will achieve the selected viscosity. Viscous compositions are normally prepared from solutions by the addition of such thickening agents.
- A pharmaceutically acceptable preservative can be employed to increase the shelf life of the pharmaceutical compositions. Benzyl alcohol can be suitable, although a variety of preservatives including, for example, parabens, thimerosal, chlorobutanol, or benzalkonium chloride can also be employed. A suitable concentration of the preservative is typically from about 0.02% to about 2% based on the total weight of the composition, although larger or smaller amounts can be desirable depending upon the agent selected. Reducing agents, as described above, can be advantageously used to maintain good shelf life of the formulation.
- The compounds of preferred embodiments can be in admixture with a suitable carrier, diluent, or excipient such as sterile water, physiological saline, glucose, or the like, and can contain auxiliary substances such as wetting or emulsifying agents, pH buffering agents, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired. See Standard texts, such as “Remington: The Science and Practice of Pharmacy”, Lippincott Williams & Wilkins; 20th edition (Jun. 1, 2003) and “Remington's Pharmaceutical Sciences,” Mack Pub. Co.; 18th and 19th editions (December 1985, and June 1990, respectively). Such preparations can include complexing agents, metal ions, polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, dextran, and the like, liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts. Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. The presence of such additional components can influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance, and are thus chosen according to the intended application, such that the characteristics of the carrier are tailored to the selected route of administration.
- For oral administration, the pharmaceutical compositions can be provided as a tablet, aqueous or oil suspension, dispersible powder or granule, emulsion, hard or soft capsule, syrup or elixir. Compositions intended for oral use can be prepared according to any method known in the art for the manufacture of pharmaceutical compositions and can include one or more of the following agents: sweeteners, flavoring agents, coloring agents and preservatives. Aqueous suspensions can contain the active ingredient in admixture with excipients suitable for the manufacture of aqueous suspensions.
- Formulations for oral use can also be provided as hard gelatin capsules, wherein the active ingredient(s) are mixed with an inert solid diluent, such as calcium carbonate, calcium phosphate, or kaolin, or as soft gelatin capsules. In soft capsules, the active compounds can be dissolved or suspended in suitable liquids, such as water or an oil medium, such as peanut oil, olive oil, fatty oils, liquid paraffin, or liquid polyethylene glycols. Stabilizers and microspheres formulated for oral administration can also be used. Capsules can include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredient in admixture with fillers such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In instances where it is desirable to maintain a compound of a preferred embodiment in a reduced form (in the case of certain active metabolites), it can be desirable to include a reducing agent in the capsule.
- Tablets can be uncoated or coated by known methods to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period of time. For example, a time delay material such as glyceryl monostearate can be used. When administered in solid form, such as tablet form, the solid form typically comprises from about 0.001 wt. % or less to about 50 wt. % or more of active ingredient(s), preferably from about 0.005, 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1 wt. % to about 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, or 45 wt. %.
- Tablets can contain the active ingredients in admixture with non-toxic pharmaceutically acceptable excipients including inert materials. For example, a tablet can be prepared by compression or molding, optionally, with one or more additional ingredients. Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active or dispersing agent. Molded tablets can be made by molding, in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
- Preferably, each tablet or capsule contains from about 10 mg or less to about 1,000 mg or more of a compound of the preferred embodiments, more preferably from about 20, 30, 40, 50, 60, 70, 80, 90, or 100 mg to about 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, or 900 mg. Most preferably, tablets or capsules are provided in a range of dosages to permit divided dosages to be administered. A dosage appropriate to the patient and the number of doses to be administered daily can thus be conveniently selected. In certain embodiments it can be preferred to incorporate two or more of the therapeutic agents to be administered into a single tablet or other dosage form (e.g., in a combination therapy); however, in other embodiments it can be preferred to provide the therapeutic agents in separate dosage forms.
- Suitable inert materials include diluents, such as carbohydrates, mannitol, lactose, anhydrous lactose, cellulose, sucrose, modified dextrans, starch, and the like, or inorganic salts such as calcium triphosphate, calcium phosphate, sodium phosphate, calcium carbonate, sodium carbonate, magnesium carbonate, and sodium chloride. Disintegrants or granulating agents can be included in the formulation, for example, starches such as corn starch, alginic acid, sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite, insoluble cationic exchange resins, powdered gums such as agar, karaya or tragacanth, or alginic acid or salts thereof.
- Binders can be used to form a hard tablet. Binders include materials from natural products such as acacia, tragacanth, starch and gelatin, methyl cellulose, ethyl cellulose, carboxymethyl cellulose, polyvinyl pyrrolidone, hydroxypropylmethyl cellulose, and the like.
- Lubricants, such as stearic acid or magnesium or calcium salts thereof, polytetrafluoroethylene, liquid paraffin, vegetable oils and waxes, sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol, starch, talc, pyrogenic silica, hydrated silicoaluminate, and the like, can be included in tablet formulations.
- Surfactants can also be employed, for example, anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate, cationic such as benzalkonium chloride or benzethonium chloride, or nonionic detergents such as polyoxyethylene hydrogenated castor oil, glycerol monostearate, polysorbates, sucrose fatty acid ester, methyl cellulose, or carboxymethyl cellulose.
- Controlled release formulations can be employed wherein the amifostine or analog(s) thereof is incorporated into an inert matrix that permits release by either diffusion or leaching mechanisms. Slowly degenerating matrices can also be incorporated into the formulation. Other delivery systems can include timed release, delayed release, or sustained release delivery systems.
- Coatings can be used, for example, nonenteric materials such as methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methylhydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl-methyl cellulose, sodium carboxy-methyl cellulose, providone and the polyethylene glycols, or enteric materials such as phthalic acid esters. Dyestuffs or pigments can be added for identification or to characterize different combinations of active compound doses.
- When administered orally in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils can be added to the active ingredient(s). Physiological saline solution, dextrose, or other saccharide solution, or glycols such as ethylene glycol, propylene glycol, or polyethylene glycol are also suitable liquid carriers. The pharmaceutical compositions can also be in the form of oil-in-water emulsions. The oily phase can be a vegetable oil, such as olive or arachis oil, a mineral oil such as liquid paraffin, or a mixture thereof. Suitable emulsifying agents include naturally-occurring gums such as gum acacia and gum tragacanth, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from fatty acids and hexitol anhydrides, such as sorbitan mono-oleate, and condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan mono-oleate. The emulsions can also contain sweetening and flavoring agents.
- When a compound of the preferred embodiments is administered by intravenous, parenteral, or other injection, it is preferably in the form of a pyrogen-free, parenterally acceptable aqueous solution or oleaginous suspension. Suspensions can be formulated according to methods well known in the art using suitable dispersing or wetting agents and suspending agents. The preparation of acceptable aqueous solutions with suitable pH, isotonicity, stability, and the like, is within the skill in the art. A preferred pharmaceutical composition for injection preferably contains an isotonic vehicle such as 1,3-butanediol, water, isotonic sodium chloride solution, Ringer's solution, dextrose solution, dextrose and sodium chloride solution, lactated Ringer's solution, or other vehicles as are known in the art. In addition, sterile fixed oils can be employed conventionally as a solvent or suspending medium. For this purpose, any bland fixed oil can be employed including synthetic mono or diglycerides. In addition, fatty acids such as oleic acid can likewise be used in the formation of injectable preparations. The pharmaceutical compositions can also contain stabilizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.
- The duration of the injection can be adjusted depending upon various factors, and can comprise a single injection administered over the course of a few seconds or less, to 0.5, 0.1, 0.25, 0.5, 0.75, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours or more of continuous intravenous administration.
- The antiviral compositions of the preferred embodiments can additionally employ adjunct components conventionally found in pharmaceutical compositions in their art-established fashion and at their art-established levels. Thus, for example, the compositions can contain additional compatible pharmaceutically active materials for combination therapy (such as supplementary antimicrobials, antipruritics, astringents, local anesthetics, anti-inflammatory agents, reducing agents, and the like), or can contain materials useful in physically formulating various dosage forms of the preferred embodiments, such as excipients, dyes, thickening agents, stabilizers, preservatives or antioxidants.
- The compounds of the preferred embodiments can be provided to an administering physician or other health care professional in the form of a kit. The kit is a package which houses a container which contains the compound(s) in a suitable pharmaceutical composition, and instructions for administering the pharmaceutical composition to a subject. The kit can optionally also contain one or more additional therapeutic agents, e.g., antiviral nucleoside analog, peptide analog, protease inhibitor, monoclonal antibody, or any other antiviral used for the treatment or prevention of virus disease (see for example Appendix 2). For example, a kit containing one or more compositions comprising compound(s) of the preferred embodiments in combination with one or more additional antiviral, antibacterial, and/or anti-infective agents can be provided, or separate pharmaceutical compositions containing a compound of the preferred embodiments and additional therapeutic agents can be provided. The kit can also contain separate doses of a compound of the preferred embodiments for serial or sequential administration. The kit can optionally contain one or more diagnostic tools and instructions for use. The kit can contain suitable delivery devices, e.g., syringes, and the like, along with instructions for administering the compound(s) and any other therapeutic agent. The kit can optionally contain instructions for storage, reconstitution (if applicable), and administration of any or all therapeutic agents included. The kits can include a plurality of containers reflecting the number of administrations to be given to a subject. In a particularly preferred embodiment, a kit for the treatment of DNA viruses, RNA viruses, and DNA and RNA reverse transcribing viruses, and combinations thereof is provided that includes amifostine or another compound of a preferred embodiment and one or more antiviral agents currently employed to treat the virus. Antiviral agents include for example, nucleoside analogs such as acyclovir, reverse transcriptase inhibitors such as zidovudine, didanosine, zalcitabine, stavudine, 3TC; non-nucleoside reverse transcriptase inhibitors; protease inhibitors; cytokines; immunomodulators, and antibodies but are not limited thereto.
- The compounds of the preferred embodiments can be administered prophylactically for the prevention of DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection. Alternatively, therapy is preferably initiated as early as possible following the onset of signs and symptoms of a viral infection, or following exposure to a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus infection. The administration route, amount administered, and frequency of administration will vary depending on the age of the patient, the severity of the infection, and any associated conditions. Contemplated amounts, dosages, and routes of administration for the compounds of preferred embodiments for treatment of a DNA virus, a RNA virus, or a DNA or RNA reverse transcribing virus infection are similar to those established for conventional antiviral agents. Detailed information relating to administration and dosages of conventional antiviral agents can be found in the Physician's Desk Reference, 47th edition. This information can be adapted in designing treatment regimes utilizing the compounds of preferred embodiments. It is expected that anti-viral therapies combining an aminothiol with another antiviral therapy will have an additive or synergistic activity, making it possible to reduce the doses of the drugs currently used to treat DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection, or making it possible to achieve higher efficacy by using therapeutic doses of two or more agents together. The combination of an aminothiol with a nucleoside analog is expected to simultaneously provide improved antiviral therapy and protection against nucleoside analog-induced side effects, especially those associated with nuclear DNA or mitochondrial DNA damage. It is further anticipated that an aminothiol could protect against deleterious side effects of another antiviral drugs, especially those that induce nuclear DNA or mitochondrial DNA damage.
- According to one embodiment, contemplated amounts of the compounds of the preferred embodiments for oral administration to treat DNA virus, RNA virus, or DNA or RNA reverse transcribing virus infection are from about 10 mg or less to about 2000 mg or more administered from about every 4 hours or less to about every 6 hours or more (or from about 4 times daily to about 6 times daily) for about 5 days or less to about 10 days or more (40 mg/day or less to about 15,000 mg/day or more) or until there is a significant improvement in the condition. For suppressive therapy to inhibit the onset of infection in susceptible individuals, doses of from about 10 mg or less to about 1000 mg or more are orally administered once, twice, or multiple times a day, typically for up to about 12 months, or, in certain circumstances, indefinitely (from about 10 mg/day to about 1,000 mg/day). When treatment is long term, it can be desirable to vary the dosage, employing a higher dosage early in the treatment, and a lower dosage later in the treatment.
- The single highest dose of amifostine administered to an adult human as documented in the literature was 1330 mg/m2. Children have been administered single doses of amifostine of up to 2700 mg/m2 with no untoward effects. The literature indicates that multiple doses (up to three times the recommended single dose of 740 to 910 mg/m2) have been safely administered within a 24-hour period. Repeated administration of amifostine at two and four hours after the initial dose does not appear to result in an increase in side effects, especially nausea, vomiting, or hypotension. It appears that the most significant deleterious side effect from the administration of amifostine is hypotension.
- According to one embodiment, contemplated amounts of the compounds of the preferred embodiments, methods of administration, and treatment schedules for individuals with infections caused by DNA viruses, RNA viruses and DNA and RNA reverse transcribing viruses are generally similar to those described above for the prevention of and/or treatment of radiation- or chemotherapy-induced cytotoxicity; they may also be similar to those used for the treatment of myelodysplastic syndrome.
- Known side effects of amifostine include decrease in systolic blood pressure, nausea, and vomiting. If such side effects are observed for the particular thiophosphate administered, it is generally preferred to administer an antiemetic medication prior to, or in conjunction with the thiophosphate. Suitable antiemetic medications include antihistamines (e.g., buclizine, cyclizine, dimenhydrinate, diphenhydramine, meclizine), anticholinergic agents (e.g., scopolamine), dopamine antagonists (e.g., chlorpromazine, droperidol, metoclopramide, prochlorperazine, promethazine), serotonin antagonists (e.g., dolasetron, granisetron, ondansetron), or other agents (e.g., dexamethasone, methylprednisolone, trimethobenzamide).
- The purity of the WR-1065 used in the experiments described below is unknown. WR-1065 is sensitive to oxidation (and possibly other reactions) and can undergo reaction to forms that appear to lack antiviral activity. Accordingly, in the tables below and in the preferred dosages provided above, the indicated concentrations represent the maximum amount of active compound that could be present; the true concentration of active compound is less than that indicated but cannot be determined definitively. In addition, for concentrations of WR-1065 of less than 100 μM, estimates of the true efficacy of the compound are limited by the fact that the compound has been found to be inactivated by a variety of medium components (Grdina et al. (2000), Drug Metabol Drug Interact 16(4): 237-79.). Below approximately 50 μM, this problem becomes especially severe.
- The impact of WR1065 treatment on replication on lung prototypic adenovirus strains in A549 cell monolayers is illustrated in Table 4. A549 or MDCK cells were used for the plaque reduction assays. Cells were grown to near confluence in Minimal Essential Medium (MEM) in six-well plates. Cells were infected with 100 plaque forming units (PFUs) of virus of virus in 100 μl and adsorption was allowed for one hour at 37° C. Cell monolayers were subsequently overlaid with 0.6% agarose in MEM. The overlay medium was prepared to contain a final concentration of 10 μM, 33 μM and 100 μM amount of WR 1065. Each concentration was assayed in triplicate. Wells overlaid with media without WR-1065 were set up as 100% infectious virus yield controls. Cultures were maintained at 37° C. in 5% CO2 for 7 days. At the end of this time period, cell cultures were fixed with 1% formaldehyde and stained with Crystal Violet for visualization of plaques and evaluation of plaque size and number. Reduction of plaque formation was expressed as a percentage of the untreated controls. Subsequently, WR 1065 was demonstrated to inhibit viral replication and development of cytotopathic effect in a dose-dependent manner in influenza A or influenza B-infected MDCK cell monolayers. Using an approach similar to that described above, MDCK cells growing in 6-well plates were infected with 100 plaque forming units (PFUs) of virus in 100 μl and adsorption was allowed for one hour at 37° C. Cell monolayers were subsequently overlaid with 0.6% agarose in MEM. The overlay medium was prepared to contain a final concentration of 10 μM, 33 μM and 100 μM amount of WR 1065. Each concentration was assayed in triplicate. Wells overlaid with media without WR-1065 were set up as 100% infectious virus yield controls. Cultures were maintained at 37° C. in 5% CO2 for 3 days. At the end of this time period, cell cultures were fixed with 1% formaldehyde and stained with Crystal Violet for visualization of plaques and evaluation of plaque size and number. Reduction of plaque formation was expressed as a percentage of the untreated controls.
-
TABLE 4 Mean infectious virus yields (PFUs × 106) 96-h post infection & WR 1065 treatmenta Susceptible 0 μM <10 μM <33 μM <100 μM Virus Species populations Trial # WR 1065 WR 1065 WR 1065 WR 1065 Ad7-p B Pediatric & military; 1 3.5 1.85 0.85 1.15 (strain respiratory infection 2 58.2 — — 30.5 Gomen) Ad5-p C Immuno-compromised; 1 12.5 9.0 5.65 2.25 (strain upper respiratory 2 665 — 600 500 Adenoid 3 1,920 — 1,660 — 75) Ad4-p E Military; conjunctivitis & 1 1,900 — 1,420 980 (strain RI- respiratory infection 2 22.5 — — 9.5 67) - Table 5 includes data from an experiment to measure WR-1065 protection against cytopathic effects in MDCK cell monolayers infected with influenza A/Puerto Rico/8/34 (H1N1), using densitometry readings of the stained cell monolayers to estimate relative cell survival. Similar results were obtained in a second independent experiment using influenza A/Puerto Rico/8/34 (H1N1), and in an experiment using the same approach with influenza B/Lee/40. In a third experiment with influenza A, WR 1065 was added to an agarose overlay following a one-hour adsorption with a different virus strain, A/HKx31 (H3N2), and a plaque reduction assay was performed (Table 5). The results illustrate protection of MDCK cells from Influenza A by WR 1065, based upon densitometry readings (experiment 1) or a plaque reduction assay (experiment 2)
- According to one aspect of the present invention, the reduced form of amifostine (WR 2721 or ethyol), was the most active moiety in the observed antiviral effects. In another aspect of the present invention, WR-1065, was the most active moiety in the observed antiviral effects. A common medium component (phenylalanine) blocks the activity of WR 1065 to some undetermined degree.
-
TABLE 5 Influenza A strain; Amount of WR 1065ª Quantitative approach 0 μM <10 μM <33 μM <100 μM Experiment 1 A/Puerto 9.57 ± 12.4 ± 13.1 ± 14.3 ± Rico/8/34(H1N1); 0.3 × 105 0.4 × 105 1.0 × 105 1.5 × 105 Densitometry readingb Experiment 2 A/HKx31(H3N2); 66.3 59.3 57 48 Mean plaque number Densitometry readings = mean counts/mm2 ± standard deviation using Quantity One (Bio-Rad) plate reader. - Protection of A549 cells from the cytopathic effects of Adenovirus 5p by WR-1065 as assessed by a plaque reduction assay is illustrated in Table 6.
-
TABLE 6 Amount of WR1065 0 μM 16.5 μM 50 μM Adenovirus 5p 64 63 41 (36% reduction) - Experiments were conducted to determine the antiretroviral effects of AZT in the presence of WR-1065, the active metabolite of amifostine. Peripheral blood mononuclear cells (PBMCs) obtained from the NIH Blood Bank were cultured with phytohemagglutinin (PHA) for 48 hours to create PHA blasts. The cells were cultured in RPMI-1640 medium with 10% fetal bovine serum (FBS), 1% penicillin/streptomycin and glutamine, and 10% Interleukin-2 (IL-2). The PHA blasts were then infected with HIV for two hours (see Perno et al. (1988), J. Exptl. Med. 168:111; Aquaro et al. (1988), J. Medical Virology 68:479-488).
- Noting the caveats outlined above regarding purity of the WR-1065 used, WR-1065 (from the NCI Chemical Carcinogen Repository; see Hoffman et al. (2001), Env. Mol. Mut. 37:117) was weighed (Mol. Wt. 134) and stored frozen in RPMI-1640 medium as a 10 mg/ml solution (13.4 μL of the solution added to 1 ml of RPMI-1640 yields 1000 μM solution). AZT (SIGMA, Mol. Wt. 267) was dissolved in phosphate-buffered saline (PBS) at a concentration of 36 mM (9.72 mg/ml), 0.01 ml (97 μg) of the resulting solution was added to 3.6 ml RPMI-1640 medium to yield a 26.9 μg/ml solution, and 0.1 ml (2.69 μg) of that resulting solution was added to 1 ml of RPMI-1640 medium to yield a 10 μM solution of AZT. The HIV-infected PHA blasts were incubated with 10 μM AZT in the presence or absence of 1000 μM WR-1065 prepared as described above.
- At 72 hours, HIV infection status was monitored in five experimental groups by measuring p24 using an ELISA kit (RETRO-TEK HIV-1, p24 Extended Range ELISA, ZMC catalog #0801137). The HIV infection status for the five experimental groups is provided in Table 3 (see Experiment #1 data).
-
TABLE 7 Estimated 10 μM HIV P24 percent viral Treatment Group AZT WR1065 pg/ml inhibition Experiment #1 PHA blasts No No 0.28 N.A. PHA blasts + HIV No No 498.62 N.A. PHA blasts + HIV Yes No 0.13 ~100% PHA blasts + HIV Yes Yes (1000 μM) 0.13 ~100% PHA blasts + HIV No Yes (1000 μM) 0.13 ~100% Experiment #2 PHA blasts No No 8.99 N.A. PHA blasts + HIV No No 176.45 N.A. PHA blasts + HIV Yes No 9.07 94.9% PHA blasts + HIV No Yes (1000 μM) 8.92 94.9% PHA blasts + HIV No Yes (330 μM) 9.31 94.7% PHA blasts + HIV No Yes (100 μM) 8.92 94.9% Experiment #3 PHA blasts No No 3.0 N.A. PHA blasts + HIV No No 44,455.8 N.A. PHA blasts + HIV Yes No 2.9 ~100% PHA blasts + HIV No Yes (66 μM) 33.2 99.9% PHA blasts + HIV No Yes (33 μM) 503.2 98.9% PHA blasts + HIV No Yes (10 μM) 911.2 98.0% PHA blasts + HIV No Yes (5 μM) 10,293.7 76.8% - Table 7 illustrates the antiviral efficacy of AZT and WR-1065 on PHA blasts. Subsequent experiments confirmed this antiretroviral effect, and demonstrated that the ability of WR 1065 to inhibit viral replication was dose-dependent and comparable in concentration and effect to that seen with AZT alone using this in vitro assay system.
- The results of these experiments illustrate that WR-1065 and some structurally-related aminothiols function as broad-spectrum antiviral agents. Taken together, these results further support the hypothesis that the antiviral activity of WR-1065 is due to its ability to bind to specific sites on viral RNA, because the only known element common to the tested viruses is their requirement for the formation and use of RNA during viral replication (Table 7).
- Experiments were conducted to determine the viability of cells exposed to various concentrations of WR-1065. Tests were conducted on PHA blasts cultured as described for the previous experiments, but were not incubated in the presence of HIV. WR-1065 was prepared from a 10 mg/ml solution in RPMI-1640 medium and diluted to the desired concentrations. Cell viability was measured 72 hours after exposure to WR-1065. Percentage of viable cells (compared to unexposed controls) is provided in Table 8.)
-
TABLE 8 WR-1065 Concentration % Viable Cells (μM) (compared to control) 1000 30 500 73 100 63 50 89 10 90 5 82 1 83
The test results demonstrated acceptable cell viability for all concentrations tested, and particularly good cell viability for concentrations of 50 μM and lower. - Cysteamine (chemical formula H2NCH2CH2SH) has been demonstrated to have anti-HIV activity (Ho et al. (1995), AIDS Res Hum Retroviruses 11(4): 451-9; Bergamini et al. (1996), J Infect Dis 174(1): 214-8). Using an in vitro assay system as described above, 200 μM cysteamine effectively suppressed (˜100%) HIV replication. In comparison, WR-1065 effectively suppressed (>99.9%) HIV replication at a concentration of less than 100 μM. Thus, the in vitro anti-HIV activity of WR-1065 is over 2-fold higher than that of cysteamine. In addition, cysteamine's duration of action was determined to be very short; to achieve an anti-HIV effect, fresh cysteamine had to be added to the cell culture system every 12 hours. WR-1065, however, had a long duration of action—it only had to be added to the culture system once in a 72 hour period.
- Cystamine (chemical formula H2N(CH2)2SS(CH2)2NH2), the oxidized form of cysteamine, has been demonstrated to have anti-HIV activity, DNA binding capacity, radioprotective capacity, and the ability to shift the equilibrium of DNA from the A-form towards the B-form (Allegra et al. (2002), Amino Acids 22(2): 155-66). WR-1065, the active form of WR-2721, has also been shown to bind to DNA in the minor groove, and also to shift the B/A-DNA equilibrium towards the B-form. Accordingly, it can be inferred that cystamine and its reduced form cysteamine are capable of binding to other nucleic acids in addition to DNA, and have some ability to bind to and/or to interact with proteins. It should be noted that the anti-HIV activity of cystamine is considered to be due, at least in large part, to its rapid in vivo conversion to cysteamine.
- A comparison of the DNA phosphate binding capacity of cystamine versus WR-1065 demonstrated that these two compounds have similar binding affinities under similar in vitro conditions (Smoluk et al. (1986), Radiat Res. 107(2):194-204). However, WR-1065 is a larger molecule than cysteamine because it contains an additional moiety: —(CH2)3NH2. Thus, it can be hypothesized that WR-1065 can bind to and thereby block a larger fragment of a nucleic acid than cysteamine. It can therefore be hypothesized that the differential binding characteristic of WR-1065 versus cysteamine may be responsible for WR-1065's improved antiviral efficacy. It is possible that WR-1065's antiviral activity is due in part to its ability to bind to and to block critical sites on nucleic acids and/or proteins. (Allegra et al. (2002), Amino Acids 22(2): 155-66; North et al., (2002), Mol. Carcinog. 33(3): 181-8). Blockage of nucleic acid sites and/or proteins is considered to be a possible mechanism for the aminothiols' modulation of enzyme function (Brekken et al. (1986), J Biol Chem 273(41): 26317-22). The mechanism by which an aminothiol could bind to and/or block nucleic acids and/or proteins without inducing significant cytotoxicity to eukaryotic cells is unknown at this time.
- As further evidence of the potential importance of WR-1065's nucleic acid binding capacity to its antiviral activity, it has been demonstrated that WR-1065 has antiviral efficacy against three different species of adenovirus and two different strains of influenza. These are dramatically different types of viruses with significantly different modes of replication. One common replication element shared by all of these viruses, as well as HIV, is a requirement for single or double-stranded RNA during part of the replication cycle. Thus, the demonstration of broad-spectrum antiviral activity supports the hypothesis that the ability of the antiviral agent WR-1065 to bind to RNA plays a role in the observed antiviral effect.
- While not wishing to be bound by any particular theory, it is believed that one or more of the compounds of the present invention function as an antiviral agent because its structure renders it bi-functional, combining two distinct properties. For example, the —(CH2)3NH2 portion of WR-1065 is believed to be responsible for binding to nucleic acids (DNA, RNA), and possibly also to some proteins. A second portion of WR-1065 —NH(CH2)2SH is believed to be largely responsible for the antiretroviral/antiviral effects that have been observed To be effective against some viruses, it is believed that the sulfhydryl group needs to be in the reduced state in order for antiretroviral/antiviral effects to be observed for the compound; however, it is possible that the sulfhydryl group may remain functional if oxidized to the disulfide (as in WR-33278). Both of these components contribute to maximizing antiretroviral/antiviral effects of the compounds of preferred embodiments (e.g., both the —(CH2)3NH2 and the —NH(CH2)2SH moieties of amifostine). According to one embodiment, the first portion of the molecule functions to align the molecule in close proximity to critical binding sites on nucleic acids and/or proteins, and the second portion of the molecule functions in a reaction that contributes to antiviral efficacy.
- The above hypothesis for the mode of action of WR-1065 and related compounds is in part based upon several compounds have been found described to inhibit replication of multiple different families of viruses (Qian-Cutrone et al., 1996, Hamasaki and Ueno, 2001, Lacourciere et al. 2000, Li et al. 2001, Nishizono and Nair, 2000, Zang and Yen, 1999, Reddy et al. 1999, Xiao et al., 2001). These compounds were hypothesized to bind to and/or otherwise block post-transcriptional regulatory elements such as the REV response element (RRE) of HIV-1 or the PRE binding site of hepatitis B virus. These elements are required for export of viral mRNA from the nucleus to the cytoplasm, formation of viral proteins, and assembly of viral components. Some compounds are hypothesized to bind directly to the RRE or PRE; others are hypothesized to interfere with protein-RNA binding through indirect mechanisms. Most of the studied compounds were known to be structurally related to the broad category of cellular compounds known as polyamines and/or to have polyamine-like functions. Polyamines are ubiquitous, naturally occurring compounds found in all cells that are hypothesized to bind to DNA and RNA and to be involved in gene expression regulation; thus, the hypothesized mode of action for the antiviral effects of these agents is consistent with their structure and what is known about their naturally occurring counterparts. It should be noted that amifostine and its analogs differ significantly in structure from the above described compounds.
- Variations to the chemical structure of the moiety —(CH2)3NH2 may be tolerated without losing or altering the antiretroviral/antiviral efficacy of the compound, so long as the resulting structure retains a binding capacity/affinity that is similar to that observed with —(CH2)3NH2 itself. Such variations may include more or less than three carbon atoms in the alkyl group, and a branched alkyl chain, lower alkyl substituents on the amino group. The potential importance of the moiety —(CH2)3NH2 (or suitable variant) to antiviral effectiveness of the compounds of preferred embodiments is supported by the work of Laayoun et al., who demonstrated a significant increase in anti-mutagenic activity when cysteamine or WR-2721 is tethered to a chromophore (quinoline or acridine) that increases DNA binding (Laayoun et al. (1994), Int J Radiat Biol 66(3): 259-66). It is hypothesized that the antimutagenic activity of the aminothiols may result from a mode of action that is also relevant for their antiviral activity, suggesting that viral affinity could be altered by changing the chemical structure of the moiety —(CH2)3NH2.
- Gutschow et al. describe two compounds that are demonstrated to display significant antiretroviral/antiviral activity (Gutschow et al. (1995), Pharmazie 50(10): 672-5.). These compounds differ in structure considerably from the compounds of preferred embodiments, but the two most efficacious compounds have, as a portion of their structure, a cysteamine-like group. Gutschow et al. tested a number of compounds, some of which differed in the number of carbon atoms separating the sulfhydryl group and its adjacent amino group, and noted that the optimal antiviral effect was observed using molecules that had a cysteamine-like group and that this effect was diminished if the number of —CH2— groups separating the sulfhydryl group and the first amino group was increased to three, as occurs in the aminothiols WR-151327 (chemical formula CH3NH(CH2)3NH(CH2)3SPO3H2) and WR-151326 (chemical formula CH3NH(CH2)3NH(CH2)3SH). Accordingly, altering the structure of the cysteamine-like group to include three —CH2— groups between the sulfhydryl group and the amino group significantly reduced the capacity of the compound to function as an antiretroviral/antiviral agent.
- In contrast to WR-151327 and WR-151326, other compounds of preferred embodiments have only two —CH2— groups separating the sulfhydryl group from the first amino group, and thus possess superior antiretroviral/antiviral properties. The antiretroviral/activity of WR-1065 is significantly greater than that reported for WR-151326, and thus WR-1065 has functional capacities that are different from those of WR-151327 and WR-151326.
- Significant questions remain as to how a molecule such as WR-1065 could interact with viral nucleic acids and proteins in such a way as to inhibit critical processes (such as replication, transcription, translation, or protein aggregation/functionality) without simultaneously exerting a similar effect upon cells of higher organisms. Some information from early work performed with viral genomes as models of DNA replication can be used to formulate testable hypotheses.
- We hypothesize that for example, WR-1065 and its analogs interfere with critical components of the viral life cycle of many/all viruses through interactions with viral nucleic acids, nucleic acid-associated proteins, and/or molecular machines involved in nucleic acid and/or protein production and maintenance. We further hypothesize that fundamental differences in nucleic acid and protein production and maintenance of viruses versus higher organisms are responsible for the differential effects of WR-1065 and its analogs. The rationale for these hypotheses is supported by the following information.
- Experimental evidence supports the hypothesis that the nucleic acids of higher organisms have a protein-to-DNA ratio of approximately 1:10. In contrast, the nucleic acids of many viruses, vegetative bacteriophage, and dinoflagellates have a protein-to-DNA ratio of approximately 1:1 (Cremisi, 1979; Kellenberger, 1988). The significantly higher ratio of proteins to nucleic acids of viruses would allow for enhanced interaction between proteins associated with nucleic acids, the nucleic acids themselves, and a molecule with nucleic acid and protein binding affinity. The enhanced opportunity for binding between WR-1065 and viral nucleic acids and proteins could result in deleterious effects upon viral nucleic acid production, maintenance, or functionality. A similar effect would not be expected to be exerted at the same magnitude upon the nucleic acids of higher organisms. The significantly lower ratio of proteins to nucleic acids in higher organisms versus viruses would be expected to reduce the opportunities for interaction/binding between WR-1065, nucleic acids, and adjacent proteins in higher organisms by a degree roughly comparable to the differences in the ratios of protein to nucleic acid found in these two different life forms.
- In addition, there are considerable differences in the types and amounts of proteins associated with viral nucleic acids as opposed to the nucleic acids of higher organisms (Cremisi, 1979). These protein-based distinctions could result in further differences in the number or types of interactions between WR-1065 and components of viruses versus cellular components of higher organisms.
- Other work suggests that WR-1065 could affect the functionality of molecular machines. For the purposes of illustration, only the molecular machines known as origin recognition complexes will be discussed, but the considerations presented below are applicable to many/all molecular machines of viruses versus higher organisms. Because the components and structure of these machines differ between simple life forms and higher life forms, WR-1065-associated effects could result in different consequences. The basis for this hypothesis comes from two separate areas of consideration. The replicon model as proposed by Jacob et al. in 1964 has been shown to be applicable to all bacterial replication systems as well as to several viral families, including simian virus-40 (SV-40). Bergsma et al. (1982) showed that initiation of replication in SV-40 required the interaction of a virally encoded initiator protein with a defined replicator sequence. In contrast to the study results, replication initiation-site selection within eukaryotic chromosomes is considered to be degenerate (Gilbert, 2004). For the latter chromosomes, multiple different replication initiation sites appear to exist, and no single obligatory site has been found.
- Thus, it can be hypothesized that WR-1065 and similar compounds could block replication initiation through interactions with a defined replicator sequence on viral nucleic acids, while not exerting a similar effect upon the nucleic acids of higher organisms because of the degenerate nature of their initiation-site selection. Binding of WR-1065 to replication initiation sites of eukaryotic cells would be hypothesized to slow down nucleic acid processing and, hence, cell division, but not block it completely. This hypothesis is supported by the fact that WR-1065 has been shown to delay the progression of cells through the cell cycle, but the exact mechanism remains unclear.
- Second, replication origin recognition complexes of higher animals share many similarities with analogous complexes in simple life forms, including viruses and archaeal cells (Gai et al., 2004). However, critical differences have been noted in the components and in the mode of action of these molecular machines (Gai et al., 2004). To use SV-40 as an example, the molecular machine known as the origin recognition complex consists of the oncogenic large tumor antigen that is formed as a monomer; six monomers then form a hexamer, and two hexamers assemble together, along with replication protein A, topoisomerase I, and polymerase-alpha/primase. These molecules form the replication initiation complex for SV-40. The functionality of this complex is highly dependent upon the conformation of individual monomers, as well as the conformation of the hexamer (Gai et al. 2004). The functionality of the entire replication initiation complex is also dependent upon the conformation of the target nucleic acids and the target initiation site (Gai et al., 2004). Because WR-1065 is known to have the capacity to alter the conformation of both nucleic acids and proteins, a complex such as that described for SV-40 would be especially vulnerable to induced conformational changes. Partial support for this hypothesis comes from studies demonstrating interactions between WR-1065 and topoisomerase enzymes. Analogous molecular machines that operate in cells of higher organisms would not be expected to share identical conformational requirements.
- In addition, because origin recognition complexes of eukaryotic cells do not appear to require an origin of replication site, but rather are capable of using a variety of sites, it is reasonable to hypothesize that these latter origin recognition complexes could potentially tolerate a degree of conformational change that SV-40 and other virally-associated origin recognition complexes cannot tolerate.
- All references cited herein, including but not limited to published and unpublished applications, patents, and literature references, as well as the references of the Appendix 1, are incorporated herein by reference in their entirety and are hereby made a part of this specification. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material. The term “comprising” as used herein is synonymous with “including,” “containing,” or “characterized by,” and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps.
- All numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth herein are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of any claims in any application claiming priority to the present application, each numerical parameter should be construed in light of the number of significant digits and ordinary rounding approaches.
- The above description discloses several methods and materials of the present invention. This invention is susceptible to modifications in the methods and materials, as well as alterations in the fabrication methods and equipment. Such modifications will become apparent to those skilled in the art from a consideration of this disclosure or practice of the invention disclosed herein. Consequently, it is not intended that this invention be limited to the specific embodiments disclosed herein, but that it cover all modifications and alternatives coming within the true scope and spirit of the invention.
-
- Allegra et al. (2002). “The ability of cystamine to bind DNA.” Amino Acids 22(2): 155-66.
- Bergamini et al. (1996). “In vitro inhibition of the replication of human immunodeficiency virus type 1 by beta-mercaptoethylamine (cysteamine).” J Infect Dis 174(1): 214-8.
- Brekken et al. (1998). “Trypanosoma brucei gamma-glutamylcysteine synthetase. Characterization of the kinetic mechanism and the role of Cys-319 in cystamine inactivation.” J Biol Chem 273(41): 26317-22.
- Clark et al. (1997). “The aminothiol WR-1065 protects T-lymphocytes from ionizing radiation-induced deletions of the HPRT gene.” Cancer Epidemiol. Biomarkers and Prevention 6:1033.
- Cremisi, C. (1979). Chromatin replication revealed by studies of animal cells and papovaviruses (simian virus 40 and polyoma virus). Microbiol Rev 43(3): 297-319.
- Gai, D., R. Zhao, D. Li, C. V. Finkielstein and X. S. Chen (2004). Mechanisms of conformational change for a replicative hexameric helicase of SV40 large tumor antigen. Cell 119(1): 47-60.
- Grdina et al. (2000). “Amifostine: mechanisms of action underlying cytoprotection and chemoprevention.” Drug Metabol Drug Interact 16(4): 237-79.
- Gutschow et al. (1995). “[Bis((2,4-dioxo-1,2,3,4-tetrahydroquinazolin-3-yl)alkyl)disulfane and 3-(mercaptoalkyl)quinazolin-2,4(1H,3H)-dione: synthesis by ring transformation and antiviral activity. 42. Heterocyclic azines with heteroatoms in the 1- and 3-positions].” Pharmazie 50(10): 672-5.
- Hamasaki et al. (2001). “Aminoglycoside antibiotics, neamine and its derivatives as potent inhibitors for the RNA-protein interactions derived from HIV-I activators.” Bioorg Med Chem Lett 11(4):591-4.
- Ho et al. (1995). “Cystamine inhibits HIV type 1 replication in cells of monocyte/macrophage and T cell lineages.” AIDS Res Hum Retroviruses 11(4): 451-9.
- Kalebic et al. (1994). “Organic thiophosphate WR-151327 suppresses expression of HIV in chronically-infected cells.” AIDS Research and Human Retroviruses 10:727.
- Kellenberger, E. (1988). About the □immer□ation of condensed and decondensed non-eukaryotic DNA and the concept of vegetative DNA (a critical review). Biophys Chem 29(1-2): 51-62.
- Laayoun et al. (1994). “Aminothiols linked to quinoline and acridine chromophores efficiently decrease 7,8-dihydro-8-oxo-2′-deoxyguanosine formation in gamma-irradiated DNA.” Int J Radiat Biol 66(3): 259-66.
- Lacourciere et al. (2000). “Mechanism of neomycin and Rev peptide binding to the Rev responsive element of HIV-1 as determined by fluorescence and NMR spectroscopy.” Biochemistry 39(19):5630-41.
- Li et al. (2001). “A heterocyclic inhibitor of the REV-RRE complex binds to RRE as a □immer.” Biochemistry 40(5):1150-8.
- List et al. (1997). “Stimulation of hematopoiesis by amifostine in patients with myelodysplastic syndrome.” Blood 90(9): 3364-9.
- Luedtke et al. (2003). “Fluorescence-based methods for evaluating the RNA affinity and specificity of HIV-1 Rev-RRE inhibitors.” Biopolymers 70(1):103-19.
- Newton et al. (1996). “Transport of aminothiol radioprotectors into mammalian cells: passive diffusion versus mediated uptake.” Radiat Res 146(2):206-15.
- Nguyen et al. (2003). “Amifostine and curative intent chemoradiation for compromised cancer patients.” Anticancer Research 23:1649.
- Nishizono et al. (2000). “Synthesis of biomimetic analogs of neomycin B: potential inhibitors of the HIV RNA Rev response element.” Nucleosides Nucleotides Nucleic Acids 19(1-2):283-95.
- North et al. (2002). “Restoration of wild-type conformation and activity of a temperature sensitive mutant of p53 (p53(V272M)) by the cytoprotective aminothiols WR1065 in the esophageal cancer cell line TE-1.” Mol. Carcinog. 33(3): 181-8.
- Oiry et al. (2004). “Synthesis and biological evaluation in human monocyte-derived macrophages of N—(N-acetyl-L-cysteinyl)-S-acetylcysteamine analogues with potent antioxidant and anti-HIV activities.” J Med Chem 47(7): 1789-95.
- Olsen et al. (1990). “Interaction of the human immunodeficiency virus type 1 Rev protein with a structured region in env mRNA is dependent on multimer formation mediated through a basic stretch of amino acids.” Genes Dev 4(8):1357-64.
- Perno et al. (1988). “Inhibition of HIF-1 replication in fresh and cultured human peripheral blood monocytes by azidothymidine.” J Experimental Medicine 168:1111.
- Qian-Cutrone et al. (1996). “Niruriside, a new HIV REV/RRE binding inhibitor from Phyllanthus niruri.” J Nat Prod 59(2):196-9.
- Rasey, J. S., N. J. Nelson, P. Mahler, K. Anderson, K. A. Krohn and T. Menard (1984). Radioprotection of normal tissues against gamma rays and cyclotron neutrons with WR-2721: LD50 studies and 35S-WR-2721 biodistribution. Radiat Res 97(3): 598-607.
- Rasey, J. S., A. M. Spence, C. C. Badger, K. A. Krohn, D. M. Vera and J. C. Livesey (1988). Specific protection of different normal tissues. Pharmacol Ther 39(1-3): 33-43.
- Reddy et al. (1999). “Inhibition of HIV replication by dominant negative mutants of Sam68, a functional homolog of HIV-1.” Rev Nat Med 5(6):635-42.
- Rossio et al. (1998). “Inactivation of HIV Type I infectivity with preservation of conformational and functional integrity of virion surface proteins.” Journal of Virology 72:7992.
- Santini et al. (1999). “The potential of amifostine: from cytoprotectant to therapeutic agent.” Haematologica 84(11): 1035-42.
- Schuchter, L. M. (1996). “Guidelines for the administration of amifostine.” Semin Oncol 23(4 Suppl 8): 40-3.
- U.S. Pat. No. 5,824,664 to Schein et al. “Suppression of HIV Expression by Organic Thiophosphate”.
- Xiao et al. (2001). “Inhibition of the HIV-I rev-RRE complex formation by unfused aromatic cations.” Bioorg Med Chem 9(5):1097-113.
- Zang et al. (1999). “Distinct export pathway utilized by the hepatitis B virus posttranscriptional regulatory element.” Virology 259(2):299-304.
- Zapp et al. (1997). “Modulation of the Rev-RRE interaction by aromatic heterocyclic compounds.” Bioorg Med Chem 5(6):1149-55.
-
APPENDIX 2 Viruses Drug Chemical Type Target/Mode Herpes virus Vidarabine (Vira-A) NRTI Viral polymerase Acyclovir (Zovirax) NRTI Viral polymerase Famcylovir (Famvir) Idoxuridine (Herplex liquifilm, Viral polymerase stoxil) Trifluridine (Viroptic) Inhibitor of thymidylate synthetase Tromantadine Derivate of Inhibits viral replication adamantane Penciclovir (Denavir) Interferes with enzymes for viral replication Docosanol (Abreva) Long chain alcohol Changes cell membrane of healthy cells Valacyclovir (Valtrex) Imiquimod (Aldara) Cytomegalo Gancyclovir (Cytovene) NRTI Viral polymerase virus Valganciclovir (Valcyte) NRTI Viral polymerase Fomivirsen (Vitravene, ISIS Antisense Prevents production of viral 2922) oligonucleotide protein Foscarnet (Foscavir) Cidofovir (Vistide, HPMPC) NRTI Viral polymerase Retrovirus Zidovudine (AZT) NRTI Reverse transcriptase Didanosine (ddI) NRTI Reverse transcriptase Zalcitabine (ddC) NRTI Reverse transcriptase Stavudine (d4T) NRTI Reverse transcriptase Lamivudine (3TC) NRTI Reverse transcriptase Abacavir (ABC) NRT Reverse transcriptase Emtricitabine (Emtriva, FTC) NRTI Reverse transcriptase Tenofovir (Viread) NRTI Reverse transcriptase Enfuvirtide (Fuzeon, T-20) Binds to HIV envelope glycoprotein gp41 prevents viral fusion with Nevirapine (Viramune) NNRTI Reverse transcriptase Delavirdine (Resciprtor) NNRTI Reverse transcriptase Efavirenz (Sustiva) NNRTI Reverse transcriptase Saquinavir (Invirase) Peptide analog Protease inhibitor Ritonavir (Norvir) Peptide analog Protease inhibitor Indinavir (Crixivan) Peptide analog Protease inhibitor Nelfinavir (Viracept) Peptide analog Protease inhibitor Amprenavir (Agenerase, Peptide analog Protease inhibitor VX-478, 141W94) Lopinavir (Kaletra) Peptide analog Protease inhibitor Atazanavir (Reyataz) Peptide analog Protease inhibitor Tipranavir (Aptivus) Peptide analog Protease inhibitor Fosamprenavir (Lexiva) Peptide analog Protease inhibitor Influenza Thiovir Adventrx virus pharmaceuticals: influenza A and avian/ human influenza virus Thiophosphono- formates Amantadine (Symmetrel) Influenza A strains: Matrix protein/haemagglutinin tricyclic amine Rimatadine (Flumadine) Influenza A strains: Matrix protein/haemagglutinin tricyclic amine Zanamivir (Relenza) Influenza A and B Neuraminidase inhibitor strains: neuraminic acid mimetic Oseltamivir (Tamiflu,GS4104) Influenza A and B Neuraminidase inhibitor strains: neuraminic acid mimetic Peramivir (BCX-1812, Influenza A and B RWJ270201) strains Hepatitis FDA approved (below) Adefovir dipivoxil (Preveon) NRTI Reverse transcriptase Interferon alfa-2a (Roferon) Protein Activates cell defense proteins Interferon alfa-2b (Intron-A) Protein Activates cell defense proteins Interferon alfa-n3 (Alferon) Protein Activates cell defense proteins Interfernon alfacon (Infergen) Protein Activates cell defense proteins Peginterferon alfa-2a (PEG- Polyethylene glycol Intron) Peginterferon alfa-2b Polyethylene glycol (Pegasys) FDA approved vaccines Hepatitis A Hepatitis A and B Hepatitis B immune globulin Hepatitis B Hepatitis B and Haemophilus influenza type b polysaccharide conjugate Hep B Investigational drugs (below) BAM-205 (NOV-205) Clevudine (L-FMAU) Pyrimidine analog Elvucitabine (ACH-126, 443, NRTI Reverse transcriptase BetaL-Fd4C) Telbivudine (L- NRTI Suppresses viral replication-- deoxythymidine, LdT) polymerase Entecavir (Baraclude) NRTI Suppresses viral replication HepeX-B Monoclonal Polymerase inhibitor antibodies Pradefovir (Remofovir) Valtorcitabine (Val-LdC) Valine ester Inhibits replication HE2000(Immunitin) Alph-epibromide Immune regulating hormone Zadaxin (Thymalfasin) Amino acid peptide Stimulates immune system Hep C Investigational drugs (below) AbXTL68 (XLT6865) Monoclonal antibody NS5B RNA polymerase Inhibits viral polymerase MN283 MN107 BILN-2061 Protease inhibitor FK788 Leflunomide analog HepeX-C Monoclonal antibodies Histamine dihydrochloride Histamine (Ceplene) Interferon gamma-1b Protein Anti-fibrotic (Actimmune) Isatoribine (ANA245) NRTI Polymerase inhibitor ISIS 14803 Phosphorothioate Inhibits replication and protein antisense expression oligonucleotide Levovirin (LVV) L-enantiomer of ribavirin--NRTI Merimempodib (VX-497) Phenyloxazole IMPDH inhibitor derivative Mycophenolate mofetil Immunosuppressive drug (Cellcept) Thymosin alpha 1 (TA1) Synthetic peptide Increases T cell function Viramidine (ICN3142) Amidine prodrug of ribavirin-- NRTI ANA971 Prodrug of isatoribine ANA245 (Isatoribine) REBIF Beta interferon Infergen/Consensus Interfernon Oral Interferon alpha IP-501 Oral phospholipids antifibrotics ANA975 Prodrug of isatoribine EMZ702 Rituximab (Rituxan, Anti-CD20 Mabthera) monoclonal antibody NM283 (Valopicitabine) Polymerase (RdRp) inhibitor HCV/MF59 Enhances immune response SCH-6 Serine protease inhibitor HCV-796 Non-nucleoside Polymerase inhibitor Civacir (Hepititis C Immune Polyclonal antibody Globulin) E-1 Prostaglandins- therapeutic vaccine Tarvacin Anti- Stimulates immune defense phosphotidylserine JTK 103 Protease inhibitor AVI-4065 (Neugene) Phosphorodiamidate morpholion oligomer Omega Interferon Multiferon Multi-subtype natural human alpha interferon Alinia (nitrazoxanide) Thiazolides IDN-6556 Capase inhibitor Albuferon Fusion protein interferon alpha- albumin Medusa Interferon Long acting interferon IC41 Therapeutic vaccine VX 950 Protease inhibitor (NS3-4A) CPG 10101 (Actilon) Immunomodulator BIVN-401 (Virastat) Monoclonal antibodies MX-3253 (Celgosivir) Prodrug of Alpha-glucosidase I inhibitor castanospermine SCH 503034 Inhibits serine protease HCV-AB68 HCV-MF59 Actilon Synthetic Selective TLR9 agonist oligonucleotide Enbrel Anti-tumor necrosis factor alpha BLX-883 Locteron-interferon AVI-4065 HCV-086 R803 Non-nucleoside Polymerase inhibitor Interleukin-10 Anti-fibrotic Heptazyme RNA inhibitor Rhinovirus Rupintrivir (AG7088) Pfizer Inc: Protease inhibitor Experimental--- BTA-798 Biota: Experimental--- MRL-2471 Rhinovirus/ Pleconaril Blocks attachment and Picornavirus uncoating Picornavirus/ WIN51711 Bind to viral capsid to prevent Poliovirus uncoating WIN Broad Rep9 Spectrum Ampligen Ribaviran (Rebetol, Rebetron) Triazol carboxamide RNA mutagen/IMPDH inhibitor
Claims (13)
1. A method of treating a virus infection in an individual in need thereof, comprising:
administering to the individual an effective antiviral amount of a compound or a pharmaceutically acceptable salt or solvate thereof, wherein the compound is of Formula (I) or Formula (II)
wherein X is selected from the group consisting of —PO3H2, hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl,
wherein each of R1, R2, and R3 is independently selected from hydrogen and C1-6 alkyl,
wherein n is an integer of from 1 to 10, and
wherein the virus is a DNA virus, an RNA virus, a DNA reverse transcribing virus or a non Retroviridae RNA reverse transcribing virus.
2. The method of claim 1 , wherein R1 is methyl, R2 is hydrogen, R3 is hydrogen, n is 3, and X is —PO3H2.
3. The method of claim 1 , wherein R1 is methyl, R2 is hydrogen, R3 is hydrogen, n is 3, and X is hydrogen.
4. The method of claim 1 , wherein R1 is hydrogen, R2 is hydrogen, R3 is hydrogen, n is 3, and X is —PO3H2.
5. The method of claim 1 , wherein R1 is hydrogen, R2 is hydrogen, R3 is hydrogen, n is 3, and X is hydrogen.
6. The method of claim 1 , wherein the compound is administered to the individual at a daily dosage of from about 200 mg/m2 to about 3000 mg/m2.
7. The method of claim 1 , wherein the step of administering is selected from the group consisting of orally administering, intravenously administering, parenterally administering, subcutaneously administering, and administering by inhalation.
8. A method of treating a virus infection in an individual in need thereof, comprising:
administering to the individual an effective antiviral amount of:
(i) amifostine, the free thiol form of amifostine, the disulfide of amifostine (WR-33278), a combination of both the free thiol and the disulfide of amifostine, or other structurally and functionally related compounds; or
(ii) phosphonol, the free thiol form of phosphonol, the disulfide of phosphonol, a combination of both the free thiol and the disulfide of phosphonol, or other structurally and functionally related compounds,
wherein the virus is a DNA virus, an RNA virus, a DNA reverse transcribing virus, or a non Retroviridae RNA reverse transcribing virus.
9. (canceled)
12. A pharmaceutical kit comprising:
a pharmaceutical composition comprising a compound or pharmaceutically acceptable salt or solvate thereof in a pharmaceutically acceptable carrier, the compound having Formula (I) or Formula (II):
wherein X is selected from the group consisting of —PO3H2, hydrogen, acetyl, isobutyryl, pivaloyl, and benzoyl,
wherein each of R1, R2, and R3 is independently selected from hydrogen and C1-6 alkyl, and
wherein n is an integer of from 1 to 10; and
directions for administering the pharmaceutical composition to a patient infected with a DNA virus, an RNA virus, a DNA reverse transcribing virus or a non Retroviridae RNA reverse transcribing virus.
13. The method of claim 1 , wherein the virus is a member of the Adenoviridae family or the Orthomyxoviridae family.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US17/959,512 US20230277566A1 (en) | 2006-04-17 | 2022-10-04 | Broad spectrum antiviral and methods of use |
Applications Claiming Priority (7)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US79273806P | 2006-04-17 | 2006-04-17 | |
US11/736,471 US9585849B2 (en) | 2006-04-17 | 2007-04-17 | Broad spectrum antiviral and methods of use |
US15/434,682 US9849143B2 (en) | 2006-04-17 | 2017-02-16 | Broad spectrum antiviral and methods of use |
US15/854,184 US20180344751A1 (en) | 2006-04-17 | 2017-12-26 | Broad Spectrum Antiviral and Methods of Use |
US16/399,186 US20200093841A1 (en) | 2006-04-17 | 2019-04-30 | Broad Spectrum Antiviral and Methods of Use |
US17/009,881 US20210228601A1 (en) | 2006-04-17 | 2020-09-02 | Broad Spectrum Antiviral and Methods of Use |
US17/959,512 US20230277566A1 (en) | 2006-04-17 | 2022-10-04 | Broad spectrum antiviral and methods of use |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US17/009,881 Continuation US20210228601A1 (en) | 2006-04-17 | 2020-09-02 | Broad Spectrum Antiviral and Methods of Use |
Publications (1)
Publication Number | Publication Date |
---|---|
US20230277566A1 true US20230277566A1 (en) | 2023-09-07 |
Family
ID=43625760
Family Applications (6)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US11/736,471 Active 2030-11-15 US9585849B2 (en) | 2006-04-17 | 2007-04-17 | Broad spectrum antiviral and methods of use |
US15/434,682 Active US9849143B2 (en) | 2006-04-17 | 2017-02-16 | Broad spectrum antiviral and methods of use |
US15/854,184 Abandoned US20180344751A1 (en) | 2006-04-17 | 2017-12-26 | Broad Spectrum Antiviral and Methods of Use |
US16/399,186 Abandoned US20200093841A1 (en) | 2006-04-17 | 2019-04-30 | Broad Spectrum Antiviral and Methods of Use |
US17/009,881 Abandoned US20210228601A1 (en) | 2006-04-17 | 2020-09-02 | Broad Spectrum Antiviral and Methods of Use |
US17/959,512 Abandoned US20230277566A1 (en) | 2006-04-17 | 2022-10-04 | Broad spectrum antiviral and methods of use |
Family Applications Before (5)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US11/736,471 Active 2030-11-15 US9585849B2 (en) | 2006-04-17 | 2007-04-17 | Broad spectrum antiviral and methods of use |
US15/434,682 Active US9849143B2 (en) | 2006-04-17 | 2017-02-16 | Broad spectrum antiviral and methods of use |
US15/854,184 Abandoned US20180344751A1 (en) | 2006-04-17 | 2017-12-26 | Broad Spectrum Antiviral and Methods of Use |
US16/399,186 Abandoned US20200093841A1 (en) | 2006-04-17 | 2019-04-30 | Broad Spectrum Antiviral and Methods of Use |
US17/009,881 Abandoned US20210228601A1 (en) | 2006-04-17 | 2020-09-02 | Broad Spectrum Antiviral and Methods of Use |
Country Status (1)
Country | Link |
---|---|
US (6) | US9585849B2 (en) |
Families Citing this family (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9585849B2 (en) | 2006-04-17 | 2017-03-07 | The Burlington Hc Research Group, Inc. | Broad spectrum antiviral and methods of use |
US20130337046A1 (en) * | 2012-06-14 | 2013-12-19 | Dale M. Walker | Methods for Improved Delivery of Aminothiols, Dimers of Aminothiols, and Heterodimers Composed of Aminothiols |
CN108495625B (en) * | 2015-11-17 | 2021-07-27 | 柏林顿Hc研究集团有限公司 | Methods for improved protection and delivery of aminothiols and analogs thereof |
WO2021231421A1 (en) * | 2020-05-13 | 2021-11-18 | The Regents Of The University Of California | Thiol-containing compounds for use in treating coronavirus |
JP2023527038A (en) * | 2020-05-25 | 2023-06-26 | ノバビオティクス・リミテッド | antiviral therapy |
WO2023028316A1 (en) * | 2021-08-27 | 2023-03-02 | The Burlington Hc Research Group, Inc. | Aminothiol-conjugates, compositions, and methods of use thereof |
Family Cites Families (11)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0473673A4 (en) | 1989-05-24 | 1995-05-17 | Us Bioscience | Method for protection from azt side effects and toxicity |
SG47101A1 (en) | 1992-07-31 | 1998-03-20 | Us Bioscience | Crystalline amifostine compositions and methods for the preparation and use of same |
US5424471A (en) | 1992-07-31 | 1995-06-13 | U.S. Bioscience, Inc. | Crystalline amifostine compositions and methods of the preparation and use of same |
US5824664A (en) | 1993-03-26 | 1998-10-20 | U.S. Bioscience, Inc. | Suppression of HIV expression by organic thiophosphate |
US5994409A (en) | 1997-12-09 | 1999-11-30 | U.S. Bioscience, Inc. | Methods for treatment of neuro--and nephro--disorders and therapeutic toxicities using aminothiol compounds |
US6489312B1 (en) * | 1999-06-15 | 2002-12-03 | Medimmune Oncology, Inc. | Pharmaceutical formulations comprising aminoalkyl phosphorothioates |
IT1313579B1 (en) | 1999-07-30 | 2002-09-09 | Acraf | PARACETAMOL-BASED LIQUID PHARMACEUTICAL COMPOSITION. |
ITMI20012135A1 (en) | 2001-10-16 | 2003-04-16 | Bioren S A | INJECTABLE SOLUTIONS READY TO USE PARACETAMOL |
US9585849B2 (en) | 2006-04-17 | 2017-03-07 | The Burlington Hc Research Group, Inc. | Broad spectrum antiviral and methods of use |
EP2010166B1 (en) | 2006-04-17 | 2013-11-06 | The Government of the United States of America as represented by the Secretary of the Department of Health and Human Services | Organic disulfide antiretroviral agents |
US20130337046A1 (en) | 2012-06-14 | 2013-12-19 | Dale M. Walker | Methods for Improved Delivery of Aminothiols, Dimers of Aminothiols, and Heterodimers Composed of Aminothiols |
-
2007
- 2007-04-17 US US11/736,471 patent/US9585849B2/en active Active
-
2017
- 2017-02-16 US US15/434,682 patent/US9849143B2/en active Active
- 2017-12-26 US US15/854,184 patent/US20180344751A1/en not_active Abandoned
-
2019
- 2019-04-30 US US16/399,186 patent/US20200093841A1/en not_active Abandoned
-
2020
- 2020-09-02 US US17/009,881 patent/US20210228601A1/en not_active Abandoned
-
2022
- 2022-10-04 US US17/959,512 patent/US20230277566A1/en not_active Abandoned
Also Published As
Publication number | Publication date |
---|---|
US20110053894A1 (en) | 2011-03-03 |
US9849143B2 (en) | 2017-12-26 |
US9585849B2 (en) | 2017-03-07 |
US20180344751A1 (en) | 2018-12-06 |
US20210228601A1 (en) | 2021-07-29 |
US20200093841A1 (en) | 2020-03-26 |
US20170157151A1 (en) | 2017-06-08 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US9849143B2 (en) | Broad spectrum antiviral and methods of use | |
IL193148A (en) | Use of combinations of agents for the preparation of medicaments for treatment of hepatitis b virus | |
JP2023547599A (en) | Phospholipid compounds and their uses | |
AU2009223064B2 (en) | Use of 4'-Thio-2'-Deoxynucleosides as Anti Orthopoxvirus Agents | |
US20230322813A1 (en) | Compounds and methods for treatment of viral infections | |
US20160374967A1 (en) | Organic thiophosphate antiretroviral agents | |
KR20010075192A (en) | Antiviral Combinations | |
US9181294B2 (en) | Nucleoside analogues for the treatment of a viral infection, and method for evaluating the sensitivity to said treatment | |
Morris-Natschke et al. | Phospholipid analogs against HIV-1 infection and disease | |
US20050113321A1 (en) | DAPD combination therapy with inosine monophosphate dehydrogenase inhibitor | |
WO2011046901A2 (en) | Broad spectrum antiviral and methods of use | |
AU2002258368A1 (en) | DAPD combination therapy with IMDPH inhibitors such as ribavirin or mycophenolic acid |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
STPP | Information on status: patent application and granting procedure in general |
Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |
|
STCB | Information on status: application discontinuation |
Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION |