WO2006019841A2 - S - Google Patents
S Download PDFInfo
- Publication number
- WO2006019841A2 WO2006019841A2 PCT/US2005/024893 US2005024893W WO2006019841A2 WO 2006019841 A2 WO2006019841 A2 WO 2006019841A2 US 2005024893 W US2005024893 W US 2005024893W WO 2006019841 A2 WO2006019841 A2 WO 2006019841A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- hiv
- cells
- subject
- cell
- egr
- Prior art date
Links
- 239000012190 activator Substances 0.000 title claims abstract description 62
- 101100444898 Mus musculus Egr1 gene Proteins 0.000 title claims abstract description 41
- 238000011282 treatment Methods 0.000 title abstract description 28
- 208000036142 Viral infection Diseases 0.000 title description 5
- 230000009385 viral infection Effects 0.000 title description 4
- 150000001875 compounds Chemical class 0.000 claims abstract description 102
- 230000014509 gene expression Effects 0.000 claims description 115
- 238000000034 method Methods 0.000 claims description 102
- 230000010076 replication Effects 0.000 claims description 65
- 230000002101 lytic effect Effects 0.000 claims description 58
- 101710205625 Capsid protein p24 Proteins 0.000 claims description 55
- 101710177166 Phosphoprotein Proteins 0.000 claims description 55
- 101710149279 Small delta antigen Proteins 0.000 claims description 55
- 102100022563 Tubulin polymerization-promoting protein Human genes 0.000 claims description 55
- CXQWRCVTCMQVQX-LSDHHAIUSA-N (+)-taxifolin Chemical group C1([C@@H]2[C@H](C(C3=C(O)C=C(O)C=C3O2)=O)O)=CC=C(O)C(O)=C1 CXQWRCVTCMQVQX-LSDHHAIUSA-N 0.000 claims description 23
- 230000007420 reactivation Effects 0.000 claims description 20
- 208000031886 HIV Infections Diseases 0.000 claims description 19
- 239000003814 drug Substances 0.000 claims description 19
- 208000037357 HIV infectious disease Diseases 0.000 claims description 17
- 239000003795 chemical substances by application Substances 0.000 claims description 17
- 208000033519 human immunodeficiency virus infectious disease Diseases 0.000 claims description 17
- 230000004913 activation Effects 0.000 claims description 16
- 238000012360 testing method Methods 0.000 claims description 13
- 230000001965 increasing effect Effects 0.000 claims description 12
- 230000008569 process Effects 0.000 claims description 12
- KQNGHARGJDXHKF-UHFFFAOYSA-N dihydrotamarixetin Natural products C1=C(O)C(OC)=CC=C1C1C(O)C(=O)C2=C(O)C=C(O)C=C2O1 KQNGHARGJDXHKF-UHFFFAOYSA-N 0.000 claims description 11
- 230000003213 activating effect Effects 0.000 claims description 8
- 239000003443 antiviral agent Substances 0.000 claims description 8
- 239000000137 peptide hydrolase inhibitor Substances 0.000 claims description 7
- 230000036436 anti-hiv Effects 0.000 claims description 6
- 239000003419 rna directed dna polymerase inhibitor Substances 0.000 claims description 6
- 229940124597 therapeutic agent Drugs 0.000 claims description 6
- 230000000527 lymphocytic effect Effects 0.000 claims description 5
- 229940122440 HIV protease inhibitor Drugs 0.000 claims description 3
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 claims description 3
- 239000004030 hiv protease inhibitor Substances 0.000 claims description 3
- 239000002777 nucleoside Substances 0.000 claims description 3
- 150000003833 nucleoside derivatives Chemical class 0.000 claims description 3
- 238000012216 screening Methods 0.000 claims description 3
- 229940042402 non-nucleoside reverse transcriptase inhibitor Drugs 0.000 claims description 2
- 239000002726 nonnucleoside reverse transcriptase inhibitor Substances 0.000 claims description 2
- 241000700605 Viruses Species 0.000 abstract description 28
- 208000029462 Immunodeficiency disease Diseases 0.000 abstract description 12
- 210000004027 cell Anatomy 0.000 description 313
- 108090000623 proteins and genes Proteins 0.000 description 146
- 230000006698 induction Effects 0.000 description 71
- 241000725303 Human immunodeficiency virus Species 0.000 description 63
- LUKBXSAWLPMMSZ-OWOJBTEDSA-N Trans-resveratrol Chemical compound C1=CC(O)=CC=C1\C=C\C1=CC(O)=CC(O)=C1 LUKBXSAWLPMMSZ-OWOJBTEDSA-N 0.000 description 51
- 230000037361 pathway Effects 0.000 description 51
- QNVSXXGDAPORNA-UHFFFAOYSA-N Resveratrol Natural products OC1=CC=CC(C=CC=2C=C(O)C(O)=CC=2)=C1 QNVSXXGDAPORNA-UHFFFAOYSA-N 0.000 description 45
- 235000021283 resveratrol Nutrition 0.000 description 42
- 229940016667 resveratrol Drugs 0.000 description 42
- 230000001105 regulatory effect Effects 0.000 description 31
- 239000000523 sample Substances 0.000 description 30
- 230000001413 cellular effect Effects 0.000 description 29
- 239000000203 mixture Substances 0.000 description 29
- 230000000694 effects Effects 0.000 description 28
- 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 28
- 229960002555 zidovudine Drugs 0.000 description 27
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 24
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 23
- PHEDXBVPIONUQT-UHFFFAOYSA-N Cocarcinogen A1 Natural products CCCCCCCCCCCCCC(=O)OC1C(C)C2(O)C3C=C(C)C(=O)C3(O)CC(CO)=CC2C2C1(OC(C)=O)C2(C)C PHEDXBVPIONUQT-UHFFFAOYSA-N 0.000 description 22
- 238000002474 experimental method Methods 0.000 description 20
- PHEDXBVPIONUQT-RGYGYFBISA-N phorbol 13-acetate 12-myristate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(OC(C)=O)C1(C)C PHEDXBVPIONUQT-RGYGYFBISA-N 0.000 description 20
- 108020004999 messenger RNA Proteins 0.000 description 18
- 238000002493 microarray Methods 0.000 description 18
- 230000029812 viral genome replication Effects 0.000 description 17
- 230000008859 change Effects 0.000 description 15
- 238000004519 manufacturing process Methods 0.000 description 14
- 238000003753 real-time PCR Methods 0.000 description 14
- 206010061598 Immunodeficiency Diseases 0.000 description 13
- 230000003612 virological effect Effects 0.000 description 13
- 208000030507 AIDS Diseases 0.000 description 11
- 238000004458 analytical method Methods 0.000 description 11
- 230000000875 corresponding effect Effects 0.000 description 11
- 238000001514 detection method Methods 0.000 description 11
- 230000007813 immunodeficiency Effects 0.000 description 11
- 208000015181 infectious disease Diseases 0.000 description 11
- 239000013610 patient sample Substances 0.000 description 11
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 10
- FWPWHHUJACGNMZ-LIPLLEKLSA-N (1r,2s)-5-(1-hydroxy-2-methylpropyl)-2-methyl-7-oxa-4-azabicyclo[3.2.0]heptane-3,6-dione Chemical compound N1C(=O)[C@@H](C)[C@H]2OC(=O)C21C(O)C(C)C FWPWHHUJACGNMZ-LIPLLEKLSA-N 0.000 description 9
- 229940079593 drug Drugs 0.000 description 9
- 239000000975 dye Substances 0.000 description 9
- 238000009472 formulation Methods 0.000 description 9
- 239000004615 ingredient Substances 0.000 description 9
- 238000002372 labelling Methods 0.000 description 9
- 239000007788 liquid Substances 0.000 description 9
- 239000000243 solution Substances 0.000 description 9
- 235000018991 trans-resveratrol Nutrition 0.000 description 9
- -1 Egr 1 activators Chemical class 0.000 description 8
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 8
- 239000004480 active ingredient Substances 0.000 description 8
- 201000010099 disease Diseases 0.000 description 8
- 238000000684 flow cytometry Methods 0.000 description 8
- 102000004169 proteins and genes Human genes 0.000 description 8
- 230000007419 viral reactivation Effects 0.000 description 8
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 7
- 230000030833 cell death Effects 0.000 description 7
- 238000006243 chemical reaction Methods 0.000 description 7
- 230000012010 growth Effects 0.000 description 7
- 239000000543 intermediate Substances 0.000 description 7
- 230000004044 response Effects 0.000 description 7
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 6
- 238000003491 array Methods 0.000 description 6
- 230000003247 decreasing effect Effects 0.000 description 6
- 150000002085 enols Chemical class 0.000 description 6
- 239000000047 product Substances 0.000 description 6
- 230000003362 replicative effect Effects 0.000 description 6
- 238000007619 statistical method Methods 0.000 description 6
- 230000000153 supplemental effect Effects 0.000 description 6
- 238000002560 therapeutic procedure Methods 0.000 description 6
- 206010001513 AIDS related complex Diseases 0.000 description 5
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 5
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 5
- 101710149951 Protein Tat Proteins 0.000 description 5
- 108020000999 Viral RNA Proteins 0.000 description 5
- 230000027455 binding Effects 0.000 description 5
- 230000033228 biological regulation Effects 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 230000037396 body weight Effects 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 239000000969 carrier Substances 0.000 description 5
- 231100000673 dose–response relationship Toxicity 0.000 description 5
- 238000001914 filtration Methods 0.000 description 5
- 230000001939 inductive effect Effects 0.000 description 5
- 238000010606 normalization Methods 0.000 description 5
- 201000003450 persistent generalized lymphadenopathy Diseases 0.000 description 5
- 239000000843 powder Substances 0.000 description 5
- 238000011160 research Methods 0.000 description 5
- 150000003839 salts Chemical class 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 239000003826 tablet Substances 0.000 description 5
- 230000008685 targeting Effects 0.000 description 5
- 241000282412 Homo Species 0.000 description 4
- 241000713340 Human immunodeficiency virus 2 Species 0.000 description 4
- 206010028980 Neoplasm Diseases 0.000 description 4
- 229940079156 Proteasome inhibitor Drugs 0.000 description 4
- IOUUIFSIQMVYKP-UHFFFAOYSA-N Tetradecyl acetate Chemical compound CCCCCCCCCCCCCCOC(C)=O IOUUIFSIQMVYKP-UHFFFAOYSA-N 0.000 description 4
- 239000000427 antigen Substances 0.000 description 4
- 108091007433 antigens Proteins 0.000 description 4
- 102000036639 antigens Human genes 0.000 description 4
- 239000003963 antioxidant agent Substances 0.000 description 4
- 235000006708 antioxidants Nutrition 0.000 description 4
- 230000015556 catabolic process Effects 0.000 description 4
- 230000003833 cell viability Effects 0.000 description 4
- 238000006731 degradation reaction Methods 0.000 description 4
- 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 4
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 102000006602 glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 4
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- 210000005260 human cell Anatomy 0.000 description 4
- 210000000987 immune system Anatomy 0.000 description 4
- 238000010348 incorporation Methods 0.000 description 4
- 239000002953 phosphate buffered saline Substances 0.000 description 4
- 239000003207 proteasome inhibitor Substances 0.000 description 4
- 230000001225 therapeutic effect Effects 0.000 description 4
- 238000013518 transcription Methods 0.000 description 4
- 230000035897 transcription Effects 0.000 description 4
- 229930185603 trichostatin Natural products 0.000 description 4
- RTKIYFITIVXBLE-QEQCGCAPSA-N trichostatin A Chemical compound ONC(=O)/C=C/C(/C)=C/[C@@H](C)C(=O)C1=CC=C(N(C)C)C=C1 RTKIYFITIVXBLE-QEQCGCAPSA-N 0.000 description 4
- 230000003827 upregulation Effects 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 3
- DYLIWHYUXAJDOJ-OWOJBTEDSA-N (e)-4-(6-aminopurin-9-yl)but-2-en-1-ol Chemical compound NC1=NC=NC2=C1N=CN2C\C=C\CO DYLIWHYUXAJDOJ-OWOJBTEDSA-N 0.000 description 3
- 102100034808 CCAAT/enhancer-binding protein alpha Human genes 0.000 description 3
- 208000003322 Coinfection Diseases 0.000 description 3
- 108020004635 Complementary DNA Proteins 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- XEKOWRVHYACXOJ-UHFFFAOYSA-N Ethyl acetate Chemical compound CCOC(C)=O XEKOWRVHYACXOJ-UHFFFAOYSA-N 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 101000945515 Homo sapiens CCAAT/enhancer-binding protein alpha Proteins 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 101710141650 Myeloid cell nuclear differentiation antigen Proteins 0.000 description 3
- 102100027994 Myeloid cell nuclear differentiation antigen Human genes 0.000 description 3
- 239000013614 RNA sample Substances 0.000 description 3
- 210000001744 T-lymphocyte Anatomy 0.000 description 3
- 102100040247 Tumor necrosis factor Human genes 0.000 description 3
- 235000009754 Vitis X bourquina Nutrition 0.000 description 3
- 235000012333 Vitis X labruscana Nutrition 0.000 description 3
- 240000006365 Vitis vinifera Species 0.000 description 3
- 235000014787 Vitis vinifera Nutrition 0.000 description 3
- 238000009825 accumulation Methods 0.000 description 3
- 230000002411 adverse Effects 0.000 description 3
- 230000000840 anti-viral effect Effects 0.000 description 3
- 230000003078 antioxidant effect Effects 0.000 description 3
- 210000003050 axon Anatomy 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 229960001467 bortezomib Drugs 0.000 description 3
- GXJABQQUPOEUTA-RDJZCZTQSA-N bortezomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-RDJZCZTQSA-N 0.000 description 3
- 230000022131 cell cycle Effects 0.000 description 3
- 239000002299 complementary DNA Substances 0.000 description 3
- 230000000120 cytopathologic effect Effects 0.000 description 3
- 238000007405 data analysis Methods 0.000 description 3
- 230000003828 downregulation Effects 0.000 description 3
- 230000001747 exhibiting effect Effects 0.000 description 3
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 3
- 239000008187 granular material Substances 0.000 description 3
- 230000006195 histone acetylation Effects 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 230000010354 integration Effects 0.000 description 3
- 230000003834 intracellular effect Effects 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 239000002502 liposome Substances 0.000 description 3
- 210000004698 lymphocyte Anatomy 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 230000008823 permeabilization Effects 0.000 description 3
- 102000013415 peroxidase activity proteins Human genes 0.000 description 3
- 108040007629 peroxidase activity proteins Proteins 0.000 description 3
- 239000008194 pharmaceutical composition Substances 0.000 description 3
- ZJAOAACCNHFJAH-UHFFFAOYSA-N phosphonoformic acid Chemical compound OC(=O)P(O)(O)=O ZJAOAACCNHFJAH-UHFFFAOYSA-N 0.000 description 3
- 230000002265 prevention Effects 0.000 description 3
- 238000003757 reverse transcription PCR Methods 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 239000000758 substrate Substances 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 208000011580 syndromic disease Diseases 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- ABZLKHKQJHEPAX-UHFFFAOYSA-N tetramethylrhodamine Chemical compound C=12C=CC(N(C)C)=CC2=[O+]C2=CC(N(C)C)=CC=C2C=1C1=CC=CC=C1C([O-])=O ABZLKHKQJHEPAX-UHFFFAOYSA-N 0.000 description 3
- 230000000699 topical effect Effects 0.000 description 3
- 230000014616 translation Effects 0.000 description 3
- PFTAWBLQPZVEMU-ZFWWWQNUSA-N (+)-epicatechin Chemical compound C1([C@@H]2OC3=CC(O)=CC(O)=C3C[C@@H]2O)=CC=C(O)C(O)=C1 PFTAWBLQPZVEMU-ZFWWWQNUSA-N 0.000 description 2
- QGVLYPPODPLXMB-UBTYZVCOSA-N (1aR,1bS,4aR,7aS,7bS,8R,9R,9aS)-4a,7b,9,9a-tetrahydroxy-3-(hydroxymethyl)-1,1,6,8-tetramethyl-1,1a,1b,4,4a,7a,7b,8,9,9a-decahydro-5H-cyclopropa[3,4]benzo[1,2-e]azulen-5-one Chemical compound C1=C(CO)C[C@]2(O)C(=O)C(C)=C[C@H]2[C@@]2(O)[C@H](C)[C@@H](O)[C@@]3(O)C(C)(C)[C@H]3[C@@H]21 QGVLYPPODPLXMB-UBTYZVCOSA-N 0.000 description 2
- XLAIWHIOIFKLEO-UHFFFAOYSA-N (E)-4-<2-(4-hydroxyphenyl)ethenyl>phenol Natural products C1=CC(O)=CC=C1C=CC1=CC=C(O)C=C1 XLAIWHIOIFKLEO-UHFFFAOYSA-N 0.000 description 2
- HOQFOJNETHAACL-UHFFFAOYSA-N 5-[2-(4-hydroxyphenyl)ethenyl]benzene-1,3-diol;stilbene Chemical compound C=1C=CC=CC=1C=CC1=CC=CC=C1.C1=CC(O)=CC=C1C=CC1=CC(O)=CC(O)=C1 HOQFOJNETHAACL-UHFFFAOYSA-N 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- LUKBXSAWLPMMSZ-UPHRSURJSA-N Cis-resveratrol Chemical compound C1=CC(O)=CC=C1\C=C/C1=CC(O)=CC(O)=C1 LUKBXSAWLPMMSZ-UPHRSURJSA-N 0.000 description 2
- 241000701022 Cytomegalovirus Species 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 102000016911 Deoxyribonucleases Human genes 0.000 description 2
- 108010053770 Deoxyribonucleases Proteins 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 108010032976 Enfuvirtide Proteins 0.000 description 2
- 108700039887 Essential Genes Proteins 0.000 description 2
- 108091060211 Expressed sequence tag Proteins 0.000 description 2
- 241000282326 Felis catus Species 0.000 description 2
- 102000003964 Histone deacetylase Human genes 0.000 description 2
- 108090000353 Histone deacetylase Proteins 0.000 description 2
- 102100039999 Histone deacetylase 2 Human genes 0.000 description 2
- 101001035011 Homo sapiens Histone deacetylase 2 Proteins 0.000 description 2
- 101000592466 Homo sapiens Proteasome subunit beta type-4 Proteins 0.000 description 2
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- JUUBCHWRXWPFFH-UHFFFAOYSA-N Hydroxytyrosol Chemical compound OCCC1=CC=C(O)C(O)=C1 JUUBCHWRXWPFFH-UHFFFAOYSA-N 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
- 208000007766 Kaposi sarcoma Diseases 0.000 description 2
- 239000005517 L01XE01 - Imatinib Substances 0.000 description 2
- 240000007472 Leucaena leucocephala Species 0.000 description 2
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 2
- 101150105878 Lman1l gene Proteins 0.000 description 2
- 206010025323 Lymphomas Diseases 0.000 description 2
- 101150058160 Lyn gene Proteins 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 102100033190 Proteasome subunit beta type-4 Human genes 0.000 description 2
- 238000002123 RNA extraction Methods 0.000 description 2
- 239000012980 RPMI-1640 medium Substances 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
- 229930006000 Sucrose Natural products 0.000 description 2
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 2
- 238000007239 Wittig reaction Methods 0.000 description 2
- 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 2
- MKUXAQIIEYXACX-UHFFFAOYSA-N aciclovir Chemical compound N1C(N)=NC(=O)C2=C1N(COCCO)C=N2 MKUXAQIIEYXACX-UHFFFAOYSA-N 0.000 description 2
- 230000029936 alkylation Effects 0.000 description 2
- 238000005804 alkylation reaction Methods 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 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 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 239000012620 biological material Substances 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 102000013515 cdc42 GTP-Binding Protein Human genes 0.000 description 2
- 108010051348 cdc42 GTP-Binding Protein Proteins 0.000 description 2
- 229940076006 cell cycle modulator Drugs 0.000 description 2
- 230000003915 cell function Effects 0.000 description 2
- 108091092328 cellular RNA Proteins 0.000 description 2
- 230000004640 cellular pathway Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 238000002648 combination therapy Methods 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 239000002552 dosage form Substances 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
- 230000002900 effect on cell Effects 0.000 description 2
- 239000000839 emulsion Substances 0.000 description 2
- 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 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 229940121372 histone deacetylase inhibitor Drugs 0.000 description 2
- 239000003276 histone deacetylase inhibitor Substances 0.000 description 2
- 238000009396 hybridization Methods 0.000 description 2
- KTUFNOKKBVMGRW-UHFFFAOYSA-N imatinib Chemical compound C1CN(C)CCN1CC1=CC=C(C(=O)NC=2C=C(NC=3N=C(C=CN=3)C=3C=NC=CC=3)C(C)=CC=2)C=C1 KTUFNOKKBVMGRW-UHFFFAOYSA-N 0.000 description 2
- 229960002411 imatinib Drugs 0.000 description 2
- 230000036737 immune function Effects 0.000 description 2
- 230000028993 immune response Effects 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 238000007918 intramuscular administration Methods 0.000 description 2
- 229960001627 lamivudine 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
- 230000007246 mechanism Effects 0.000 description 2
- 238000002483 medication Methods 0.000 description 2
- 150000007522 mineralic acids Chemical class 0.000 description 2
- 238000000465 moulding Methods 0.000 description 2
- NQDJXKOVJZTUJA-UHFFFAOYSA-N nevirapine Chemical compound C12=NC=CC=C2C(=O)NC=2C(C)=CC=NC=2N1C1CC1 NQDJXKOVJZTUJA-UHFFFAOYSA-N 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 238000012898 one-sample t-test Methods 0.000 description 2
- 150000007524 organic acids Chemical class 0.000 description 2
- 239000002245 particle Substances 0.000 description 2
- 239000006072 paste Substances 0.000 description 2
- 238000001558 permutation test Methods 0.000 description 2
- 150000002989 phenols Chemical class 0.000 description 2
- QGVLYPPODPLXMB-QXYKVGAMSA-N phorbol Natural products C[C@@H]1[C@@H](O)[C@]2(O)[C@H]([C@H]3C=C(CO)C[C@@]4(O)[C@H](C=C(C)C4=O)[C@@]13O)C2(C)C QGVLYPPODPLXMB-QXYKVGAMSA-N 0.000 description 2
- 210000002381 plasma Anatomy 0.000 description 2
- 239000013641 positive control Substances 0.000 description 2
- 230000001566 pro-viral effect Effects 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 229940002612 prodrug Drugs 0.000 description 2
- 239000000651 prodrug Substances 0.000 description 2
- 239000003528 protein farnesyltransferase inhibitor Substances 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 239000011541 reaction mixture Substances 0.000 description 2
- 238000012755 real-time RT-PCR analysis Methods 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 230000022983 regulation of cell cycle Effects 0.000 description 2
- 230000002441 reversible effect Effects 0.000 description 2
- 238000005070 sampling Methods 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 230000002123 temporal effect Effects 0.000 description 2
- 230000036962 time dependent Effects 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 238000011200 topical administration Methods 0.000 description 2
- 230000007704 transition Effects 0.000 description 2
- 238000011269 treatment regimen Methods 0.000 description 2
- 238000000870 ultraviolet spectroscopy Methods 0.000 description 2
- 238000011144 upstream manufacturing Methods 0.000 description 2
- 238000012795 verification Methods 0.000 description 2
- 230000007733 viral latency Effects 0.000 description 2
- PFTAWBLQPZVEMU-DZGCQCFKSA-N (+)-catechin Chemical compound C1([C@H]2OC3=CC(O)=CC(O)=C3C[C@@H]2O)=CC=C(O)C(O)=C1 PFTAWBLQPZVEMU-DZGCQCFKSA-N 0.000 description 1
- 229930013915 (+)-catechin Natural products 0.000 description 1
- 235000007219 (+)-catechin Nutrition 0.000 description 1
- 235000007246 (+)-epicatechin Nutrition 0.000 description 1
- XTYSXGHMTNTKFH-BDEHJDMKSA-N (2s)-1-[(2s,4r)-4-benzyl-2-hydroxy-5-[[(1s,2r)-2-hydroxy-2,3-dihydro-1h-inden-1-yl]amino]-5-oxopentyl]-n-tert-butyl-4-(pyridin-3-ylmethyl)piperazine-2-carboxamide;hydrate Chemical compound O.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 XTYSXGHMTNTKFH-BDEHJDMKSA-N 0.000 description 1
- HSTZMXCBWJGKHG-UHFFFAOYSA-N (E)-piceid Natural products OC1C(O)C(O)C(CO)OC1OC1=CC(O)=CC(C=CC=2C=CC(O)=CC=2)=C1 HSTZMXCBWJGKHG-UHFFFAOYSA-N 0.000 description 1
- 125000005273 2-acetoxybenzoic acid group Chemical group 0.000 description 1
- WREGKURFCTUGRC-UHFFFAOYSA-N 4-Amino-1-[5-(hydroxymethyl)oxolan-2-yl]pyrimidin-2-one Chemical compound O=C1N=C(N)C=CN1C1OC(CO)CC1 WREGKURFCTUGRC-UHFFFAOYSA-N 0.000 description 1
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 description 1
- 229940124718 AIDS vaccine Drugs 0.000 description 1
- 206010000807 Acute HIV infection Diseases 0.000 description 1
- 241000416162 Astragalus gummifer 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
- 108010019625 Atazanavir Sulfate Proteins 0.000 description 1
- JMGZEFIQIZZSBH-UHFFFAOYSA-N Bioquercetin Natural products CC1OC(OCC(O)C2OC(OC3=C(Oc4cc(O)cc(O)c4C3=O)c5ccc(O)c(O)c5)C(O)C2O)C(O)C(O)C1O JMGZEFIQIZZSBH-UHFFFAOYSA-N 0.000 description 1
- 208000003174 Brain Neoplasms Diseases 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 208000026310 Breast neoplasm Diseases 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 210000004366 CD4-positive T-lymphocyte Anatomy 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
- 241000283707 Capra Species 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 102000011068 Cdc42 Human genes 0.000 description 1
- 108050001278 Cdc42 Proteins 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 101000876610 Dictyostelium discoideum Extracellular signal-regulated kinase 2 Proteins 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
- OTMSDBZUPAUEDD-UHFFFAOYSA-N Ethane Chemical compound CC OTMSDBZUPAUEDD-UHFFFAOYSA-N 0.000 description 1
- 244000004281 Eucalyptus maculata Species 0.000 description 1
- 108010007457 Extracellular Signal-Regulated MAP Kinases Proteins 0.000 description 1
- 102000007665 Extracellular Signal-Regulated MAP Kinases Human genes 0.000 description 1
- 206010017533 Fungal infection Diseases 0.000 description 1
- 108091006027 G proteins Proteins 0.000 description 1
- 102000030782 GTP binding Human genes 0.000 description 1
- 108091000058 GTP-Binding Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 108010024124 Histone Deacetylase 1 Proteins 0.000 description 1
- 102100039996 Histone deacetylase 1 Human genes 0.000 description 1
- 108010033040 Histones Proteins 0.000 description 1
- 102000006947 Histones Human genes 0.000 description 1
- 101001136753 Homo sapiens 26S proteasome regulatory subunit 8 Proteins 0.000 description 1
- 101000917826 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-a Proteins 0.000 description 1
- 101001052493 Homo sapiens Mitogen-activated protein kinase 1 Proteins 0.000 description 1
- 101000611202 Homo sapiens Peptidyl-prolyl cis-trans isomerase B Proteins 0.000 description 1
- 101001124667 Homo sapiens Proteasome subunit alpha type-5 Proteins 0.000 description 1
- 101001090813 Homo sapiens Proteasome subunit alpha type-6 Proteins 0.000 description 1
- 101000933607 Homo sapiens Protein BTG3 Proteins 0.000 description 1
- 101000587434 Homo sapiens Serine/arginine-rich splicing factor 3 Proteins 0.000 description 1
- 101000987310 Homo sapiens Serine/threonine-protein kinase PAK 2 Proteins 0.000 description 1
- 101000713585 Homo sapiens Tubulin beta-4A chain Proteins 0.000 description 1
- 108010084873 Human Immunodeficiency Virus nef Gene Products Proteins 0.000 description 1
- 108700020121 Human Immunodeficiency Virus-1 rev Proteins 0.000 description 1
- 102000006992 Interferon-alpha Human genes 0.000 description 1
- 108010047761 Interferon-alpha Proteins 0.000 description 1
- 102000014150 Interferons Human genes 0.000 description 1
- 108010050904 Interferons Proteins 0.000 description 1
- 102000000588 Interleukin-2 Human genes 0.000 description 1
- 108010002350 Interleukin-2 Proteins 0.000 description 1
- OFFWOVJBSQMVPI-RMLGOCCBSA-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.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 OFFWOVJBSQMVPI-RMLGOCCBSA-N 0.000 description 1
- 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 1
- 208000032420 Latent Infection Diseases 0.000 description 1
- HLFSDGLLUJUHTE-SNVBAGLBSA-N Levamisole Chemical compound C1([C@H]2CN3CCSC3=N2)=CC=CC=C1 HLFSDGLLUJUHTE-SNVBAGLBSA-N 0.000 description 1
- 102100029204 Low affinity immunoglobulin gamma Fc region receptor II-a Human genes 0.000 description 1
- 208000008771 Lymphadenopathy Diseases 0.000 description 1
- 206010027336 Menstruation delayed Diseases 0.000 description 1
- 102100024193 Mitogen-activated protein kinase 1 Human genes 0.000 description 1
- 208000034578 Multiple myelomas Diseases 0.000 description 1
- 208000031888 Mycoses Diseases 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- 101100384865 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) cot-1 gene Proteins 0.000 description 1
- 101100445499 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) erg-1 gene Proteins 0.000 description 1
- 206010067482 No adverse event Diseases 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 102100040283 Peptidyl-prolyl cis-trans isomerase B Human genes 0.000 description 1
- 208000037581 Persistent Infection Diseases 0.000 description 1
- 206010049025 Persistent generalised lymphadenopathy Diseases 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 102100038374 Pinin Human genes 0.000 description 1
- 101710173952 Pinin Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 206010036790 Productive cough Diseases 0.000 description 1
- 102100029270 Proteasome subunit alpha type-5 Human genes 0.000 description 1
- 102100034664 Proteasome subunit alpha type-6 Human genes 0.000 description 1
- 102100026035 Protein BTG3 Human genes 0.000 description 1
- 101000933967 Pseudomonas phage KPP25 Major capsid protein Proteins 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- VSWDORGPIHIGNW-UHFFFAOYSA-N Pyrrolidine dithiocarbamic acid Chemical compound SC(=S)N1CCCC1 VSWDORGPIHIGNW-UHFFFAOYSA-N 0.000 description 1
- 230000006819 RNA synthesis Effects 0.000 description 1
- 108700005075 Regulator Genes Proteins 0.000 description 1
- 206010038997 Retroviral infections Diseases 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
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 102100029665 Serine/arginine-rich splicing factor 3 Human genes 0.000 description 1
- 102100027939 Serine/threonine-protein kinase PAK 2 Human genes 0.000 description 1
- 241001137926 Solea Species 0.000 description 1
- 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 1
- 238000000692 Student's t-test Methods 0.000 description 1
- PPBRXRYQALVLMV-UHFFFAOYSA-N Styrene Natural products C=CC1=CC=CC=C1 PPBRXRYQALVLMV-UHFFFAOYSA-N 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 108010046075 Thymosin Proteins 0.000 description 1
- 102000007501 Thymosin 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
- 229920001615 Tragacanth Polymers 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 239000007984 Tris EDTA buffer Substances 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 102100036788 Tubulin beta-4A chain Human genes 0.000 description 1
- 108020005202 Viral DNA Proteins 0.000 description 1
- 229960004748 abacavir Drugs 0.000 description 1
- 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 1
- 238000002835 absorbance Methods 0.000 description 1
- FGDQGIKMWOAFIK-UHFFFAOYSA-N acetonitrile;phosphoric acid Chemical compound CC#N.OP(O)(O)=O FGDQGIKMWOAFIK-UHFFFAOYSA-N 0.000 description 1
- 229960004150 aciclovir Drugs 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 150000007513 acids Chemical class 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 238000007818 agglutination assay Methods 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 229960001830 amprenavir Drugs 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 210000003484 anatomy Anatomy 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 230000000843 anti-fungal effect Effects 0.000 description 1
- 230000000798 anti-retroviral effect Effects 0.000 description 1
- 238000011225 antiretroviral therapy Methods 0.000 description 1
- 229940124522 antiretrovirals Drugs 0.000 description 1
- 239000003903 antiretrovirus agent Substances 0.000 description 1
- 229960003277 atazanavir Drugs 0.000 description 1
- 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 1
- 239000002585 base Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid group Chemical group C(C1=CC=CC=C1)(=O)O WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000036983 biotransformation Effects 0.000 description 1
- 210000001124 body fluid Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 239000006227 byproduct Substances 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- OSQPUMRCKZAIOZ-UHFFFAOYSA-N carbon dioxide;ethanol Chemical compound CCO.O=C=O OSQPUMRCKZAIOZ-UHFFFAOYSA-N 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 230000036996 cardiovascular health Effects 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000004656 cell transport Effects 0.000 description 1
- 230000023549 cell-cell signaling Effects 0.000 description 1
- 230000007248 cellular mechanism Effects 0.000 description 1
- 210000003169 central nervous system Anatomy 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- CXQWRCVTCMQVQX-UHFFFAOYSA-N cis-dihydroquercetin Natural products O1C2=CC(O)=CC(O)=C2C(=O)C(O)C1C1=CC=C(O)C(O)=C1 CXQWRCVTCMQVQX-UHFFFAOYSA-N 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 230000000295 complement effect Effects 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
- 239000000470 constituent Substances 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 238000002425 crystallisation Methods 0.000 description 1
- 230000008025 crystallization Effects 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 230000007423 decrease Effects 0.000 description 1
- 230000007850 degeneration Effects 0.000 description 1
- WHBIGIKBNXZKFE-UHFFFAOYSA-N delavirdine mesylate Natural products 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 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 230000000779 depleting effect Effects 0.000 description 1
- 230000000994 depressogenic effect Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 238000002405 diagnostic procedure Methods 0.000 description 1
- 229960002656 didanosine Drugs 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 229960003804 efavirenz Drugs 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 229960002062 enfuvirtide Drugs 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- IVTMALDHFAHOGL-UHFFFAOYSA-N eriodictyol 7-O-rutinoside Natural products OC1C(O)C(O)C(C)OC1OCC1C(O)C(O)C(O)C(OC=2C=C3C(C(C(O)=C(O3)C=3C=C(O)C(O)=CC=3)=O)=C(O)C=2)O1 IVTMALDHFAHOGL-UHFFFAOYSA-N 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000002349 favourable effect Effects 0.000 description 1
- 239000012467 final product Substances 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 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
- 239000012458 free base Substances 0.000 description 1
- 229940099052 fuzeon Drugs 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 229930182478 glucoside Natural products 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 150000004677 hydrates Chemical class 0.000 description 1
- UWYVPFMHMJIBHE-OWOJBTEDSA-N hydroxymaleic acid group Chemical group O/C(/C(=O)O)=C/C(=O)O UWYVPFMHMJIBHE-OWOJBTEDSA-N 0.000 description 1
- 229940095066 hydroxytyrosol Drugs 0.000 description 1
- 235000003248 hydroxytyrosol Nutrition 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 230000008102 immune modulation Effects 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 229960001936 indinavir Drugs 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 230000001524 infective effect Effects 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 238000013383 initial experiment Methods 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229940079322 interferon Drugs 0.000 description 1
- 229940112586 kaletra Drugs 0.000 description 1
- 229940043355 kinase inhibitor Drugs 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 229960001614 levamisole Drugs 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 206010025482 malaise Diseases 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 238000011880 melting curve analysis Methods 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 230000006679 metabolic signaling pathway Effects 0.000 description 1
- 238000010208 microarray analysis Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 239000007932 molded tablet Substances 0.000 description 1
- 230000004879 molecular function Effects 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 239000002324 mouth wash Substances 0.000 description 1
- 239000007923 nasal drop Substances 0.000 description 1
- 229940100662 nasal drops Drugs 0.000 description 1
- 239000007922 nasal spray Substances 0.000 description 1
- 229940097496 nasal spray Drugs 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 229960000884 nelfinavir Drugs 0.000 description 1
- 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 1
- 229960000689 nevirapine Drugs 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 230000012223 nuclear import Effects 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 235000010603 pastilles Nutrition 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 238000003068 pathway analysis Methods 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 235000020030 perry Nutrition 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- WLJVXDMOQOGPHL-UHFFFAOYSA-N phenylacetic acid Chemical compound OC(=O)CC1=CC=CC=C1 WLJVXDMOQOGPHL-UHFFFAOYSA-N 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 230000001323 posttranslational effect Effects 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 230000037452 priming Effects 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 239000002510 pyrogen Substances 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- FDRQPMVGJOQVTL-UHFFFAOYSA-N quercetin rutinoside Natural products OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(OC=2C(C3=C(O)C=C(O)C=C3OC=2C=2C=C(O)C(O)=CC=2)=O)O1 FDRQPMVGJOQVTL-UHFFFAOYSA-N 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 229960000311 ritonavir Drugs 0.000 description 1
- 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 1
- 235000005493 rutin Nutrition 0.000 description 1
- IKGXIBQEEMLURG-BKUODXTLSA-N rutin Chemical compound O[C@H]1[C@H](O)[C@@H](O)[C@H](C)O[C@@H]1OC[C@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](OC=2C(C3=C(O)C=C(O)C=C3OC=2C=2C=C(O)C(O)=CC=2)=O)O1 IKGXIBQEEMLURG-BKUODXTLSA-N 0.000 description 1
- ALABRVAAKCSLSC-UHFFFAOYSA-N rutin Natural products CC1OC(OCC2OC(O)C(O)C(O)C2O)C(O)C(O)C1OC3=C(Oc4cc(O)cc(O)c4C3=O)c5ccc(O)c(O)c5 ALABRVAAKCSLSC-UHFFFAOYSA-N 0.000 description 1
- 229960004555 rutoside Drugs 0.000 description 1
- YGSDEFSMJLZEOE-UHFFFAOYSA-M salicylate Chemical compound OC1=CC=CC=C1C([O-])=O YGSDEFSMJLZEOE-UHFFFAOYSA-M 0.000 description 1
- 229960001860 salicylate Drugs 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 229960001852 saquinavir Drugs 0.000 description 1
- 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 1
- 238000007423 screening assay Methods 0.000 description 1
- 210000000582 semen Anatomy 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 230000001568 sexual effect Effects 0.000 description 1
- 238000007493 shaping process Methods 0.000 description 1
- 239000012453 solvate Substances 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 210000003802 sputum Anatomy 0.000 description 1
- 208000024794 sputum Diseases 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000000528 statistical test Methods 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000008093 supporting effect Effects 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- LCJVIYPJPCBWKS-NXPQJCNCSA-N thymosin Chemical compound SC[C@@H](N)C(=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(C)C)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(C)C)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)=O LCJVIYPJPCBWKS-NXPQJCNCSA-N 0.000 description 1
- 229960000838 tipranavir Drugs 0.000 description 1
- 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 1
- 235000010487 tragacanth Nutrition 0.000 description 1
- 239000000196 tragacanth Substances 0.000 description 1
- 229940116362 tragacanth Drugs 0.000 description 1
- 108091008023 transcriptional regulators Proteins 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000006648 viral gene expression Effects 0.000 description 1
- 239000011534 wash buffer Substances 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000008215 water for injection Substances 0.000 description 1
- 208000016261 weight loss Diseases 0.000 description 1
- 230000004580 weight loss Effects 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 229960000523 zalcitabine Drugs 0.000 description 1
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/569—Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
- G01N33/56983—Viruses
- G01N33/56988—HIV or HTLV
-
- 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
-
- 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
- A61K31/045—Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
- A61K31/05—Phenols
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/35—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
- A61K31/352—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline
- A61K31/353—3,4-Dihydrobenzopyrans, e.g. chroman, catechin
-
- 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
- A61P31/14—Antivirals for RNA viruses
- A61P31/18—Antivirals for RNA viruses for HIV
Definitions
- the human immunodeficiency virus type 1 (HIV-I, also referred to as HTLV- III LAV or HTLV-III/LAV) and, to a lesser extent, human immunodeficiency virus type 2 (HIV-2) is the etiological agent of the acquired immune deficiency syndrome (AIDS) and related disorders.
- AIDS acquired immune deficiency syndrome
- AIDS While AIDS, itself, does not necessarily cause death, in many individuals the immune system is so severely depressed that various other diseases (secondary infections or unusual tumors) such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus related lymphomas among others occur, which ultimately results in death. These secondary infections may be treated using other medications. However, such treatment can be adversely affected by the weakened immune system.
- Some humans infected with the AIDS virus seem to live many years with little or no symptoms, but appear to have persistent infections.
- Another group of humans suffers mild immune system depression with various symptoms such as weight loss, malaise, fever and swollen lymph nodes. These syndromes have been called persistent generalized lymphadenopathy syndrome (PGL) and AIDS related complex (ARC) and may or may not develop into AIDS. In all cases, those infected with the HIV are believed to be persistently infective to others.
- PDL persistent generalized lymphadenopathy syndrome
- ARC AIDS related complex
- LTR long terminal repeat
- TNF ⁇ is present in markedly enhanced levels in HIV infected individuals, suggesting that the cytokine plays an important role in the pathogenesis of AIDS.
- Lahdevirta J., Am. J. Med., 85:289-291 (1988).
- the compounds are those of formula I (trans-resveratrol), formula II (cis-resveratrol) or formula III (taxifolin):
- aspects of the invention are methods of treating or preventing HIV infection in a subject comprising administration of an effective amount of an Egr 1 activator, including those having a structure represented by the formulae herein.
- compounds e.g., compounds represented by the formulae herein
- the compounds of the present invention can induce lytic replication in cells latently infected with an immunodeficiency virus such as HIV by targeting a product of a gene, which is differentially expressed in latently infected cells and lytic replicating cells.
- the compounds of the present invention can treat cells infected acutely and chronically by immunodeficiency viruses, for example, HIV, preferably HIV-I, and thus can be used to treat humans infected by HIV.
- immunodeficiency viruses for example, HIV, preferably HIV-I
- the cell is preferably a lymphocytic cell or a monocytic cell. In preferred embodiments, the cell is a human cell.
- Other aspects are a method of reducing latent HIV reservoirs in an HIV- infected subject comprising administration to the subject of an effective amount of an Egr 1 activator (e.g., a compound of the formulae herein); a method of increasing (e.g., 2-20 fold relative to cells treated with AZT) expression of p24 in a latently HIV- infected cell comprising administration to the cell of an effective amount of an Egr 1 activator (e.g., a compound of the formulae herein); a method of activating latent HIV-provirus in a cell in a subject comprising administration to the subject of an effective amount of an Egr 1 activator (e.g., a compound of the formulae herein); and a method of activating latent HIV-provirus in a cell in a subject comprising administration to the subject of an effective amount of an Egr 1 activator (e.g., a compound of the formulae herein).
- an Egr 1 activator e.g.,
- the methods delineated herein can further include administration of one or more additional anti-HIV therapeutic agents.
- the additional agent(s) can be one or more reverse transcriptase inhibitors, one or more protease inhibitors r, or combination thereof.
- the Egr 1 activator in the methods herein is resveratrol.
- Another aspect is a method of screening for a compound capable of activating latent HIV-infected cells comprising contacting an ACH-2 cell or J 1.1 cell or Ul cell with a test compound and determining the level of p24 expression; the method can be wherein increased expression of p24 in cells treated with a test compound relative to non-treated cells indicates a compound capable of activating or reactivating latent HIV-infected cells.
- the test compound can include those of any of the formulae delineated herein.
- Another aspect is a method of reducing latent HIV-reservoirs in a subject by controlled activation of viral replication including administration of an effective amount of a Egr 1 activator.
- Controlled activation is that activation initiated by administration of an effective amount of Egr 1 activator such that the Egr 1 activator reactivates (directly or indirectly) replication processes in latent HIV-infected cells.
- the latent HIV-reservoirs are collections of latent HIV-infected cells, that is cells in which the HIV-replication is considered to be in a latent state.
- the method can further include administration with one or more antiviral agents, thus both depleting the latent cell reservoir and inducing their lytic replication whereupon the cells in that state are subjected to and susceptible to the antiretroviral therapy, which controls their proliferation.
- the compounds of the present invention can treat cells infected acutely and chronically by immunodeficiency viruses, for example, HIV, preferably HIV-I, and thus can be used to treat humans infected by HIV.
- immunodeficiency viruses for example, HIV, preferably HIV-I
- Other aspects are a method of inhibiting HIV replication in a cell comprising administration to the cell of an effective amount of an Ergl activator; a method of treating latently HIV-infected cells in a subject comprising administration to the subject of an effective amount of an Erg 1 inhibitor; and a method of modulating lytic replication in an HIV-infected cell in a subject comprising administration to the subject of an effective amount of a Ergl inhibitor.
- the cell is a lymphocytic cell or a monocytic cell.
- the invention provides a method of treating or preventing HIV infection in a subject.
- the method comprises identifying the subject as in need of Egr 1 activation, and administration of an effective amount of an Egr 1 activator.
- the method comprises administration of an effective amount of an Egr 1 activator having the structure of formula (I), (II), or (III):
- the method further comprises administration to the subject of an effective amount of an HIV protease inhibitor.
- the invention provides a method of treating latently HIV- infected cells in a subject comprising administration to the subject of an effective amount of an Egr 1 activator that is taxifolin.
- the invention provides a method of modulating lytic replication in an HIV-infected cell in a subject comprising administration to the subject of an effective amount of an Egr 1 activator that is taxifolin.
- the cell is a lymphocytic cell. In other preferred embodiments, the cell is a monocytic cell. In certain preferred embodiments, the cell is a human cell.
- the invention provides a method of reducing latent HIV reservoirs in an HIV-infected subject comprising administration to the subject of an effective amount of an Egr 1 activator that is taxifolin.
- the invention provides a method of increasing (e.g., fold increase relative to cells treated with AZT) expression of p24 in a latently HIV- infected cell comprising administration to the cell of an effective amount of an Egr 1 activator that is taxifolin.
- the invention provides a method of activating latent HIV-provirus in a cell in a subject comprising administration to the subject of an effective amount of an Egr 1 activator that is taxifolin.
- the method further comprises administration of one or more additional anti-HIV therapeutic agents to the subject.
- the additional agent(s) are one or more reverse transcriptase inhibitors, one or more protease inhibitors, or combination thereof.
- the invention provides a method of treating HIV infection in a subject.
- the method comprises the steps of identifying a subject as in need of reactivation of a replication process in latent HIV-infected cells; administering to the subject an effective amount of an Egrl activator to reactivate the replication process; and administering to the subject one or more HIV antiviral agents to inhibit the induced lytic replication in the subject and/or the cells.
- the method provides a means for decreasing or eliminating the pool of latently infected cells and also decreasing the viral population.
- the invention provides a method of screening for a compound capable of activating latent HIV-infected cells comprising contacting an ACH-2 cell or Jl.1 cell or Ul cell with a test compound and determining the level of p24 expression in the cell or cells compared to a control.
- the test compound comprises an Egr 1 activator.
- the invention provides a method of treating HIV infection in a subject comprising identifying the subject as in need of Egr 1 activation, administering an effective amount of an Egr 1 activator to reactivate HIV replication in infected cells, and further administering an effective amount of an additional antiviral agent that is a non-nucleoside reverse transcriptase inhibitor, a protease inhibitor, or other non-nucleoside antiviral agent.
- the methods delineated herein include administering to a subject (e.g., a human or an animal) in need thereof an effective amount of one or more Egrl activators, e.g., compounds as delineated herein.
- the methods can also include the step of identifying that the subject is in need of treatment of diseases or disorders described herein, e.g., identifying that the subject is in need of reactivation of a replication process or processes in latent HIV-infected cells.
- the identification can be in the judgment of a subject or a health professional and can be subjective (e.g., opinion) or objective (e.g., measurable by a test or a diagnostic method).
- Tests for HIV infection include polymerase chain reaction-based (PCR-based) amplification and detection of viral RNA; Western blot detection of anti-HIV antibodies; agglutination assays for anti-HIV antibodies; ELISA-based detection of HIV-specific antigens (e.g., p24); line immunoassay (LIA); and other methods known to one of ordinary skill in the art.
- PCR-based polymerase chain reaction-based
- Western blot detection of anti-HIV antibodies e.g., agglutination assays for anti-HIV antibodies
- ELISA-based detection of HIV-specific antigens e.g., p24
- line immunoassay line immunoassay
- the methods of the invention can include the step of obtaining a sample of biological material (such as a bodily fluid) from a subject; testing the sample to determine the presence or absence of detectable HIV infection, HIV particles, or HIV nucleic acids; and determining whether the subject is in need of treatment according to the invention, i.e., identifying whether the subject is in need of reactivation of a replication process or processes in latent HIV-infected cells.
- a sample of biological material such as a bodily fluid
- the methods delineated herein can further include the step of assessing or identifying the effectiveness of the treatment or prevention regimen in the subject by assessing the presence, absence, increase, or decrease of a marker, including a marker or diagnostic measure of HIV infection, HIV replication, viral load, or expression of an HIV infection marker; preferably this assessment is made relative to a measurement made prior to beginning the therapy.
- a marker including a marker or diagnostic measure of HIV infection, HIV replication, viral load, or expression of an HIV infection marker; preferably this assessment is made relative to a measurement made prior to beginning the therapy.
- Such assessment methodologies are known in the art and can be performed by commercial diagnostic or medical organizations, laboratories, clinics, hospitals and the like.
- the methods can further include the step of taking a sample from the subject and analyzing that sample.
- the sample can be a sampling of cells, genetic material, tissue, or fluid (e.g., blood, plasma, sputum, etc.) sample.
- the methods can further include the step of reporting the results of such analyzing to the subject or other health care
- the terms “subject” and “patient” are used interchangeably.
- the terms “subject” and “subjects” refer to an animal, preferably a mammal including a non-primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey, ape, monkey, or human), and more preferably a human.
- the subject is an immunocompromised or immunosuppressed mammal, preferably a human (e.g., an HIV infected patient).
- the subject is a farm animal (e.g., a horse, a cow, a pig, etc.) or a pet (e.g., a dog or a cat).
- the subject is a human.
- the invention also provides pharmaceutical compositions comprising a compound of any of the formulae herein and a suitable carrier therefore for use in the methods and conditions referred to above.
- Figure 1 shows the flow cytometric analysis of chronically infected ACH-2 cells before and after induction. Uninduced cells and cells from serial time points were fixed and permeabilized for intracellular p24 labeling. As an isotype control, cell samples were labeled with mouse IgGl. For each sample, 100,000 events were collected. In the figure, each sample histogram labeled for p24 (darker color) is overlaid with the control histogram labeled for the isotype control (lighter color).
- A Uninduced ACH-2 cells, showing minimal p24 accumulation with 8.2% of cells infected
- B ACH-2 cells at 0.5 hours post induction (p.i.) with 7.4% of cells positive for p24
- C ACH-2 cells at 6 hours p.i, with 61.6% cells infected
- D ACH-2 cells at 12, 18, and 24 hours p.i., respectively, showing complete infection.
- Flow cytometric analysis was performed on all batches of cells to ensure active replication of HIV following induction with PMA. Data from one induction experiment is shown. Data indicate that viral replication occurs in an ordered manner post induction, and complete infection of cells is achieved within 12 hours post induction of chronically infected ACH-2 cells.
- Figure 2 shows the levels of expression of multiply spliced (MS HIV-I) and unspliced (US HIV-I) mRNA prior and post induction of chronically infected ACH-2 cells.
- Real time RT-PCR reactions were carried out using Taqman probes specific for early (multiply spliced) and late (unspliced) transcripts of HIV-I, tagged with FAM and TAMRA fluorescent dyes at the 5' and 3' ends respectively. Reactions were performed in triplicate for each time point as described in the Methods section and average values are shown. Maximal fold change in mRNA levels for early transcripts (MS HIV-I) was observed 8 hours post induction. Fold change for late transcripts (US HIV-I) showed maximal increase 18 hours post induction.
- Figure 3 shows the hierarchical clustering of differentially expressed cellular genes before and after induction of chronically infected ACH-2 cells.
- the figure shows the hierarchical clustering of the cellular genes that showed significant differential expression (p ⁇ 0.001) across the time course (before induction up to 96 hours post induction), following reactivation of chronically infected ACH-2 cells as per the criteria described in the Methods.
- Genes that are shown on the color scale >1 showed up regulation, those on the color scale ⁇ 1 were down regulated, while those that did not show any change with respect to normalized matched control are shown in black.
- the gray areas indicate missing data for the given gene and time point.
- the magnified panels indicate selected kinetic profiles that are seen before and following induction into active viral replication.
- A Up regulation of selected genes observed before induction;
- B Up regulation of genes immediately following induction;
- C Genes that are up regulated prior to induction and down regulated 12-24 hours post induction;
- D Genes that are up regulated in the early stage following reactivation, but are down regulated in the intermediate stage;
- E Genes that are down regulated before induction but are up regulated in the intermediate stage followed by down regulation in the late stage (48-96 hours p.i.).
- Figure 4 shows the trends seen in pathways that show differential expression before and after induction of chronically infected ACH-2 cells. Pathway profiles observed prior to induction and following reactivation of ACH-2 cells with PMA over a period of 96 hours. The figure shows the number of genes in each pathway that were differentially expressed in a particular pathway, (A) indicates the pathways that were maximally altered prior to induction. (B) includes the pathways that showed maximum change during the early phase of the lytic cycle, (0.5-8 hours p.i.). (C) represents the pathways that showed maximal change during the period of 12-24 hours post induction. Most pathways did not show any change during the period of 48-96 hours post induction. The groups above are a selected representation of the various pathways that changed differentially prior to induction and/or over the time course studied. Classification of the altered genes into various pathways was performed using the CGAP pathway databases.
- Figure 5 shows the hierarchical clustering of genes that show differential expression across three chronically infected cell lines prior to induction.
- Hierarchical clustering of differentially expressed genes that show a significant change in expression p ⁇ 0.001
- Genes shown on the color scale >1 are up regulated, those on the color scale ⁇ 1 exhibit down regulation, while black indicates normal expression.
- dark gray areas indicate missing values.
- Many genes are altered similarly across the cell lines.
- Each cell line also shows some unique patterns of cellular expression. Data are the average of values from eight independent samples per cell line. The magnified portions of the cluster highlight some of the patterns of gene expression across the cell lines.
- FIG. 6 shows the effects of specific agents on HIV p24 production in chronically infected ACH-2 cells.
- Different concentrations of a proteasome inhibitor (A) clastolactacystin-beta-lactone or (B) Resveratrol, an Egrl activator or (C) Trichostatin, a HDAC inhibitor were tested in chronically infected ACH-2 cells treated with 250 nM AZT.
- Figure 7 shows the effect of resveratrol on latently infected Jurkat clones.
- Figure 8 shows the effect of resveratrol on reactivation of aviremic patient sample #1.
- Figure 9 shows the effect of resveratrol (lOO ⁇ M) on reactivation of aviremic patient samples.
- Egrl activators e.g., compounds of the formulae herein
- an immunodeficiency virus preferably human cells infected with HIV and thus can be used for treatment in HIV infected individuals.
- an "Egrl activator” means a compound or composition capable of increasing the amount or activity of Egrl in a cell or in a subject.
- the compound or composition may increase transcription or translation of the Egrl protein, reduce or prevent degradation of the Egrl protein or reduce or prevent degradation of the Egrl protein transcript.
- an Egrl activator may desirably increase translational or even post-translational processing of Egrl protein.
- the compounds of the invention also include compounds which affect other factors downstream of Egrl .
- Egrl controls p21 c ⁇ ; expression; thus, Egrl activators can include compounds that upregulate p21 Q - p; expression.
- resveratrol acts upstream of the ERK pathway, and thus Egrl activators include compounds that act upstream of ERKl and/or ERK2.
- Resveratrol is an Egrl activator (see, e.g., F.D. Ragione et al, J. Biol. Chem., Jun 2003; 278: 23360 - 23368).
- Other Egrl activators include the anti-oxidants hydroxytyrosol and pyrrolidine dithiocarbamate (see, e.g., F.D. Ragione et al, FEBS Lett. 2002 Dec 18;532(3):289-94).
- Egrl activators can be identified using screening assays, e.g., as described in the previosuly-cited references.
- prodrugs include esters and other pharmaceutically acceptable derivatives, which, upon administration to a subject, are capable of providing the parent compounds described herein (see Goodman and Gilman's, The Pharmacological basis of Therapeutics, 8 th ed., McGraw-Hill, Int. Ed. 1992, "Biotransformation of Drugs”).
- Resveratrol (3,5,4'-trihydroxystilbene) has been identified as a constituent not only of grape skins (Soleas et al . (1995) Am. J. Enol. Vitic. 46(3):346-352) but has also been found to be present in ground nuts, eucalyptus, and other plant species. Goldberg et al. (1995), Am. J. Enol. Vitic. 46(2):159-165. A great deal of interest has been focused on the compound's antifungal activity and its correlation with resistance to fungal infection. Id at 159. Resveratrol also has been shown to effective for cardiovascular health due to its anti-oxidant properties (see, e.g., Sieman, E. H., and Creasy, L. L.
- Resveratrol may be obtained commercially (typically as the trans isomer, e.g. from the Sigma Chemical Company, St. Louis, Mo.), or it may be isolated from wine or grape skins, or it may be chemically synthesized.
- Synthesis is typically carried out by a Wittig reaction linking two substituted phenols through a styrene double bond, as described by Moreno-Manas et al. (1985) Anal. Quim.
- resveratrol is intended to mean either the cis-isomer of resveratrol, the trans-isomer of resveratrol, or a mixture of the two isomers.
- the term is also intended to include both the naturally occurring active agent and the compound as it may be chemically synthesized in the laboratory.
- Resveratrol may be administered in natural form, i.e., as isolated from grape skins, wine or other plant-derived compositions, or it may be administered as chemically synthesized in the laboratory (e.g., using the methods of Moreno-Manas et al., Jeandet et al., or Goldberg et al. (1994), cited earlier herein), or as obtained commercially, e.g., from the Sigma Chemical Company (St. Louis, Mo.).
- Trans-resveratrol may be isolated from wine using the procedure of Goldberg et al. (1995) Am. J. Enol. Vitic. 46(2): 159-165, as follows: selected wine is maintained at 4. degree. Cs and tightly stoppered in the dark prior to the isolation procedure. Preferably, isolation of resveratrol from the wine is conducted within two weeks of obtaining the wine. Briefly, a predetermined quantity of wine is passed through a suitable cartridge, e.g., a preconditioned C-18 SP cartridge (Supelco), and the retained trans-resveratrol is eluted with ethyl acetate. The product may be purified using conventional crystallization or chromatographic techniques.
- a suitable cartridge e.g., a preconditioned C-18 SP cartridge (Supelco)
- the product may be purified using conventional crystallization or chromatographic techniques.
- trans-resveratrol is synthesized from appropriately substituted phenols by means of a Wittig reaction modified by Waterhouse from the method of Moreno-Manas and Pleixats, cited supra. The final product is greater than approximately 95% pure as may be confirmed using NMR and UV spectroscopy.
- trans-resveratrol as isolated from wine, or may be chemically synthesized, and may also be converted to the cis isomer by preparing a solution of the trans isomer in a suitable solvent, e.g., 0.2 M phosphoric acid-acetonitrile (4:1 v/v). The solution is then irradiated for 5-10 min at a wavelength of 254 nm and an intensity of 990 LuW/cm 2 . Ultraviolet spectroscopy will confirm that the trans-resveratrol peak, at 306 nm, to be substantially reduced following this procedure. (See Goldberg et al. (1995) J. Chromatog. 708:89-98.).
- the compounds of the present invention can provide effective therapy of latently infected cells (i.e. cells infected by a virus which is an immunodeficiency virus such as FIV, SIV, HIV, etc.) as evidenced by the induction of lytic replication in latently infected cells.
- latently infected cells i.e. cells infected by a virus which is an immunodeficiency virus such as FIV, SIV, HIV, etc.
- the present invention can be used in treating those diagnosed as having AIDS as well as those having ARC, PGL and those seropositive but asymptomatic patients.
- an effective amount of a compound can also be used prophylactically as a preventative for high risk individuals.
- Compounds of the present invention can be used to treat cells, especially mammalian cells and in particular human cells, infected by an immunodeficiency virus such as HIV.
- an immunodeficiency virus such as HIV.
- P24 a major structural protein (product of gag), has been widely used for monitoring HTV-I replication in cells and vireamia in individuals.
- Egrl activators e.g., resveratrol
- concentrations that do not adversely affect cells can dramatically reduce the number of cells latently infected with HIV, e.g.
- a reduction of HIV-I latently infected cells as shown by an increase (e.g., X-fold increase, where x is 2, 3, 4, 5, 10, 15, 20, 30, 40, or any integer between about 2 and about 50) in p24 levels in cells treated with test compound (e.g., Egr 1 activators) relative to levels in untreated latent HTV-infected cells, hi another aspect the increase can be determined relative to untreated uninfected cells.
- an increase e.g., X-fold increase, where x is 2, 3, 4, 5, 10, 15, 20, 30, 40, or any integer between about 2 and about 50
- test compound e.g., Egr 1 activators
- the effective amount of a compound of the present invention used to obtain such a result can be at micromolar concentrations. Furthermore, the administration of the compounds of the present invention at effective concentrations, which enhance HIV expression, has not been found to adversely affect treated cells.
- Resveratrol is an Egrl activator, and causes an increase in p24 expression, which may be the cause of the latency reactivation from latently HIV infected cells.
- the compounds of the present invention can be administered to HTV infected individuals or to individuals at high risk for HIV infection, for example, those having sexual relations with an HTV infected partner, intravenous drug users, etc. Because of its effect of inducing lytic replication, the compounds of the present invention and a pharmaceutical compositions comprising one or more compounds of the formulae herein can be used prophylactically as a method of prevention for such individuals to minimize their risk of cells becoming latently infected. The compound is administered at an effective amount as set forth below by methodology such as described herein.
- compounds of the present invention induce lytic replication of HIV-I LTR and HIV-I in latently infected cells.
- compounds of the present invention in a dose- dependent fashion cause latently infected cells to lyrically replicate.
- such induction is provided with essentially no adverse effects on cell survival or cellular mRNA or total cellular RNA synthesis.
- compounds of the present invention will have utility in clearing latent infections of an HIV infection and other retroviral infections in cells and in a human, and (in preferred embodiments) to ultimately entirely clear virus from an infected subject.
- one or more compounds of the invention is administered in an amount sufficient to activate lytic replication in at least about 25 percent of infected cells, more preferably an amount sufficient to induce lytic replication in at least about 50 percent of the infected cells and still more preferably induce lytic replication in at least about 75 percent of latently infected cells.
- a preferred effective dose of one or more compounds of the present invention will be in the range 0.1 mg to 5g per kilogram body weight of recipient per day, more preferably in the range of 0.1 mg to 1,000 mg per kilogram body weight per day, and still more preferably in the range of 1 to 600 mg per kilogram of body weight per day.
- a preferred effective dose of one or more therapeutic compounds can be readily determined based on known factors such as efficacy of the particular therapeutic agent used, age, weight and gender of the patient, and the like. See dosage guidelines as set forth e.g. in
- the desired dose is suitably administered once or several more sub-doses administered at appropriate intervals throughout the day, or other appropriate schedule.
- These sub-doses may be administered as unit dosage forms, for example, containing 100 to 4,000 mg, preferably 100 to 2,000 mg.
- a compound e.g., of the formulae herein
- a compound is used in accordance with the present invention in an isolated form distinct as it may be naturally found and in a comparatively pure form, e.g., at least 85% by weight pure, more preferably at least 95% pure.
- the administered compound of the formulae herein be at least 98% or even greater than 99% pure.
- Such a material would be considered sterile for pharmaceutical purposes.
- Potential contaminants include side products that may result upon synthesis of a compound of the invention or materials that may be otherwise associated with the compound prior to its isolation and purification.
- the present compounds should preferably be sterile and pyrogen free. Purification techniques known in the art may be employed, for example chromatography.
- Administration of the compounds of the invention may be by any suitable route including oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous and intradermal) with oral or parenteral being preferred. It will be appreciated that the preferred route may vary with, for example, the condition and age of the recipient.
- the administered ingredients may be used in therapy in conjunction with other medicaments such as reverse transcriptase inhibitors such as dideoxynucleosides, e.g. zidovudine (AZT), 2',3 t -dideoxyinosine (ddl) and 2',3'-dideoxycytidine (ddC), lamivudine (3TC), stavudine (d4T), and TRIZrVTR (abacavir + zidovudine + lamivudine), nonnucleosides, e.g., efavirenz (DMP-266, DuPont Pharmaceuticals/Bristol Myers Squibb), nevirapine (Boehringer Ingleheim), and delaviridine (Pharmacia-Upjohn), TAT antagonists such as Ro 3-3335 and Ro 24- 7429, protease inhibitors, e.g., indinavir (Merck), ritonavir (Abbott), s
- Egrl activators can be combined with the use of other lytic replication activators, for example, proteasome inhibitors or farnesyl transferase inhibitors, as disclosed in U.S. provisional patent application Nos. 60/587,810 and 60/587,771, respectively, both filed on July 13, 2004 and incorporated herein by reference.
- one or more compounds of the invention are used in conjunction with one or more therapeutic agents useful for treatment or prevention of HIV, a symptom associated with HIV infection, or other disease or disease symptom such as a secondary infection or unusual tumor such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus-related lymphomas among others, that can result in HIV immuno-compromised subjects.
- a symptom associated with HIV infection or other disease or disease symptom
- a secondary infection or unusual tumor such as herpes, cytomegalovirus, Kaposi's sarcoma and Epstein-Barr virus-related lymphomas among others, that can result in HIV immuno-compromised subjects.
- one or more compounds of the formulae herein are used in conjunction with a standard HIV antiviral treatment regimens (e.g., HAART).
- a standard HIV antiviral treatment regimens e.g., HAART.
- This combination is advantageous in that the compound(s) of the formulae herein can activate latent HIV infected cells to replicate by stimulating lytic viral replication, thus making them susceptible to the co ⁇ administered standard HIV antiviral treatment regimens.
- the latent or secondary reservoirs of HTV-infected cells are depleted through "controlled” activation (rather then serendipitous or uncontrolled activation), resulting in more complete elimination of virus, while controlling the spread of viral infection..
- the treatment methods herein include administration of a so-called HIV-drug "cocktail" or combination therapy, wherein a combination of reverse transcriptase inhibitor(s) and HIV protease inhibitor(s) is co-administered.
- an Egrl activator is not combined with a nucleoside analog (such as ddl, zalcitabine (ddC), and zidovudine (AZT)).
- a combination therapy according to the invention includes administration of an Egrl activator together with an abl kinase inhibitor such as imatinib (use of imatinib for HIV treatment is described more fully in co-pending U.S. Provisional Patent Application No. 60/588,015, filed June 13, 2004).
- the methods involve modulation of any gene that exhibits altered expression in chronically HPV-infected cells compared to uninfected parental cells, prior to induction into lytic replication.
- the methods herein can involve, or target, any of the genes listed in the tables herein. This modulation can be direct or indirect, that is, it can be by direct control of expression or binding activity of the target, or by indirect control of the expression or binding activity of the target.
- another aspect is modulation of replication activity of latent HIV-infected cells.
- the methods involve modulation of lyn, cdc42, MNDA, CEBP alpha or Meisl by administration of the compounds of the formulae herein.
- Egrl (early growth response 1) is a cell cycle modulator. It has recently been shown that resveratrol, an anti-oxidant stilbene (3, 5, 4'-trihydroxystilbene), can activate Egrl, thereby modulating p21cip expression and thus exert an effect on cell cycle regulation. Egrl expression also causes growth arrest (Gl-S, G2-M phases)
- Egi 1 was down regulated during viral latency in ACH-2 cells.
- Resveratrol an Egrl activator, caused viral reactivation.
- Increased levels of Egrl leads to growth arrest (Gl/S, G2M) (52), similar to that observed during viral replication.
- Down regulation of Egrl may lead to decreased Egrl levels thus preventing growth arrest and creating conditions unfavorable for viral replication.
- the effect observed with resveratrol may be a combination of manipulating the latent cellular environment and mimicking the effects of active HIV replication on the cell cycle, thus providing favorable conditions for viral replication.
- Resveratrol has been shown to not affect the levels of NFkappa-B in other cell lines studied (52), suggesting that mechanisms other than NFkappa-B modulation may contribute to viral latency and reactivation.
- the present invention includes use of both racemic mixtures and optically active stereoisomers of compounds of the formulae herein.
- compositions of compounds of the invention used in combination with other compounds (e.g., reverse transcriptase inhibitors, protease inhibitors, and the like) may be employed alone or in combination with acceptable carriers such as those described below.
- a suitable effective dose of a compound in such a composition will be in the range of 1 to 5,000 mg per kilogram body weight of recipient per day, preferably in the range of 10 to 4,000 mg per kilogram body weight of recipient per day. When multiple compounds having complementary activity are administered it is expected one can use the lower portion of these ranges (or even less).
- the pharmaceutically acceptable salts of compounds for use in the methods of the invention can be synthesized from the corresponding compound of this invention which contain a basic moiety by conventional chemical methods.
- the salts are prepared by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
- One or more compounds of the formulae herein may be administered alone, or as part of a pharmaceutical composition, comprising at least one Egrl activator (e.g., a compound of the formulae herein) together with one or more acceptable carriers thereof and optionally other therapeutic ingredients, including those therapeutic agents discussed supra.
- the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
- stable refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds).
- the compounds delineated herein are commercially available or readily synthesized by one of ordinary skill in the art using methodology know in the art.
- Some of the compounds of this invention have one or more double bonds, or one or more asymmetric centers. Such compounds can occur as racemates, racemic mixtures, single enantiomers, individual diastereomers, diastereomeric mixtures, and cis- or trans- or E- or Z- double isomeric forms. All such isomeric forms of these compounds are expressly included in the present invention.
- the compounds of this invention may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein (e.g., alkylation of a ring system may result in alkylation at multiple sites, the invention expressly includes all such reaction products). All such isomeric forms of such compounds are expressly included in the present invention. All crystal forms of the compounds described herein are expressly included in the present invention.
- compositions include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
- the formulations may conveniently be presented in unit dosage form, e.g., tablets and sustained release capsules, and in liposomes, and may be prepared by any methods well known in the art of pharmacy.
- Such methods include the step of bringing into association the active ingredients to be administered and the carrier which constitutes one or more accessory ingredients.
- the compositions are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers, liposomes or finely divided solid carriers or both, and then if necessary shaping the product.
- compositions of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion, or packed in liposomes and as a bolus, etc.
- a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
- Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent.
- Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
- the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein.
- compositions suitable for topical administration include lozenges comprising the ingredients in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the ingredient to be administered in a suitable liquid carrier.
- compositions suitable for topical administration to the skin may be presented as ointments, creams, gels and pastes comprising one or more compounds of the present invention and a pharmaceutically acceptable carrier.
- a suitable topical delivery system is a transdermal patch containing the ingredient to be administered.
- compositions suitable for rectal administration may be presented as a suppository with a suitable base comprising, for example, cocoa butter or a salicylate.
- compositions suitable for nasal administration wherein the carrier is a solid include a coarse powder having a particle size, for example, in the range 20 to 500 microns which is administered in the manner in which snuff is taken, i.e., by rapid inhalation through the nasal passage from a container of the powder held close up to the nose.
- Suitable formulations wherein the carrier is a liquid, for administration, as for example, a nasal spray or as nasal drops, include aqueous or oily solutions of the active ingredient.
- compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
- Compositions suitable for parenteral administration include aqueous and non ⁇ aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
- the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
- sterile liquid carrier for example water for injections, immediately prior to use.
- Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
- formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example, those suitable for oral administration may include flavoring agents.
- ACH-2, A3.01, Jl.1, and Ul cells were obtained through the NIH AIDS Research and Reference Reagent Program, Division of AIDS, NIAID, NIH.
- U-937 cells were obtained from American Type Culture Collection (Manassas, VA).
- ACH-2, Jl.1 and Ul are chronically infected cell lines harboring HIV-I LAV strain, while A3.01, Jurkat, and U-937 are the corresponding parental uninfected cell lines.
- Cells were grown in RPMI-1640 (Invitrogen, San Diego, CA) with 10% fetal bovine serum (FBS, Invitrogen), 5% penicillin-streptomycin (Invitrogen), and 2mM glutamine (Invitrogen). Cells were maintained at a concentration of 1x10 6 cells/ml in T-175 flasks. Cell concentrations and cell viability were monitored throughout the experiment at all time points studied. Cells were induced by addition of 20 ng/mL of phorbol myristyl acetate (PMA or TPA, Sigma, St Louis, MO) for one hour, after which the cells were washed with phosphate buffered saline (PBS).
- PMA or TPA phorbol myristyl acetate
- ACH-2 and A3.01 were washed twice with ice-cold PBS and suspended in 50 ⁇ L ice-cold permeabilization buffer (BD Biosciences, San Jose, CA), and incubated at 4 0 C in the dark for 30 minutes. The cells were fixed using the CytoFix/CytoPerm kit (BD Biosciences) and 5 ⁇ L KC57-FITC-labeled p24 antibody (Beckman Coulter), was added to detect intracellular p24.
- RNA from the samples was subjected to DNase treatment to remove contaminating DNA, and the DNAse was inactivated using the DNase Free kit (Amersham Biosciences, Piscataway, NJ) according to manufacturer's protocols. 2 ⁇ g of RNA was reverse transcribed using the Taqman RT kit from ABI per manufacturer's specifications.
- reaction mixture 50 ⁇ l was incubated at 65 0 C for 5 minutes followed by 37 0 C for 45 minutes, 94 0 C for 5 minutes, and then cooled on ice. 1/40 th aliquots of the corresponding samples were then used in a real- time PCR reaction using Taqman probes labeled with FAM and TAMRA at the 5' and 3' ends respectively. Primer probe pairs were designed using PrimerExpress (ABI). The reactions were carried out in triplicate for each time point and the fold changes observed were normalized to GAPDH, for each time point.
- sequence detection primers for unspliced or late RNA were SK38 and SK39 from the HIV-I Gene Amplimer kit (ABI).
- a TAMRA labeled probe with sequence identical to SK 19 (ABI) was used for Real time PCR quantitation of the late viral RNA species.
- Standards from the kit were diluted to calculate copy number of virus based on gag mRNA concentrations.
- Real time RT-PCR analysis was also carried out for selected cellular genes using gene specific primer probe pairs and Taqman detection primers.
- RNA obtained from induced chronically infected and corresponding uninfected parental cells were used for microarray experiments. For each time point, RNA from the induced chronically infected ACH-2 cells and RNA from the corresponding induced, uninfected A3.01 cells were compared to minimize effects due to PMA induction.
- Microarray s were obtained from the National Cancer Institute Microarray Facility, Advanced Technology Center (Gaithersburg, MD). The microarrays (Hs. UniGem2) contained 10,395 cDNA spots on each glass slide.
- cDNAs were selected for spotting on the slides based on their known or probable involvement in oncogenesis, signal transduction, apoptosis, immune function, inflammatory pathways, cellular transport, transcription, protein translation and other important cellular functions.
- a number of expressed sequence tags (ESTs) from unknown genes homologous to known genes and cDNAs encoding housekeeping genes were also included in these gene sets.
- ESTs expressed sequence tags
- 50 ⁇ g of total RNA from PMA induced ACH-2 cells and 70 ⁇ g of total RNA from PMA induced A3.01 cells was labeled with Cy-3-dUTP and Cy-5-dUTP respectively as previously described (34, 60). Higher amounts of RNA were used for Cy-5 labeling to minimize the disparities in dye incorporation.
- RNA from PMA- induced, infected cells from a particular time point was compared with RNA from the corresponding PMA-induced, uninfected cells from the same time point for subsequent hybridization to the same array to ensure accurate comparisons and to eliminate inter-array variability.
- the labeled cDNAs were then combined and purified using MicroCon YM-30 (Millipore, Bedford, MA) spin column Filters, to remove any unincorporated nucleotides. 8-10 ⁇ g each of Cot-1 DNA, (Boehringer Mannheim, Indianapolis, IN), yeast tRNA (Sigma) and polyA (Amersham Biosciences) were added to the reaction mixture and heated at 100 0 C for 1 minute.
- Hybridization of the labeled cDNA to the microarray was carried out at 65 0 C overnight, followed by washes with IX SSC, 0.2X SSC and 0.05X SSC respectively. The slides were dried by centrifugation at 1000 rpm for 3 minutes and then scanned as described below. RNA samples from three identical but independently conducted time course experiments were tested. Microarray experiments were performed at least twice for each time point (technical replicates) of each experiment. We also compared AZT-treated ACH-2 cells to untreated ACH-2 cells to determine whether , any differences in gene expression might be solely due to AZT.
- RNA from the same samples labeled with Cy5 (70 ⁇ g RNA) and Cy3 (50 ⁇ g RNA) were co-hybridized to the same array, scanned, and data were analyzed for all the cell lines studied, using identical filtering and statistical tests, and genes showing dye incorporation bias were eliminated from further analysis as described below.
- Microarray Scanning and Data Analysis The slides were scanned using an Axon GenePix 4000 scanner (Axon Instruments, Union City, CA). The photomultiplier tube values (PMT) were adjusted to obtain equivalent intensities at both wavelengths used, 635 nm and 532 nm for the Cy5 and Cy3 channels respectively. Image analysis was performed using GenePix analysis software (Axon Instruments) and data analysis was performed using the microArray Database (mAdb) system hosted by the Center for Information Technology and Center for Cancer Research at NIH (http://nciarray.nci.nih.gov). Each array was normalized using Lowess normalization (71).
- Filtering criteria were as follows: a) For each spot, signal intensity must be at least twice that of the background intensity; b) Each gene must have values in at least 70% of the arrays; c). Each array must have values for at least 70% of the gene spots. Genes that showed dye labeling bias in a particular cell line after normalization were excluded from that gene set prior to further analysis. This was determined using a one sample t-test on mean log ratios for replicate arrays with the same sample labeled with both Cy3 and Cy5.
- Hierarchical clustering analyses on the resulting data sets were done using the mAdb system as well as Cluster and TreeView software programs (Stanford University, CA). Since statistically significant gene classes need not necessarily indicate the biological relevance of genes in a particular class, pathway analysis of the various genes that showed significant differential expression was performed by utilizing analysis tools provided by the NIH mAdb database (http://nciarray.nci.nih.gov) and querying the database of the Cancer Genome Anatomy Project (CGAP), (http://cgap.nci.nih.gov ⁇ with pathway information provided by KEGG (www, genome.ad.i p/kegg ⁇ and Biocarta (www.biocarta.com) pathway databases.
- CGAP Cancer Genome Anatomy Project
- AZT (250 nM) was added to the chronically infected cells in order to inhibit p24 production that may be caused due to low levels of actively replicating virus present along with the chronically infected cells and to ensure that any increases in p24 expression would be attributable to activation of latent provirus and not due to subsequent amplification via additional rounds of viral replication.
- Cells were incubated with different concentrations of either CLBL, resveratrol, or trichostatin at 37 0 C. 200 ⁇ L samples of cell supernatant were collected at 24 hours after treatment.
- TNF-8& 0.5 ⁇ g/mL tumor necrosis factor alpha
- the plates were washed, and rabbit anti-HIV p24 antibody was added at 1:400 dilution. Following incubation for one hour, the plates were washed and goat anti-rabbit IgG peroxidase labeled antibody at 1:300 dilution was added. The plates were incubated for one hour at 37 0 C, followed by washing and addition of a two-component substrate.
- Substrate solution consisted of equal volumes of TMB peroxidase substrate and peroxidase solution B (Kirkegaard and Perry Laboratories, Gaithersburg, MD). Samples were incubated for 30 minutes at room temperature and reactions were stopped by addition of IN hydrochloric acid solution. The absorbance was measured at 450 nm using a SpectraMax250 spectrophotometer (Molecular Devices Corporation, Sunnyvale, CA). The samples were assayed in duplicate and experiments were performed at least thrice using independent cell samples.
- the late time period (48-96 hours p.i) showed the least change with 566 genes exhibiting significant altered expression (p ⁇ 0.001).
- Many of the genes that were differentially expressed in the early time period also showed either similar or the opposite trend in their expression patterns during the other time periods, hence some genes were included in the analysis of both the time periods.
- a number of discrete patterns of gene regulation were observed.
- Several cellular genes showed distinct temporal expression patterns during the lytic replication cycle, an expected finding, but more interestingly, a smaller number of genes appeared to be differentially expressed in the latently infected ACH-2 cells compared to their parental, uninfected cells, even before induction of the lytic cycle.
- genes encoding transcription factors, components of proteasomes, factors that control immune function, apoptosis and other functional classes were included.
- gene classes that were annotated in the gene ontology database GO database, www.geneontology.org
- observed/expected ratio for the number of genes within a functional class that were differentially expressed was set at greater than one (O/E > 1), so as to eliminate functional classes where the number of genes differentially expressed was not greater than that randomly expected.
- O/E > 1 the gene ontology database
- Table 1 An abbreviated listing of the genes grouped according to known functions that were differentially expressed before induction is given in Table 1.
- Table 1 Functionally related genes that were differentially expressed prior to induction in chronically infected ACH-2 cells.
- Table 1 Functionally related genes that were differentially expressed prior to induction in chronically infected ACH-2 cells. List of selected classes of genes based on known function that are differentially expressed in latently infected ACH-2 cells, relative to uninfected parental cell line, A3.01. A number of genes involved in similar cellular functions previously not associated with presence of proviral HIV were altered coordinately even during the latent non-replicative stage. EXAMPLE 6
- PSMB4 has peptidase activity, which is inhibited by Tat during viral replication.
- Tat competes with the 11 S regulatory subunit, for binding to the 2OS core complex due to presence of a common binding site in Tat and the 1 IS regulator alpha subunit (32, 59).
- Proteasomes are also involved in processing certain regions of HIV-I Nef preferentially, which leads to production of Nef-specific CTLs (cytotoxic T-lymphocytes) (44).
- Many other classes of genes encoding immune response modulators, integrins, cell cycle modulators (such as Egrl), nuclear import factors, and G-protein signaling molecules were also differentially expressed.
- a listing of genes that were differentially expressed prior to induction, based on their functional classification is given (Table 1).
- a list of pathways that were affected in the uninduced, chronically infected cells is given (supplemental data, Table S2).
- the proteins encoded by these genes are known to be critical in the progress of certain leukemias (18, 54, 65), but have not been hitherto related to HIV latency. Certain genes show similar differential expression in ACH-2 and J 1.1 but not in Ul cells. Also, some genes show opposite trends in the cell lines tested. For example, proteasome subunits are up regulated in ACH-2, while they are down regulated in Ul. A list of common pathways affected and some pathways that change selectively is given (supplemental data, Table S4). The list of differentially expressed genes common to all three cell lines is given along with their expression ratios (supplemental data, Table S5).
- HDACl histone deacetylases
- HDAC2 histone deacetylases
- proteasome class of genes showed increased expression in ACH-2 cells even prior to induction, but not so in J 1.1 and Ul cell lines. Hence, we sought to determine whether inhibition of proteasomes would induce latent provirus into lytic replication.
- a proteasome inhibitor clastolactacystin-beta-lactone (19)
- Egr 1 activators are capable of increasing the efficiency of an acute HIV infection (58).
- PS-341 a highly specific Egr 1 activator that binds to the Beta-5 subunit of the proteasome and is approved for clinical treatment of multiple myeloma (1, 3).
- PS-341 due to the extreme cytotoxicity of PS-341 even at low concentration (100 nM) we were unable to observe any changes in p24 expression under our experimental conditions (1-1000 nM) in latently infected cells (data not shown).
- Egrl (early growth response 1) was down regulated in ACH-2 cells prior to induction and more importantly, up regulated during lytic replication. Egrl is involved in cell cycle regulation and cell differentiation, in response to a number of different growth factors (56, 64). Egrl activity is suppressed in many cancers including breast cancer and brain tumors, indicating that its activity is essential in cell cycle regulation (42),(38). It has recently been shown that resveratrol, an anti-oxidant stilbene (3, 5, 4'-trihydroxystilbene), can activate Egrl, thereby modulating p21cip expression and thus exert an effect on cell cycle regulation. Egrl expression also causes growth arrest (Gl-S, G2-M phases) (52).
- Table Sl List of sequence detection primers and probe pairs for real time RT- PCR quantitation of selected genes.
- Table Sl List of sequence detection primers and probe pairs for real time RT- PCR quantitation of selected genes.
- PrimerExpress software from Applied Biosystems (ABI).
- Table S2 Pathways that changed significantly prior and post induction of ACH-2 cells.
- Table S3 Verification of differentially expressed gene expression levels by real time RT-PCR quantitation.
- Table S3 Verification of differentially expressed gene expression levels by real time RT-PCR quantitation.
- RT-PCR data for the selected genes was normalized to data for GAPDH for each cell line, before assessing fold change in ACH-2 cells with respect to A3.01 cells. Differential expression of the selected genes was confirmed by RT-PCR quantitation.
- Table S4 Pathways that change significantly in three chronically infected cell lines.
- Table S4 Pathways that changed significantly in three chronically infected cell lines.
- Table S5 Genes that showed similar differential expression in all three chronically infected cell lines.
- Table S5 Genes that showed similar differential expression in all three chronically infected cell lines.
- Enriched CD4+ T-cells were obtained from patients (see, e.g., Chun TW, Finzi D. Margolick J, Chadwick K. Schwartz D. Siliciano RF. In vivo fate of HIV-I - infected T cells: quantitative analysis of the transition to stable latency .Nat Med. 1995 Dec;l (12): 1284-90) who had been treated with highly active antiretroviral threrapy (HAART) for some time and had viral loads below the limits of detection, but presumably had reservoirs of cells latently infected with HIV.
- HAART highly active antiretroviral threrapy
- resveratrol caused reactivation of latent provirus from latently infected cell lines as well as from aviremic patient samples.
- MNDA human myeloid cell nuclear differentiation antigen
- Tumor necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected T-cell clone. Proc Natl Acad Sci U S A. 86:2365-8.
- HIV-I human immunodeficiency virus type 1
- T lymphocyte epitopes of HIV-I Nef Generation of multiple definitive major histocompatibility complex class I ligands by proteasomes. J Exp Med 191:239-52.
- ERK MAP kinase links cytokine signals to activation of latent HIV-I infection by stimulating a cooperative interaction of AP-I and NF-kappaB. J Biol Chem 274:27981-8.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Virology (AREA)
- Immunology (AREA)
- Molecular Biology (AREA)
- Engineering & Computer Science (AREA)
- Urology & Nephrology (AREA)
- Tropical Medicine & Parasitology (AREA)
- AIDS & HIV (AREA)
- Hematology (AREA)
- Biomedical Technology (AREA)
- Communicable Diseases (AREA)
- Food Science & Technology (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biotechnology (AREA)
- Cell Biology (AREA)
- General Chemical & Material Sciences (AREA)
- Microbiology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Biochemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
Abstract
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US58801304P | 2004-07-13 | 2004-07-13 | |
US60/588,013 | 2004-07-13 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2006019841A2 true WO2006019841A2 (fr) | 2006-02-23 |
WO2006019841A3 WO2006019841A3 (fr) | 2007-02-22 |
Family
ID=35907884
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2005/024893 WO2006019841A2 (fr) | 2004-07-13 | 2005-07-13 | S |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2006019841A2 (fr) |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2015114584A1 (fr) * | 2014-01-31 | 2015-08-06 | Ghimas S.P.A. | Polydatine pour le traitement du vih chez des patients |
EP2931040A4 (fr) * | 2012-12-11 | 2016-07-13 | Univ Vanderbilt | Méthodes et compositions pour le traitement d'une infection par le vih |
US9453017B2 (en) | 2011-09-30 | 2016-09-27 | Vanderbilt University | Antiviral therapies with phospholipase D inhibitors |
WO2023069981A1 (fr) * | 2021-10-19 | 2023-04-27 | Phyto Tech Corp. | Utilisations de la taxifoline pour la santé respiratoire |
WO2023235826A3 (fr) * | 2022-06-02 | 2024-02-01 | The Regents Of The University Ofcolorado A Body Corporate | Procédés de modulation d'endonucléase g à l'aide de resvératrol et de ses dérivés |
Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DE4340438A1 (de) * | 1993-11-27 | 1995-06-01 | Erwin Backhaus | Mittel zur Inaktivierung von Retroviren |
WO1999056737A1 (fr) * | 1998-05-05 | 1999-11-11 | Institut National De La Sante Et De La Recherche Medicale (Inserm) | Antagonistes des ligands du recepteur des arylhydrocarbures |
WO2001012228A2 (fr) * | 1999-08-13 | 2001-02-22 | University Of Maryland Biotechnology Institute | Compositions pour le traitement des infections virales, et procedes correspondants |
WO2003002126A1 (fr) * | 2001-06-28 | 2003-01-09 | University Of Pretoria | Agent antiretroviral en combinaison avec du polyphenol de the pour le traitement d'infections virales |
WO2003054160A2 (fr) * | 2001-12-19 | 2003-07-03 | The Regents Of The University Of California | Lignees cellulaires a virus de l'immunodeficience latent, et procedes d'utilisation |
WO2006017346A1 (fr) * | 2004-07-13 | 2006-02-16 | GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH A ND HUMAN SERVICES OFFICE OF TECHNOLOGY TRANSFER | Traitement d'infections virales au moyen d'inhibiteurs de proteasome |
-
2005
- 2005-07-13 WO PCT/US2005/024893 patent/WO2006019841A2/fr active Application Filing
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DE4340438A1 (de) * | 1993-11-27 | 1995-06-01 | Erwin Backhaus | Mittel zur Inaktivierung von Retroviren |
WO1999056737A1 (fr) * | 1998-05-05 | 1999-11-11 | Institut National De La Sante Et De La Recherche Medicale (Inserm) | Antagonistes des ligands du recepteur des arylhydrocarbures |
WO2001012228A2 (fr) * | 1999-08-13 | 2001-02-22 | University Of Maryland Biotechnology Institute | Compositions pour le traitement des infections virales, et procedes correspondants |
WO2003002126A1 (fr) * | 2001-06-28 | 2003-01-09 | University Of Pretoria | Agent antiretroviral en combinaison avec du polyphenol de the pour le traitement d'infections virales |
WO2003054160A2 (fr) * | 2001-12-19 | 2003-07-03 | The Regents Of The University Of California | Lignees cellulaires a virus de l'immunodeficience latent, et procedes d'utilisation |
WO2006017346A1 (fr) * | 2004-07-13 | 2006-02-16 | GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH A ND HUMAN SERVICES OFFICE OF TECHNOLOGY TRANSFER | Traitement d'infections virales au moyen d'inhibiteurs de proteasome |
Non-Patent Citations (6)
Title |
---|
DELLA RAGIONE FULVIO ET AL: "Antioxidants induce different phenotypes by a distinct modulation of signal transduction." FEBS LETTERS, vol. 532, no. 3, 18 December 2002 (2002-12-18), pages 289-294, XP004398419 ISSN: 0014-5793 cited in the application * |
HEREDIA, ALONSO ET AL: "Synergistic inhibition of HIV -1 in activated and resting peripheral blood mononuclear cells, monocyte-derived macrophages, and selected drug-resistant isolates with nucleoside analogues combined with a natural product, resveratrol" JAIDS, JOURNAL OF ACQUIRED IMMUNE DEFICIENCY SYNDROMES , 25(3), 246-255 CODEN: JJASFJ, 2000, XP000982355 * |
KRISHNAN VYJAYANTHI ET AL: "Host cell gene expression during human immunodeficiency virus type I latency and reactivation and effects of targeting genes that are differentially expressed in viral latency" JOURNAL OF VIROLOGY, vol. 78, no. 17, September 2004 (2004-09), pages 9458-9473, XP008067579 ISSN: 0022-538X * |
MAHMOOD, N. ET AL: "Constituents of Cuscuta reflexa are anti- HIV agents" ANTIVIRAL CHEMISTRY & CHEMOTHERAPY , 8(1), 70-74 CODEN: ACCHEH; ISSN: 0956-3202, 1997, XP008067586 * |
RABBI MOHAMMED F ET AL: "Interleukin-10 enhances tumor necrosis factor-alpha activation of HIV-1 transcription in latently infected T cells" JOURNAL OF ACQUIRED IMMUNE DEFICIENCY SYNDROMES AND HUMAN RETROVIROLOGY, vol. 19, no. 4, 1 December 1998 (1998-12-01), pages 321-331, XP008067583 ISSN: 1077-9450 * |
VAN 'T WOUT ANGELIQUE B ET AL: "Cellular gene expression upon human immunodeficiency virus type 1 infection of CD4+-T-cell lines." JOURNAL OF VIROLOGY, vol. 77, no. 2, January 2003 (2003-01), pages 1392-1402, XP002396149 ISSN: 0022-538X * |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9453017B2 (en) | 2011-09-30 | 2016-09-27 | Vanderbilt University | Antiviral therapies with phospholipase D inhibitors |
EP2931040A4 (fr) * | 2012-12-11 | 2016-07-13 | Univ Vanderbilt | Méthodes et compositions pour le traitement d'une infection par le vih |
WO2015114584A1 (fr) * | 2014-01-31 | 2015-08-06 | Ghimas S.P.A. | Polydatine pour le traitement du vih chez des patients |
WO2023069981A1 (fr) * | 2021-10-19 | 2023-04-27 | Phyto Tech Corp. | Utilisations de la taxifoline pour la santé respiratoire |
WO2023235826A3 (fr) * | 2022-06-02 | 2024-02-01 | The Regents Of The University Ofcolorado A Body Corporate | Procédés de modulation d'endonucléase g à l'aide de resvératrol et de ses dérivés |
Also Published As
Publication number | Publication date |
---|---|
WO2006019841A3 (fr) | 2007-02-22 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Krishnan et al. | Host cell gene expression during human immunodeficiency virus type 1 latency and reactivation and effects of targeting genes that are differentially expressed in viral latency | |
Imai et al. | The periodontal pathogen Porphyromonas gingivalis induces the Epstein–Barr virus lytic switch transactivator ZEBRA by histone modification | |
US20240261307A1 (en) | Inhibitors of sars-cov-2 viral replication and uses thereof | |
Lai et al. | Gnidimacrin, a potent anti-HIV diterpene, can eliminate latent HIV-1 ex vivo by activation of protein kinase C β | |
Ferrucci et al. | Extracellular HIV-1 viral protein R affects astrocytic glyceraldehyde 3-phosphate dehydrogenase activity and neuronal survival | |
Leneva et al. | Umifenovir susceptibility monitoring and characterization of influenza viruses isolated during ARBITR clinical study | |
Sanchez et al. | Antiretrovirals, methamphetamine, and HIV-1 envelope protein gp120 compromise neuronal energy homeostasis in association with various degrees of synaptic and neuritic damage | |
Roy | Potential of small-molecule fungal metabolites in antiviral chemotherapy | |
Brogdon et al. | In vitro effects of the small-molecule protein kinase C agonists on HIV latency reactivation | |
EP3145513B1 (fr) | 3-(4-((4-(morpholinomethyl-benzyl)oxy)-1-oxoisoindolin-2-yl)piperidine-2,6-dione pour le traitement du lupus érythémateux systémique | |
Scopelliti et al. | Comparative antiviral activity of integrase inhibitors in human monocyte-derived macrophages and lymphocytes | |
McNulty et al. | iPSC neuronal assay identifies amaryllidaceae pharmacophore with multiple effects against herpesvirus infections | |
Alachkar et al. | L-methionine enhances neuroinflammation and impairs neurogenesis: implication for Alzheimer's disease | |
Boutolleau et al. | Resistance pattern of cytomegalovirus (CMV) after oral valganciclovir therapy in transplant recipients at high-risk for CMV infection | |
KR20210137989A (ko) | 바이오마커, 및 바이러스 감염, 염증 또는 암의 치료에서의 용도 | |
Mori et al. | Structure-based identification of HIV-1 nucleocapsid protein inhibitors active against wild-type and drug-resistant HIV-1 strains | |
Samikkannu et al. | HIV-1 Subtypes B and C Tat Differentially Impact Synaptic Plasticity Expression and Implicates HIV-Associated Neurocognitive Disorders § | |
WO2006019841A2 (fr) | S | |
WO2014201426A9 (fr) | Analogues triazol-1-ol médicaments de latence antirétroviral | |
WO2006017346A1 (fr) | Traitement d'infections virales au moyen d'inhibiteurs de proteasome | |
Geffin et al. | Fingolimod induces neuronal-specific gene expression with potential neuroprotective outcomes in maturing neuronal progenitor cells exposed to HIV | |
Shin et al. | Identification of aristolactam derivatives that act as inhibitors of human immunodeficiency virus type 1 infection and replication by targeting tat-mediated viral transcription | |
Zhou et al. | Bcl-2 Antagonist Obatoclax Reactivates Latent HIV-1 via the NF-κB Pathway and Induces Latent Reservoir Cell Apoptosis in Latently Infected Cells | |
Esimone et al. | Broad spectrum antiviral fractions from the lichen Ramalina farinacea (L.) Ach. | |
Pomerantz | Effects of HIV-1 Vpr on neuroinvasion and neuropathogenesis |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A2 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A2 Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG |
|
NENP | Non-entry into the national phase in: |
Ref country code: DE |
|
WWW | Wipo information: withdrawn in national office |
Country of ref document: DE |
|
122 | Ep: pct application non-entry in european phase |