WO2000008043A2 - Prevention et traitement de pathologie virale - Google Patents
Prevention et traitement de pathologie virale Download PDFInfo
- Publication number
- WO2000008043A2 WO2000008043A2 PCT/US1999/017487 US9917487W WO0008043A2 WO 2000008043 A2 WO2000008043 A2 WO 2000008043A2 US 9917487 W US9917487 W US 9917487W WO 0008043 A2 WO0008043 A2 WO 0008043A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- virus
- cell
- hiv
- envelope protein
- molecular structure
- Prior art date
Links
- 230000003612 virological effect Effects 0.000 title claims abstract description 79
- 238000011282 treatment Methods 0.000 title claims abstract description 36
- 230000002265 prevention Effects 0.000 title claims description 18
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 title description 12
- 201000010099 disease Diseases 0.000 title description 11
- 241000700605 Viruses Species 0.000 claims abstract description 305
- 238000000034 method Methods 0.000 claims abstract description 262
- 229960005486 vaccine Drugs 0.000 claims abstract description 167
- 230000004927 fusion Effects 0.000 claims abstract description 132
- 230000003472 neutralizing effect Effects 0.000 claims abstract description 79
- 239000000203 mixture Substances 0.000 claims abstract description 74
- 230000003053 immunization Effects 0.000 claims abstract description 57
- 239000003433 contraceptive agent Substances 0.000 claims abstract description 21
- 230000002254 contraceptive effect Effects 0.000 claims abstract description 21
- 210000003754 fetus Anatomy 0.000 claims abstract description 12
- 238000000746 purification Methods 0.000 claims abstract description 10
- 210000004027 cell Anatomy 0.000 claims description 531
- 101710091045 Envelope protein Proteins 0.000 claims description 235
- 101710188315 Protein X Proteins 0.000 claims description 235
- 241000725303 Human immunodeficiency virus Species 0.000 claims description 214
- 108090000623 proteins and genes Proteins 0.000 claims description 167
- 230000001413 cellular effect Effects 0.000 claims description 156
- 208000015181 infectious disease Diseases 0.000 claims description 154
- 241000282414 Homo sapiens Species 0.000 claims description 117
- 102000004169 proteins and genes Human genes 0.000 claims description 100
- 102000005962 receptors Human genes 0.000 claims description 96
- 108020003175 receptors Proteins 0.000 claims description 96
- 230000002163 immunogen Effects 0.000 claims description 94
- 108010052285 Membrane Proteins Proteins 0.000 claims description 74
- 108010003533 Viral Envelope Proteins Proteins 0.000 claims description 72
- 241001465754 Metazoa Species 0.000 claims description 67
- 150000007523 nucleic acids Chemical class 0.000 claims description 67
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 claims description 65
- 230000027455 binding Effects 0.000 claims description 64
- 108020004707 nucleic acids Proteins 0.000 claims description 63
- 102000039446 nucleic acids Human genes 0.000 claims description 63
- 230000035772 mutation Effects 0.000 claims description 47
- 239000013598 vector Substances 0.000 claims description 45
- 101100005713 Homo sapiens CD4 gene Proteins 0.000 claims description 44
- 208000031886 HIV Infections Diseases 0.000 claims description 42
- 210000000170 cell membrane Anatomy 0.000 claims description 40
- 210000002966 serum Anatomy 0.000 claims description 38
- 238000009472 formulation Methods 0.000 claims description 36
- 241000713772 Human immunodeficiency virus 1 Species 0.000 claims description 35
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 35
- 230000009261 transgenic effect Effects 0.000 claims description 34
- 101800001690 Transmembrane protein gp41 Proteins 0.000 claims description 32
- 210000004369 blood Anatomy 0.000 claims description 29
- 239000008280 blood Substances 0.000 claims description 29
- 210000004779 membrane envelope Anatomy 0.000 claims description 29
- 238000004519 manufacturing process Methods 0.000 claims description 28
- 208000030507 AIDS Diseases 0.000 claims description 26
- 239000013603 viral vector Substances 0.000 claims description 26
- 108010067390 Viral Proteins Proteins 0.000 claims description 25
- 230000006870 function Effects 0.000 claims description 23
- 108700019146 Transgenes Proteins 0.000 claims description 22
- 230000007910 cell fusion Effects 0.000 claims description 22
- 230000007423 decrease Effects 0.000 claims description 22
- 238000000338 in vitro Methods 0.000 claims description 20
- 238000012258 culturing Methods 0.000 claims description 19
- 239000002245 particle Substances 0.000 claims description 19
- 239000003431 cross linking reagent Substances 0.000 claims description 18
- 239000003937 drug carrier Substances 0.000 claims description 17
- 239000013592 cell lysate Substances 0.000 claims description 16
- 239000004365 Protease Substances 0.000 claims description 15
- 238000012216 screening Methods 0.000 claims description 15
- 208000037357 HIV infectious disease Diseases 0.000 claims description 14
- 241000124008 Mammalia Species 0.000 claims description 14
- 108091005804 Peptidases Proteins 0.000 claims description 14
- 208000033519 human immunodeficiency virus infectious disease Diseases 0.000 claims description 14
- 101000946926 Homo sapiens C-C chemokine receptor type 5 Proteins 0.000 claims description 13
- 238000003501 co-culture Methods 0.000 claims description 12
- 102000018697 Membrane Proteins Human genes 0.000 claims description 11
- 239000006071 cream Substances 0.000 claims description 11
- 239000006260 foam Substances 0.000 claims description 11
- 230000002401 inhibitory effect Effects 0.000 claims description 11
- 235000015110 jellies Nutrition 0.000 claims description 11
- 239000002674 ointment Substances 0.000 claims description 11
- 206010046865 Vaccinia virus infection Diseases 0.000 claims description 10
- 239000008274 jelly Substances 0.000 claims description 10
- 208000007089 vaccinia Diseases 0.000 claims description 10
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 claims description 9
- 102000048160 human CCR5 Human genes 0.000 claims description 8
- 101000914479 Homo sapiens CD81 antigen Proteins 0.000 claims description 7
- 241000712464 Orthomyxoviridae Species 0.000 claims description 7
- 102100027221 CD81 antigen Human genes 0.000 claims description 6
- 241000711950 Filoviridae Species 0.000 claims description 6
- 241000712907 Retroviridae Species 0.000 claims description 6
- 241000710924 Togaviridae Species 0.000 claims description 6
- 238000012544 monitoring process Methods 0.000 claims description 6
- 230000004853 protein function Effects 0.000 claims description 6
- 241000710781 Flaviviridae Species 0.000 claims description 5
- 241000711504 Paramyxoviridae Species 0.000 claims description 5
- 241000150350 Peribunyaviridae Species 0.000 claims description 5
- 241000700625 Poxviridae Species 0.000 claims description 5
- 241000711931 Rhabdoviridae Species 0.000 claims description 5
- 230000001900 immune effect Effects 0.000 claims description 4
- 239000006166 lysate Substances 0.000 claims description 4
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 claims description 3
- 238000002967 competitive immunoassay Methods 0.000 claims description 3
- 230000002934 lysing effect Effects 0.000 claims description 3
- 101000716102 Homo sapiens T-cell surface glycoprotein CD4 Proteins 0.000 claims 8
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 claims 2
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 claims 1
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 claims 1
- 230000015572 biosynthetic process Effects 0.000 abstract description 78
- 238000005755 formation reaction Methods 0.000 abstract description 78
- 238000002649 immunization Methods 0.000 abstract description 39
- 239000003814 drug Substances 0.000 abstract description 19
- 230000001404 mediated effect Effects 0.000 abstract description 18
- 238000002955 isolation Methods 0.000 abstract description 15
- 239000000654 additive Substances 0.000 abstract description 12
- 239000010836 blood and blood product Substances 0.000 abstract description 6
- 230000037361 pathway Effects 0.000 abstract description 6
- 244000052613 viral pathogen Species 0.000 abstract description 6
- 239000003443 antiviral agent Substances 0.000 abstract description 5
- 210000000987 immune system Anatomy 0.000 abstract description 5
- 229940125691 blood product Drugs 0.000 abstract description 4
- 230000003990 molecular pathway Effects 0.000 abstract description 3
- 102100034349 Integrase Human genes 0.000 description 191
- 238000006386 neutralization reaction Methods 0.000 description 106
- 235000018102 proteins Nutrition 0.000 description 85
- 238000003556 assay Methods 0.000 description 79
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 69
- 230000014509 gene expression Effects 0.000 description 46
- 108020004414 DNA Proteins 0.000 description 37
- 241000699670 Mus sp. Species 0.000 description 37
- 238000012360 testing method Methods 0.000 description 37
- 230000010530 Virus Neutralization Effects 0.000 description 34
- 230000000694 effects Effects 0.000 description 33
- 108010061299 CXCR4 Receptors Proteins 0.000 description 31
- 102000012000 CXCR4 Receptors Human genes 0.000 description 31
- 230000001225 therapeutic effect Effects 0.000 description 31
- 238000011830 transgenic mouse model Methods 0.000 description 29
- 241000699666 Mus <mouse, genus> Species 0.000 description 26
- 238000001415 gene therapy Methods 0.000 description 25
- 230000003993 interaction Effects 0.000 description 25
- 239000012528 membrane Substances 0.000 description 23
- 210000004379 membrane Anatomy 0.000 description 23
- 239000000523 sample Substances 0.000 description 23
- 238000002965 ELISA Methods 0.000 description 19
- 230000028993 immune response Effects 0.000 description 19
- 239000000047 product Substances 0.000 description 19
- 241000699660 Mus musculus Species 0.000 description 18
- 210000001744 T-lymphocyte Anatomy 0.000 description 18
- 239000002502 liposome Substances 0.000 description 18
- 238000012546 transfer Methods 0.000 description 18
- 208000036142 Viral infection Diseases 0.000 description 17
- 241000701161 unidentified adenovirus Species 0.000 description 17
- 230000009385 viral infection Effects 0.000 description 17
- 150000001875 compounds Chemical class 0.000 description 16
- 239000012634 fragment Substances 0.000 description 16
- 108020001507 fusion proteins Proteins 0.000 description 16
- 210000004408 hybridoma Anatomy 0.000 description 16
- 102000037865 fusion proteins Human genes 0.000 description 15
- 239000013612 plasmid Substances 0.000 description 15
- 239000013604 expression vector Substances 0.000 description 14
- 239000000543 intermediate Substances 0.000 description 14
- 229940096437 Protein S Drugs 0.000 description 13
- 108010031318 Vitronectin Proteins 0.000 description 13
- 102100035140 Vitronectin Human genes 0.000 description 13
- QSHGUCSTWRSQAF-FJSLEGQWSA-N s-peptide Chemical compound C([C@@H](C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=1C=CC(OS(O)(=O)=O)=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(O)=O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCCN)C(O)=O)NC(=O)[C@@H](NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC(O)=O)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C1=CC=C(OS(O)(=O)=O)C=C1 QSHGUCSTWRSQAF-FJSLEGQWSA-N 0.000 description 13
- 210000002845 virion Anatomy 0.000 description 13
- 241000710831 Flavivirus Species 0.000 description 12
- 238000001727 in vivo Methods 0.000 description 12
- 235000019419 proteases Nutrition 0.000 description 12
- 230000001177 retroviral effect Effects 0.000 description 12
- 241001430294 unidentified retrovirus Species 0.000 description 12
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 12
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 11
- 238000004132 cross linking Methods 0.000 description 11
- 238000011161 development Methods 0.000 description 11
- 238000002347 injection Methods 0.000 description 11
- 239000007924 injection Substances 0.000 description 11
- 210000005105 peripheral blood lymphocyte Anatomy 0.000 description 11
- 108010043277 recombinant soluble CD4 Proteins 0.000 description 11
- 230000001105 regulatory effect Effects 0.000 description 11
- 230000035945 sensitivity Effects 0.000 description 11
- 210000000130 stem cell Anatomy 0.000 description 11
- 229920000936 Agarose Polymers 0.000 description 10
- 239000002671 adjuvant Substances 0.000 description 10
- 239000000427 antigen Substances 0.000 description 10
- 108091007433 antigens Proteins 0.000 description 10
- 102000036639 antigens Human genes 0.000 description 10
- 230000002238 attenuated effect Effects 0.000 description 10
- 230000001419 dependent effect Effects 0.000 description 10
- 238000002360 preparation method Methods 0.000 description 10
- 102000004196 processed proteins & peptides Human genes 0.000 description 10
- 239000006228 supernatant Substances 0.000 description 10
- VOTJUWBJENROFB-UHFFFAOYSA-N 1-[3-[[3-(2,5-dioxo-3-sulfopyrrolidin-1-yl)oxy-3-oxopropyl]disulfanyl]propanoyloxy]-2,5-dioxopyrrolidine-3-sulfonic acid Chemical compound O=C1C(S(=O)(=O)O)CC(=O)N1OC(=O)CCSSCCC(=O)ON1C(=O)C(S(O)(=O)=O)CC1=O VOTJUWBJENROFB-UHFFFAOYSA-N 0.000 description 9
- 101710154606 Hemagglutinin Proteins 0.000 description 9
- 241000711549 Hepacivirus C Species 0.000 description 9
- 101710093908 Outer capsid protein VP4 Proteins 0.000 description 9
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 description 9
- 101710176177 Protein A56 Proteins 0.000 description 9
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 9
- 229940079593 drug Drugs 0.000 description 9
- 230000005764 inhibitory process Effects 0.000 description 9
- 210000002540 macrophage Anatomy 0.000 description 9
- 239000002953 phosphate buffered saline Substances 0.000 description 9
- 230000008569 process Effects 0.000 description 9
- 238000001890 transfection Methods 0.000 description 9
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 8
- 101710205625 Capsid protein p24 Proteins 0.000 description 8
- 241000282693 Cercopithecidae Species 0.000 description 8
- 229940033330 HIV vaccine Drugs 0.000 description 8
- 241000714177 Murine leukemia virus Species 0.000 description 8
- 108091028043 Nucleic acid sequence Proteins 0.000 description 8
- 101710177166 Phosphoprotein Proteins 0.000 description 8
- 108020004511 Recombinant DNA Proteins 0.000 description 8
- 101710149279 Small delta antigen Proteins 0.000 description 8
- 102100022563 Tubulin polymerization-promoting protein Human genes 0.000 description 8
- 150000001413 amino acids Chemical class 0.000 description 8
- 239000010432 diamond Substances 0.000 description 8
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 8
- 239000000185 hemagglutinin Substances 0.000 description 8
- 230000002209 hydrophobic effect Effects 0.000 description 8
- 206010022000 influenza Diseases 0.000 description 8
- 150000002632 lipids Chemical class 0.000 description 8
- 238000011160 research Methods 0.000 description 8
- 239000000243 solution Substances 0.000 description 8
- 238000002255 vaccination Methods 0.000 description 8
- 241000283973 Oryctolagus cuniculus Species 0.000 description 7
- 241000700618 Vaccinia virus Species 0.000 description 7
- 238000001042 affinity chromatography Methods 0.000 description 7
- 235000001014 amino acid Nutrition 0.000 description 7
- 125000002091 cationic group Chemical group 0.000 description 7
- 238000005202 decontamination Methods 0.000 description 7
- 230000003588 decontaminative effect Effects 0.000 description 7
- 238000003018 immunoassay Methods 0.000 description 7
- 230000001939 inductive effect Effects 0.000 description 7
- 210000004185 liver Anatomy 0.000 description 7
- 239000000463 material Substances 0.000 description 7
- 230000003641 microbiacidal effect Effects 0.000 description 7
- 229940124561 microbicide Drugs 0.000 description 7
- 239000002855 microbicide agent Substances 0.000 description 7
- 238000002560 therapeutic procedure Methods 0.000 description 7
- 210000001519 tissue Anatomy 0.000 description 7
- 238000013518 transcription Methods 0.000 description 7
- 230000035897 transcription Effects 0.000 description 7
- 238000012384 transportation and delivery Methods 0.000 description 7
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 6
- PXHVJJICTQNCMI-UHFFFAOYSA-N Nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 6
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 6
- 229940024606 amino acid Drugs 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 230000036436 anti-hiv Effects 0.000 description 6
- 230000000840 anti-viral effect Effects 0.000 description 6
- 230000008901 benefit Effects 0.000 description 6
- 238000004113 cell culture Methods 0.000 description 6
- 230000009089 cytolysis Effects 0.000 description 6
- 210000001163 endosome Anatomy 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 6
- 230000005847 immunogenicity Effects 0.000 description 6
- 238000011065 in-situ storage Methods 0.000 description 6
- 238000011534 incubation Methods 0.000 description 6
- 230000008611 intercellular interaction Effects 0.000 description 6
- 210000004698 lymphocyte Anatomy 0.000 description 6
- 230000004048 modification Effects 0.000 description 6
- 238000012986 modification Methods 0.000 description 6
- 244000052769 pathogen Species 0.000 description 6
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 6
- 239000008194 pharmaceutical composition Substances 0.000 description 6
- 241000894007 species Species 0.000 description 6
- 238000010561 standard procedure Methods 0.000 description 6
- 230000003068 static effect Effects 0.000 description 6
- 239000000126 substance Substances 0.000 description 6
- 230000017960 syncytium formation Effects 0.000 description 6
- 102000009410 Chemokine receptor Human genes 0.000 description 5
- 108050000299 Chemokine receptor Proteins 0.000 description 5
- 108090000288 Glycoproteins Proteins 0.000 description 5
- 102000003886 Glycoproteins Human genes 0.000 description 5
- 241000282412 Homo Species 0.000 description 5
- 239000000232 Lipid Bilayer Substances 0.000 description 5
- 241000700159 Rattus Species 0.000 description 5
- 101710120037 Toxin CcdB Proteins 0.000 description 5
- 239000013543 active substance Substances 0.000 description 5
- 238000010171 animal model Methods 0.000 description 5
- 239000000969 carrier Substances 0.000 description 5
- 230000008859 change Effects 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 238000010276 construction Methods 0.000 description 5
- 238000013270 controlled release Methods 0.000 description 5
- -1 ddl Chemical compound 0.000 description 5
- 230000002950 deficient Effects 0.000 description 5
- 238000001514 detection method Methods 0.000 description 5
- 239000000839 emulsion Substances 0.000 description 5
- 238000000684 flow cytometry Methods 0.000 description 5
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 5
- 230000002779 inactivation Effects 0.000 description 5
- 239000004615 ingredient Substances 0.000 description 5
- 238000001990 intravenous administration Methods 0.000 description 5
- 230000007774 longterm Effects 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- 239000002609 medium Substances 0.000 description 5
- 230000034217 membrane fusion Effects 0.000 description 5
- 244000005700 microbiome Species 0.000 description 5
- 238000002156 mixing Methods 0.000 description 5
- 230000007935 neutral effect Effects 0.000 description 5
- 230000001717 pathogenic effect Effects 0.000 description 5
- 230000009467 reduction Effects 0.000 description 5
- 230000010076 replication Effects 0.000 description 5
- 230000002441 reversible effect Effects 0.000 description 5
- 238000002741 site-directed mutagenesis Methods 0.000 description 5
- 239000011780 sodium chloride Substances 0.000 description 5
- 239000007790 solid phase Substances 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- 230000029812 viral genome replication Effects 0.000 description 5
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- 108700011778 CCR5 Proteins 0.000 description 4
- 108010041397 CD4 Antigens Proteins 0.000 description 4
- 108091026890 Coding region Proteins 0.000 description 4
- 239000004971 Cross linker Substances 0.000 description 4
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 4
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 4
- 101710125507 Integrase/recombinase Proteins 0.000 description 4
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 4
- 241000282560 Macaca mulatta Species 0.000 description 4
- 241000712079 Measles morbillivirus Species 0.000 description 4
- 108010006232 Neuraminidase Proteins 0.000 description 4
- 102000005348 Neuraminidase Human genes 0.000 description 4
- 241000580858 Simian-Human immunodeficiency virus Species 0.000 description 4
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 4
- 108700005077 Viral Genes Proteins 0.000 description 4
- 239000003708 ampul Substances 0.000 description 4
- 238000013459 approach Methods 0.000 description 4
- 210000003719 b-lymphocyte Anatomy 0.000 description 4
- 230000001580 bacterial effect Effects 0.000 description 4
- 239000011324 bead Substances 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 239000003795 chemical substances by application Substances 0.000 description 4
- 230000001086 cytosolic effect Effects 0.000 description 4
- 239000003599 detergent Substances 0.000 description 4
- 238000010790 dilution Methods 0.000 description 4
- 239000012895 dilution Substances 0.000 description 4
- 238000004520 electroporation Methods 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 150000002148 esters Chemical class 0.000 description 4
- 230000002068 genetic effect Effects 0.000 description 4
- 235000011187 glycerol Nutrition 0.000 description 4
- 229960001438 immunostimulant agent Drugs 0.000 description 4
- 239000003022 immunostimulating agent Substances 0.000 description 4
- 230000003308 immunostimulating effect Effects 0.000 description 4
- 239000008101 lactose Substances 0.000 description 4
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 4
- WSFSSNUMVMOOMR-NJFSPNSNSA-N methanone Chemical compound O=[14CH2] WSFSSNUMVMOOMR-NJFSPNSNSA-N 0.000 description 4
- 238000002703 mutagenesis Methods 0.000 description 4
- 231100000350 mutagenesis Toxicity 0.000 description 4
- 239000003921 oil Substances 0.000 description 4
- 230000003389 potentiating effect Effects 0.000 description 4
- 239000002243 precursor Substances 0.000 description 4
- 230000006337 proteolytic cleavage Effects 0.000 description 4
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 4
- 230000003362 replicative effect Effects 0.000 description 4
- 230000000241 respiratory effect Effects 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 238000012552 review Methods 0.000 description 4
- 210000003491 skin Anatomy 0.000 description 4
- 150000003384 small molecules Chemical class 0.000 description 4
- 239000007787 solid Substances 0.000 description 4
- 238000007920 subcutaneous administration Methods 0.000 description 4
- 241000712461 unidentified influenza virus Species 0.000 description 4
- 230000007502 viral entry Effects 0.000 description 4
- 229960004854 viral vaccine Drugs 0.000 description 4
- 238000001262 western blot Methods 0.000 description 4
- 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 4
- KWVJHCQQUFDPLU-YEUCEMRASA-N 2,3-bis[[(z)-octadec-9-enoyl]oxy]propyl-trimethylazanium Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(C[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC KWVJHCQQUFDPLU-YEUCEMRASA-N 0.000 description 3
- 108010042708 Acetylmuramyl-Alanyl-Isoglutamine Proteins 0.000 description 3
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 3
- 241000710929 Alphavirus Species 0.000 description 3
- 241000283690 Bos taurus Species 0.000 description 3
- QYOVMAREBTZLBT-KTKRTIGZSA-N CCCCCCCC\C=C/CCCCCCCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCO Chemical compound CCCCCCCC\C=C/CCCCCCCCOCCOCCOCCOCCOCCOCCOCCOCCOCCOCCO QYOVMAREBTZLBT-KTKRTIGZSA-N 0.000 description 3
- 241000282472 Canis lupus familiaris Species 0.000 description 3
- 241000283707 Capra Species 0.000 description 3
- 102000019034 Chemokines Human genes 0.000 description 3
- 108010012236 Chemokines Proteins 0.000 description 3
- 108010035563 Chloramphenicol O-acetyltransferase Proteins 0.000 description 3
- 208000035473 Communicable disease Diseases 0.000 description 3
- 241000711573 Coronaviridae Species 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 3
- 241000282326 Felis catus Species 0.000 description 3
- 241000287828 Gallus gallus Species 0.000 description 3
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 3
- 108010024636 Glutathione Proteins 0.000 description 3
- 101000961414 Homo sapiens Membrane cofactor protein Proteins 0.000 description 3
- 102100034353 Integrase Human genes 0.000 description 3
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 3
- 241000713666 Lentivirus Species 0.000 description 3
- 102100039373 Membrane cofactor protein Human genes 0.000 description 3
- 206010028980 Neoplasm Diseases 0.000 description 3
- 108700026244 Open Reading Frames Proteins 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 3
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 3
- 241000725643 Respiratory syncytial virus Species 0.000 description 3
- 229920002684 Sepharose Polymers 0.000 description 3
- 229920002472 Starch Polymers 0.000 description 3
- 241000282887 Suidae Species 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 241000710959 Venezuelan equine encephalitis virus Species 0.000 description 3
- 230000000996 additive effect Effects 0.000 description 3
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 210000004899 c-terminal region Anatomy 0.000 description 3
- 239000001506 calcium phosphate Substances 0.000 description 3
- 229910000389 calcium phosphate Inorganic materials 0.000 description 3
- 235000011010 calcium phosphates Nutrition 0.000 description 3
- 239000001913 cellulose Substances 0.000 description 3
- 229920002678 cellulose Polymers 0.000 description 3
- 235000013330 chicken meat Nutrition 0.000 description 3
- 238000003776 cleavage reaction Methods 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 238000013461 design Methods 0.000 description 3
- 239000003085 diluting agent Substances 0.000 description 3
- 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 3
- 239000002552 dosage form Substances 0.000 description 3
- 239000003623 enhancer Substances 0.000 description 3
- 108010078428 env Gene Products Proteins 0.000 description 3
- 238000007499 fusion processing Methods 0.000 description 3
- 238000001502 gel electrophoresis Methods 0.000 description 3
- 229960003180 glutathione Drugs 0.000 description 3
- 239000001963 growth medium Substances 0.000 description 3
- RAXXELZNTBOGNW-UHFFFAOYSA-N imidazole Natural products C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 3
- 230000000984 immunochemical effect Effects 0.000 description 3
- 238000001114 immunoprecipitation Methods 0.000 description 3
- 238000000099 in vitro assay Methods 0.000 description 3
- 239000000411 inducer Substances 0.000 description 3
- 230000002458 infectious effect Effects 0.000 description 3
- 238000001802 infusion Methods 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 230000037431 insertion Effects 0.000 description 3
- 230000010354 integration Effects 0.000 description 3
- 230000003834 intracellular effect Effects 0.000 description 3
- 238000007918 intramuscular administration Methods 0.000 description 3
- 238000007912 intraperitoneal administration Methods 0.000 description 3
- 238000007914 intraventricular administration Methods 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- MYWUZJCMWCOHBA-VIFPVBQESA-N methamphetamine Chemical compound CN[C@@H](C)CC1=CC=CC=C1 MYWUZJCMWCOHBA-VIFPVBQESA-N 0.000 description 3
- 239000011859 microparticle Substances 0.000 description 3
- BSOQXXWZTUDTEL-ZUYCGGNHSA-N muramyl dipeptide Chemical compound OC(=O)CC[C@H](C(N)=O)NC(=O)[C@H](C)NC(=O)[C@@H](C)O[C@H]1[C@H](O)[C@@H](CO)O[C@@H](O)[C@@H]1NC(C)=O BSOQXXWZTUDTEL-ZUYCGGNHSA-N 0.000 description 3
- 229910052759 nickel Inorganic materials 0.000 description 3
- 239000002773 nucleotide Substances 0.000 description 3
- 125000003729 nucleotide group Chemical group 0.000 description 3
- 235000019198 oils Nutrition 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 3
- 238000012545 processing Methods 0.000 description 3
- 230000009257 reactivity Effects 0.000 description 3
- 230000006798 recombination Effects 0.000 description 3
- 238000011084 recovery Methods 0.000 description 3
- 238000012340 reverse transcriptase PCR Methods 0.000 description 3
- 150000003839 salts Chemical class 0.000 description 3
- 230000007017 scission Effects 0.000 description 3
- 230000009870 specific binding Effects 0.000 description 3
- 210000000952 spleen Anatomy 0.000 description 3
- 210000004988 splenocyte Anatomy 0.000 description 3
- 238000010186 staining Methods 0.000 description 3
- 239000008107 starch Substances 0.000 description 3
- 235000019698 starch Nutrition 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 229940031626 subunit vaccine Drugs 0.000 description 3
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 3
- 230000001960 triggered effect Effects 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- 210000000605 viral structure Anatomy 0.000 description 3
- 229960002555 zidovudine Drugs 0.000 description 3
- ASWBNKHCZGQVJV-UHFFFAOYSA-N (3-hexadecanoyloxy-2-hydroxypropyl) 2-(trimethylazaniumyl)ethyl phosphate Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(O)COP([O-])(=O)OCC[N+](C)(C)C ASWBNKHCZGQVJV-UHFFFAOYSA-N 0.000 description 2
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 2
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 2
- 108010068327 4-hydroxyphenylpyruvate dioxygenase Proteins 0.000 description 2
- 239000013607 AAV vector Substances 0.000 description 2
- 206010001513 AIDS related complex Diseases 0.000 description 2
- 102000007698 Alcohol dehydrogenase Human genes 0.000 description 2
- 108010021809 Alcohol dehydrogenase Proteins 0.000 description 2
- 108010017088 CCR5 Receptors Proteins 0.000 description 2
- 102000004274 CCR5 Receptors Human genes 0.000 description 2
- 102000014914 Carrier Proteins Human genes 0.000 description 2
- 241000699800 Cricetinae Species 0.000 description 2
- 241000701022 Cytomegalovirus Species 0.000 description 2
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 2
- 241000725619 Dengue virus Species 0.000 description 2
- 241000702421 Dependoparvovirus Species 0.000 description 2
- 238000012286 ELISA Assay Methods 0.000 description 2
- 201000011001 Ebola Hemorrhagic Fever Diseases 0.000 description 2
- 241001115402 Ebolavirus Species 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- 241000283086 Equidae Species 0.000 description 2
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 2
- 108700039691 Genetic Promoter Regions Proteins 0.000 description 2
- 108010070675 Glutathione transferase Proteins 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 108010027044 HIV Core Protein p24 Proteins 0.000 description 2
- 102100029100 Hematopoietic prostaglandin D synthase Human genes 0.000 description 2
- 241000700586 Herpesviridae Species 0.000 description 2
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 2
- 206010061598 Immunodeficiency Diseases 0.000 description 2
- 208000029462 Immunodeficiency disease Diseases 0.000 description 2
- 102000000588 Interleukin-2 Human genes 0.000 description 2
- 108010002350 Interleukin-2 Proteins 0.000 description 2
- 101710125418 Major capsid protein Proteins 0.000 description 2
- 229930195725 Mannitol Natural products 0.000 description 2
- 101710169105 Minor spike protein Proteins 0.000 description 2
- 101710081079 Minor spike protein H Proteins 0.000 description 2
- 108010085220 Multiprotein Complexes Proteins 0.000 description 2
- 102000007474 Multiprotein Complexes Human genes 0.000 description 2
- 241000711386 Mumps virus Species 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 108010021466 Mutant Proteins Proteins 0.000 description 2
- 102000008300 Mutant Proteins Human genes 0.000 description 2
- 108091061960 Naked DNA Proteins 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 241000282579 Pan Species 0.000 description 2
- 108020002230 Pancreatic Ribonuclease Proteins 0.000 description 2
- 102000005891 Pancreatic ribonuclease Human genes 0.000 description 2
- 208000002606 Paramyxoviridae Infections Diseases 0.000 description 2
- 241001494479 Pecora Species 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 108010047620 Phytohemagglutinins Proteins 0.000 description 2
- 206010035226 Plasma cell myeloma Diseases 0.000 description 2
- 239000004698 Polyethylene Substances 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 108700008625 Reporter Genes Proteins 0.000 description 2
- 108010073443 Ribi adjuvant Proteins 0.000 description 2
- 241000710799 Rubella virus Species 0.000 description 2
- 241000713311 Simian immunodeficiency virus Species 0.000 description 2
- 241000710960 Sindbis virus Species 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine kinase Proteins 0.000 description 2
- 229920004890 Triton X-100 Polymers 0.000 description 2
- 239000013504 Triton X-100 Substances 0.000 description 2
- 108020005202 Viral DNA Proteins 0.000 description 2
- 108010059722 Viral Fusion Proteins Proteins 0.000 description 2
- 108070000030 Viral receptors Proteins 0.000 description 2
- 206010052428 Wound Diseases 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
- 238000009825 accumulation Methods 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 150000007513 acids Chemical class 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 229940037003 alum Drugs 0.000 description 2
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 2
- 150000001412 amines Chemical class 0.000 description 2
- 210000004102 animal cell Anatomy 0.000 description 2
- 230000005875 antibody response Effects 0.000 description 2
- 239000012062 aqueous buffer Substances 0.000 description 2
- 108091008324 binding proteins Proteins 0.000 description 2
- 238000004166 bioassay Methods 0.000 description 2
- 230000005540 biological transmission Effects 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 210000000601 blood cell Anatomy 0.000 description 2
- 210000001185 bone marrow Anatomy 0.000 description 2
- 102220481133 cAMP-dependent protein kinase catalytic subunit beta_G10V_mutation Human genes 0.000 description 2
- 210000000234 capsid Anatomy 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 2
- 230000006037 cell lysis Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 229940030156 cell vaccine Drugs 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 230000002759 chromosomal effect Effects 0.000 description 2
- 210000000349 chromosome Anatomy 0.000 description 2
- 230000002860 competitive effect Effects 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 239000012141 concentrate Substances 0.000 description 2
- 210000004748 cultured cell Anatomy 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 239000008121 dextrose Substances 0.000 description 2
- 210000001840 diploid cell Anatomy 0.000 description 2
- 235000021186 dishes Nutrition 0.000 description 2
- 239000003995 emulsifying agent Substances 0.000 description 2
- 238000005538 encapsulation Methods 0.000 description 2
- 210000002889 endothelial cell Anatomy 0.000 description 2
- 229940088598 enzyme Drugs 0.000 description 2
- 210000002919 epithelial cell Anatomy 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 230000001605 fetal effect Effects 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 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 2
- 125000000524 functional group Chemical group 0.000 description 2
- 230000000799 fusogenic effect Effects 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 238000010363 gene targeting Methods 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 238000003306 harvesting Methods 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 210000003494 hepatocyte Anatomy 0.000 description 2
- 125000000487 histidyl group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C([H])=N1 0.000 description 2
- 238000002744 homologous recombination Methods 0.000 description 2
- 230000006801 homologous recombination Effects 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 230000007813 immunodeficiency Effects 0.000 description 2
- 230000016784 immunoglobulin production Effects 0.000 description 2
- 238000009169 immunotherapy Methods 0.000 description 2
- 239000007943 implant Substances 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 229910052500 inorganic mineral Inorganic materials 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 239000008176 lyophilized powder Substances 0.000 description 2
- ZLNQQNXFFQJAID-UHFFFAOYSA-L magnesium carbonate Chemical compound [Mg+2].[O-]C([O-])=O ZLNQQNXFFQJAID-UHFFFAOYSA-L 0.000 description 2
- 239000001095 magnesium carbonate Substances 0.000 description 2
- 229910000021 magnesium carbonate Inorganic materials 0.000 description 2
- 235000019359 magnesium stearate Nutrition 0.000 description 2
- 239000000594 mannitol Substances 0.000 description 2
- 235000010355 mannitol Nutrition 0.000 description 2
- 229910052751 metal Inorganic materials 0.000 description 2
- 239000002184 metal Substances 0.000 description 2
- 239000000693 micelle Substances 0.000 description 2
- 244000000010 microbial pathogen Species 0.000 description 2
- 239000003094 microcapsule Substances 0.000 description 2
- 238000000520 microinjection Methods 0.000 description 2
- 230000003278 mimic effect Effects 0.000 description 2
- 239000011707 mineral Substances 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 201000000050 myeloid neoplasm Diseases 0.000 description 2
- 210000004897 n-terminal region Anatomy 0.000 description 2
- 210000004940 nucleus Anatomy 0.000 description 2
- 238000011275 oncology therapy Methods 0.000 description 2
- 239000006179 pH buffering agent Substances 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 230000001885 phytohemagglutinin Effects 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- 229920001983 poloxamer Polymers 0.000 description 2
- 229920002401 polyacrylamide Polymers 0.000 description 2
- 229920000447 polyanionic polymer Polymers 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 229920005862 polyol Polymers 0.000 description 2
- 150000003077 polyols Chemical class 0.000 description 2
- 230000002064 post-exposure prophylaxis Effects 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 238000001556 precipitation Methods 0.000 description 2
- 229940080818 propionamide Drugs 0.000 description 2
- 230000006333 protein structural change Effects 0.000 description 2
- 230000004850 protein–protein interaction Effects 0.000 description 2
- 238000003259 recombinant expression Methods 0.000 description 2
- GKBMIFPNPOSTHB-BJBKLNMKSA-N recombinant soluble cd4 Chemical compound NC(=O)C[C@H](N)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CS)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCC(O)=O)C(O)=O GKBMIFPNPOSTHB-BJBKLNMKSA-N 0.000 description 2
- 238000005215 recombination Methods 0.000 description 2
- CVHZOJJKTDOEJC-UHFFFAOYSA-N saccharin Chemical compound C1=CC=C2C(=O)NS(=O)(=O)C2=C1 CVHZOJJKTDOEJC-UHFFFAOYSA-N 0.000 description 2
- 239000012679 serum free medium Substances 0.000 description 2
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 2
- 230000010473 stable expression Effects 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 230000001629 suppression Effects 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 238000013268 sustained release Methods 0.000 description 2
- 239000012730 sustained-release form Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 208000011580 syndromic disease Diseases 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 125000003396 thiol group Chemical group [H]S* 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 238000011269 treatment regimen Methods 0.000 description 2
- 230000010415 tropism Effects 0.000 description 2
- 241001515965 unidentified phage Species 0.000 description 2
- 229940125575 vaccine candidate Drugs 0.000 description 2
- 230000001018 virulence Effects 0.000 description 2
- 238000009736 wetting Methods 0.000 description 2
- 239000000080 wetting agent Substances 0.000 description 2
- QMXCRMQIVATQMR-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) 3-pyridin-2-ylsulfanylpropanoate Chemical compound O=C1CCC(=O)N1OC(=O)CCSC1=CC=CC=N1 QMXCRMQIVATQMR-UHFFFAOYSA-N 0.000 description 1
- IEUUDEWWMRQUDS-UHFFFAOYSA-N (6-azaniumylidene-1,6-dimethoxyhexylidene)azanium;dichloride Chemical compound Cl.Cl.COC(=N)CCCCC(=N)OC IEUUDEWWMRQUDS-UHFFFAOYSA-N 0.000 description 1
- XKQYCEFPFNDDSJ-UHFFFAOYSA-N 1-[3-[2-[(4-azido-2-hydroxybenzoyl)amino]ethyldisulfanyl]propanoyloxy]-2,5-dioxopyrrolidine-3-sulfonic acid Chemical compound OC1=CC(N=[N+]=[N-])=CC=C1C(=O)NCCSSCCC(=O)ON1C(=O)C(S(O)(=O)=O)CC1=O XKQYCEFPFNDDSJ-UHFFFAOYSA-N 0.000 description 1
- PYVHLZLQVWXBDZ-UHFFFAOYSA-N 1-[6-(2,5-dioxopyrrol-1-yl)hexyl]pyrrole-2,5-dione Chemical compound O=C1C=CC(=O)N1CCCCCCN1C(=O)C=CC1=O PYVHLZLQVWXBDZ-UHFFFAOYSA-N 0.000 description 1
- UPNUQQDXHCUWSG-UHFFFAOYSA-N 1-[6-(4-azido-2-nitroanilino)hexanoyloxy]-2,5-dioxopyrrolidine-3-sulfonic acid Chemical compound O=C1C(S(=O)(=O)O)CC(=O)N1OC(=O)CCCCCNC1=CC=C(N=[N+]=[N-])C=C1[N+]([O-])=O UPNUQQDXHCUWSG-UHFFFAOYSA-N 0.000 description 1
- UFBJCMHMOXMLKC-UHFFFAOYSA-N 2,4-dinitrophenol Chemical compound OC1=CC=C([N+]([O-])=O)C=C1[N+]([O-])=O UFBJCMHMOXMLKC-UHFFFAOYSA-N 0.000 description 1
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- QQZOUYFHWKTGEY-UHFFFAOYSA-N 4-azido-n-[2-[2-[(4-azido-2-hydroxybenzoyl)amino]ethyldisulfanyl]ethyl]-2-hydroxybenzamide Chemical compound OC1=CC(N=[N+]=[N-])=CC=C1C(=O)NCCSSCCNC(=O)C1=CC=C(N=[N+]=[N-])C=C1O QQZOUYFHWKTGEY-UHFFFAOYSA-N 0.000 description 1
- BRLRJZRHRJEWJY-VCOUNFBDSA-N 5-[(3as,4s,6ar)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]-n-[3-[3-(4-azido-2-nitroanilino)propyl-methylamino]propyl]pentanamide Chemical compound C([C@H]1[C@H]2NC(=O)N[C@H]2CS1)CCCC(=O)NCCCN(C)CCCNC1=CC=C(N=[N+]=[N-])C=C1[N+]([O-])=O BRLRJZRHRJEWJY-VCOUNFBDSA-N 0.000 description 1
- AYXZIZMZXAORLO-UFLZEWODSA-N 5-[(3as,4s,6ar)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoic acid;1-hydroxypyrrolidine-2,5-dione Chemical class ON1C(=O)CCC1=O.N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 AYXZIZMZXAORLO-UFLZEWODSA-N 0.000 description 1
- 101150094949 APRT gene Proteins 0.000 description 1
- 101000768957 Acholeplasma phage L2 Uncharacterized 37.2 kDa protein Proteins 0.000 description 1
- 101000823746 Acidianus ambivalens Uncharacterized 17.7 kDa protein in bps2 3'region Proteins 0.000 description 1
- 101000916369 Acidianus ambivalens Uncharacterized protein in sor 5'region Proteins 0.000 description 1
- 101000769342 Acinetobacter guillouiae Uncharacterized protein in rpoN-murA intergenic region Proteins 0.000 description 1
- 241001455214 Acinonyx jubatus Species 0.000 description 1
- 101000823696 Actinobacillus pleuropneumoniae Uncharacterized glycosyltransferase in aroQ 3'region Proteins 0.000 description 1
- 102100029457 Adenine phosphoribosyltransferase Human genes 0.000 description 1
- 108010024223 Adenine phosphoribosyltransferase Proteins 0.000 description 1
- 101000786513 Agrobacterium tumefaciens (strain 15955) Uncharacterized protein outside the virF region Proteins 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 101000618005 Alkalihalobacillus pseudofirmus (strain ATCC BAA-2126 / JCM 17055 / OF4) Uncharacterized protein BpOF4_00885 Proteins 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 241001128034 Amphotropic murine leukemia virus Species 0.000 description 1
- 208000031295 Animal disease Diseases 0.000 description 1
- 102100020724 Ankyrin repeat, SAM and basic leucine zipper domain-containing protein 1 Human genes 0.000 description 1
- 241000712892 Arenaviridae Species 0.000 description 1
- 241000712891 Arenavirus Species 0.000 description 1
- OMLWNBVRVJYMBQ-YUMQZZPRSA-N Arg-Arg Chemical compound NC(N)=NCCC[C@H](N)C(=O)N[C@@H](CCCN=C(N)N)C(O)=O OMLWNBVRVJYMBQ-YUMQZZPRSA-N 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 101000967489 Azorhizobium caulinodans (strain ATCC 43989 / DSM 5975 / JCM 20966 / LMG 6465 / NBRC 14845 / NCIMB 13405 / ORS 571) Uncharacterized protein AZC_3924 Proteins 0.000 description 1
- 210000002237 B-cell of pancreatic islet Anatomy 0.000 description 1
- 101000823761 Bacillus licheniformis Uncharacterized 9.4 kDa protein in flaL 3'region Proteins 0.000 description 1
- 101000819719 Bacillus methanolicus Uncharacterized N-acetyltransferase in lysA 3'region Proteins 0.000 description 1
- 101000789586 Bacillus subtilis (strain 168) UPF0702 transmembrane protein YkjA Proteins 0.000 description 1
- 101000792624 Bacillus subtilis (strain 168) Uncharacterized protein YbxH Proteins 0.000 description 1
- 101000790792 Bacillus subtilis (strain 168) Uncharacterized protein YckC Proteins 0.000 description 1
- 101000819705 Bacillus subtilis (strain 168) Uncharacterized protein YlxR Proteins 0.000 description 1
- 101000948218 Bacillus subtilis (strain 168) Uncharacterized protein YtxJ Proteins 0.000 description 1
- 101000718627 Bacillus thuringiensis subsp. kurstaki Putative RNA polymerase sigma-G factor Proteins 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 1
- 101000641200 Bombyx mori densovirus Putative non-structural protein Proteins 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 241000710780 Bovine viral diarrhea virus 1 Species 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-M Bromide Chemical compound [Br-] CPELXLSAUQHCOX-UHFFFAOYSA-M 0.000 description 1
- 102100024167 C-C chemokine receptor type 3 Human genes 0.000 description 1
- 101710149862 C-C chemokine receptor type 3 Proteins 0.000 description 1
- 102100037853 C-C chemokine receptor type 4 Human genes 0.000 description 1
- POYURJRVIHUPJY-UHFFFAOYSA-N C1=CC(C(=O)O)=CC=C1N(C(=O)CI)N1C(=O)CCC1=O Chemical compound C1=CC(C(=O)O)=CC=C1N(C(=O)CI)N1C(=O)CCC1=O POYURJRVIHUPJY-UHFFFAOYSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000712083 Canine morbillivirus Species 0.000 description 1
- 108090000565 Capsid Proteins Proteins 0.000 description 1
- 101710132601 Capsid protein Proteins 0.000 description 1
- 241000701489 Cauliflower mosaic virus Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 102100023321 Ceruloplasmin Human genes 0.000 description 1
- 102000001327 Chemokine CCL5 Human genes 0.000 description 1
- 108010055166 Chemokine CCL5 Proteins 0.000 description 1
- 101000947633 Claviceps purpurea Uncharacterized 13.8 kDa protein Proteins 0.000 description 1
- 241000581364 Clinitrachus argentatus Species 0.000 description 1
- 241000283716 Connochaetes Species 0.000 description 1
- 241000186216 Corynebacterium Species 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 238000001712 DNA sequencing Methods 0.000 description 1
- 208000001490 Dengue Diseases 0.000 description 1
- 206010012310 Dengue fever Diseases 0.000 description 1
- 238000009007 Diagnostic Kit Methods 0.000 description 1
- BXZVVICBKDXVGW-NKWVEPMBSA-N Didanosine Chemical compound O1[C@H](CO)CC[C@@H]1N1C(NC=NC2=O)=C2N=C1 BXZVVICBKDXVGW-NKWVEPMBSA-N 0.000 description 1
- 102100024746 Dihydrofolate reductase Human genes 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 101000948901 Enterobacteria phage T4 Uncharacterized 16.0 kDa protein in segB-ipI intergenic region Proteins 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 1
- 101000805958 Equine herpesvirus 4 (strain 1942) Virion protein US10 homolog Proteins 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 101000790442 Escherichia coli Insertion element IS2 uncharacterized 11.1 kDa protein Proteins 0.000 description 1
- 101000788354 Escherichia phage P2 Uncharacterized 8.2 kDa protein in gpA 5'region Proteins 0.000 description 1
- LYCAIKOWRPUZTN-UHFFFAOYSA-N Ethylene glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 1
- 208000004729 Feline Leukemia Diseases 0.000 description 1
- 241000713800 Feline immunodeficiency virus Species 0.000 description 1
- 101000770304 Frankia alni UPF0460 protein in nifX-nifW intergenic region Proteins 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 101710177291 Gag polyprotein Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 101000797344 Geobacillus stearothermophilus Putative tRNA (cytidine(34)-2'-O)-methyltransferase Proteins 0.000 description 1
- 101000748410 Geobacillus stearothermophilus Uncharacterized protein in fumA 3'region Proteins 0.000 description 1
- 241000282816 Giraffa camelopardalis Species 0.000 description 1
- 208000032612 Glial tumor Diseases 0.000 description 1
- 206010018338 Glioma Diseases 0.000 description 1
- SXRSQZLOMIGNAQ-UHFFFAOYSA-N Glutaraldehyde Chemical compound O=CCCCC=O SXRSQZLOMIGNAQ-UHFFFAOYSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 229920002683 Glycosaminoglycan Polymers 0.000 description 1
- 229940126544 HIV-1 protease inhibitor Drugs 0.000 description 1
- 101000772675 Haemophilus influenzae (strain ATCC 51907 / DSM 11121 / KW20 / Rd) UPF0438 protein HI_0847 Proteins 0.000 description 1
- 101000631019 Haemophilus influenzae (strain ATCC 51907 / DSM 11121 / KW20 / Rd) Uncharacterized protein HI_0350 Proteins 0.000 description 1
- 101000768938 Haemophilus phage HP1 (strain HP1c1) Uncharacterized 8.9 kDa protein in int-C1 intergenic region Proteins 0.000 description 1
- 108091005904 Hemoglobin subunit beta Proteins 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 101000785414 Homo sapiens Ankyrin repeat, SAM and basic leucine zipper domain-containing protein 1 Proteins 0.000 description 1
- 101000738584 Homo sapiens C-C chemokine receptor type 4 Proteins 0.000 description 1
- 108090000144 Human Proteins Proteins 0.000 description 1
- 102000003839 Human Proteins Human genes 0.000 description 1
- 241000714260 Human T-lymphotropic virus 1 Species 0.000 description 1
- 241000714259 Human T-lymphotropic virus 2 Species 0.000 description 1
- 241000701109 Human adenovirus 2 Species 0.000 description 1
- 241000701085 Human alphaherpesvirus 3 Species 0.000 description 1
- 108010091358 Hypoxanthine Phosphoribosyltransferase Proteins 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- 102100029098 Hypoxanthine-guanine phosphoribosyltransferase Human genes 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 1
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 1
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 1
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 241000371980 Influenza B virus (B/Shanghai/361/2002) Species 0.000 description 1
- 102100023915 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 241000701377 Iridoviridae Species 0.000 description 1
- 101000782488 Junonia coenia densovirus (isolate pBRJ/1990) Putative non-structural protein NS2 Proteins 0.000 description 1
- 101000811523 Klebsiella pneumoniae Uncharacterized 55.8 kDa protein in cps region Proteins 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- 125000001176 L-lysyl group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C([H])([H])C([H])([H])C([H])([H])C(N([H])[H])([H])[H] 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- 101000818409 Lactococcus lactis subsp. lactis Uncharacterized HTH-type transcriptional regulator in lacX 3'region Proteins 0.000 description 1
- 101000878851 Leptolyngbya boryana Putative Fe(2+) transport protein A Proteins 0.000 description 1
- NNJVILVZKWQKPM-UHFFFAOYSA-N Lidocaine Chemical compound CCN(CC)CC(=O)NC1=C(C)C=CC=C1C NNJVILVZKWQKPM-UHFFFAOYSA-N 0.000 description 1
- 102000003960 Ligases Human genes 0.000 description 1
- 108090000364 Ligases Proteins 0.000 description 1
- 101000897431 Macaca mulatta C-C chemokine receptor type 5 Proteins 0.000 description 1
- 101100005716 Macaca mulatta CD4 gene Proteins 0.000 description 1
- 241000701076 Macacine alphaherpesvirus 1 Species 0.000 description 1
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 1
- PEEHTFAAVSWFBL-UHFFFAOYSA-N Maleimide Chemical compound O=C1NC(=O)C=C1 PEEHTFAAVSWFBL-UHFFFAOYSA-N 0.000 description 1
- 101710175625 Maltose/maltodextrin-binding periplasmic protein Proteins 0.000 description 1
- 241001115401 Marburgvirus Species 0.000 description 1
- 201000005505 Measles Diseases 0.000 description 1
- 108010090054 Membrane Glycoproteins Proteins 0.000 description 1
- 102000012750 Membrane Glycoproteins Human genes 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- 101000758828 Methanosarcina barkeri (strain Fusaro / DSM 804) Uncharacterized protein Mbar_A1602 Proteins 0.000 description 1
- 101001122401 Middle East respiratory syndrome-related coronavirus (isolate United Kingdom/H123990006/2012) Non-structural protein ORF3 Proteins 0.000 description 1
- 241000712045 Morbillivirus Species 0.000 description 1
- 241000713333 Mouse mammary tumor virus Species 0.000 description 1
- 208000005647 Mumps Diseases 0.000 description 1
- 241000711408 Murine respirovirus Species 0.000 description 1
- 101000723939 Mus musculus Transcription factor HIVEP3 Proteins 0.000 description 1
- 101001055788 Mycolicibacterium smegmatis (strain ATCC 700084 / mc(2)155) Pentapeptide repeat protein MfpA Proteins 0.000 description 1
- 101710107068 Myelin basic protein Proteins 0.000 description 1
- GXCLVBGFBYZDAG-UHFFFAOYSA-N N-[2-(1H-indol-3-yl)ethyl]-N-methylprop-2-en-1-amine Chemical compound CN(CCC1=CNC2=C1C=CC=C2)CC=C GXCLVBGFBYZDAG-UHFFFAOYSA-N 0.000 description 1
- 208000010359 Newcastle Disease Diseases 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 101710163270 Nuclease Proteins 0.000 description 1
- 102000011931 Nucleoproteins Human genes 0.000 description 1
- 108010061100 Nucleoproteins Proteins 0.000 description 1
- 101710087110 ORF6 protein Proteins 0.000 description 1
- 208000001388 Opportunistic Infections Diseases 0.000 description 1
- 101000740670 Orgyia pseudotsugata multicapsid polyhedrosis virus Protein C42 Proteins 0.000 description 1
- 241000713112 Orthobunyavirus Species 0.000 description 1
- 241000150452 Orthohantavirus Species 0.000 description 1
- 240000007594 Oryza sativa Species 0.000 description 1
- 235000007164 Oryza sativa Nutrition 0.000 description 1
- 108010067372 Pancreatic elastase Proteins 0.000 description 1
- 241000282320 Panthera leo Species 0.000 description 1
- 241000282373 Panthera pardus Species 0.000 description 1
- 108090000526 Papain Proteins 0.000 description 1
- 241000282520 Papio Species 0.000 description 1
- 241000237988 Patellidae Species 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108010033276 Peptide Fragments Proteins 0.000 description 1
- 102000007079 Peptide Fragments Human genes 0.000 description 1
- 108091000080 Phosphotransferase Proteins 0.000 description 1
- 101000769182 Photorhabdus luminescens Uncharacterized protein in pnp 3'region Proteins 0.000 description 1
- 241000711902 Pneumovirus Species 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 239000004952 Polyamide Substances 0.000 description 1
- 229920001030 Polyethylene Glycol 4000 Polymers 0.000 description 1
- 108010076039 Polyproteins Proteins 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 241000283080 Proboscidea <mammal> Species 0.000 description 1
- 101000961392 Pseudescherichia vulneris Uncharacterized 29.9 kDa protein in crtE 3'region Proteins 0.000 description 1
- 101000731030 Pseudomonas oleovorans Poly(3-hydroxyalkanoate) polymerase 2 Proteins 0.000 description 1
- 101001065485 Pseudomonas putida Probable fatty acid methyltransferase Proteins 0.000 description 1
- 241001112090 Pseudovirus Species 0.000 description 1
- 206010037742 Rabies Diseases 0.000 description 1
- 241000711798 Rabies lyssavirus Species 0.000 description 1
- 101000711023 Rhizobium leguminosarum bv. trifolii Uncharacterized protein in tfuA 3'region Proteins 0.000 description 1
- 101000948156 Rhodococcus erythropolis Uncharacterized 47.3 kDa protein in thcA 5'region Proteins 0.000 description 1
- 101000917565 Rhodococcus fascians Uncharacterized 33.6 kDa protein in fasciation locus Proteins 0.000 description 1
- 108010003581 Ribulose-bisphosphate carboxylase Proteins 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
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 241001533467 Rubulavirus Species 0.000 description 1
- 238000011579 SCID mouse model Methods 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 101000790284 Saimiriine herpesvirus 2 (strain 488) Uncharacterized 9.5 kDa protein in DHFR 3'region Proteins 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- 108010071811 Simian immunodeficiency virus Gag protein p27 Proteins 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 238000002105 Southern blotting Methods 0.000 description 1
- 101000936719 Streptococcus gordonii Accessory Sec system protein Asp3 Proteins 0.000 description 1
- 101000788499 Streptomyces coelicolor Uncharacterized oxidoreductase in mprA 5'region Proteins 0.000 description 1
- 101001102841 Streptomyces griseus Purine nucleoside phosphorylase ORF3 Proteins 0.000 description 1
- 101000708557 Streptomyces lincolnensis Uncharacterized 17.2 kDa protein in melC2-rnhH intergenic region Proteins 0.000 description 1
- 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 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric Acid Chemical class [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 108020005038 Terminator Codon Chemical group 0.000 description 1
- 101000649826 Thermotoga neapolitana Putative anti-sigma factor antagonist TM1081 homolog Proteins 0.000 description 1
- 241000710771 Tick-borne encephalitis virus Species 0.000 description 1
- 102100023935 Transmembrane glycoprotein NMB Human genes 0.000 description 1
- 102000001742 Tumor Suppressor Proteins Human genes 0.000 description 1
- 108010040002 Tumor Suppressor Proteins Proteins 0.000 description 1
- 101710095001 Uncharacterized protein in nifU 5'region Proteins 0.000 description 1
- 101000827562 Vibrio alginolyticus Uncharacterized protein in proC 3'region Proteins 0.000 description 1
- 101000778915 Vibrio parahaemolyticus serotype O3:K6 (strain RIMD 2210633) Uncharacterized membrane protein VP2115 Proteins 0.000 description 1
- 108020000999 Viral RNA Proteins 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 208000003152 Yellow Fever Diseases 0.000 description 1
- 241000710772 Yellow fever virus Species 0.000 description 1
- HMNZFMSWFCAGGW-XPWSMXQVSA-N [3-[hydroxy(2-hydroxyethoxy)phosphoryl]oxy-2-[(e)-octadec-9-enoyl]oxypropyl] (e)-octadec-9-enoate Chemical compound CCCCCCCC\C=C\CCCCCCCC(=O)OCC(COP(O)(=O)OCCO)OC(=O)CCCCCCC\C=C\CCCCCCCC HMNZFMSWFCAGGW-XPWSMXQVSA-N 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 210000001789 adipocyte Anatomy 0.000 description 1
- 230000001919 adrenal effect Effects 0.000 description 1
- 238000001261 affinity purification Methods 0.000 description 1
- 238000007818 agglutination assay Methods 0.000 description 1
- 108010050122 alpha 1-Antitrypsin Proteins 0.000 description 1
- 108010026331 alpha-Fetoproteins Proteins 0.000 description 1
- VREFGVBLTWBCJP-UHFFFAOYSA-N alprazolam Chemical compound C12=CC(Cl)=CC=C2N2C(C)=NN=C2CN=C1C1=CC=CC=C1 VREFGVBLTWBCJP-UHFFFAOYSA-N 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229940126575 aminoglycoside Drugs 0.000 description 1
- 229960003896 aminopterin Drugs 0.000 description 1
- 238000000540 analysis of variance Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 230000000798 anti-retroviral effect Effects 0.000 description 1
- 230000002421 anti-septic effect Effects 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 238000011225 antiretroviral therapy Methods 0.000 description 1
- 229940121357 antivirals Drugs 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 108010068380 arginylarginine Proteins 0.000 description 1
- 239000013602 bacteriophage vector Substances 0.000 description 1
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 1
- UCMIRNVEIXFBKS-UHFFFAOYSA-N beta-alanine Chemical compound NCCC(O)=O UCMIRNVEIXFBKS-UHFFFAOYSA-N 0.000 description 1
- 230000001588 bifunctional effect Effects 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 239000003124 biologic agent Substances 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000001851 biosynthetic effect Effects 0.000 description 1
- 238000001815 biotherapy Methods 0.000 description 1
- 108091006004 biotinylated proteins Proteins 0.000 description 1
- NXVYSVARUKNFNF-UHFFFAOYSA-N bis(2,5-dioxopyrrolidin-1-yl) 2,3-dihydroxybutanedioate Chemical compound O=C1CCC(=O)N1OC(=O)C(O)C(O)C(=O)ON1C(=O)CCC1=O NXVYSVARUKNFNF-UHFFFAOYSA-N 0.000 description 1
- LNQHREYHFRFJAU-UHFFFAOYSA-N bis(2,5-dioxopyrrolidin-1-yl) pentanedioate Chemical compound O=C1CCC(=O)N1OC(=O)CCCC(=O)ON1C(=O)CCC1=O LNQHREYHFRFJAU-UHFFFAOYSA-N 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 229960001506 brilliant green Drugs 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 210000004413 cardiac myocyte Anatomy 0.000 description 1
- 210000004970 cd4 cell Anatomy 0.000 description 1
- 239000006143 cell culture medium Substances 0.000 description 1
- 230000015861 cell surface binding Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 239000013522 chelant Substances 0.000 description 1
- SIOLHBFVZMHKPF-MOXYJHBNSA-N chembl436727 Chemical compound C([C@@H](C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CO)C(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](N)CCCCN)[C@@H](C)O)C1=CNC=N1 SIOLHBFVZMHKPF-MOXYJHBNSA-N 0.000 description 1
- 238000010382 chemical cross-linking Methods 0.000 description 1
- 239000003638 chemical reducing agent Substances 0.000 description 1
- 108091006116 chimeric peptides Proteins 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 238000012875 competitive assay Methods 0.000 description 1
- 230000000536 complexating effect Effects 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 239000000599 controlled substance Substances 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000003412 degenerative effect Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 208000025729 dengue disease Diseases 0.000 description 1
- 230000000368 destabilizing effect Effects 0.000 description 1
- 229940039227 diagnostic agent Drugs 0.000 description 1
- 239000000032 diagnostic agent Substances 0.000 description 1
- 229960002656 didanosine Drugs 0.000 description 1
- 238000002050 diffraction method Methods 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 108020001096 dihydrofolate reductase Proteins 0.000 description 1
- 125000000118 dimethyl group Chemical group [H]C([H])([H])* 0.000 description 1
- ILKCDNKCNSNFMP-UHFFFAOYSA-N dimethyl octanediimidate;hydron;dichloride Chemical compound Cl.Cl.COC(=N)CCCCCCC(=N)OC ILKCDNKCNSNFMP-UHFFFAOYSA-N 0.000 description 1
- MWRBNPKJOOWZPW-CLFAGFIQSA-N dioleoyl phosphatidylethanolamine Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC(COP(O)(=O)OCCN)OC(=O)CCCCCCC\C=C/CCCCCCCC MWRBNPKJOOWZPW-CLFAGFIQSA-N 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 229940126534 drug product Drugs 0.000 description 1
- 239000003596 drug target Substances 0.000 description 1
- 238000002651 drug therapy Methods 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 210000002257 embryonic structure Anatomy 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 210000003979 eosinophil Anatomy 0.000 description 1
- 210000000981 epithelium Anatomy 0.000 description 1
- 210000000267 erythroid cell Anatomy 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 239000013613 expression plasmid Substances 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 150000004665 fatty acids Chemical group 0.000 description 1
- 210000004700 fetal blood Anatomy 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000000835 fiber Substances 0.000 description 1
- 235000013312 flour Nutrition 0.000 description 1
- 239000011888 foil Substances 0.000 description 1
- 238000010230 functional analysis Methods 0.000 description 1
- 238000002825 functional assay Methods 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000012817 gel-diffusion technique 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
- 238000001476 gene delivery Methods 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- YQEMORVAKMFKLG-UHFFFAOYSA-N glycerine monostearate Natural products CCCCCCCCCCCCCCCCCC(=O)OC(CO)CO YQEMORVAKMFKLG-UHFFFAOYSA-N 0.000 description 1
- SVUQHVRAGMNPLW-UHFFFAOYSA-N glycerol monostearate Natural products CCCCCCCCCCCCCCCCC(=O)OCC(O)CO SVUQHVRAGMNPLW-UHFFFAOYSA-N 0.000 description 1
- 230000001456 gonadotroph Effects 0.000 description 1
- 210000003714 granulocyte Anatomy 0.000 description 1
- 108060003552 hemocyanin Proteins 0.000 description 1
- 208000010710 hepatitis C virus infection Diseases 0.000 description 1
- FUZZWVXGSFPDMH-UHFFFAOYSA-N hexanoic acid Chemical compound CCCCCC(O)=O FUZZWVXGSFPDMH-UHFFFAOYSA-N 0.000 description 1
- 238000013537 high throughput screening Methods 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- DCPMPXBYPZGNDC-UHFFFAOYSA-N hydron;methanediimine;chloride Chemical compound Cl.N=C=N DCPMPXBYPZGNDC-UHFFFAOYSA-N 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- 210000003016 hypothalamus Anatomy 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 208000026278 immune system disease Diseases 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 230000000951 immunodiffusion Effects 0.000 description 1
- 238000000760 immunoelectrophoresis Methods 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 238000012151 immunohistochemical method Methods 0.000 description 1
- 239000012133 immunoprecipitate Substances 0.000 description 1
- 239000003547 immunosorbent Substances 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 230000000415 inactivating effect Effects 0.000 description 1
- 238000009399 inbreeding Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 239000012678 infectious agent Substances 0.000 description 1
- 208000037797 influenza A Diseases 0.000 description 1
- 208000037798 influenza B Diseases 0.000 description 1
- 208000037799 influenza C Diseases 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 238000002743 insertional mutagenesis Methods 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 230000034184 interaction with host Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 210000004347 intestinal mucosa Anatomy 0.000 description 1
- 210000004020 intracellular membrane Anatomy 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000005342 ion exchange Methods 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 125000000741 isoleucyl group Chemical group [H]N([H])C(C(C([H])([H])[H])C([H])([H])C([H])([H])[H])C(=O)O* 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 210000002510 keratinocyte Anatomy 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 229960001627 lamivudine Drugs 0.000 description 1
- 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 1
- 238000011031 large-scale manufacturing process Methods 0.000 description 1
- 229960004194 lidocaine Drugs 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 210000005229 liver cell Anatomy 0.000 description 1
- 239000003589 local anesthetic agent Substances 0.000 description 1
- 230000005923 long-lasting effect Effects 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 230000006674 lysosomal degradation Effects 0.000 description 1
- 230000032575 lytic viral release Effects 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 238000007726 management method Methods 0.000 description 1
- 230000008774 maternal effect Effects 0.000 description 1
- 210000003593 megakaryocyte Anatomy 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- RUAIJHHRCIHFEV-UHFFFAOYSA-N methyl 4-amino-5-chlorothiophene-2-carboxylate Chemical compound COC(=O)C1=CC(N)=C(Cl)S1 RUAIJHHRCIHFEV-UHFFFAOYSA-N 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 239000011325 microbead Substances 0.000 description 1
- 210000001589 microsome Anatomy 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 238000000302 molecular modelling Methods 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 210000002200 mouth mucosa Anatomy 0.000 description 1
- 208000010805 mumps infectious disease Diseases 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 210000000663 muscle cell Anatomy 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- 108010065781 myosin light chain 2 Proteins 0.000 description 1
- 210000000822 natural killer cell Anatomy 0.000 description 1
- 239000006199 nebulizer Substances 0.000 description 1
- 210000001178 neural stem cell Anatomy 0.000 description 1
- 230000009251 neurologic dysfunction Effects 0.000 description 1
- 208000015015 neurological dysfunction Diseases 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 210000002569 neuron Anatomy 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 238000011587 new zealand white rabbit Methods 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 210000004882 non-tumor cell Anatomy 0.000 description 1
- 230000036963 noncompetitive effect Effects 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 210000004248 oligodendroglia Anatomy 0.000 description 1
- 238000002515 oligonucleotide synthesis Methods 0.000 description 1
- 229920001542 oligosaccharide Polymers 0.000 description 1
- 150000002482 oligosaccharides Chemical class 0.000 description 1
- 210000000287 oocyte Anatomy 0.000 description 1
- KHPXUQMNIQBQEV-UHFFFAOYSA-N oxaloacetic acid Chemical compound OC(=O)CC(=O)C(O)=O KHPXUQMNIQBQEV-UHFFFAOYSA-N 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 229940055729 papain Drugs 0.000 description 1
- 235000019834 papain Nutrition 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 230000009984 peri-natal effect Effects 0.000 description 1
- 210000003668 pericyte Anatomy 0.000 description 1
- 210000005259 peripheral blood Anatomy 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 102000013415 peroxidase activity proteins Human genes 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 230000002688 persistence Effects 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- CTRLRINCMYICJO-UHFFFAOYSA-N phenyl azide Chemical compound [N-]=[N+]=NC1=CC=CC=C1 CTRLRINCMYICJO-UHFFFAOYSA-N 0.000 description 1
- 150000003905 phosphatidylinositols Chemical class 0.000 description 1
- 102000020233 phosphotransferase Human genes 0.000 description 1
- 230000000243 photosynthetic effect Effects 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 229920002647 polyamide Polymers 0.000 description 1
- 229920002704 polyhistidine Polymers 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 230000002516 postimmunization Effects 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 125000001844 prenyl group Chemical group [H]C([*])([H])C([H])=C(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 150000003141 primary amines Chemical class 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000000644 propagated effect Effects 0.000 description 1
- KUAPETJWSCRJET-UHFFFAOYSA-N propane-1,1,3-tricarbaldehyde Chemical compound O=CCCC(C=O)C=O KUAPETJWSCRJET-UHFFFAOYSA-N 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 235000013772 propylene glycol Nutrition 0.000 description 1
- 210000004777 protein coat Anatomy 0.000 description 1
- 230000002797 proteolythic effect Effects 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 238000012797 qualification Methods 0.000 description 1
- 238000012207 quantitative assay Methods 0.000 description 1
- 238000003127 radioimmunoassay Methods 0.000 description 1
- 238000003156 radioimmunoprecipitation Methods 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 1
- 239000003488 releasing hormone Substances 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 210000003660 reticulum Anatomy 0.000 description 1
- 108010066533 ribonuclease S Proteins 0.000 description 1
- 235000009566 rice Nutrition 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
- 201000005404 rubella Diseases 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 238000007790 scraping Methods 0.000 description 1
- 238000006748 scratching Methods 0.000 description 1
- 230000002393 scratching effect Effects 0.000 description 1
- 238000007423 screening assay Methods 0.000 description 1
- 239000006152 selective media Substances 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 230000005582 sexual transmission Effects 0.000 description 1
- 108010061514 sialic acid receptor Proteins 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 230000003584 silencer Effects 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 238000004513 sizing Methods 0.000 description 1
- 210000002027 skeletal muscle Anatomy 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 210000004927 skin cell Anatomy 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- RYYKJJJTJZKILX-UHFFFAOYSA-M sodium octadecanoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCC([O-])=O RYYKJJJTJZKILX-UHFFFAOYSA-M 0.000 description 1
- 229940054269 sodium pyruvate Drugs 0.000 description 1
- JJGWLCLUQNFDIS-GTSONSFRSA-M sodium;1-[6-[5-[(3as,4s,6ar)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoylamino]hexanoyloxy]-2,5-dioxopyrrolidine-3-sulfonate Chemical compound [Na+].O=C1C(S(=O)(=O)[O-])CC(=O)N1OC(=O)CCCCCNC(=O)CCCC[C@H]1[C@H]2NC(=O)N[C@H]2CS1 JJGWLCLUQNFDIS-GTSONSFRSA-M 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 238000000527 sonication Methods 0.000 description 1
- 238000001179 sorption measurement Methods 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 239000000934 spermatocidal agent Substances 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 239000008227 sterile water for injection Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 238000012385 systemic delivery Methods 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 230000002381 testicular Effects 0.000 description 1
- 229940104230 thymidine Drugs 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 230000001131 transforming effect Effects 0.000 description 1
- 238000011820 transgenic animal model Methods 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 108091007466 transmembrane glycoproteins Proteins 0.000 description 1
- 102000035160 transmembrane proteins Human genes 0.000 description 1
- 108091005703 transmembrane proteins Proteins 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- 239000000225 tumor suppressor protein Substances 0.000 description 1
- 238000010396 two-hybrid screening Methods 0.000 description 1
- 238000000108 ultra-filtration Methods 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 239000012646 vaccine adjuvant Substances 0.000 description 1
- 229940124931 vaccine adjuvant Drugs 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 230000007501 viral attachment Effects 0.000 description 1
- 230000007486 viral budding Effects 0.000 description 1
- 230000019540 viral envelope fusion with host membrane Effects 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000004584 weight gain Effects 0.000 description 1
- 235000019786 weight gain Nutrition 0.000 description 1
- 229940051021 yellow-fever virus Drugs 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K19/00—Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/08—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
- C07K16/10—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
- C07K16/1036—Retroviridae, e.g. leukemia viruses
- C07K16/1045—Lentiviridae, e.g. HIV, FIV, SIV
- C07K16/1063—Lentiviridae, e.g. HIV, FIV, SIV env, e.g. gp41, gp110/120, gp160, V3, PND, CD4 binding site
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
- A61K39/21—Retroviridae, e.g. equine infectious anemia virus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
- A61P31/16—Antivirals for RNA viruses for influenza or rhinoviruses
-
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
- A61P31/22—Antivirals for DNA viruses for herpes viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/525—Virus
- A61K2039/5256—Virus expressing foreign proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/53—DNA (RNA) vaccination
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/545—Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55566—Emulsions, e.g. Freund's adjuvant, MF59
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/60—Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
- A61K2039/6031—Proteins
- A61K2039/605—MHC molecules or ligands thereof
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/24011—Poxviridae
- C12N2710/24111—Orthopoxvirus, e.g. vaccinia virus, variola
- C12N2710/24141—Use of virus, viral particle or viral elements as a vector
- C12N2710/24143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16111—Human Immunodeficiency Virus, HIV concerning HIV env
- C12N2740/16134—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- the inventions consisting of monoclonal antibodies to the FRMS and affinity tagged viral envelope protein, respectively, were made with government support under grant numbers R01 AI44669-01 and R21 AI44312-02 awarded by the National Institutes of Health. The Government has certain rights in these inventions.
- the present invention relates to the surprising discovery that highly effective vaccines may be constructed which present transitional fusion-related determinants to the immune system that engender unique antibodies capable of potently neutralizing a broad range of primary isolates, from worldwide locations and from different phylogenetic viral clades.
- the invention relates to the generation of Fusion-Related Molecular Structures (FRMS) which comprise one or more fusion-related determinants.
- FRMS Fusion-Related Molecular Structures
- the invention further relates to the discovery that the broad and uniform neutralization of diverse primary isolates indicates that the critical determinants presented by FRMS is highly conserved and may be intimately tied to the basic functioning of the envelope protein in binding and fusion.
- the present invention provides methods for formation, isolation and purification of the FRMS as well as the use of such FRMS in a variety of compositions and methods, including, for example, as vaccine immunogens, diagnostics, and therapeutics.
- the present invention concerns FRMS capable of eliciting neutralizing antibodies to viral pathogens and primary isolates of a virus. Antibodies raised to the FRMS can be used to study the molecular pathway toward fusion of the virus and host cell and to identify points of a possible antibody-mediated blockade to that pathway.
- the invention also relates to the use of antibodies of the invention for anti-viral agents, blood product additives, contraceptive additives passive immunization in post-exposure treatments or fetus immunization.
- Viruses are infectious agents responsible for many diseases in humans, animals, bacteria and plants, and are therefore of great medical, and commercial importance.
- a free virus particle called a virion
- a virion is made up of a genome (which can be RNA or DNA), associated proteins or polyamides, and a protein coat called a capsid.
- a capsid Some virions are further surrounded by a membranous envelope. Capsid and envelope structures serve to protect the genome from nucleases present in the environment and also serve to facilitate viral attachment and entry into the cell in which it will infect and replicate.
- a virus must utilize the biosynthetic machinery of the host cell to replicate. Viruses use the synthetic pathways and substrates available in animal cells to maintain and propagate their own genetic information. To gain access to the cell's biomachinery, the virus must enter or infect the cell. Entrance is facilitated by the presence of one or more viral receptor proteins on host cell membranes. Viral proteins bind to these receptors
- enveloped viruses infection is facilitated by fusion of the viral envelope with a cellular membrane such as a cell surface membrane or an internal membrane.
- a cellular membrane such as a cell surface membrane or an internal membrane.
- enveloped viruses are implicated in human and animal disease.
- HIV HIV
- HIV human immunodeficiency virus
- AIDS acquired immune deficiency syndrome
- HIV-1 Barre- Sinoussi, F., et al., 1983, Science 220:868-870; Gallo, R., et al., 1984, Science 224:500- 503
- HIV-2 Clavel, F., et al., 1986, Science 233:343-346; Guyader, M., et al., 1987, Nature 326:662-669).
- significant genetic heterogeneity exists within populations of each types.
- Retroviruses are small enveloped viruses that contain a single-stranded RNA genome, and replicate via a DNA intermediate produced by a virally-encoded reverse transcriptase, an RNA-dependent DNA polymerase (Varmus, H., 1988, Science 240:1427-1439).
- the HIV viral particle comprises a viral core, composed in part of capsid proteins, together with the viral RNA genome and those enzymes required for early replicative events.
- Myristylated gag protein forms an outer shell around the viral core, which, in turn, is surrounded by a lipid membrane envelope derived from an infected cell membrane.
- the HIV envelope surface glycoproteins are synthesized as a single 160 kilodalton precursor protein which is cleaved by a cellular protease during viral budding into two glycoproteins, gp41 and gpl20.
- gp41 is a transmembrane glycoprotein and gpl20 is an extracellular glycoprotein which remains non-covalently associated with gp41, possibly in a trimeric or multimeric form (Hamrnarskjold, M., et al., 1989, Biochem. Biophys. Acta 989:269-280).
- CD4 cellular membrane protein acts as a cellular receptor for the HIV-1 virus (Dalgleish, A., et al., 1984, Nature
- the viral envelope protein Upon interaction of the viral envelope protein and cellular receptor, the
- envelope protein-CD4 complex undergoes conformational changes facilitating subsequent interaction with host cellular co-receptors to form a trimolecular complex. Further conformational changes in the trimolecular complex allow fusion of the apposed cell membrane and virus envelope releasing viral genetic material into the cell.
- reverse transcriptase has been one focus of drug development.
- a number of reverse-transcriptase-targeted drugs including 2',3'-dideoxynucleoside analogs such as AZT, ddl, ddC, and d4T have been developed which have been shown to been active against HIV (Mitsuya, H., et al., 1991, Science 249: 1533-1544).
- the new treatment regimens for HIV-1 demonstrate that a combination of anti-HIV compounds, which target reverse transcriptase (RT), such as azidothymidine (AZT), lamivudine (3TC), dideoxyinosine (ddl), dideoxycytidine (ddC) used in combination with an HIV-1 protease inhibitor have a far greater effect (2 to 3 logs reduction) on viral load compared to AZT alone (about 1 log reduction).
- RT reverse transcriptase
- AZT azidothymidine
- ddl dideoxyinosine
- ddC dideoxycytidine
- improved results have recently been obtained with a combination of AZT, ddl, 3TC and ritonavir (Perelson, A.S., et al., 1996, Science 15:1582-1586).
- cytotoxic therapy may also lead to suppression of CD8 + T cells, which are essential to the control of HIV, via killer cell activity (Blazevic, V., et al., 1995, AIDS Res. Hum. Retroviruses 11:1335-1342) and by the release of suppressive factors, notably the chemokines Rantes, MLP-lc and MlP-l ⁇ (Cocchi, F., et al., 1995, Science 270: 1811-1815).
- Recombinant soluble CD4 has been shown to inhibit infection of CD4 + T cells by some HIV-1 strains (Smith, D.H., et al., 1987, Science 238:1704-1707). Certain primary HIV-1 isolates, however, are relatively less sensitive to inhibition by recombinant CD4 (Daar, E., et al, 1990, Proc. Natl. Acad. Sci. USA 87:6574- 6579). In addition, recombinant soluble CD4 clinical trials have produced inconclusive 0 results (Schooley, R., et al., 1990, Ann. Int. Med.
- Late stage processing is dependent on the activity of a viral protease, and drugs are being developed which inhibit this protease (Erickson, J., 1990, Science 249:527- 533).
- drugs are being developed which inhibit this protease (Erickson, J., 1990, Science 249:527- 533).
- chemokines produced by CD8 + T cells have been implicated in suppression of HIV infection (Paul, W.E., 1994, Cell 82:177; Bolognesi, D.P., 1993, Semin. Immunol. 5:203).
- chemokines RANTES, MLP-l ⁇ and MlP-l ⁇ which are secreted by CD8 + T cells, were shown to suppress HIV-1 p24 antigen production in cells infected with HIV-1 or HIV-2 isolates in vitro (Cocchi, F, et al., 1995, Science 270:1811-1815). Thus, these and other chemokines may prove useful in therapies for HTV infection. The clinical outcome, however, of all these and other candidate drugs is still in question.
- inactivated or attenuated pathogens Traditional methods for preparing vaccines include the use of inactivated or attenuated pathogens.
- a suitable inactivation of the pathogenic 1 ⁇ microorganism renders it harmless as a biological agent but does not destroy its immunogenicity. Injection of these "killed" particles into a host will then elicit an immune response capable of preventing a future infection with a live microorganism.
- a major concern in the use of killed vaccines (using inactivated pathogen) is failure to inactivate all the microorganism particles. Even when this is accomplished, since killed n pathogens do not multiply in their host, or for other unknown reasons, the immunity achieved is often incomplete, short lived and requires multiple immunizations. Finally, the inactivation process may alter the microorganism's antigens, rendering them less effective as immunogens.
- Attenuation refers to the production of strains of pathogenic microorganisms which have essentially lost their disease-producing ability.
- One way to accomplish this is to subject the microorganism to unusual growth conditions and/or frequent passage in cell culture. Mutants are then selected which have lost virulence but yet are capable of eliciting an immune response. Attenuated pathogens often make good irnrnunogens as they actually replicate in the host cell and elicit long lasting immunity.
- problems are _ n encountered with the use of live vaccines, the most worrisome being insufficient attenuation and the risk of reversion to virulence.
- envelope proteins gpl 60, gpl20, gp41
- envelope proteins gpl 60, gpl20, gp41
- these proteins have been the most common candidates to act as immunogens for anti-HIV vaccine development.
- Several groups have begun to use various portions of gpl60, g ⁇ l20, and/or gp41 as immunogenic targets for the host immune system. See for example, Ivanoff, L., et al., U.S. Pat. No. 5,141,867; Saith, G., et al., WO92/22,654; Shafferman, A., WO91/09,872; Formoso, C, et al, WO90/07,119.
- PB Is peripheral blood lymphocytes
- Laboratory adapted strains are those which are persistently grown in established T cell lines in the laboratory (See Wrin et al., 1995, J. of Virology 69:39). Previous vaccine candidates have elicited neutralizing antibodies to laboratory-adapted strains. For example, Wrin et al., 1995, Journal of Virology 69:39, tested HIV envelope proteins produced by recombinant
- Neutralizing antibodies are important to prevent binding and fusion of the virus to the host cell.
- the action of antibodies that neutralize laboratory-adapted isolates have been analyzed.
- Neutralizing antibodies directed specifically to the CD4 binding domain on gpl20 interfere with CD4 binding.
- some neutralizing antibodies inhibit interaction with the co-receptor (Trkola et al., 1998, Journal of Virology 72:1876; Wu, et al., 1996, Nature 384:179-83).
- the present invention is based on the surprising discovery that highly effective vaccines may be constructed which present transitional fusion-related determinants to the immune system that engender unique antibodies capable of potently neutralizing a broad range of primary isolates of enveloped viruses, from worldwide locations and from different phylogenetic viral clades.
- the vaccine of the invention comprise as immunogens Fusion-Related Molecular Structures (FRMS) which comprise one or more fusion-related determinants.
- FRMS Fusion-Related Molecular Structures
- the invention is further based on the discovery that the broad and uniform neutralization of diverse primary isolates indicates that the critical determinants presented by FRMS are highly conserved and may be intimately tied to the basic functioning of the envelope protein in binding and fusion.
- the present invention provides the use of such FRMS in a variety of compositions and methods, including, for example, as vaccine immunogens, blood product additives, anti-viral agents, diagnostics and therapeutics.
- the present invention concerns FRMS capable of eliciting neutralizing antibodies to enveloped viral pathogens. Epitopes which elicit these neutralizing antibody result from conformational changes during the process of binding and fusion of the viral envelope and host cell membranes.
- the complexes result from the interaction of viral proteins with at least one host cellular receptor or co-receptor and are created by co-culturing cells transformed with a nucleic acid expressing a viral protein and cells expressing host cellular receptor(s).
- the cells recombinantly express the host cell receptor(s).
- cultures are fixed at the onset of cell-cell interaction to preserve fusion-competent immunogens.
- the fusion-related immunogens are formed as a result of the interaction of the major human immunodeficiency virus type 1 (HIV-1) envelope protein, the host cellular receptor CD4 and the host cellular co-receptor CCR5.
- HIV-1 major human immunodeficiency virus type 1
- Vaccinating with the FRMS of the subject invention as immunogens raises an immune response to the viral pathogen and elicits the production of neutralizing antibodies in the vaccinated host.
- antibodies raised to the FRMS can be used to study the molecular pathway toward fusion of the virus and host cell and to identify points of a possible antibody-mediated blockade to that pathway.
- specific high-affinity "tags" may be expressed as part of the envelope protein or host cellular receptor(s) that faclilitates isolation and purification of the FRMS of the invention.
- the present invention further relates to the development of monoclonal antibodies (mAb) elicited by these FRMS immunogens.
- Monoclonal antibodies which are _ n capable of neutralization of primary isolates are provided.
- Neutralizing mAbs to functionally conserved fusion-dependent epitopes may also be useful for passive immunization in post-exposure treatments or fetus immunization.
- the present invention provides an isolated molecular structure comprising an epitope formed as a result of association of (a) an envelope protein of an enveloped virus, with (b) one or more cellular membrane proteins, which envelope protein and cellular membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins.
- the present invention provides an isolated molecular structure comprising an epitope formed as a result of association of (a) an HIV envelope protein, or a mutant thereof - n that assembles into the viral envelope; with (b) human CD4 and a co-receptor for HIV fusion.
- co-receptor is the chemokine receptor CCR5 or CXCR4.
- the molecular structure is formed by association of a mutant of HIV gp41 that is fusion-defective.
- the mutant contains one or more mutations selected from the group consisting of V2E, G10V, V570R, and Y586E.
- the molecular structure is formed by association of wild-type HIV envelope protein. s
- the present invention provides an isolated molecular structure comprising an epitope formed as a result of association of (a) a mutant envelope protein of an enveloped virus, which envelope protein in wild-type form functions in fusion of the viral envelope with a host cell membrane, and which mutant envelope protein is fusion-defective; and (b) one or more host cellular membrane proteins which function as receptors for said envelope protein.
- said virus is from a viral family is selected from the group consisting of Retro viridae, Rhabdoviridae, Caronaviridae, Filoviridae, Poxviridae, Bunyaviridae, Flaviviridae, Togaviridae, Orthomyxoviridae, Paramyxoviridae, and n Herpesviridae.
- the envelope protein is El and E2 of HCV and the cellular membrane protein is CD81.
- the molecular structure is a cross-linked cellular molecular structure.
- the molecular structure is isolated from a cell lysate.
- the cellular molecular structure comprises cells recombinantly expressing the envelope protein.
- the cell lysate is from a plurality of cells comprising cells recombinantly expressing the envelope protein.
- the cellular molecular structure further comprises cells recombinantly expressing the one or more host cellular membrane proteins.
- the present invention provides a recombinant enveloped virus, wherein said virus recombinantly expresses on its envelope a cell receptor for a native envelope protein n of said virus.
- said cell receptor is human CD4 or a co-receptor for HIV, or said virus recombinantly expresses both human CD4 and said co-receptor.
- said suitable conditions comprise a lowering of pH.
- the cross-linked cellular molecular structure further comprises a cross-linked viral particle of said virus, containing said envelope protein.
- the present invention provides a vaccine formulation comprising an immunogenic amount of the molecular structure and a pharmaceutically acceptable carrier.
- the present invention provides a monoclonal antibody to the molecular structure.
- the antibody is labeled.
- the present invention provides a purified polyclonal antiserum specific to - n the molecular structure.
- the present invention provides a contraceptive jelly, foam, cream, or ointment comprising an amount of the antibody effective to inhibit or decrease infection by the virus.
- the present invention provides a contraceptive jelly, foam, cream, or ointment comprising an amount of the antibody effective to inhibit or decrease infection by HIV.
- the present invention provides a contraceptive jelly, foam, cream, or ointment comprising an amount of the antiserum effective to inhibit or decrease infection by HIV.
- the present invention provides a sample of mammalian blood, to which an amount of the antibody has been added effective to inhibit or decrease infection by the virus.
- the present invention provides a sample of human blood, to which an amount of the antibody has been added effective to inhibit or decrease infection by HIV.
- said envelope protein or host cellular membrane proteins further comprises an affinity tag.
- said envelope protein, CD4, or co- receptor further comprises an affinity tag.
- the antibody is labeled.
- the present invention provides a kit comprising in one or more containers a labeled monoclonal antibody to the molecular structure.
- the present invention provides a kit comprising in one or more containers a labeled monoclonal antibody to the molecular structure.
- the kit comprises the molecular structure in a separate container.
- the present invention provides a cell line that recombinantly expresses an envelope protein of an enveloped virus that functions in fusion of the viral envelope with a ⁇ host cell membrane, or a mutant form of said envelope protein that is fusion-defective, which cell line expresses one or more cellular membrane proteins that function as receptors for said envelope protein.
- said one or more proteins that function as receptors are recombinantly expressed.
- the present invention provides a cell line that recombinantly expresses HIV ? , gpl60, which cell line expresses CD4 and a co-receptor for HIV; said cell line lacking a functional protease that cleaves gpl60 to produce gpl20 and gp41.
- the present invention provides a method of treating or preventing infection by a virus in a subject comprising administering to the subject an immunogenic amount of the molecular structure effective to treat or prevent infection by the virus.
- the subject is a human.
- the subject is a domestic animal.
- the present invention provides a method of treating or preventing infection by HIV in a human comprising administering to the human an immunogenic amount of the molecular structure effective to treat or prevent infection by HIV.
- the present invention provides a method of treating or preventing infection by a virus in a subject comprising administering to the subject an amount of the monoclonal antibody effective to treat or prevent infection by the virus. ;
- the present invention provides a method of treating or preventing infection by HIV in a human comprising administering to the human an amount of the monoclonal antibody effective to treat or prevent infection by HIV.
- said human has a high risk of HIV infection.
- the method is for treatment of AIDS in said human.
- the present invention provides a method for treating or preventing infection by HIV in a human fetus comprising administering to a pregnant human containing said fetus an amount of the monoclonal antibody effective to treat or prevent infection by HIV in said fetus.
- the present invention provides a method of inhibiting infection by a virus in a sample of blood comprising contacting said sample of blood with an amount of the monoclonal antibody effective to inhibit infection by said virus.
- the present invention provides a method of inhibiting infection by HIV in a sample of human blood comprising contacting said sample of human blood with an amount of the monoclonal antibody effective to inhibit infection by HIV.
- the present invention provides a method of decontaminating surgical or dental tools comprising contacting said tools with an amount of the monoclonal antibody effective to inhibit infection by said virus.
- the present invention provides a method of decontaminating surgical or dental tools comprising contacting said tools with an amount of the monoclonal antibody effective to inhibit infection by HIV.
- the present invention provides a method for monitoring the production of antibody to the molecular structure in a subject previously administered an amount of the molecular structure comprising isolating from said subject a sample comprising serum; and detecting the presence of any antibodies to the molecular structure in said serum.
- said detecting is carried out by a method comprising performing a competitive immunoassay with labeled antibody to the molecular structure.
- the present invention provides a method of producing an immunogen for use in a vaccine for the treatment or prevention of infection by a virus comprising the following steps in the order stated: (a) contacting an envelope protein or chimeric form thereof of an enveloped virus, which envelope protein functions in fusion of the viral envelope with a cell membrane, or a mutant form or chimeric form thereof of said envelope protein that is fusion-defective, with one or more cell proteins or chimeric forms thereof that function as receptors for said envelope protein; (b) exposing said envelope protein or chimeric form thereof or mutant form or chimeric form thereof, and said one or more host cell proteins or chimeric forms thereof, to a cross-linking agent; and (c) isolating a cross-linked structure comprising said envelope protein or chimeric form thereof or mutant form or chimeric form thereof.
- the present invention provides a method of producing an immunogen for use in a vaccine for the treatment or prevention of infection by HIV comprising the following steps in the order stated: (a) co-culturing a first cell recombinantly expressing HIV envelope protein or a chimeric form thereof, or a mutant form or chimeric form thereof of said envelope protein that is fusion-defective, with a second cell that expresses (i) human CD4 or a chimeric form thereof, and (ii) a co-receptor for HIV or a chimeric form thereof; (b) exposing said envelope protein or chimeric form thereof or mutant form or chimeric form thereof, and said CD4 or chimeric form thereof and co-receptor or chimeric form thereof, to a cross-linking agent; and (c) isolating a cross-linked structure comprising said envelope protein or chimeric form thereof or mutant form or chimeric form thereof.
- said virus is HIV and said host cell proteins are human CD4 and a co-receptor for HIV.
- said second cell recombinantly expresses CD4 or said co- receptor or both CD4 and said co-receptor or chimeric forms of any of the foregoing.
- said first and second cell are the same cell type.
- said envelope protein or chimeric form thereof or mutant form or chimeric form thereof is present on a viral particle or virus-like particle.
- said cross-linked structure is a cross-linked cellular complex.
- the virus is selected from the group consisting of Retroviridae, Rhabdoviridae, Coronaviridae, Filoviridae, Poxviridae, Bunyaviridae, Flaviviridae, Togaviridae, Orthomyxoviridae, Paramyxoviridae, and Herpesviridae.
- said contacting step occurs by infecting cells expressing said host cell proteins with said virus expressing said envelope protein or chimeric form thereof or mutant form or chimeric form thereof.
- a chimeric form of said envelope protein or one of said host cell proteins is contacted, said chimeric form comprising an affinity tag.
- a chimeric form of said envelope protein or CD4 or said co-receptor is contacted, said chimeric form comprising an affinity tag.
- the present invention provides a method of producing an immunogen for use in a vaccine for the treatment or prevention of infection by HIV comprising the following steps in the order stated: (a) co-culturing a first cell recombinantly expressing HIV envelope protein or a chimeric form thereof, or a mutant form or chimeric form thereof of said envelope protein that is fusion-defective, with a second cell that expresses (i) human CD4 or a chimeric form thereof, and (ii) a co-receptor for HIV or a chimeric form thereof wherein at least one of said chimeric forms comprising an affinity tag is expressed; (b) lysing said co-cultured cells to form a cell lysate under non-denaturing conditions; and (c) isolating from said cell lysate a molecular structure comprising said envelope protein or chimeric form thereof or mutant form or chimeric form thereof by a method comprising contacting said cell lysate with a binding partner to said affinity tag and recovering
- the present invention provides a cross-linked structure that is the product of the method.
- the present invention provides a monoclonal antibody to the structure of that neutralizes in vitro the following primary isolates of HIV: 92US657, 92US660, 92RW023, 93IN101, 92UG035, and 92TH023.
- the present invention provides a contraceptive, jelly, foam, cream or ointment comprising an amount of the antibody to inhibit or decrease infection by HIV.
- the present invention provides a method of treating or preventing infection by a virus in a subject comprising administering to the subject an immunogenic amount of the structure effective to treat or prevent infection by the virus.
- the present invention provides a method of treating or preventing infection by HIV in a human comprising administering to the human an immunogenic amount of the molecular structure effective to treat or prevent infection by HIV.
- the present invention provides a method of decontaminating surgical or dental tools comprising contacting said tools with an amount of the monoclonal antibodylay effective to inhibit infection by HIV.
- the present invention provides a method of decontaminating surgical or dental tools comprising contacting said tools with an amount of the monoclonal antibody effective to inhibit infection by said virus.
- the present invention provides a method of screening a molecular structure for vaccine efficacy comprising immunizing a transgenic non-human mammal with the molecular structure, wherein said transgenic non-human mammal expresses from one or more transgenes both human CD4 and a co-receptor for HIV, and detecting any neutralizing antibodies to HIV that are produced by said mammal.
- the present invention provides a method of screening a molecular structure for vaccine efficacy comprising immunizing a transgenic non-human mammal with the molecular structure, wherein said transgenic non-human mammal expresses from one or more transgenes said one or more host cellular membrane proteins; and detecting any neutralizing antibodies to said virus that are produced by said mammal.
- the mammal is a mouse.
- said first cell recombinantly expresses HIV envelope protein or a chimeric form thereof.
- mice hu CD4+, hu CCR5+, mouse CD4+
- Sera were tested for neutralization of 168P using U87-CD4 cells expressing either CXCR4 (black symbols) or CCR5 (white symbols).
- Data represent averages of three to six neutralization assays using serum obtained 2 weeks following second and third immunization.
- FIG. 2A-B Neutralization of P168 by FC but not FI, vaccine sera.
- PBLs were isolated, stimulated with phytohemagglutinin, and grown in the presence of interleukin-2; neutralization . was determined. HIV p24 antigen was determined after 5 days of culture by ELISA (Coulter Corporation) and values were normalized to the virus control
- FC vaccine serum does not neutralize pseudotyped HIV virions bearing - discipline amphotropic MLV envelope protein (A) or primary SIVmac251 (B).
- A amphotropic MLV envelope protein
- B primary SIVmac251
- FC Immunogen black squares
- FI immunogen Cos-env + U87 cells, gray circles
- FIG. 4A-B Neutralization of TCLA 168C virus by FI vaccine sera.
- Neutralization sensitivity of the 168P PI virus (A) and its TCLA derivative 168C (B) were tested in U87- CD4-CXCR4 cells with pooled sera: FC immunogen (black squares), FI immunogens (COS-env + U87 cells, gray circles; COS-env + U87-CD4 cells, gray diamonds; COS-env +sCD4, white diamonds), and mock-transfected cell controls (white circles).
- FIG. 1 Neutralizing activity of two HIV Clade B primary isolates with hybridoma supernatants.
- FIG. 9 Serum titers of mice immunized with fusion-competent vaccine immunogens.
- White square represents fusion-competent (FRMS) immunogen (env+with CD4/CCR5); black circles represent immunogen from cells expressing envelope protein immunogen and cells expressing neither CD4 nor CCR5 (env); white circles represent immunogen from cells expressing envelope protein and cells expressing CD4 immunogen (env+CD4); white triangle represent cell controls expressing CD4 and CCR5 immunogen (CD4/CCR5).
- FRMS fusion-competent
- FRMS fusion-competent
- black circles represent immunogen from cells expressing envelope protein immunogen and cells expressing neither CD4 nor CCR5 (env)
- white circles represent immunogen from cells expressing envelope protein and cells expressing CD4 immunogen (env+CD4)
- white triangle represent cell controls expressing CD4 and CCR5 immunogen (CD4/CCR5).
- Figure 10 Cross-neutralization of the 320SI primary virus isolate by antibodies obtained from mice immunized with a fusion-competent immunogen complex of the subject invention.
- Black circles represent fusion-competent (FRMS) immunogen (env with CD4/CCR5); white squares represent immunogen from cells expressing envelope protein (env); white circles represent immunogen from cells expressing envelope protein and CD4 (env + CD4).
- FRMS fusion-competent
- env fusion-competent
- env envelope protein
- CD4 env + CD4
- Envelope protein co-purified with CD4-Spep and CCR5-Spep detected by Western blot analysis, a) 168P envelope protein isolated following co-culture with cells expressing CD4-Spep b) 168P envelope protein isolated following co-culture with cells expressing CD4 and CCR5-Spep c) 168P envelope protein was undetectable following co-culture with mock-transfected cells.
- the present invention concerns fusion-related molecular-structures (FRMS) capable of eliciting neutralizing antibodies to enveloped viral pathogens.
- FRMS of the invention comprise epitopes formed as a result of the association of one or more cellular molecules with one or more viral envelope molecules.
- the process of viral envelope fusion n with a cell membrane is initiated following binding of a viral envelope protein to specific cell receptors and/or co-receptors on the host cell and involves fusion of the viral envelope with surface or internal cell membranes. Fusion is driven by conformational changes which occur within the envelope-receptor/co-receptor complex.
- the FRMS of the invention may be used for a variety of pu ⁇ oses including but not limited to those presented in Sections 5.5 and 5.7, herein.
- the FRMS when used to vaccinate may be used for a variety of pu ⁇ oses including but not limited to those presented in Sections 5.5 and 5.7, herein.
- the FRMS when used to vaccinate may be used for a variety of pu ⁇ oses including but not limited to those presented in Sections 5.5 and 5.7, herein.
- the present invention provides an isolated molecular structure comprising an epitope formed as a result of association of (a) an envelope protein of an enveloped virus, with (b) one or more cellular membrane proteins, which envelope protein and cellular membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins.
- a cellular membrane protein includes proteins of the cell surface membrane and internal membranes (including but not limited to the endoplasmic reticulum membrane).
- Such cellular membrane protein may be an integral membrane protein (e.g., a trans-membrane protein), or may be attached to the membrane by an attached lipid (e.g., fatty acid chain or prenyl group) or by an oligosaccharide (e.g., to a phopholipid, phosphatidylinositol), or the cellular membrane protein may also be associated with the membrane by non-covalent interactions (e.g., by association with an integral membrane protein).
- an integral membrane protein e.g., a trans-membrane protein
- an attached lipid e.g., fatty acid chain or prenyl group
- an oligosaccharide e.g., to a phopholipid, phosphatidylinositol
- the cellular membrane protein may also be associated with the membrane by non-covalent interactions (e.g., by association with an integral membrane protein).
- the present invention also provides an isolated molecular structure comprising an epitope formed as a result of association of (a) a mutant envelope protein of an enveloped virus, which envelope protein in wild-type form functions in fusion of the viral envelope with a host cell membrane, and which mutant envelope protein is fusion- defective; and (b) one or more host cellular membrane proteins which function as receptors for said envelope protein.
- the present invention provides an isolated molecular structure comprising an epitope formed as a result of association of (a) an HIV envelope protein, or a mutant thereof that assembles into the viral envelope; with (b) human CD4 and a co-receptor for HIV fusion.
- the co-receptor is the chemokine receptor CCR5 or CXCR4.
- the molecular structure is formed by association of a mutant of HIV gp41 that is fusion-defective.
- the mutant contains one or more mutations selected from the group consisting of V2E, G10V, V570R, and Y586E.
- the molecular structure is formed by association of wild-type HIV envelope protein.
- the envelope protein is El and E2 of Flavivirus HCV and the cellular
- membrane protein is CD81.
- the present invention also provides a method of producing an immunogen for use in a vaccine for the treatment or prevention of infection by a virus comprising the following steps in the order stated: (a) contacting an envelope protein or chimeric form thereof of an enveloped virus, which envelope protein functions in fusion of the viral
- said virus is HIV and said host cell proteins are human CD4 and a co-receptor for HIV.
- the present invention also provides a method of producing an immunogen for use in a vaccine for the treatment or prevention of infection by HIV comprising the following steps in the order stated: (a) co-culturing a first cell recombinantly expressing HIV envelope protein(s) or a chimeric form thereof, or a mutant
- form or chimeric form thereof of said envelope protein that is fusion-defective with a second cell that expresses (i) human CD4 or a chimeric form thereof, and (ii) a co-receptor for HIV or a chimeric form thereof; (b) exposing said envelope protein or chimeric form thereof or mutant form or chimeric form thereof, and said CD4 or chimeric form thereof and co-receptor or chimeric form thereof, to a cross-linking agent; and (c) isolating a cross-
- n linked structure comprising said envelope protein or chimeric form thereof or mutant form or chimeric form thereof.
- said second cell recombinantly expresses CD4 or said co-receptor or both CD4 and said co-receptor or chimeric forms of any of the foregoing.
- the present invention provides a method of
- producing an immunogen for use in a vaccine for the treatment or prevention of infection by HIV comprising the following steps in the order stated: (a) co-culturing a first cell recombinantly expressing HIV envelope protein or a chimeric form thereof, or a mutant form or chimeric form thereof of said envelope protein that is fusion-defective, with a second cell that expresses (i) human CD4 or a chimeric form thereof, and (ii) a co-receptor
- the present invention relates to FRMS which comprise an epitope formed as a result of the association of one or more viral envelope molecule(s) and one or more host _ ⁇ cell molecule(s).
- the FRMS of the invention encompass structures resulting from the association of at least one viral envelope protein and at least one host cell protein.
- a FRMS of the invention may encompass fusion-competent complexes (such as complexes formed during fusion events of the viral envelope with a cellular membrane), fusion-defective complexes (such as those formed during pre-fusion events between a fusion-defective viral envelope and a cellular membrane) as well as complexes formed by the association of one or more viral envelope molecules with one or more cellular receptor and or co-receptor for the virus.
- the FRMS result from the association of HIV envelope protein with CD4 and a chemokine receptor.
- the FRMS of the invention thus comprise epitopes capable of eliciting neutralizing antibodies to the virus.
- the FRMS of the invention are formed by a variety of methods as described herein.
- cells expressing one or more components whose association results in the FRMS are cultured to form the FRMS. All the components may be expressed on one cell, or a first component may be expressed on one cell, and a second component may be expressed on a different cell, and a third component (if any) may be expressed on one of the foregoing cells or a different cell.
- one of the cells may be replaced by a viral particle or pseudovirus or recombinant virus or virus-like particle containing the component(s) on its surface.
- the components may each be endogenously or recombinantly expressed by the cells.
- FRMS may be formed by the interaction of
- a soluble form of a viral envelope protein of a envelope virus is used in the formation of a FRMS of the invention.
- the soluble envelope protein may be added t o host cells in n vitro which host cells express the rece ⁇ tor(s) for the virus.
- the association of the soluble viral envelope protein and host cell receptor(s) results in the formation of a FRMS.
- one or more soluble cell recptor(s) are used in formation of the FRMS.
- soluble cellular receptor(s) whose association with the viral envelope protein(s) results in the formation of a r FRMS, may be used in the formation of a FRMS by contacting the viral envelope protein(s) with said soluble cell receptor(s).
- all of the components whose association result in the formation of the FRMS are in a soluble form and the FRMS is reconstitutional in vitro.
- the reconstitution of a FRMS may optionally be aided by the addition of a mAb to the FRMS.
- the FRMS is forced upon a decrease in pH.
- pH-medicated cell fusion allows for the formulation of the FRMS for enveloped viruses which cellular membrane proteins are located in internal cellular membranes.
- the cellular components involved in the formation of a FRMS for a particular virus include the cell receptors and/or co-receptors for the particular virus.
- At least some of these components are endogenously expressed by the cell.
- any cell known in the art which expresses a cellular component of the FRMS may be used in the formation of a FRMS.
- Cells expressing native cellular receptors and/or co-receptors for viral proteins can be used.
- human CD4 is known in the art as a
- cells endogenously expressing CD4 are used in the formation of a FRMS.
- Cells endogenously expressing a component of the complexes of the invention may comprise one or more of the cellular components used to form the FRMS.
- cells may be used which express all of the required cellular components of the
- FRMS or cells may be used which do not express all of the required cellular components.
- any cell which expresses both CD4 and an HIV co-receptor, such as CCR5 may be used in the formation of an HIV FRMS.
- any cell which endogenously express CD4 but does not endogenously express an HIV co- receptor in this embodiment, and as discussed below, components of the complex which
- the co-receptor CXCR4 is predominantly located on T cell lines.
- the ⁇ . co-receptor CCR5 is predominantly located on macrophage cells.
- the co-receptor CXCR4 is predominantly located on T cell lines.
- the co-receptor CCR5 is predominantly located on macrophage cells.
- Exogenous components may be introduced into the cell by any method known in the art, including those described in Section 5.1.2.
- a cell transformed by means of a viral gene or genome integration, which expresses a viral component of the FRMS may be used in the formation of a fusion related complex.
- the cell may be said to endogenously express a viral component important in the construction of an FRMS.
- virally-transformed cells may also endogenously express one or more cellular components of the FRMS.
- viral particles may be used as a source of endogenous expression of viral components of a FRMS.
- Any of the components important in the construction of a FRMS or fragments, analogs or derivatives thereof may be introduced into a cell such that the
- exogenous component is expressed in the cell.
- the present invention encompasses expression systems, both eucaryotic and procaryotic expression vectors, which may be used to express a component important in the formation of a FRMS or fragment, analog or derivative thereof of the present invention.
- the viral envelope protein is
- the exogenous viral envelope protein may be introduced into the cell by transfection or viral vectors as described herein, or by infection with the live or attenuated virus comprising the viral envelope protein.
- viral infection can lead to production of a viral envelope protein in preferred embodiments, the expression of a viral envelope protein on the cell membrane is enhanced by the modification (e.g. by
- ⁇ recombinant means of the viral envelope protein so as to increase its expression on the cell surface, or other cell membrane.
- targeting signals are known in the art that enhance delivery of cellular proteins to particular subcellular compartments or locations.
- an endoplasimic reticulum signal, KDEL, (one letter amino acid codes) could be engineered into the viral envelope protein to promote targetry of the protein to the ER ⁇ when a cell is infected with the virus.
- the invention also provides a cell expressing the viral and cellular components important for the formation of a FRMS.
- a cell is constructed to express the cell receptor and/or co-receptor for a virus and to recombinantly express the viral envelope protein.
- Such cell may endogenously express the host cell ⁇ - receptors and/or co-receptors for the virus or recombinantly express one or both of such protein(s).
- the nucleic acids encoding such proteins may be expressed from different vectors or from a single vector (e.g., wherein a vector comprises multiple genes operably linked to a single promoter).
- ⁇ ⁇ formation of a FRMS are expressed on an individual cell.
- a cell is constructed to recombinantly express CD4, a co-receptor CCR5 or CXCR4 and HIV gpl60.
- gpl20 and gp41 may be used in place of gpl60.
- the viral envelope protein is also constructed such that it is lacking the
- native protease site responsible for cleavage of gp 160 to gp 120 and gp41.
- an alternative protease site is constructed into the viral envelope protein which protease is not native to the host cell (such as a bacterial protease). Formation of the FRMS is triggered on the cell expressing the three proteins, by contacting the cell with the non- native protease (such as by adding an amount effective to induce cleavage, to the cell media).
- a variety of proteases and protease sites are known in the art and may be used in accordance with the invention.
- nucleotide sequence coding for a viral envelope protein or cellular receptor protein of the invention or a functionally active analog or fragment or other derivative thereof can be inserted into an appropriate expression vector, e.g., a vector which contains the necessary elements for the transcription and translation of the inserted protein- coding sequence and/or for carrying out the methods of the invention.
- the invention encompasses the use of DNA expression vectors and/or viral vectors that contain coding sequences of a component of FRMS or analog, derivative or fragment thereof operatively associated with a regulatory element that directs expression of the coding sequences and genetically engineered host cells that contain any of the foregoing coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences in the host cell.
- the DNA expression vectors and viral vectors containing the nucleic acids encoding a component important in the formation of a FRMS of the present invention may be produced by recombinant DNA technology using techniques well known in the art.
- methods for preparing the expression vectors and viral vectors of the invention by n expressing nucleic acid containing sequences encoding a component important in the formation of a FRMS or analog, derivative or fragment thereof are described herein.
- Methods which are well known to those skilled in the art can be used to construct expression vectors containing gene product coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in
- gene product sequences may be chemically synthesized using, for example, oligonucleotide synthesizers. See, for example, the techniques described in "Oligonucleotide Synthesis", 1984, Gait, M.J. ed., LRL Press, Oxford, which is inco ⁇ orated by reference herein in its entirety.
- Gene expression may be regulated by a variety of methods known in the art including but not limited to those presented in Mizuno, T. et al.,
- the nucleic acids or DNA encoding one or more components which result in the formation of the FRMS may be introduced into a cell may be accomplished by a variety of methods, such as liposomes, electroporation, transfection, viral vectors, bacteriophage, etc.
- nucleic acids encoding one or more components important in the formation of a FRMS is accomplished by packaging plasmid DNA
- ⁇ - comprising the nucleic acids that code for the component(s) into liposomes or by complexing the plasmid DNA comprising nucleic acids that code for the component(s) with lipids or liposomes to form DNA-lipid or DNA-liposome complexes.
- the liposome is composed of cationic and neutral lipids commonly used to transfect cells in vitro. The cationic lipids complex with the plasmid DNA and form liposomes.
- Cationic liposomes can be complexed with the a negatively-charged biologically active molecule (e.g., DNA) by mixing these components and allowing them to charge-associate.
- Cationic liposomes are particularly useful with a nucleic acid because of the nucleic acids negative charge.
- cationic liposomes include lipofectin, lipofectamine, lipofectace and DOTAP (Hawley-Nelson et al., 1992, Focus 15(3):73-83; Feigner et al., 1992, Proc. Natl. Acad. Sci. U.S.A.
- cationic lipids are also available, e.g., dimethyldioctadeclammonium bromide (DDAB); a biodegradable lipid 1, 2-bis(oleoyloxy)- 3-(trimethylammonio) propane (DOTAP); these liposomes may be mixed with a neutral ⁇ lipid, e.g., L- dioleoyl phosphatidylethanolamine (DOPE) or cholesterol (Choi), two commonly used neutral lipids for systemic delivery.
- DOPE L- dioleoyl phosphatidylethanolamine
- Choi cholesterol
- DNA:liposome ratios may be optimized using the methods used by those of skill in the art (see e.g., Nicolau et al., 1987, Methods Enzymol. 149:157) and can readily be utilized herein by one of ordinary skill in the art to encase the complex of this invention.
- the plasmid DNA coding for the genes or nucleic acids encoding one or more components resulting in the formation of a FRMS of the invention may be delivered via polycations, molecules which carry multiple positive charges and are used to achieve gene transfer in vitro, in vivo and ex vivo. Polycations, such as polyethilenimine, may be used to achieve successful gene
- Recombinant viruses can also be used to deliver the components whose association results in the FRMS. Cells infected by these viruses or transformed by the viral genome will thus express the desired components.
- vector expressing one or more component(s) described herein can be engineered.
- a variety of viruses may be genetically engineered to express a component important in the formation of a FRMS.
- it may be desired that the recombinant viruses are either cold adapted, temperature sensitive, or attenuated.
- viruses and viral vectors may be used to deliver the nucleotide sequences encoding one or more
- Retroviral vectors are commonly used to deliver genes to host cells. Retroviral vectors are extremely efficient gene delivery vehicles that cause no detectable harm as they enter the cells. The retroviral nucleic acid may integrate into host
- a vector encoding a packagable RNA vector genome
- ⁇ - operably linked to a promoter in which all the functional retroviral auxiliary genes are absent, is used to transfer the DNA encoding a component important in the formation of a FRMS of the present invention.
- examples of such vectors are described in WO 98/17815, WO 98/17816 and WO 98/17817, each of which is inco ⁇ orated herein by reference in their entirety.
- non-integrating viral vectors which infect and transduce non-dividing cells such as adenoviral vectors may be used to deliver a component important in the formation of a FRMS.
- Adenoviral vectors have several advantages since such vectors avoid risks associated with permanently altering the host cell genome or promoting insertional mutagenesis. Adenoviruses are one of the best developed
- Recombinant adenoviruses can be produced at very high titers is highly infectious and efficiently transfer genes to a wide variety of non-replicating and replicating cells and is further ideal for in vivo mammalian gene transfer.
- Adenovirus-based vectors are relatively safe and can be manipulated to
- n encode the desired component(s) and at the same time to be inactivated in terms of its ability to replicate in a normal lytic viral life cycle.
- Adenovirus has a natural tropism for airway epithelia. Therefore, adenovirus-based vectors are particularly preferred for epithelial delivery applications.
- the adenovirus-based gene therapy vector comprises an adenovirus 2 serotype genome in which the Ela and the Elb
- the adenovirus —based gene therapy vector contains only the essential open reading frame (ORF3 or ORF6 of adenoviral early region 4 (E4) and is deleted of all other E4 open reading frames, or may additionally contain deletions in the E3 regions (see e.g. U.S. Patent No. 5,670,488, inco ⁇ orated herein by reference in its entirety).
- the adenovirus-based therapy vector used may be a pseudo-adenovirus (PAV), which contain no harmful viral genes and a theoretical capacity for foreign material of nearly 36 kb.
- PAV pseudo-adenovirus
- adeno-associated virus (AAV) systems may be used to deliver the component important in the formation of a FRMS of the present invention.
- AAV has a wide host range and AAV vectors have currently have been designed which do not require helper virus. Examples of such AAV vectors are described in WO 97/17458.
- n Vaccinia viral vectors may be used in accordance with the present invention, as large fragments of DNA are easily cloned into its genome and recombinant attenuated vaccinia variants have been described (Meyer, et al., 1991, J. Gen. Virol. 72:1031-1038).
- a vaccinia viral vector is used to deliver the components important to the formation of an FRMS such that the components are expressed in the c recombinant vaccinia infected cell.
- the components are human CD4, a co-receptor (e.g., CCR5 or CXCR4), and the HIV envelope protein.
- infection of a cell with the recombinant vaccinia results in the expression of the components and formation of an HIV FRMS.
- two different recombinant vaccinia viruses n are constructed such that the first virus encodes the HIV env protein, and the second virus encodes human CD4 and a chemokine receptor.
- the first virus is used to infect a first population of cells such that the infection results in the expression of the HIV env protein.
- the second virus is used to infect a second population of cells such that the infection results in the expression of CD4 and the chemokine receptor.
- Co-culturing of the first infected population of cells with the second infected population of cells results in the formation of an HIV FRMS.
- the co-cultured cells may be cross-linked.
- Orthomyxoviruses including influenza
- Paramyxoviruses including respiratory syncytial virus and Sendai virus
- Rhabdoviruses may be engineered to
- Reverse genetic techniques can be applied to manipulate negative and positive strand RNA viral genomes to introduce mutations which result in attenuated phenotypes, as demonstrated in influenza virus, measles virus, Sindbis virus and poliovirus (see Palese et al., 1996, Proc. Natl. Acad. Sci. USA 93:11354-11358). These techniques may also be utilized to introduce foreign DNA, i.e., encoding a component or protein important in the formation of a FRMS to create recombinant viral vectors to be used in accordance with the present invention.
- attenuated adenoviruses and retroviruses may be engineered to express a component important in the formation of a FRMS. Therefore, a wide variety of viruses may be engineered to deliver a component important in the formation of a FRMS of the present invention.
- Bacteriophage may be used to specifically infects a bacterial cell (Soothill,
- the non- viral DNA e.g., encoding a cell receptor
- non-native viral DNA e.g., encoding a viral envelope protein
- receptor(s), and/or co-receptor(s) used to construct the FRMS of the subject invention can be provided to cells by virions, virus-infected cells or chemically/genetically inactivated virions, viral vectors, viral replicons (i.e. non-replicating viral constructs, e.g. VEE replicons), virus-like particles produced by recombinant DNA methods or as naked DNA.
- recombinant expression systems may employ regulatory
- 9 ⁇ elements including not limited to, inducible and non-inducible promoters, enhancers, operators. Any of the methods previously described for the insertion of DNA or nucleic acid fragments into a vector may be used to construct expression vectors containing a gene or chimeric gene consisting of appropriate transcriptional/translational control signals and the protein coding sequences. These methods may include in vitro recombinant DNA and
- expression of a nucleic acid sequence encoding a protein or peptide fragment may be regulated by a second nucleic acid sequence so that the protein or peptide is expressed in a host cell transformed with the recombinant DNA molecule.
- expression of a viral envelope protein may be controlled by any promoter/enhancer element known in the art.
- a promoter/enhancer may be homologous
- Promoters which may be used to control the expression of a protein include, but are not limited to, the SV40 early promoter region (Benoist et al., 1981, Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto et al., 1980, Cell 22:787-797), the he ⁇ es thymidine kinase promoter (Wagner et al., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:1441-
- promoter of the photosynthetic enzyme ribulose biphosphate carboxylase (Herrera-Estrella et al., 1984, Nature 310:115-120)
- promoter elements from yeast or other fungi such as the Gal4- responsive promoter, the ADC (alcohol dehydrogenase) promoter, PGK (phosphoglycerol
- elastase I gene control region which is active in pancreatic acinar cells (Swift et al., 1984, Cell 38:639-646; Ornitz et al., 1986, Cold Spring Harbor Symp. Quant. Biol.
- mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., 1986, Cell 45:485-495), albumin gene control region which is active in liver (Pinkert et al, 1987, Genes and Devel. 1 :268-276), alpha-fetoprotein gene control region which is active in liver (Krumlauf et al., 1985, Mol. Cell. Biol. 5:1639- 1648; Hammer et al., 1987, Science 235:53-58), alpha 1-antitrypsin gene control region which is active in the liver (Kelsey et al., 1987, Genes and Devel.
- beta-globin gene control region which is active in erythroid cells (Mogram et al., 1985, Nature 315:338- n 340; Kollias et al., 1986, Cell 46:89-94), myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-712); myosin light chain-2 gene control region which is active in skeletal muscle (Sani, 1985, Nature 314:283-286), and gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason et al., 1986, Science 234:1372-1378).
- a vector is used that comprises a promoter 5 operably linked to a nucleic acid encoding a viral or cellular protein or chimeric protein and one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic resistance gene).
- a promoter 5 operably linked to a nucleic acid encoding a viral or cellular protein or chimeric protein and one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic resistance gene).
- two or more promoters may be used to direct expression of two or more genes within a single plasmid.
- the CMV immediate early (IE) promoter is used to direct expression of a nucleic acid encoding a viral or cellula ⁇ rotein or chimeric protein.
- Promoters can be inducible, or constitutive. Expression from certain promoters can be elevated in the presence of certain inducers; thus, for example, expression of the genetically engineered protein chimeras may be controlled. Inducible promoters may 5 be used to control expression of the proteins of the invention, such that the protein is produced only in the presence of the inducer.
- the present invention also provides methods for stable expression of the components which result in the formation of an FRMS of the invention. For long term, high-yield production of recombinant proteins, stable expression is possible. For example, cell lines which stably express one or more cellular receptors and co-receptors for the virus, may be engineered.
- host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter sequences, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
- expression control elements e.g., promoter sequences, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
- engineered cells may be allowed to grow for 1-2 days in ⁇ an enriched media, and then are switched to a selective media.
- the selectable marker in the recombinant plasmid confers resistance to the selection foci (e.g., by stably integrating the plasmid into their chromosomes) and allows cells to and grow to form which in turn can be cloned and expanded into cell lines.
- This method may advantageously be used to engineer cell lines.
- This method may advantageously be used to engineer cell lines which express the selected gene products. Such cell lines would be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the selected gene product.
- a number of selection systems may be used in generating stably-expressing cell lines, including but not limited to the he ⁇ es simplex virus thymidine kinase (Wigler, et al., 1977, Cell 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska et al., n 1962, Proc. Natl. Acad. Sci. USA 48:2026), and adenine phosphoribosyltransferase (Lowy, et al., 1980, Cell 22:817) genes can be employed in tk " , hgprt " or aprt " cells, respectively.
- antimetabolite resistance can be used as the basis of selection for the following genes: DHFR, which confers resistance to methotrexate (Wigler, et al., 1980, Natl. Acad. Sci. USA 77:3567; O ⁇ are, et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan et al., 1981, Proc. Natl. Acad. Sci. USA 5 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin, et al., 1981, J. Mol. Biol. 150:1); and hygro, which confers resistance to hygromycin (Santerre, et al., 1984, Gene 30:147).
- methotrexate Wigler, et al., 1980, Natl. Acad. Sci. USA 77:35
- the present invention encompasses the expression of component(s) whose association results in the formation of the FRMS.
- expression may be in primary cells, animal cells, and insect cell lines.
- primary or secondary cells or cell strains may be used including but not limited to cells isolated from skin, bone marrow, liver, spleen, pancreas, kidney, adrenal and neurological tissue to name a few.
- Other cells types that may be used in accordance with the present invention are immune cells (such as T-cells, B-cells, natural killer cells, etc.), macrophages/monocytes, adipocytes, pericytes, fibroblasts, neuronal cells, reticular cells etc.
- secondary cell lines may be used, including, but not limited to hepatic cell lines, such as CWSV, NR, Chang liver cells, or other cell lines such as CHO, VERO, BHK, Hela, COS, MDCK, 293, 373, CaSki and W138 cell lines.
- host cells comprise one or more cell receptors that facilitate binding or viral infection.
- the cell is eukaryotic, preferably a cell line, preferably mammalian, and most preferably human may be used for the method
- Cells may be derived from human (e.g., HeLa cells), primate, mouse, rabbit, chicken, etc., although may also be from a transgenic non-human animal.
- Numerous eukaryotic cell lines may be purchased from ATCC (American Type Culture Collection, Rockville, MD).
- ATCC American Type Culture Collection, Rockville, MD
- eukaryotic cell is derived from a human.
- the cell is derived from a mouse, monkey, or rat.
- non-tumor cell lines and autologous permissive cells are used in preparing the FRMS of the subject invention.
- the present invention provides cells comprising one or more components which result in the formation of an FRMS of the invention.
- the present invention also provides cells comprising all of the components required for the formation of
- the present invention provides a cell that recombinantly expresses an envelope protein of an enveloped virus that functions in fusion of the viral envelope with a host cell membrane, or a mutant form of said envelope protein that is fusion-defective.
- the cell expresses one or more cellular membrane proteins that function as receptors for said envelope protein.
- the invention provides a cell that recombinantly expresses HIV gpl60, human CD4 and a co-receptor for HIV.
- the present invention also provides a cell line that recombinantly expresses an envelope protein of an enveloped virus that functions in fusion of the viral envelope with a host cell membrane, or a mutant form of said envelope protein that is fusion-defective.
- the cell line expresses one or more cellular membrane proteins that function as receptors for said envelope protein.
- the invention provides a cell line that recombinantly expresses HIV gpl60, which cell line expresses CD4 and a co-receptor for HIV; said cell line lacking a functional protease that cleaves gpl60 to produce gpl20 and gp41.
- cells comprising the FRMS of the invention are administered as an immunogen(s) to a subject (see Section 5.5, herein).
- the cells are human primary cells.
- the cells are autologous.
- any viral envelope protein containing wild type fusion activity is used to make the FRMS.
- a fusion-defective envelope protein is used, as described in this section.
- the present invention provides for the use of mutations to viral envelope proteins or host cell receptor or co-receptor proteins which mutations inhibit the completion of process of viral envelope and cell membrane fusion.
- the invention provides fusion- defective mutations which do not allow completion of the process of viral envelope-cell membrane fusion, but which do result in the formation of a fusion-related molecular structure of the invention.
- the fusion-defective mutations are used to construct epitopes comprising an FRMS.
- association of the fusion defective envelope protein with the cellular receptor(s) results in the accumulation of the FRMS.
- the fusion-defective mutations of the invention may be used in the formation of an FRMS.
- Any technique for mutagenesis known in the art can be used to modify individual nucleotides in a DNA sequence, for pu ⁇ ose of making amino acid substitution(s) in the expressed peptide sequence, for creating/deleting restriction sites, or for adding affinity tags.
- Such techniques include but are not limited to, chemical mutagenesis, in vitro site-directed mutagenesis (Hutchinson, C, et al., 1978, J. Biol. Chem 253:6551), oligonucleotide-directed mutagenesis (Smith, 1985, Ann. Rev. Genet. 19:423-463; Hill et al., 1987, Methods Enzymol. 155:558-568), PCR-based overlap extension (Ho et al., 1989,
- a fusion-defective mutation may be constructed for any enveloped virus.
- mutations are constructed that affect the ability to complete fusion.
- Mutations known not to detrimentally affect envelope protein expression and transit to the cell surface will be preferred.
- a fusion-defective mutant is confirmed by expressing the mutated gene in a cell (e.g., the methods of the invention) such as COS,
- mutants may then be assayed for cell surface expression of the mutant component (by methods known in the art such as, e.g., by flow cytometry or by live cell indirect immunofluorescence). Additionally, proteolytic processing of a mutant protein such as a mutant of HIV gpl60 may be assayed by Western blot analysis or any other methods known in the art. Fusion competency of mutant components may be assayed as described in Section 5.7, herein, (e.g., by syncytium formation assay, e.g., in the case of HIV, U87-CD4-co-receptor cell fusion assay).
- binding-defective, fusion-defective or fusion-arrested mutant envelope proteins are tested for the ability to elicit primary isolate virus neutralizing antibodies in the transgenic mouse vaccination assay.
- immunogens may comprise cells expressing the test mutant envelope protein cocultured with e.g. U87-CD4-CCR5 cells (when testing HIV related mutants).
- mutations that elicit broad PI virus neutralization are developed as subunit and recombinant virus-based immunogens.
- Table I provides a list of exemplary mutations which are provided for HIV. As will be apparent to one skilled in the art, similar mutations can be constructed for other enveloped viruses.
- the bridging sheet of the gpl20 core structure includes those of Kwong et al. (Kwong PD, et al., 1998, Nature 393:648-59; Kwong PD, et al., 1999, J Biol Chem. 274:4115-23) and regions of putative gp41/gpl20 interaction.
- the FRMS of the subject invention are prepared using viral envelope proteins containing binding and fusion-related mutations.
- the viral proteins bind host cell receptors, however, mutation causes the fusion process to be arrested before or during fusion of the viral envelope and host cell membrane.
- the FRMS of the subject invention include epitopes resulting from the association of one or more viral envelope proteins and one or more cell membrane proteins, including those proteins which are fusion-defective, fusion-arrested or binding-defective so long as the mutant proteins when used in the methods of the invention result in an epitope elicits PI neutralizing antibodies.
- Protein complexes arrested during the process of fusion can be isolated and used in the methods and compositions of the invention including but not limited to immunogens, diagnostics, kits, vaccines and compositions.
- the FRMS comprise the major viral envelope protein of HIV-1.
- HIV-1 the major viral envelope protein of HIV-1.
- fusion-related mutations which may be used in constructing the complexes of the subject invention.
- the oligomeric envelope protein complex gpl60 comprising the gpl20 surface and gp41 transmembrane portions, binds initially to
- the CD4 receptor on the host cell Conformational changes in both the envelope protein and CD4 receptor facilitate interaction with a co-receptor molecule on the host cell surface. Further conformational changes in the viral protein-CD4-co-receptor trimolecular complex allow exposure of a viral gp41 pre-hai ⁇ in intermediate and the insertion of the N-terminal gp41 hydrophobic fusion domain into the host cell membrane. Helical heptad-repeat regions within the pre-hai ⁇ in intermediate subsequently collapse to form the trimeric, coiled-coil core of fusion active gp41. The coiled-coil core drives the ultimate fusion of the apposed cell and virus membranes.
- Viral envelope proteins with mutations that abrogate proteolytic cleavage of the g ⁇ l60 precursor protein preventing liberation of the gp41 hydrophobic fusion domain n can be used to construct the subject complexes.
- certain mutations that alter the highly conserved lys/arg-X-lys/arg-arg site at the C-terminus of gpl20 abrogates fusogenicity (see e.g., Lee CN, et al., 1994, AIDS Res. Hum. Retroviruses.10: 1065-9).
- mutations that affect the N-terminal gp41 fusion peptide can be
- the hydrophobic N-terminal region of gp41 mediates fusion by inserting into the cell membrane and destabilizing the lipid bilayer, perhaps by adopting a helical structure. Certain amino acid changes within this region render the envelope protein nonfusogenic. Synthetic peptides bearing these changes are either unable to bind to lipid bilayers unable to elicit fusion.
- n fusion-defective mutations in g ⁇ 41 have been well characterized: V2E and GlOV (Kliger Y, et al., 1997, J Biol Chem.272: 13496-505).
- the former involves a polar substitution in the hydrophobic peptide, whereas the later may affect the helical structure assumed in the lipid bilayer.
- These mutations can be introduced into the 168P envelope gene (gp41: ala-yal-gly-ile-gly-val-leu-phe-leu-gl -phe-leu-gly]. by site-directed mutagenesis or any method known in the art and the envelope proteins tested for expression and fusogenicity.
- envelope proteins having mutations that alter the coiled-coil core region of gp41 can be used to construct the structures of the subject invention.
- the i highly-conserved coiled-coil motif is found in proteins involved in fusion of many virus families, and similar structures have been identified in cellular proteins that mediate vesicular fusion.
- Synthetic peptides that mimic either the N- or C-helical region e.g. DP107 and DP178
- potently neutralize virus infectivity presumably by binding the cognate helical region and interfering with the collapses of the pre-hai ⁇ in intermediate.
- N- and C-helical regions of gp41 have also been saturated by mutagenesis.
- Several mutations have been identified that retain normal envelope protein expression and assembly yet are non-fusogenic, presumably though disruption of the interface between N- and C-helical coils (e.g. V570R and Y586E) (Weng et al., 1998, Journal of Virology 72:9676-9682).
- Competent or mutated viral envelope protein as well as cell receptors and/or co-receptors can be inco ⁇ orated into viral vectors or diploid cells by methods known in the art including those described herein.
- Viral genes encoding viral proteins are routinely cloned for expression in bacterial cells, in eucaryotic or procaryotic cells, or in viral or DNA vectors.
- human cellular receptors can be cloned and expressed to produce models by which to study dangerous pathogenic diseases.
- Viral genes or host receptors can be inco ⁇ orated into foreign DNA by transfection or through the use of viral vectors.
- FRMS of the subject invention can be formed as a result of the interaction of viral proteins and a variety of animal cellular components including but not limited by those listed above.
- FRMS FRMS
- methods may be used to preserve or "capture" the FRMS in an immunogenic form that provides vaccine efficacy for primary viral isolates.
- the FRMS of the subject invention result from interaction of one or more viral envelope proteins with one or more host cellular receptors and/or co-receptors.
- complexes are "captured" by fixing or cross- linking the FRMS.
- a FRMS formed in co-cultured cells expressing the viral i and cellular components respectively, which result in a FRMA may be captured by fixation of the cells with a cross-linking agent.
- cells transformed with a nucleic acid expressing a viral envelope protein and cells transformed with nucleic acids expressing host cellular receptors and co-receptors are cultured together.
- the viral protein expressed on the surface of a first cell binds to the receptor(s) expressed on the surface of a second cell.
- the cells are fixed at the initiation of this cell-cell interaction.
- crosslinking "captures" the intermediate fusion complex and provides fusion-related determinants which may be isolated and use as, for example, vaccine components. Any
- fusing cells or fusion-defective mutant cells can be fixed or cross-linked by any method known in the art.
- Cross-linking reagents that can be used include but are not limited to formaldehyde glutaraldehyde, formalin, p-Azidobenzoyl
- the cross-linking reagent is formaldehyde.
- low levels of formaldehyde (0.2%) are used in order to conserve antigenicity. In other embodiments, higher concentrations however can be used to optimize stability.
- Formaldehyde in concentrations of from about 0.01% to about 8% can be used in preparing the complexes of the subject invention.
- - that can be used to cross-link fusing cells include but are not limited to glutaraldehyde (0.05-0.5%).
- fixing or cross-linking conditions i.e., reagent used, concentration of reagents, time and temperature
- concentration of reagents, time and temperature can be varied to determine optimal conditions for immunogenicity of the FRMS.
- the complexes of the subject invention can be prepared by infecting human diploid cells in cell culture with, for example, recombinant vaccinia virus expressing viral envelope proteins and CD4/co-receptor (if not endogenously present). During fusion, the cells are cross-linked to capture the fusion-related structures and determinants.
- the cells undergoing infection may be cross-linked in order to capture the FRMS.
- a single cell may be engineered to express all of the components important for the formation of a FRMS.
- such a cells or cells may be cross-linked or fixed in order to capture the FRMS.
- the invention provides viral particles which are engineered to express in the viral envelope the cellular components or proteins important for FRMS formation.
- the recombinant viral particles may be cross-linked to capture the FRMS formed withing the viral particle envelope.
- the complexes need not be fixed or cross-linked. It has been found that the complexes of the subject invention form in cell lysates, in solution. Therefore, creation of the subject complexes by, for example, the simple lysis of cells transfected or infected with HIV-1 envelope protein as well as CD4/co- receptor where the FRMS form in solution.
- Cell lysis protocols are well known in the art (see, e.g., Sambrook et al., supra). Cell lysis protocols may involve detergent lysis (e.g., 1% NP40) freeze thaw lysis, sonication lysis, or any method known in the art.
- cell cultures are fixed at the onset of cell-cell interactions by fixation in ice cold 0.2% formaldehyde in phosphate buffered saline.
- detergent lysis of cells involves the use of the detergent Brij 97.
- the invention provides a molecular structure which is a cross- linked cellular molecular structure or is isolated from a cell lysate.
- a molecular structure which is a cross- linked cellular molecular structure or is isolated from a cell lysate.
- fusion-defective mutations which allow accumulation of the FRMS and allows capturing of the FRMS.
- cells e.g. cross-linked cells
- FRMS binds to antibodies
- the FRMS may be isolated and purified by standard methods including chromatography (e.g., ion exchange, affinity, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
- chromatography e.g., ion exchange, affinity, and sizing column chromatography
- centrifugation e.g., centrifugation
- differential solubility e.g., differential solubility
- the components important in the formation of a FRMS can be synthesized comprising an affinity tag which facilitates recovery and purification.
- the peptide tag can be associated with any portion of the protein, so long as such association does not alter the epitope formation generated by association of the modified component with other members of the complex.
- such a chimeric protein comprising an affinity tag can be made by ligating a gene sequence coding for the component to the sequence encoding the peptide tag in the proper reading frame and recombinantly expressing the protein. Care should be taken to ensure that the modified gene remains within the same translational reading frame, uninterrupted by translational
- peptide tags known in the art may be used in the modification of a protein, such as but not limited to the polyhistidine sequence (Petty, 1996, Metal-chelate affinity chromatography, in Current Protocols in Molecular Biology, Vol. 2, Ed. Ausubel et al., Greene Publish. Assoc. & Wiley Interscience), glutathione S-transferase (GST; Smith, 1993, Methods Mol. Cell Bio. 4:220-229), the E. coli maltose binding protein (Guan et al., 1987, Gene 67:21-30), and various cellulose binding domains (U.S. Patent Nos.
- peptide tags are short amino acid sequences to which monoclonal antibodies are available, such as but not limited to the following well known f) examples, the FLAG epitope, the myc epitope at amino acids 408-439, the influenza virus hemagglutinin (HA) epitope.
- Other peptide tags are recognized by specific binding partners and thus facilitate isolation by affinity binding to the binding partner, which is preferably immobilized and/or on a solid phase surface.
- peptide r tags many methods can be used to obtain the coding region of the above-mentioned peptide r tags, including but not limited to, DNA cloning, DNA amplification, and synthetic methods. Some of the peptide tags and reagents for their detection and isolation are available commercially.
- DNA sequences encoding desired peptide tags which are known or readily available from libraries or commercial suppliers are suitable in the practice of this
- peptide tags are recognized by specific binding partners and thus facilitate isolation by affinity binding to the binding partner which can be immobilized onto a solid phase surface.
- the chimeric protein gene product can be prepared using recombinant DNA techniques. For example, gene sequence encoding a component important in the formation of a FRMS can be introduced into a vector containing the sequence of a peptide tag, such that the component gene is expressed as a peptide-tagged chimeric protein.
- Peptide tags which may be recognized by specific binding partners, may be used for affinity binding to the binding partner immobilized on a solid phase surface.
- a poly-histidine tagged fusion-related protein is constructed by insertion of a fusion-related protein gene or gene fragment into an expression vector such as one of the pET15 series of vectors (Novagen), that express inserted sequences as chimeric proteins with N-terminal poly-histidine tags. Proteins which have a succession of six or more histidine residues at their amino or carboxyl terminus have a strong binding affinity to nickel. Poly-histidine-tagged chimeric proteins will bind specifically to the surface of a solid phase coated with chelated nickel. In a specific preferred embodiment, microtiter plates coated with metal chelates are used to isolate the poly-histidine-tagged chimeric proteins (Pierce).
- a system described by Janknecht, et al. allows for the ready purification of non-denatured chimeric proteins expressed in human cell lines (Janknecht, et al., 1991, Proc. Natl. Acad. Sci. USA 88, 8972-8976).
- the gene of interest is subcloned into a vaccinia recombination plasmid such that the gene's open reading frame is translationally fused to an amino-terminal tag consisting of six
- Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni 2+ nitriloacetic acid-agarose columns and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
- an expression construct can be made by subcloning a fusion-related protein into the EcoRI restriction site of each of the three pGEX n vectors (glutathione S-transferase expression vectors; Smith et al., 1988, Gene 7:31-40). This allows for the expression of a chimeric protein comprising the fusion-related protein linked to the GST binding domain in all three reading frames, such that, in one frame, the GST binding activity is maintained in the resulting chimeric protein.
- a GST chimeric peptide has a strong binding affinity for its substrate, glutathione.
- FRMS comprising a GST tag is separated from a binding mixture comprising a cellular lysate or fixed cells by contacting such mixture with a glutathione-linked solid phase surface, such as glutathione sepharose beads.
- a glutathione-linked solid phase surface such as glutathione sepharose beads.
- the GST-chimeric protein can be anchored to glutathione-agarose beads or a glutathione-sepharose column. The mixture is then added to the column or beads in a manner that allows interaction and
- a chimeric protein may be readily purified by utilizing an antibody specific for the chimeric protein being expressed.
- an affinity-tagged FRMS can be constructed by conjugation of an affinity compound to the fusion-related protein.
- Affinity compounds can be used, such as, but not limited to, biotin, photobiotin, or other compounds known in the art.
- affinity compounds or affinity tags can be conjugated to the fusion-related protein through a polyfunctional crosslinker, and preferably a bifunctional molecule.
- polyfunctional crosslinker encompasses molecules having more than one functional group that reacts with a functional group on the fusion-
- crosslinker forms covalent bonds with an amino or sulfhydryl group on a polypeptide.
- biotin N-hydroxysuccinimide esters may be used.
- This STAGTM system was developed commercially by Novagen, Inc. for use in the detection and isolation of recombinant chimeric proteins expressed in bacterial and baculovirus systems, and has recently been extended to mammalian chimeric proteins.
- the fusion-related proteins of the FRMS is tagged with S-peptides.
- the invention provides examples of components of complexes which were tagged with the S-peptide at the C-terminal ends of the CD4, CCR5 or envelope protein molecules. Tagged complexes were readily isolated and purified by S-protein
- membrane-impermeable cross-linking reagents can be used to fix cells containing tagged complexes.
- fixing cells containing tagged complexes with a membrane impermeable cross-linking agent facilitates avoiding inactivation of the cytoplasmic S-peptide tag.
- _ - sequence may be altered (ala-glu-thr-ala-ala-ala-ala- phe-glu-arg-gln-his-met-asp-ser) so that it no longer contains vulnerable lysine residues and thus can be fixed with formaldehyde without resulting in inactivation of the cytoplasmic tag.
- Fusing cells can be fixed with a wide range of membrane impermeable cross-linking reagents including, but not limited to BS 3 and DTSSP (homobifunctional N-hydroxysuccinimidyl (NHS) esters that
- _ n react through amines to form either non-cleavable or reversible linkages, respectively); Sulfo-SMPB (heterobifunctional NHS ester and maleimide to irreversibly cross-link amine and sulfhydryl groups); Sulfo-SANPAH and SASD (heterobifunctional NHS ester and photoreactive phenylazide cross-linkers which form either non-cleavable or reversible linkages, respectively) (Pierce Chemical Company).
- Sulfo-SMPB heterofunctional NHS ester and maleimide to irreversibly cross-link amine and sulfhydryl groups
- Sulfo-SANPAH and SASD heterofunctional NHS ester and photoreactive phenylazide cross-linkers which form either non-cleavable or reversible linkages, respectively
- Tagged complexes are readily purified providing an isolated preparation of the fusion-related components which may be used for example, for inclusion in vaccine formulations. Purified or isolated tagged complexes can also be used as diagnostic standards for in vitro assays. Further, tagged complexes permit the functional analysis of envelope-mediated fusion.
- the invention provides a method of purifying a protein complex comprising an envelope protein of human immunodeficiency virus type 1 functionally interacting with human CD4 and human CCR5 including the steps of: a) tagging the complex with a peptide sequence to facilitate subsequent purification; and b) isolating the tagged complex.
- the invention provides an isolated protein complex comprising an envelope protein of human immunodeficiency virus type 1 functionally interacting with human CD4 and human CCR5.
- TYPES OF FRMS As will be apparent to one skilled in the art, any enveloped virus may be used in the methods of the invention in order to construct or use the FRMS. For example, an important use of the FRMS of the invention is as vaccine immunogens.
- the enveloped viruses that can be used include but are not limited to the families of Retro viridae, Rhabdoviridae, Coronaviridae, Filoviridae, Arenaviridae, Poxviridae, Bunyaviridae, Flaviviridae, Togaviridae, Orthomyxoviridae, ⁇ Paramyxoviridae, He ⁇ esviridae, and Iridoviridae.
- a retroviral FRMS is formed by the methods of the invention.
- the FRMS of the invention comprises the major viral envelope immunogen of human immunodeficiency virus type 1 (HIV-1) and arises from interaction with the host cellular receptor for HIV-1
- HIV-1 human immunodeficiency virus type 1
- the host cellular co-receptor can be CCR5, CXCR4, CCR3, CCR2b or any other known in the art.
- An exemplified FRMS of the present invention results from interaction of the HIV-1 isolate 168P viral envelope protein arising from interaction with the host cellular receptor CD4, and the host cellular co-receptor CCR5. It should be apparent to those skilled in the art that other recombinant HIV envelope proteins,
- _ n including those that act independently of CD4, could be used to prepare FRMS within the scope of the subject invention.
- the immunogens of the subject invention are prepared by co-culturing envelope-protein expressing COS-7 cells (LaCasse et al. 1998, Science, 283:357-360) and
- the envelope-expressing cells are harvested using 0.5 mM EDTA 24 hr after transfection (30-60% transfection efficiency) and co-cultured with an equal number of I
- U87-CD4-CCR5 target cells progress towards fusion is assessed by staining envelope-expressing cells and monitoring their inco ⁇ oration into multinucleated syncytia microscopically. Fusion is complete within 12-24 hr. Complexes are formed immediately. Cells fixed after 1-5 hrs of co-culture are at an estimated 10-30% of maximal syncytium - formation. This early time point in fusion was chosen initially in order to capture the transitional intermediates that lead to fusion. Other time points (e.g. 10 hr, 24 hr or greater) can also be used to obtain the fusion-related complexes of the subject invention.
- Flavivirus FRMS is generated by the methods of the invention.
- Viruses within the Flavivirus family include, for example, Hepatitis C Virus (HCV), Dengue virus, Yellow Fever virus, Tick-borne Encephalitis virus, and Bovine Viral Diarrhea virus.
- HCV Hepatitis C Virus
- the Flaviviruses are similar to the Togaviridae which include the alphaviruses (e.g. , Venezuelan Equine Encephalitis virus, Sindbis virus, Semliki Forest ⁇ virus) and Rubella virus.
- the Flaviviruses enter target cells via receptor-mediated endocytosis followed by pH-induced fusion within the endosome.
- Viruses within the Flavivirus family encode an envelope (E) protein which is proteolytically cleaved in many family members to mature El and E2 proteins.
- E envelope
- the E2 protein is the receptor binding moiety arriving (binding the cellular membrane protein CD81) and El is the putative viral fusion protein.
- a Flavivirus or Togavirus FRMS is formed by the methods of the invention.
- an HCV FRMS of the invention results from the association of El, E2 and CD81.
- the HCV El and E2 proteins can be co-expressed in an acceptable cell substrate and will become - localized in the cell endoplasmic reticulum (ER) and not significantly on the cell surface.
- these cell cultures are incubated in medium buffered to pH 5 to 6.8 in order to trigger E1E2 fusion of the intracellular membrane.
- the cultures are treated with a cross-linking agent to capture the intermediate fusion-competent structures, and used directly (or upon isolation of intracellular ER membranes) as vaccine immunogen.
- the disrupted intracellular ER membranes containing E1E2 protein are isolated as everted micelles and then incubated with an appropriate permissive cell expressing the E2 receptor, CD81.
- ElE2-containing micelles are internalized and El -mediated fusion occurs in the endosome. Cells are then cross-linked and used for example, as vaccine immunogens. Alternatively, E1E2 proteins are engineered - to alter ER retention signals and thus allow expression of El E2 proteins on the cell surface thus obviating the need to isolate ER membranes. i
- recombinant DNA derived HCV virus-like particles or HCV E1E2 containing pseudotyped virions can be produced and triggered for fusion by incubation with receptor-expressing cells in medium adjusted to pH 5-6.8.
- the cultures are treated with a cross-linking agent such as c - formaldehyde and the FRMS is used as a vaccine immunogen.
- virions can be incubated with CD81 -expressing cells; internalized virions will be induced to fuse within the endosome and can be captured by cross-linking treatment for use as a vaccine immunogen of the invention.
- an orthomyxo virus FRMS is
- influenza FRMS elicits neutralizing antibodies against a wide variety of flu strains and would allow for a drift-independent flu vaccine.
- the Orthomyxoviruses enter target
- NA viral neuraminidase
- HA hemagglutinin
- influenza FRMS is generated by the methods of the invention.
- Influenza FRMS is generated by the methods similar to those described above for the Flaviviruses and include pH-triggered HA-mediated fusion between appropriate cells or between cells and isolated cell membranes or virion particles, capturing the complex may ⁇ - be performed, for example, with a cross-linking agent allowing NA-mediated endocytosis and subsequent endosomal fusion to be captured.
- a paramyxovirus FRMS is generated by the methods of the invention.
- the Paramyxovirus family encompasses several subgroups of significant medical and veterinary importance, including but not limited to,
- Morbilliviruses (measles virus, Canine Distemper virus, Rinde ⁇ est virus), Rubulaviruses (Mumps virus, Newcastle Disease), paramyxoviruses (human and bovine parainfluenza), and Pneumoviruses (Respiratory Syncytium virus). These viruses are similar to the Orthomyxoviruses, except that Paramyxovirus fusion occurs at the cell surface rather than in the endosome, and is pH independent. For the Paramyxoviruses, the neuraminidase-
- HN ⁇ ⁇ - hemagglutinin (HN) (or simply hemagglutinin (H)) protein mediates cell attachment whereas the fusion (F) protein mediates virus-cell or cell-cell fusion.
- F virus-cell or cell-cell fusion.
- the H protein binds the cellular CD46 receptor and the F protein mediates membrane fusion.
- cells expressing H and F proteins from MV are co-cultured with cells expressing CD46 (and currently unidentified co- receptors) and result in cell-cell fusion that can be captured by cross-linking or other methods described herein.
- the co-receptor may be
- a he ⁇ esvirus FRMS is generated by the methods of the invention.
- the He ⁇ esviruses comprise a large family that include members of significant medical and veterinary importance.
- the human he ⁇ esviruses include, but are
- HSV 1 and HSV 2 Varicella-zoster virus, Epstein-Barr virus, Cytomegalovirus, and Kaposi's He ⁇ esvirus HHV 8.
- binding and entry is a complex process involving several viral glycoproteins (gD, gC, and gH/gL) and several cell receptors and adhesion molecules (glycosaminoglycans and He ⁇ esvirus entry mediator (Hve) proteins
- gD viral glycoproteins
- gC cell receptors and adhesion molecules
- Hve He ⁇ esvirus entry mediator
- Binding and fusion occur on the cell surface in a pH-independent manner.
- HSV FRMS is constructed using a cell expressing the cellular receptors and adhesion molecules (e.g. human MRC5 cells) and co-culturing such cells with cells expressing the HSV glycoproteins gD, gC, gH, and gL.
- Cell-cell fusion is preferably arrested by fixation with a cross-linking agent, to allow capturing of the 0 FRMS -
- viral proteins and host cellular receptors can be combined to form the subject FRMS for viruses including He ⁇ es virus, pox viruses, paramyxovirus, measles, mumps, rubella, respiratory syncytial virus, influenza, Hepatitis C, ebola and flaviviruses such as Dengue and Yellow Fever.
- viruses including He ⁇ es virus, pox viruses, paramyxovirus, measles, mumps, rubella, respiratory syncytial virus, influenza, Hepatitis C, ebola and flaviviruses such as Dengue and Yellow Fever.
- the FRMS of the - subject invention can include viral proteins from viruses of veterinary importance including rabies, feline leukemia, feline immunodeficiency virus and rinde ⁇ est.
- Table II presents an exemplary list of the cellular receptor(s) for particular enveloped viruses, which when expressed along with the viral envelope protein and allowed to associate with the envelope protein results in FRMS formation.
- Table III presents an exemplary list of envelope proteins for particular families of enveloped viruses, which when expressed along with the cellular receptor protein(s) and allowed to associate with the cellular receptor protein(s) results in FRMS formation.
- a triple-stranded coiled-coil structure of Orthomyxoviridae (such as Influenza), Filoviridae (such as Ebola), or Retroviridae (such as HIV) facilitates the formation of FRMS.
- Orthomyxoviridae such as Influenza
- Filoviridae such as Ebola
- Retroviridae such as HIV
- synthetic peptides may be used to inhibit viral infection.
- synthetic peptides that comprise either of the gp41 helical coils are able to bind the cognate helical region and broadly inhibit viral infectivity (see e.g., Wild, C, et al., 1992, Proceedings of the National Academy of Sciences USA 89:10537; Furuta, R.A., et al., 1998, Nature Structural Biology 5:276).
- neutralizing antibodies to FRMS immunogens may likewise target structures involved in the activation of fusion.
- viral strains may be used which do not require a host cell receptor, but require only a host cell co-receptor for association with the virus.
- an HIV strain which does not require CD4 for viral binding may be used in the methods of the invention (see e.g., Hoxie, et al., 1998, J. Reprod. Immunol. 41:197-211.). Therefore, when using such a strain, expression of CD4 is not necessary.
- the subject invention also concerns methods for vaccinating an individual or animal to raise an immune response to and prevent infection by a virus.
- the FRMS of the subject invention can be presented to the vaccinee by several means.
- the FRMS described above can be administered to a vaccinee as fixed cells combined with an adjuvant. Further, isolated and purified FRMS can be administered to a vaccinee.
- FRMS can be prepared in situ by the simultaneous immunization of a vaccinee with transfected cells expressing viral protein and transfected cells expressing host cellular receptors and or co-receptors.
- vectors containing DNA encoding viral proteins and vectors containing DNA encoding receptor proteins can be used in this immunization strategy.
- Vectors can include viral vectors such as vaccinia and vectors used in DNA immunization. After simultaneous immunization, interaction between the viral protein and host cellular receptor protein occurs in vivo forming the FRMS of the subject invention and thereby exposing the vaccinee to the unique epitopes capable of eliciting neutralizing antibodies.
- Another method by which the FRMS can be formed in situ includes utilizing the receptors and co-receptors on host cells.
- transfected cells expressing the HIV glycoprotein or vectors containing DNA encoding the protein are injected into or targeted (e.g. by VEE or other vectors) the lymph node of a host.
- the viral protein binds and initiates fusion in vivo to host cells having viral receptors and co-receptors to form the FRMS whereby the vaccinee is exposed to the newly formed neutralizing epitopes.
- a vaccine formulation comprises an isolated protein complex comprising an envelope protein of human immunodeficiency virus type 1 functionally interacting with human CD4 and human CCR5, and a pharmaceutically acceptable carrier.
- the invention provides a method of immunizing an animal to a virus comprising the steps of administering to the animal a vaccine formulation comprising a protein complex comprising one or more viral proteins functionally interacting with one or more host cellular receptors or co-receptors to mediate viral binding, entry and/or infection; whereby neutralizing antibodies to the virus is generated.
- the invention provides a vaccine formulation comprising (a) a first nucleic acid encoding an envelope protein of an enveloped virus; and (b) a second nucleic acid encoding one or more cellular membrane proteins, which envelope protein and cellular membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins, such that the envelope protein and cellular membrane proteins are expressed in the subject and neutralizing antibodies to the virus are produced; and (c) a pharmaceutically acceptable carrier.
- the invention provides a method of treating a host that has been exposed to a virus, or preventing infection of a host by said virus, the method comprising the steps of administering to the host antibodies generated by immunizing an animal with an isolated protein complex comprising an envelope protein of human immunodeficiency virus type 1 functionally interacting with human CD4 and human CCR5. in an amount effective to treat or prevent infection of said host.
- the invention provides a method of preparing a protein complex comprising one or more viral proteins functionally interacting with one or more host cellular receptors or co-receptors to mediate viral binding, entry and/or infection, including the steps of: a) culturing a first cell expressing one or more viral proteins; b) culturing a second cell expressing one or more host cellular receptors or co-receptors for said one or more viral proteins' c) co-culturing the first and second cells; d) fixing said co- culture during cell-cell fusion; and e) isolating the fixed cells.
- vaccine formulations of the invention include but are not limited to oral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal routes, and via scarification (scratching through the top layers of skin, e.g., using a bifurcated needle).
- the patient or subject to which the vaccine is administered is preferably a mammal, most preferably a human, but can also be a non-human animal including but not limited to cows, horses, sheep, pigs, fowl (e.g., chickens), goats, cats, dogs, hamsters, mice and rats.
- cows horses, sheep, pigs, fowl (e.g., chickens), goats, cats, dogs, hamsters, mice and rats.
- the patient or subject to which the vaccine is administered can also be a non-
- the invention provides a means of wildlife management, and provides a method of preventing or treating a wild animal or animal population which has a high degree of inbreeding such that said populations are highly susceptible to viral disease.
- the subject is a human.
- said human has a high risk of HIV infection.
- the subject is a domestic animal.
- the present invention thus provides a method of immunizing an animal, or treating or preventing viral diseases or disorders in an animal, comprising administering to n the animal an effective immunizing dose of a vaccine of the present invention.
- the vaccine formulations of the present invention can also be used to produce antibodies for use in passive immunotherapy, in which short-term protection of a host is achieved by the administration of pre-formed antibody directed against a heterologous organism.
- Vaccination of mice with immunogens comprising FRMS of the subject invention elicits the production of neutralizing antibodies by immunized animals.
- the complexes are administered as a vaccine comprising fixed whole cells formulated with an adjuvant. It would be apparent to one skilled in the art however that other vaccine strategies can be used in practicing the subject invention.
- isolated and purified FRMS and/or epitopes thereof can be administered as subunit vaccines.
- genes encoding the functional complexes can be constructed and placed in vectors or plasmids for use in live vector or DNA plasmid vaccines.
- vaccine compositions of the invention can comprise "pharmaceutically acceptable carriers".
- pharmaceutically acceptable carriers are substances which do not interfere with the operation of the immunogen and are non-toxic to the animal.
- Pharmaceutically acceptable carriers include but are not limited to saline, buffered saline, dextrose, water, glycerol, sterile isotonic aqueous buffer, oil-on-water or water-in-oil emulsions, aqueous compositions, liposomes, microbeads, microsomes or adjuvant compounds and combinations thereof.
- a physiologically balanced culture medium containing one or more stabilizing agents such as stabilized, hydrolyzed proteins, lactose, etc.
- the carrier is preferably sterile.
- the formulation should suit the mode of administration.
- composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
- the composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder.
- Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
- the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
- a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
- an ampoule of sterile diluent can be provided so that the ingredients may be mixed prior to administration.
- a lyophilized fusion-competent complex of the invention is provided in a first container; a second container comprises diluent consisting of an aqueous solution of 50% glycerin, 0.25% phenol, and an antiseptic (e.g., 0.005% brilliant green).
- diluent consisting of an aqueous solution of 50% glycerin, 0.25% phenol, and an antiseptic (e.g., 0.005% brilliant green).
- An effective immunizing amount is that amount sufficient to produce an immune response to the complex in the host to which the fusion-dependent complex, or vaccine immunogen, is administered.
- an effective immunizing amount of a vaccine immunogen for a human subject of the present invention is within the range of 10 to 100 mg per kilogram body weight, more preferably 0.1 tolOmg per kilogram body weight. Boosting is possible but not preferred.
- the exact amount of vaccine immunogen utilized in a given preparation is not critical, provided that the minimum amount necessary to provoke an immune response is given.
- individual dosages may range from about 50-650 ⁇ g per immunization.
- dosing is dependent upon the formulation of the vaccine immunogen (e.g. purified mimitope, vs. purified FRMS, vs. whole cell preparations).
- the vaccine formulation comprises an effective immunizing amount of the FRMS immunogen, preferably in combination with an immunostimulant; and a pharmaceutically acceptable carrier.
- immunostimulant is intended to encompass any compound or composition which has the ability to enhance the activity of the immune system, whether it be a specific
- n potentiating effect in combination with a specific immunogen, or simply an independent effect upon the activity of one or more elements of the immune response.
- immunostimulant compounds in vaccine compositions are the adjuvants alum or muramyl dipeptide (MDP) and its analogues. Methods of utilizing these materials are known in the art, and it is well within the ability of the skilled artisan to determine an
- purified immunogens or complexes vaccines can be carried out by standard methods.
- the purified protein(s) should be adjusted to an appropriate concentration, formulated with any suitable vaccine adjuvant and packaged for use.
- Suitable adjuvants may include, but are not limited to: mineral gels, e.g., aluminum hydroxide; surface active substances such as lysolecithin, pluronic polyols; polyanions; peptides; oil emulsions; alum, and MDP.
- the immunogen may also be inco ⁇ orated into liposomes, or conjugated to polysaccharides and/or other polymers for use in a vaccine formulation.
- the complex is a hapten, i.e., a molecule that is antigenic in that it can react selectively with cognate antibodies, but not immunogenic in that it cannot elicit an immune response
- the hapten may be covalently bound to a carrier or immunogenic molecule; for instance, a large protein such as serum albumin will confer immunogenicity to the hapten coupled to it.
- the hapten-carrier may be formulated for use as a vaccine.
- Effective doses (immunizing amounts) of the vaccines of the invention may be any suitable vaccines of the invention.
- the present invention provides a method of treating or preventing infection by a virus in a subject comprising administering to the subject an immunogenic amount of the FRMS effective to treat or prevent infection by the virus.
- the present invention provides a method of treating or preventing infection by HIV in a human comprising administering to the human an immunogenic amount of the FRMS effective to treat or prevent infection by HIV.
- the present invention also provides a method of treating or preventing infection by a virus in a subject comprising administering to the subject an amount of the FRMS effective to treat or prevent infection by the virus.
- the invention provides a method of treating or preventing infection by HIV in a human comprising administering to the human an amount of the monoclonal antibody effective to treat or prevent infection by HIV.
- FRMS FRMS
- Such antibodies may be used as an immunogen to generate antibodies which immunospecifically bind such an immunogen.
- Such antibodies include but are not limited to polyclonal, monoclonal, chimeric, single chain, Fab fragments, and an Fab expression library.
- the subject invention also concerns methods for generating an immune
- an animal is administered a FRMS of the invention.
- the FRMS comprises the HIV envelope protein and arises from interaction with CD4 and CCR5.
- the FRMS is administered in a manner such that an immune response is produced in the animal to the immunogen.
- antibodies to epitopes on the FRMS are produced. More preferably, the antibodies produced include neutralizing antibodies. In a highly preferred embodiment, the antibodies produced are capable of neutralizing a wide variety of primary isolates of the virus.
- ⁇ - polyclonal antibodies to a FRMS or derivative or analog may be obtained.
- various host animals can be immunized by injection with the native FRMS, or a synthetic version, or derivative (e.g., fragment or chimera) thereof, including but not limited to rabbits, mice, rats, etc.
- Various adjuvants may be used to
- _ ⁇ increase the immunological response, depending on the host species, and including but not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum.
- Freund's complete and incomplete
- mineral gels such as aluminum hydroxide
- surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants
- BCG Bacille Calmette-Guerin
- corynebacterium parvum include BCG (bacille Calmette-Guerin) and corynebacterium parvum.
- any technique which provides for the production of antibody molecules by continuous cell lines in culture may be used.
- the hybridoma technique originally developed by Kohler and Milstein, (Kohler et al., 1975, Nature 256:495-497), as well as the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4:72), and the EBV-hybridoma technique to produce human monoclonal antibodies Colde et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96).
- monoclonal antibodies can be produced in germ-free animals (see e.g., PCT/US90/022548).
- human antibodies may be used and can be obtained by using human hybridomas (Cole et al., 1983, Proc. Natl. Acad. Sci. U.S.A. 80:2026-2030) or by transforming human B cells with EBV virus in vitro (Cole et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, pp. 77-96).
- spleens from mice immunized with FRMS are harvested and splenocytes are isolated by teasing the spleen apart in culture medium.
- Splenocytes are fused with HPRT-negative mouse myeloma cells using polyethylene glycol (PEG).
- PEG polyethylene glycol
- Cell pellets containing a 4:1 ratio of splenocytes to myeloma cells are resuspended and treated in serum-free medium containing 50% pretested PEG-4000 (2.5 min contact).
- the cell suspension is then slowly diluted in serum-free medium, and cells are plated in 96-well culture dishes in HAT selection medium: DMEM culture medium containing 20% pretested fetal bovine serum, 10% NCTC-109 (Gibco), 1% nonessential amino acids, IX glutamine and IX Pen-Strep, IX OPI (15 mg/ml oxaloacetate, 5 mg/ml sodium pyruvate, and 20 units/ml insulin), 100 ⁇ M hypoxanthine, 0.4 ⁇ M aminopterin, and 16 ⁇ M thymidine. Wells containing hybridomas are monitored microscopically and supernatants harvested as appropriate/necessary for assay of antibody production.
- HAT selection medium DMEM culture medium containing 20% pretested fetal bovine serum, 10% NCTC-109 (Gibco), 1% nonessential amino acids, IX glutamine and IX Pen-Strep, IX OPI (15 mg/ml oxaloacetate
- Antibody fragments which contain the idiotype of the molecule can be generated by known techniques.
- such fragments include but are not limited to, the F(ab') 2 fragment which can be produced by pepsin digestion of the antibody molecule, the Fab' fragments which can be generated by reducing the disulfide bridges of the F(ab') 2 fragment, the Fab fragments which can be generated by treating the antibody molecule with papain and a reducing agent, and Fv fragments.
- Fab fragments from combinatorial libraries are also contemplated in the invention (see e.g., Chanock et al., 1993, Infect Agents Dis. 2:118-31).
- screening for the desired antibody can be accomplished by techniques known in the art (e.g., enzyme- linked immunosorbent assay or ELISA). For example, to select antibodies which specifically recognize FRMS, one may
- ⁇ performed by functional assays such as virus neutralization assay, such as those known in the art or described herein.
- Antibodies specific to a domain of a FRMS is also provided.
- Antibodies specific to an epitope of a FRMS protein are also provided.
- the generated antibodies may be isolated by standard techniques known in n the art (e.g., immunoaffinity chromatography, centrifugation, precipitation, etc.) and used in diagnostic immunoassays.
- the antibodies may also be used to monitor treatment and/or disease progression.
- Any immunoassay system known in the art, such as those listed supra, may be used for this pu ⁇ ose including but not limited to competitive and noncompetitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme-linked
- the FRMS of the subject invention when used as immunogens are capable
- the FRMS elicit neutralizing antibody to a wide variety of primary isolates of the virus.
- the FRMS elicit neutralizing antibody to primary isolates of HIV-1.
- Neutralizing antibodies while not elicited by static HIV envelope protein, are able to bind at least weakly to static envelope and are carried with the envelope protein through the
- viral envelope proteins can serve as a vector to bring antibodies (e.g. linked to e.g. toxin, other protein, vaccine immunogens) into the cell, into the cytoplasm or into the histocompatibility complex presentation pathway.
- Hybridoma cell supernatants of the invention can be assayed for the ability to neutralize the homologous virus as well as for the ability to neutralize a range of genetically diverse viruses.
- the ability to neutralize genetically diverse viruses suggests recognition by the mAb of a conserved determinant. Further, mAb can be tested for
- MAb 1 n inhibition of envelope-mediated cell-cell fusion.
- MAb can also be assayed for the ability to bind and/or immunoprecipitate envelope protein and envelope protein-containing receptor-associated complexes.
- mAb that are specific for fusion-dependent epitopes and bind only weakly, if at all, to static envelope protein.
- hybridomas are identified using high-throughput
- the antibodies produced by the methods of the invention are used individually (e.g. a single mAb) or in combination (e.g., one or more mAbs directed to the same or different epitopes). Combinatorial use of
- 9n antibodies may include multiple mAbs directed to the same virus or to different viruses. In some embodiments of the invention it may be advantageous to use multiple mAbs directed to the same virus or multiple mAbs directed too the same FRMS of a virus.
- antibodies of the invention or the derivatives or analogues thereof to bind FRMS of the invention and thereby interfere with viral infection can be assayed by various methods.
- Binding can be assayed by means well-known in the art. For example,
- bioassays may be performed in which cells known to be expressing a chemokine receptor are exposed to the mAb derivative or analogue to be tested and assayed for a known effect (e.g., signal transduction).
- mAb, derivatives or analogues can be tested for the ability to bind chemokine receptors, host cell receptors or viral envelope proteins by procedures, including but not limited to, protein affinity chromatography, affinity blotting,
- High throughput screening for mAb, derivative or analogue binding may be performed by methods known in the art, including but not limited to flow cytometry.
- cells that express human CD4 and one of the HIV co-receptors are treated with biotinylated mAb, derivative, or analogue and cell surface binding to the FRMS is detected with an avidin FITC conjugate.
- flow cytometry system may be used in a competitive binding assays using the monoclonal antibodies of the invention in the following manner or by any method known in the art.
- a mAb of the invention is labeled and examined for the ability to bind an FRMS of the invention compared with the ability of a
- Test antibodies of the invention may be derived from samples such as serum from a subject, or manufacturing samples such as hybridoma supernatants.
- the anti-viral activity exhibited by the mAb, mAb derivative and/or analogue of the invention may be measured, for example, by easily
- ⁇ performed in vitro assays, which can test the compound's ability to inhibit syncytia formation or to inhibit infection by cell- free virus and assess the effects of the compound on cell proliferation and viability. Applying these assays, the relative anti-viral activity that a mAb, derivative and/or analogue exhibits against a given virus or strain of immunodeficiency virus formulation best suited for viral and strain specific inhibitory n activity can be determined.
- a cell fusion assay is used to test the ability of mAb, derivative or analogue, to inhibit virus-induced syncytia formation in vitro.
- a cell fusion assay is used to test the ability of a mAb derivate or analog of the invention to inhibit HIV-induced syncytia formation in vitro.
- such an assay involves culturing uninfected CD4 + cells in the presence of chronically HIV-infected cells and the composition containing the mAb, derivative or analogue to be assayed. For each, a range of concentrations may be tested. This range should include a control culture wherein no mAb, derivative and/or analogue has been added. Standard conditions for culturing, well known to those of ordinary skill in the art, are used. After incubation for an
- the culture is examined microscopically for the presence of multinucleated giant cells, which are indicative of cell fusion and syncytia formation.
- an in vitro infectivity assay is performed using primary macrophages and the macrophage-tropic isolate HIV-l BaL , the first described macrophage-tropic HIV-1 isolate (see, Gartner et al., 1986, Science 233:215).
- primary macrophage cells isolated according to methods known in the art are infected with HIV-l BaL that has been propagated and maintained only in primary macrophages.
- the input immunodeficiency virus is incubated with primary macrophages in the presence of concentrations of the mAb, derivative, or analogue to be tested. After a defined period of infection, unbound virus is removed by washing, and the cells are placed in culture.
- the level of virus replication in this assay may be assessed by techniques known
- RT reverse transcriptase
- a constant level of inhibition of viral infection or replication is determined by measuring output HIV p24 levels (or another indicator of viral infection or replication, such as for example, RT) relative to control assays performed in the absence of the mAb, derivative or analogue.
- the ⁇ mAb derivative or analogue reduces levels of virus, as measured by, for example, p24, by ⁇ 50% relative to control assays carried out in the absence of test compound.
- telomeres The presence of p24 may be determined using methods known in the art, such as commercially available enzyme-linked immunosorbent assays (Coulter, Hialeah, Florida; Abbott Laboratories, Hvalstad, Norway). Alternatively, RT activity may be tested by monitoring cell-free enzyme-linked immunosorbent assays (Coulter, Hialeah, Florida; Abbott Laboratories, Hvalstad, Norway). Alternatively, RT activity may be tested by monitoring cell-free
- a mAb of the invention is assayed in a primary isolate neutralization assay.
- a variety of neutralization assays are known in the
- mAbs of the invention are assayed in a focus assay.
- test antibody is incubated with a measured amount of virus prior to the addition of the virus to host cells. Infection of the host cells is measured by staining said cells by immunohistochemical methods known in the art using an antibody specific for the viral protein(s).
- a focus assay In one embodiment of the focus assay, test antibody is incubated with a measured amount of virus prior to the addition of the virus to host cells. Infection of the host cells is measured by staining said cells by immunohistochemical methods known in the art using an antibody specific for the viral protein(s).
- control assay is performed in parallel in which no test antibody is used.
- Test antibody which inhibits (e.g., prevents or decreases) infection compared to the control infection is indicative of a neutralizing antibody.
- a PBL assay can be used to assay neutralization by a test antibody.
- test antibody is incubated with a measured amount of virus prior to the addition of
- _ n the virus to host cells.
- Host cell cultures are allowed to incubate for a period of days, and infection is assayed by measuring the virions present in a sample of host cell supernatant.
- a control assay is performed in parallel in which no test antibody is used.
- Test antibody which inhibits (e.g., prevents or decreases) infection compared to the control infection is indicative of a neutralizing antibody.
- the present invention encompasses the use of a non-human transgenic animal which expresses one or more components whose association results in the formation of the FRMS of the invention.
- the components whose association results in the formation of the FRMS may include those listed in Table II and Table III, herein.
- the transgenic animal models of the invention may be used as Tolerance models and/or Infection models. In the case of Tolerance models, it is preferred that all of the cellular components important to the formation of the FRMS are expressed in the transgenic animal. In the case of Infection models, one or more of the components important for the formation of the FRMS may be expressed such that the transgenic animal is capable of infection by the virus to which the FRMS is derived.
- mice Animals of any species, including, but not limited to, mice, rats, rabbits, guinea pigs, pigs, micro-pigs, goats, sheep, and non-human primates, e.g., baboons, monkeys, and chimpanzees may be used to generate transgenic animals expressing a
- transgenic refers to animals expressing one or more components whose association results in the formation of the FRMS gene sequences from a different species (e.g., mice expressing human sequences encoding one or more component whose association results in the formation of a FRMS), as well as animals that have been genetically engineered to over n express endogenous (i.e., same species) sequences encoding one or more component whose association results in the formation of a FRMS or animals that have been genetically engineered to no longer express endogenous gene sequences encoding one or more component whose association results in the formation of a FRMS (i.e., "knock-out" animals), and their progeny.
- Any technique known in the art may be used to introduce an gene encoding one or more component whose association results in the formation of a FRMS transgene into animals to produce the founder lines of transgenic animals.
- Such techniques include, but are not limited to pronuclear microinjection (Hoppe and Wagner, 1989, U.S. Pat. No. 4,873,191); retrovirus mediated gene transfer into germ lines (Van der Putten, et al, 1985,
- transgenic animal clones containing a one or more component whose association results in the formation of a FRMS transgene for example, nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal or adult cells induced to quiescence (Campbell, et al, 1996, Nature 380, 64-66; Wilmut, et al, Nature 385, 810-813).
- the present invention provides for transgenic animals that carry a transgene of one or more component whose association results in the formation of a FRMS in all their cells, as well as animals that carry the transgene in some, but not all their cells, i.e., mosaic animals.
- the transgene may be integrated as a single transgene or in concatamers, e.g., head-to-head tandems or head-to-tail tandems.
- the transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Lasko, et al, 1992, Proc. Natl. Acad. Sci. USA 89, 6232-6236).
- n The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
- gene targeting is preferred. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous gene are designed for
- the transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous gene encoding one or more component whose association results in the formation of a FRMS in only that cell type, by following, for example, the teaching of Gu, perennial et al. (Gu, et al, 1994, Science 265, 103-106).
- the regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
- the expression of the recombinant component whose association results in the formation of a FRMS gene may be ⁇ e . assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to assay whether integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques that include but are not limited to Northern blot analysis of tissue samples obtained from the animal, in situ
- Samples of the component gene-expressing tissue may also be evaluated immunocytochemically using antibodies specific for the component transgene product.
- the invention encompasses the use of a transgenic mouse for the development of model system which allows screening of a test vaccines intended as vaccines for a species other than mouse.
- the test vaccine is a vaccine which is intended for a human.
- the transgenic mouse model of the invention encompasses a mouse which is constructed to express the host cell receptors and/or co-receptors of another species for a virus which is not native to a mouse. Such mice are advantageous for the analysis of virus neutralization, since the effect of neutralizing antibodies directed to the host cell receptors is eliminated. These mice provide a model system for viral tolerance.
- a transgenic mouse may be constructed or used which expresses human host cell receptor(s) for a virus capable of infecting a human (such as HIV).
- vaccine studies utilize transgenic mice expressing human CD4 and CCR5 co-receptor under the control of CD4 regulatory elements which restrict co-expression to thymocytes and T helper lymphocytes (Killeen et al., 1993, EMBO J., 12:1547-53). These mice are used in order to allow for the analysis of virus neutralization without the confounding effect of neutralizing antibodies directed to CD4 or CCR5.
- the immune response mimics that in humans; immunogenic epitopes are restricted to envelope and to HIV-dependent conformations of CD4 and co- receptor.
- the present invention provides a novel use as a system for the analysis of HIV vaccines.
- the transgenes function only to provide tolerance to the human CD4 and CCR5 components of the vaccine.
- transgenic mice are immunized one or more times with the FRMS of the invention.
- control immunogens may be used to vaccinate other mice which serve as a control.
- the FRMS immunogen may be in any of the forms described herein including in cell lysate, in solution, in a cross-linked structure, isolated, purified, etc.
- the FRMS immunogen may also be introduced to the mouse in a vaccine formulation.
- sera or antibodies produced from mice immunized the FRMS or control immunogens are collected and assayed for the ability to neutralized PI virus.
- Serum is collected at an appropriate time following immunization to allow for the production of an immune response in the mouse. Such times are well known in the art. In one embodiment, sera is obtained 2 weeks following each immunization.
- sera or antibodies are tested for the ability to neutralize two, three or four primary isolates of the virus. More preferably, sera or antibodies are tested for the ability to neutralize four, six, eight primary isolates of the virus. Most preferably, sera or antibodies are tested for the ability to neutralize 10-15, 15-25, or more primary isolates of the virus.
- Neutralization of a primary isolate may be assayed by methods known in the art including those presented herein.
- neutralization is determined by the inhibition of infectivity associated with incubating the test antibody with a sample of the virus to be used in infection.
- An antibody which inhibits (e.g. decreases or blocks) viral infection is indicative of a neutralizing antibody.
- sera or antibodies are tested for the ability to inhibit syncytia formation or to inhibit infection by cell-free virus and assess the effects of the compound on cell proliferation and viability.
- the test antibody is a monoclonal antibody.
- the test antibody is a human monoclonal antibody produced in response to a FRMS immunogen.
- FRMS immunogen a monoclonal antibody produced in response to a FRMS immunogen.
- virus neutralization assay is performed on more than one primary isolates.
- virus neutralization assay are performed on 2-5, 5-10, 10-15 primary isolates.
- neutralization assay are performed on 15-20, 20-30, or 30 or more primary isolates.
- the primary isolates assayed are from more than
- Neutralization may characterized in terms of percent neutralization of a number of primary isolates. Generally, in this embodiment, a single assay is used to assess antibody neutralization. In one embodiment, an antibody of the invention neutralizes 30- 40%, 40-50%, or 50-60% of the primary isolates of the virus. In a preferred embodiment, ⁇ an antibody of the invention neutralizes 60-75%, 70-85%, or 80-90% of the primary isolates of the virus. In a most preferred embodiment, an antibody of the invention neutralizes 90- 95%, 95-98%, or 99-100% of the primary isolates of the virus. In preferred embodiments, when mAb are tested for neutralization of primary isolates, neutralization is expressed as the number of foci in the presence of mAb (or hybridoma supernatant) relative to the ⁇ . number in the presence of medium alone.
- Transgenic mice (hu CD4+, hu CCR5+, mouse CD4+) are immunized with an HIV FRMS immunogen (cross-linked COS-env co-cultured with U87-CD4-CCR5 cells) or with cell controls (U87-CD4-CCR5 cells alone or cocultured with mock-transfected COS cells). Sensitivity of the homologous 168P virus to 0 neutralization by vaccine sera is determined with U87-CD4 cells expressing either CCR5 or CXCR4 co-receptor (see, LaCasse, et al., 1998, Science 72:2491; Follis, K.E., et al.,1998, Journal of Virology 72:7603).
- HIV primary isolates may include but are not 5 limited to 92US657, 92US660, 92TH014, 89.6, 320NSI, 320SI, SHIV89.6P, 168P, 92RW023, 92UG031, 92UG037, 92RW008, 93IN101, 93IN999, 93IN905, 93IN904, 92UG035, 92UG021, 92UG024, 92UG046, 92TH023, 92TH024, 93TH051, and 93TH053.
- an antibody produced to an HIV FRMS by the methods of the invention is capable of neutralization of 60-70%, 70-80%, 80-85% of such primary isolates of HIV.
- an antibody produced to an HIV FRMS is capable of neutralization of 80-90%, 90-95%, or 95-99% of
- an antibody produced to an HIV FRMS is capable of neutralization of 100% of such primary isolates of HTV.
- primary isolates of HIV may include one or more isolates of clade A, B, C, D, E, F, G, H, or I. In other embodiments, primary isolates of HIV may include one or more isolates an M-group, O-group, or N-group.
- neutralization activity can be demonstrated as antibody-mediated by methods known in the art. For example, sera can be adsorbed to a solid support containing Protein-A and Protein-G. In the case that an antibody is responsible for the virus neutralization activity of the sera, sera depleted of Protein-A and Protein-G binding-proteins will be expected to contain little or no virus neutralization
- the present invention provides a method of screening a molecular structure for vaccine efficacy comprising immunizing a transgenic non-human mammal with the molecular structure, wherein said transgenic non-human mammal
- 9 discloses from one or more transgenes both human CD4 and a co-receptor for HIV, and detecting any neutralizing antibodies to HIV that are produced by said mammal.
- the invention also provides a method of screening a molecular structure for vaccine efficacy comprising immunizing a transgenic non-human mammal with the molecular structure, wherein said transgenic non-human mammal expresses from one or ⁇ . more transgenes said one or more host cellular membrane proteins; and detecting any neutralizing antibodies to said virus that are produced by said mammal.
- Test animals may include mice, hamsters, dogs, cats, monkeys, rabbits, chimpanzees, etc., and eventually human subjects.
- methods of introduction of the vaccine may include oral, intracerebral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal or any other standard routes of immunization.
- the immune response of the test subjects can be analyzed by various approaches such as: the reactivity of the resultant immune serum to the immunogen of the invention, as assayed by known techniques, e.g., enzyme linked immunosorbent assay (ELISA), immunoblots, radioimmune assays (RIA) radioimmunoprecipitations, etc.
- ELISA enzyme linked immunosorbent assay
- RIA radioimmune assays
- protection of immunized hosts from infection by the pathogen and or attenuation of symptoms due to infection by the pathogen in immunized hosts can serve as evidence of vaccine efficacy.
- the vaccine of the invention may be tested in rabbits for the ability to induce an antibody response to the FRMS immunogen.
- Male specific-pathogen- free (SPF) young adult New Zealand White rabbits may be used.
- the test group each receives a fixed concentration of the vaccine.
- a control group receives an injection of 1 mM Tris-HCl pH 9.0 without the FRMS immunogen.
- Blood samples may be drawn from the rabbits every one or two weeks, and serum analyzed for antibodies to FRMS. The presence of antibodies specific for the immunogen may be assayed, e *., using an ELISA.
- animal testing of viral vaccine effacy is by use of a transgenic mouse model as described herein in Section 5.7.1.
- the invention also provides methods for optimizing and quantitating vaccines comprising the subject FRMS.
- the subject complexes are prepared using autologous cells negating reactivity toward host cellular components.
- Vaccine 9n compositions comprising complexes prepared using non-autologous cells, however, can be assessed using transgenic animals genetically engineered to be tolerant to the host cellular components of the complexes. Using these transgenic animals it is possible to determine whether the success achieved by vaccinating with the fusion-complexes of the subject invention is due to novel fusion-competent epitopes presented by the complexes.
- the FRMS result from the interaction of HIV(168P) viral envelope protein and the human proteins CD4 and CCR5.
- transgenic mice genetically engineered to be tolerant to the human components of the vaccine.
- Mice expressing both the human CD4 receptor and the human CCR5 co-receptor were used to evaluate the complex and - n demonstrate that the neutralizing antibody generated was to immunologic epitopes restricted to envelope protein or HIV-dependent conformations of CD4 and co-receptor. It is noted that these transgenic mice were originally used as a mouse model for HIV infection. Utilizing transgenic animals to screen vaccine immunogens is a novel use of these mice. Transgenic mice exhibiting tolerance to host portions of vaccines can be used to optimize vaccine components, test variants and evaluate adjuvants.
- transgenic animals other than transgenic mice can be used to screen candidate vaccines.
- the invention provides a method of screening a molecular structure for vaccine efficacy comprising immunizing a transgenic non-human mammal with a molecular structure comprising an epitope formed as a result of association of (a) an envelope protein of an enveloped virus, with (b) one or more cellular membrane proteins, which envelope protein and cellular membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins, wherein said transgenic non-human n mammal expresses from one or more transgenes said one or more host cellular membrane proteins; and detecting any neutralizing antibodies to said virus that are produced by said mammal.
- the molecular structure is isolated.
- the invention provides a method of screening a molecular structure for vaccine efficacy comprising immunizing a transgenic non-human mammal with a molecular structure comprising an epitope formed as a result of association
- the subject invention also encompasses antibodies that are elicited by the novel epitopes of the FRMS of the subject invention contain antibodies useful as reference standards and components in diagnostic assays and kits. Additionally, mAb to the FRMS of the subject invention can be used as reference standards, diagnostic agents for in vitro and in vivo bioassays and as markers for critical determinants in binding and fusion. For example, during large scale production of the FRMS of the invention, the mAb of the
- the mAb may be used to determine the amount to lot-to-lot variation (e.g., by ELISA, or RIA using the mAb).
- labeled FRMS may be used, by way of a competition assay with the test FRMS for binding a mAb specific for the FRMS.
- Similar assays may be used to determine the antibody titer of a sample of serum from a subject.
- a sample of serum from a subject is taken at a time
- the sample serum is then tested for ability to neutralize virus.
- the serum may also be tested for the ability to bind to the immunogen FRMS, or to compete with a neutralizing mAb for binding to the immunogen FRMS.
- the inability or weak ability of the serum sample to neutralize virus or bind FRMS provides an indication that booster vaccinations with the FRMS immunogen are desired for
- the present invention provides a method for monitoring the production of antibody to the molecular structure in a subject previously administered an amount of the molecular structure comprising isolating from said subject a sample comprising serum; and detecting the presence of any antibodies to the molecular structure in said serum.
- detecting is carried out by a method comprising
- polyclonal sera or monoclonal antibodies can be administered to individuals for pu ⁇ oses of treatment or prevention of viral infection and its sequences.
- such antibody can be administered for pu ⁇ oses post-exposure prophylaxis or to prevent maternal transfer of virus to the neonate by way of passive f) immunization.
- Humanized mAb of the invention can be used in protection against maternal-infant virus transmission or in post-exposure prophylaxis or treatment.
- MAb of the present invention are also useful in passive immunization studies in a hu-PBL-scid mouse model of HIV infection and in SHIV studies in rhesus macaques. Accordingly, the present invention provides a method for treating or preventing infection by HIV in a human
- 9 - fetus comprising administering to a pregnant human containing said fetus an amount of the monoclonal antibody effective to treat or prevent infection by HIV in said fetus.
- Production of mAb to the FRMS of the subject invention can be used to further delineate the biochemical and immunochemical pathway to infection, and define critical epitopes for PI virus neutralization.
- Monoclonal antibodies (mAb) generated to the FRMS of the subject invention can be used to further delineate the biochemical and immunochemical pathway to infection, and define critical epitopes for PI virus neutralization.
- n FRMS of the subject invention can be used to dissect the biochemical basis for binding and entry, and the immunochemical basis for primary isolate virus neutralization.
- immunogens should be chosen for MAb development that excel in immunogenicity studies in vaccination models, are potent and elicit a broad range of primary isolate virus neutralizing antibodies.
- the antibodies of the present invention can also be used in the production of antiidiotypic antibody.
- the antiidiotypic antibody can then in turn be used for immunization, in order to produce a subpopulation of antibodies that bind the initial antigen of the pathogenic microorganism (Jerne, 1974, Ann. Immunol. (Paris) 125c:373; Jerne, et al., 1982. EMBO J. 1:234).
- mAb can also be used as an anti-viral agent in blood and blood products.
- the present invention provides neutralizing antibodies that are useful as an
- the mAb of the invention is useful as an additive to donor blood (e.g., blood within a blood bank) which is to be used in the treatment of a subject.
- the mAb of the invention is useful as an additive to blood collection bags, gloves, blood sample collection containers, etc.
- one or more mAb(s) of the invention are used to cleanse the birth canal prior to delivery of a child in order to prevent perinatal infection.
- Effective amounts of the mAb of the invention for blood and blood-product additives include 1 ⁇ g/ml to lOmg/ml. As will be apparent to one skilled in the art, effective amounts will be influences by antibody affinity, additives, and formulations. Any amount may be used such that the effective
- ⁇ - amount is capable of inhibiting (e.g. reducing or preventing) viral infection.
- a mixture of neutralizing mAb against a variety of viruses is used in decontamination.
- an effective amount of a neutralizing mAb of the invention which is immunospecific for HIV is added to a sample of human blood before such blood is transfused into a recipient subject.
- the HIV is neutralized n by the mAb.
- the present invention provides a sample of mammalian blood, to which an amount of the antibody has been added effective to inhibit or decrease infection by the virus.
- the present invention provides a sample of human blood, to which an amount of the antibody has been added effective to inhibit or decrease infection by HIV.
- the present invention also provides a method of inhibiting infection by a
- the present invention provides a method of inhibiting infection by HIV in a sample of human blood comprising contacting said sample of human blood with an amount of the monoclonal antibody effective to inhibit infection by HIV.
- the neutralizing antibodies of the invention are also useful in decontaminating any object exposed to the virus to which the antibody is directed.
- Objects which may be decontaminated with the mAb of the invention include but are not limited to surgical and dental tools (such as drills, picks, scalpels, etc).
- a mixture of neutralizing mAb against a variety of viruses is used in decontamination.
- objects which are not readily disposable are decontaminated with one or more mAbs of the invention.
- Effective amounts of the mAb of the invention for decontamination include 1 ⁇ g/ml to lOmg/ml.
- effective amounts will be influences by antibody affinity, additives and formulations. Any amount may be used such that the effective amount is capable inhibiting viral infection following decontamination.
- HIV is neutralized in the decontamination. Accordingly, the present
- ⁇ - invention provides a method of decontaminating surgical or dental tools comprising contacting said tools with an amount of the monoclonal antibody effective to inhibit infection by said virus.
- the present invention provides a method of decontaminating surgical or dental tools comprising contacting said tools with an amount of the monoclonal antibody effective to inhibit infection by HIV.
- antibodies directed to an FRMS may be used to screen molecules which comprise an epitope recognized by the antibody.
- a mAb oft he invention can be used to screen small molecules for the presence of an antibody-recognized epitope. In one binding of the mAb to the small molecules indicates the presence of such epitope on the small molecule.
- Such small molecules in turn
- the mAbs of the invention may be tested as potential immunogens or therapeutics.
- the mAbs of the invention may be used to identify peptides which bind to the antibody (e.g., from a phage display library). Such peptides may be tested, for example, for immunogenicity (i.e. as mimetopes).
- the mAb of the invention are also useful as an anti-viral agent in n contraceptive or microbicide products.
- the addition of one or more mAb of the invention to a contraceptive or microbicide product is particularly preferred for viruses which are known to be sexually transmitted.
- the invention provides a contraceptive product comprising an effective amount of one or more neutralizing mAb(s) of the invention.
- the contraceptive products may further comprise of spermicidal agent.
- spermicidal agent a mixture of neutralizing mAbs against a variety of viruses is used in the contraceptive product.
- Effective amounts of the mAb of the invention for contraceptive or microbicide products include 1 ⁇ g/ml to lOmg/ml.
- the effective amounts will be influences by antibody affinity, additives and formulations. Any amount may be used such that the effective amount is capable of inhibiting (e.g. reducing or preventing) viral infection.
- an effective amount is that which is capable of inhibiting infection upon exposure by sexual transmission of a virus.
- the contraceptive or microbicide products comprises mAb of the invention immunospecific to HIV or other sexually transmitted virus.
- the present invention provides a contraceptive or microbicide in the form of a jelly, foam, cream, or ointment comprising an amount of the antibody of the invention effective to inhibit or decrease infection by the virus.
- the present invention provides a contraceptive or microbicide in the form of a jelly, foam, cream, or ointment comprising an amount of the antibody of the ineffective to inhibit or decrease infection by HIV.
- the present invention provides methods of eliciting production of anti-
- FRMS antibodies in a subject by the administration of a FRMS.
- compositions comprise a therapeutically effective amount of a Therapeutic, and a
- the term "pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
- carrier refers to a diluent, adjuvant, excipient, or vehicle with which the Therapeutic is administered.
- Such pharmaceutical 0 carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical
- excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like.
- the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills,
- compositions can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
- Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E.W. Martin.
- Such compositions will contain a therapeutically effective amount of the Therapeutic, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
- the formulation should suit the mode of administration.
- the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
- compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
- the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
- the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a n hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
- composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
- an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
- the Therapeutics of the invention can be formulated as neutral or salt forms.
- Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino 9n ethanol, histidine, procaine, etc.
- GENE THERAPY Gene therapy refers to therapy performed by the administration of a nucleic acid molecule to a subject.
- the nucleic acid ⁇ - molecule(s) produces its encoded protein(s) and mediates a therapeutic effect by forming a FRMS in vivo.
- a nucleic acid molecule comprising a sequence encoding one or more components whose association results in the FRMS of the invention or a functional derivative thereof, are administered to produce an immunological response - nest to the FRMS, by way of gene therapy.
- a nucleic acid or nucleic acids encoding a viral envelope protein of an envelope virus, a host cell receptor, and a host cell co-receptor or functional derivatives thereof are administered by way of gene therapy.
- a viral envelope protein of an envelope virus administered by way of gene therapy.
- the Therapeutic comprises an HIV
- a nucleic acid molecule is used in which the HIV envelope protein coding sequence is delivered to a subject, which subject expresses native CD4 and a native co-receptor.
- Delivery of the nucleic acid into a patient may be either direct, in which case
- the patient is directly exposed to the nucleic acid or nucleic acid-carrying vector, or indirect, in which case, cells are first transformed with the nucleic acid in vitro, then transplanted into the patient.
- nucleic acid or nucleic acid-carrying vector or indirect, in which case, cells are first transformed with the nucleic acid in vitro, then transplanted into the patient.
- the nucleic acid is directly administered in vivo, n where it is expressed to produce the encoded product.
- This can be accomplished by any of numerous methods known in the art, e.g., by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by infection using a defective or attenuated retroviral or other viral vector (see, U.S. Patent No.
- microparticle bombardment - ( e -g- > a ⁇ ene g un i Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by encapsulation in liposomes, microparticles, or microcapsules, or by administering it in linkage to a peptide which is known to enter the nucleus, or by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu and Wu, 1987, J. Biol. Chem. 262:4429-4432), which can be used to target cell types
- a nucleic acid-ligand complex can be formed in which the ligand comprises a fusogenic viral peptide that disrupts endosomes, preventing lysosomal degradation of the nucleic acid.
- the nucleic acid can be targeted in vivo for cell specific uptake and expression by targeting a specific receptor (see, e.g., International Patent Publications WO 92/06180 by
- nucleic acid can be introduced intracellularly and inco ⁇ orated within host cell DNA for expression, by homologous recombination (Koller and Smithies, 1989, Proc. Natl. Acad. Sci. USA 86:8932-8935, Zijlstra et al., 1989, Nature 342:435-438).
- a viral vector that contains a nucleic acid encoding a viral envelope protein, and host cellular membrane protein(s) are used.
- Any of the viral vectors described in Section 5.1.2, may be used for gene therapy pu ⁇ oses.
- a retroviral vector can be used (see Miller et al., 1993, Meth. Enzymol. 217:581-599). These retroviral vectors have been modified to delete retroviral sequences that are not necessary for packaging of the viral genome and integration into host cell DNA.
- Other references illustrating the use of retroviral vectors in gene therapy include:
- Adenoviruses are other viral vectors that can be used in gene therapy. Adenoviruses are especially attractive vehicles for delivering - genes to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson, 1993, Cu ⁇ ent Opinion in Genetics and Development 3:499-503 present a review of adenovirus-based gene therapy.
- Adeno-associated virus has also been proposed for use in gene therapy (Walsh et al., 1993, Proc. Soc. Exp. Biol. Med. 204:289-300).
- Another approach to gene therapy involves transferring a gene into cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection. Any method known in the art to transfer a gene into a cell
- the method of transfer includes the transfer of a selectable marker to the cells.
- the cells are then placed under selection to isolate those cells that have taken up and are expressing the transfe ⁇ ed gene. Those cells are then delivered to a patient.
- the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
- introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell- mediated gene transfer, spheroplast fusion, etc.
- Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Loeffler and Behr, 1993, Meth. Enzymol. 217:599-618, Cohen et al., 1993, Meth. Enzymol.
- the technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell, and is heritable and expressible by
- the resulting recombinant cells can be delivered to a patient by various methods known in the art.
- epithelial cells are injected, e.g., subcutaneously.
- recombinant skin cells may be applied as a skin graft onto the patient.
- Recombinant blood cells e.g., hematopoietic stem or progenitor _ cells
- the amount of cells envisioned for use depends on the desired effect, patient state, etc., and can be determined by one skilled in the art.
- Cells into which a nucleic acid can be introduced for pu ⁇ oses of gene therapy encompass any desired, available cell type, and include but are not limited to
- the cell used for gene therapy is autologous to the patient.
- nucleic acid molecule encoding the components whose association results in the formation of a FRMS is used.
- the nucleic acid encodes an
- encoding nucleic acid molecule is/are introduced into the cells such that the gene or genes are expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect.
- stem or progenitor cells are used. Any stem and/or progenitor cells which can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention.
- stem cells include but are not limited to hematopoietic stem cells (HSC), stem cells of epithelial tissues such as the skin and the lining of the gut, embryonic heart muscle cells, liver stem cells (International Patent Publication WO 94/08598), and neural stem cells (Stemple and Anderson, 1992, Cell 71:973-985).
- HSC hematopoietic stem cells
- stem cells of epithelial tissues such as the skin and the lining of the gut
- embryonic heart muscle cells embryonic heart muscle cells
- liver stem cells International Patent Publication WO 94/08598
- neural stem cells Stemple and Anderson, 1992, Cell 71:973-985.
- the nucleic acid to be introduced for pu ⁇ oses of gene therapy comprises an inducible promoter operably linked to the coding region (see Section 5.1.2), such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription.
- the invention provides a method of treating or preventing infection by a virus in a subject comprising administering to the subject (a) a first nucleic acid encoding an envelope protein of an enveloped virus; and (b) a second nucleic acid encoding one or more cellular membrane proteins, which envelope protein and cellular .
- membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins, such that the envelope protein and cellular membrane proteins are expressed in the subject and neutralizing antibodies to the virus are produced.
- the method wherein the first and second nucleic acids are the same.
- the first ⁇ and second nucleic acid are different nucleic acid vectors.
- the envelope protein is en envelope protein of HIV
- the cellular membrane proteins are CD4 and an HIV co-receptor.
- the invention provides methods of treatment and prevention of viral infection and disease by administration to a subject in need of such treatment of a therapeutically or prophylactically effective amount of a Therapeutic of the invention.
- the subject is preferably an animal, including, but not limited to, animals such as monkeys, cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most _ n preferably human.
- the subject is a human not afflicted with HIV infection.
- a Therapeutic of the invention e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the Therapeutic, receptor-mediated endocytosis (see, e.g., Wu et al., 1987, J. Biol. Chem. 262:4429-4432), construction of a Therapeutic nucleic acid as part of a retroviral or other vector, etc.
- Methods of introduction include but are not limited to intradeimal, intramuscular, intraperitoneal, intravenous, s subcutaneous, intranasal, epidural, and oral routes.
- the compounds may be administered by any convenient route, for example by infusion or bolus injection, by abso ⁇ tion through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
- Administration can be any convenient route, for example by infusion or bolus injection, by abso ⁇ tion through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
- Administration can be any convenient route, for example by infusion or bolus injection, by abso ⁇ tion through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents.
- Administration can be any convenient route, for example by infusion or bolus injection, by abso ⁇ tion through epithelial
- ⁇ be systemic or local.
- Pulmonary administration can also be employed, e.g., by use of
- compositions of the invention comprising antibody locally to the area in need of treatment; this may be achieved, for example and not by way of limitation, by topical application, by injection, by means of a catheter, by means of a suppository, or by
- the pharmaceutical compositions of the invention is administered to an open wound of a human, which wound is suspected of being exposed to HTV.
- the Therapeutic can be delivered in a vesicle, in
- the Therapeutic can be delivered in a controlled - release system.
- a pump may be used (see Langer, supra; Sefton, 1987, CRC Crit. Ref. Biomed. Eng. 14:201; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321 :574).
- polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and
- a controlled release system can be placed in proximity of the therapeutic target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in
- compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
- the pack may for example comprise metal or plastic foil, such as a blister pack.
- the pack or dispenser device may be accompanied by instructions for administration.
- Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labelled for treatment of an indicated condition.
- the present invention provides assays to test the anti-HIV efficacy of
- the invention further provides methods to monitor FRMS vaccine efficacy in patients or subjects by assaying antibodies in a sample of sera derived from the patient or subject following vaccination
- the Therapeutics of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or
- prophylactic activity prior to use in humans.
- Any in vitro or in vivo assay known in the art to measure viral infection or production can be used to test the efficacy of a Therapeutic of the invention.
- a method of screening a preparation comprising a mAb for anti-HIV activity comprises assaying said n preparation or fraction for the ability to inhibit HIV infection.
- cultured hematopoietic cells e.g., primary PBMCs, isolated macrophages, isolated CD4 + T cells or cultured H9 human T cells
- titers known in the art to
- n available HTV ELISA may be used as a comparative assay for pre- and post- treatment with the Therapeutic.
- a reporter gene i.e., a gene the protein or RNA product of which is readily detected, such as, but not limited to, the gene for chloramphenicol acetyltransferase (CAT)
- CAT chloramphenicol acetyltransferase
- the resulting construct is then introduced by transfection, or any other method s known in the art, into a cultured cell line, such as, but not limited to, the human CD4 + T cell line HUT 78.
- transcription from the HIV-1 LTR is determined by measurement of CAT activity using techniques which are routine in the art. Reduction in HIV-1 LTR driven transcription demonstrates utility of the
- Therapeutics of the invention can also be determined in SIV infected rhesus monkeys (see, Letrin, N.L., et al., 1990, J. AIDS 3:1023-1040), particularly rhesus monkeys infected with SrV mac251 , which SIV strain induces a syndrome in SIV infected rhesus monkeys (see, Letrin, N.L., et al., 1990, J. AIDS 3:1023-1040), particularly rhesus monkeys infected with SrV mac251 , which SIV strain induces a syndrome in
- monkeys which is very similar to human AIDS (Kestler, H., et al., 1990, Science 248: 1109-1112).
- monkeys can be infected with cell free SrV mac251 , for example, with virus at a titer of 10 45 TCID J0 /ml. Infection is monitored by the appearance of SIV p27 antigen in PBMCs.
- Utility of the Therapeutic is characterized by normal weight gain, decrease in SIV titer in PBMCs and an increase in CD4 + T cells.
- a SHIV model is used in which an HIV env protein is constructed into the backbone of SIV.
- monkeys are immunized using a FRMS vaccine comprising cells expressing HIV env and cells expressing rhesus CD4 and CCR5 coreceptor. If adequate in vitro neutralization is obtained, then animals will be challenged with an infectious SHIV virus bearing a neutralization-sensitive HIV env.
- a FRMS vaccine comprising cells expressing HIV env and cells expressing rhesus CD4 and CCR5 coreceptor.
- the utility of the Therapeutic can be determined in human subjects.
- the efficacy of treatment with a Therapeutic can be assessed by measurement of various ⁇ . parameters of HIV infection and HTV associated disease.
- the change in viral load can be determined by quantitative assays for plasma HIV-1 RNA using quantitative RT-PCR (Van Gemen, B., et al., 1994, J. Virol. Methods 49:157-168; Chen, Y.H., et al., 1992, AIDS 6:533-539) or by assays for viral production from isolated PBMCs. Viral production from PBMCs is determined by co-culturing PBMCs from the subject with non-
- kits comprising the fusion-competent protein complexes or antibodies generated by those complexes. Kits including the subject complexes or antibodies can be used to analyze vaccine lots, conduct stability studies or
- Antibodies and the complexes can also be used in diagnostic kits to detect the presence of antibody or virus in the host system.
- antibodies or the subject complexes can be used as substrate or reagent in a kit providing a standard diagnostic enzyme-linked immunosorbent assay (ELISA) or competitive ELISA.
- ELISA enzyme-linked immunosorbent assay
- the invention also provides a pharmaceutical pack or kit comprising one or . 0 more containers comprising one or more of the ingredients of the vaccine formulations of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
- the invention provides a kit comprising in one or more containers a labeled monoclonal antibody to a molecular structure comprising an epitope formed as a result of association of (a) an envelope protein of an enveloped virus, with (b) one or more cellular membrane proteins, which envelope protein and cellular membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of
- the invention provides a kit which further comprises in a separate container a molecular structure comprising an epitope formed as a result of association of (a) an envelope protein of an enveloped virus, with (b) one or more cellular membrane proteins, which envelope protein and cellular membrane proteins are necessary and sufficient under _ suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins.
- the invention provides a kit comprising in one or more containers a labeled monoclonal antibody to a molecular structure comprising an epitope formed as a result of association of (a) an HIV envelope protein, or a mutant thereof
- the invention provides a kit which further comprises in a separate container an epitope formed as a result of association of (a) an HIV envelope protein, or a mutant thereof that assembles into the viral envelope; with (b) human CD4 and a co-receptor for HIV fusion.
- the invention provides a kit comprising in one or more containers, a molecular structure comprising an epitope formed as a result of association of (a) an envelope protein of an enveloped virus, with (b) one or more cellular membrane proteins, which envelope protein and cellular membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins.
- the molecular structure is isolated.
- the invention provides a kit which further comprises a pharmaceutically acceptable carrier, and wherein said molecular structure is present in an immunological amount.
- the invention provides a kit comprising in one or more containers (a) a first nucleic acid encoding an envelope protein of an enveloped virus; and (b) a second nucleic acid encoding one or more cellular membrane proteins, which . ⁇ envelope protein and cellular membrane proteins are necessary and sufficient under suitable conditions for fusion of said envelope of the virus with a cell membrane containing said cellular membrane proteins, such that the envelope protein and cellular membrane proteins are expressed in the subject and neutralizing antibodies to the virus are produced.
- the inventors of the present invention have made the su ⁇ rising discovery that the ability to neutralize PI viruses is related to the presentation of functioning envelope protein in active infection, as compared to the static, non- functioning presentation of the envelope protein in rgpl20 vaccines.
- the HIV envelope protein orchestrates a complex series of protein-protein interactions and structural changes that ultimately results in fusion of the virus and cell membranes, and infection of the cell.
- CD4 CC chemokine receptor 5
- CCR5 CC chemokine receptor 5
- CXC chemokine receptor 4 (Berger, E.A., 1997, AIDS 11 (suppl A), S3; Moore, J.P., et al., 1997, Cu ⁇ ent Opinions in Immunology 9:551; Doranz, B.J., et al, 1997, Immunology Research 16:15). Without limitation as to mechanism, interaction with either co-receptor induces further conformational change in the envelope protein and exposure of the hydrophobic fusion domain of the transmembrane gp41 subunit, which then mediates
- HIV vaccine immunogens were developed by the methods of the invention that explicitly inco ⁇ orate these functional intermediate structures.
- HTV vaccine immunogens were generated that capture the transient structures that arise during HIV binding and fusion.
- formaldehyde-fixed whole cell vaccines elicited antibodies
- the HIV envelope protein orchestrates a complex series of protein-protein interactions and structural changes that ultimately results in fusion of the
- the envelope protein upon binding to CD4, the envelope protein undergoes conformational change that facilitates subsequent interaction with one of several co-receptor molecules, predominantly the CC chemokine receptor 5 (CCR5) or the CXC chemokine receptor 4 (CXCR4) (Reviewed in Berger, E.A., 1997, AIDS l l(suppl A), S3; Moore, J.P., et al, 1997, Current Opinions in
- envelope protein function is the ability to mediate cell-cell fusion.
- cells expressing envelope protein are cocultured with cells expressing CD4 and co-receptor, multinucleated syncytia form over the course of 6-24 hr.
- the process of binding and fusion was captured in progress by formaldehyde- , crosslinking prior to extensive syncytium-formation.
- the functioning envelope protein was derived from a T- lymphocytropic PI virus obtained from the Amsterdam Cohort (ACH168.10; 168P).
- _ n molecularly cloned envelope gene of ACH168.10 was isolated by PCR using the pCR3.1- Uni plasmid (Invitrogen) (Tersmette, M., et al., 1989, Journal of Virology 63:2118; Wrin, T., et al., 1995, Science 69:39; LaCasse, R.A., et al., 1998, Journal of Virology 72:2491).
- the molecularly cloned envelope protein, as well as the parental syncytium-inducing(SI) virus utilizes both CCR5 and CXCR4 co-receptors.
- Cos-7 cells were transfected to express the envelope protein (COS-env) and subsequently cocultured with human U87 glioma cells that express CD4 and CCR5 co- receptor (U87-CD4-CCR5).
- the functional 168P (168P23) envelope gene was transfected into COS-7 cells (American Type Culture Collection, Manassus, VA) by calcium phosphate precipitation (20 ⁇ g DNA/10 6 cells/10 cm culture dish) by methods known in the art (see e.g., Jordan, M., et al., 1996, Nucleic Acids Research 24:596).
- Transiently-expressing COS- 7 cells were harvested 2 days later using 0.5 mM EDTA in phosphate-buffered saline
- CCR5 transgenic mouse is summarized as follows: a 1.15 kb hu CCR5 cDNA was molecularly cloned into an engineered Sail site in exon 2 of a murine CD4 expression cassette (construct c in (Sawada, et al., 1994, Cell 77:917). This minigene contained the murine CD4 enhancer, the CD4 promoter, the first (noncoding) exon, and intron 1 with an internal deletion that eliminated the CD4 silencer. Transgenic founders were identified by flow cytometry using a mAb by methods known in the art.
- mice were bred to hu CD4 expressing transgenic mice to yield progeny expressing hu CD4, hu CCR5, and mouse CD4.
- Pups were screened for expression of hu CD4, hu CCR5, and mouse CD4 by flow cytometry using a Coulter EPICS ELITE flow cytometer.
- the following antibody reagents were used: mouse ⁇ -human CD4/CyChrome (Pharmingen), mouse oc -human CCR5 MAB 180 (R&D Systems) with goat ⁇ -mouse Ig/FITC (Caltag), and rat ⁇ -mouse CD4 L3T4/PE.
- mice expressing hu CD4, hu CCR5, and mouse CD4 were immunized with either FRMS immunogen or with cell controls (U87-CD4-CCR5 cells, alone or cocultured with mock-transfected COS cells).
- Vaccines comprised formaldehyde- fixed whole cells (3xl0 6 cells/0.1 ml) formulated with an equal volume of Ribi adjuvant (R- 700; reconstituted in half the recommended volume of PBS); in some experiments, the initial immunization was with adjuvant containing cell wall material (R-730). Mice received 0.05 ml vaccine in four subcutaneous sites.
- Booster immunizations were at three week intervals, and mice were bled at 10-28 days post-immunizations from the tail. Serum antibodies directed to gpl20 were quantitated by gpl20 ELISA as per methods known in the art (see, Moore, J., et al., 1989, AIDS 3:155).
- Sensitivity of the homologous 168P virus to neutralization by vaccine sera was determined with U87-CD4 cells expressing either CCR5 or CXCR4 co-receptor.
- This PI virus neutralization assay has been validated relative to standard neutralization assay in PBL culture (LaCasse, R.A., et al., 1998, Science 72:2491; Follis, K.E., et al., 1998, Journal of Virology 72:7603) and was determined to perform well in the presence of mouse serum. All sera were heat-inactivated prior to use in neutralization assays.
- Sera were tested for neutralization of 168P using U87- CD4 cells expressing either CXCR4 (black symbols) or CCR5 (white symbols). Data represent averages of three to six neutralization assays using serum obtained 2 weeks following second and third immunization.
- results of virus neutralization assays indicated that no inhibition of infectivity was observed in sera from mice immunized with cell controls, indicating that the transgenic mice were in fact tolerant to hu CD4 and CCR5 and that other adventitious cellular reactivities did not interfere with the virus infectivity assay (Fig 1).
- Sera from mice immunized with FRMS immunogens neutralized the homologous 168P PI virus. Neutralization activity was further demonstrated to be antibody-mediated and as such
- activity could be adsorbed to, and subsequently eluted from, a solid support containing Protein-A and Protein-G .
- serum was adsorbed sequentially to Protein-A Sepharose (Sigma) and Protein-G agarose (Sigma) at 4°. Adso ⁇ tion of antibody was confirmed by gpl20 ELISA.
- the solid supports were combined and antibodies were eluted using 100 mM glycine pH 2.5. The eluate was neutralized and dialyzed by centrifugal
- Serotypically-distinct determinants may arise independently in multiple clades.
- 'fusion-competent' envs derived from clades other than B may be used to determine empirically the number of prototypic envs that define 'fusion-competent' neutralization serotypes. Regardless of the total number of HIV serotypes, it is significant that 80% of PI - viruses tested could be strongly neutralized (>70%) using a single representative but appropriately presented clade B env.
- CXCR4 co-receptor may substitute for CCR5
- CXCR4 a co-receptor to which the animal had not been exposed, argues that neutralization may not directly target the CCR5 component of the vaccine. Further, the CXCR4 co-receptor may substitute for CCR5.
- Fusion-incompetent (FI) immunogens - cocultures that express env but do not undergo cell-cell fusion. These include COS-env cocultured with U87 cells (no CD4 or CCR5 co-receptor), COS-env cocultured with U87-CD4 cells, and COS-env cells to which soluble CD4 (sCD4) was complexed. Specifically, envelope-incompetent (FI) immunogens - cocultures that express env but do not undergo cell-cell fusion. These include COS-env cocultured with U87 cells (no CD4 or CCR5 co-receptor), COS-env cocultured with U87-CD4 cells, and COS-env cells to which soluble CD4 (sCD4) was complexed. Specifically, envelope-
- n expressing cultures were incubated with sCD4 (Berger, E.A., et al., 1988, Proceedings of the National Academy of Sciences USA 85:2357) (5 ⁇ g/ml; 1 hr at 37°) and subsequently washed to remove unbound sCD4. All FI immunogens were fixed with formaldehyde as above. An additional FI immunogen comprised COS-env and U87-CD4-CCR5 cells that were separately fixed with formaldehyde prior to mixing during the formulation of the
- Neutralization was independent _ of specific co-receptor use and data here represent averages of three to six neutralization assays in U87-CD4-CXCR4 or -CCR5 cells. In some cases, sera from all animals within each experimental group were pooled. These results are consistent with the well- documented failure of rgpl20 vaccines to elicit PI virus neutralization. The difference in neutralization by FC and FI vaccine sera was also observed in assays utilizing human
- PBLs primary blood lymphocytes
- FI immunogens Neutralization of the homologous 168P PI virus in human PBL culture by FC, but not FI immunogens was also demonstrated. As shown in Fig 2B, lymphocytes were isolated, stimulated with phytohemagglutinin, and grown in the presence of interleukin-2; neutralization was determined as described herein. HIV p24 antigen was determined after 5
- Vaccine groups were as defined in Fig 2A, and sera were pooled for this assay.
- FC vaccine serum did not neutralize pseudotyped HIV 1 , virions bearing amphotropic MLV envelope protein or primary SIVmac251.
- HIV bearing an amphotropic MLV envelope protein (ampho MLV pseudotype) was used in neutralization sensitivity using pooled FC and FI antisera determined in U87-CD4-CXCR4 cells.
- Primary isolate SIVmac251 neutralization was determined in U87-CD4-CCR5 cells. Symbols are as defined in Fig 2 FC immunogen (black squares) and FI immunogen (Cos- 0 env + U87 cells, black circles).
- FI vaccines were able to elicit neutralization of a related laboratory-adapted isolate of HIV, the T-cell line adapted
- HIV89.6 2Q Cold-dextrative virus
- SHIV89.6, and SHIV89.6P Reimann, K.A., et al., 1996, Journal of Virology 70:3198
- All other primary isolates were obtained through the NTH AIDS Research and Reference Reagent Program and the UNAIDS Network for HIV -1 Isolation and Characterization.
- PI viruses were subjected to limited expansion in PHA-activated PBLs (Wrin, T., et al., 1995, Science
- FC sera elicited by a functioning clade B envelope protein were able to neutralize 23 of 24 PI viruses tested - monocytropic/NSI and T- lymphocytropic/SI viruses from North America/Europe (clade B), Africa (clades A and D),
- - - FC immunogens of the invention may target primarily viral determinants.
- FI sera were uniformly unable to neutralize these heterologous PI viruses.
- the broad and uniform neutralization of diverse PI viruses by the FRMS immunogens suggests that the critical determinants presented by FRMS immunogens (such as FC immunogens) are highly conserved, and may be intimately tied to the basic functioning of the envelope protein in binding and fusion.
- FC vaccine serum was sequentially adsorbed four times with approximately 10 6 formaldehyde-fixed COS cells expressing 168P envelope. Incubations were for 1 hr at 4° with rocking. Controls included prebleed serum and formaldehyde-fixed mock-
- FC vaccine serum was removed by incubation with envelope-expressing cells, but only minimally reduced by incubation with COS cell controls (Fig 6 and Fig 7). Specifically, FC vaccine serum was repeatedly incubated with
- hybridoma supernatants were made by the methods of the invention (using cross-linked COS-env + U87-CD4-CCR5 as an immunogen) were capable of neutralizing primary isolates. Briefly, hybridomas assayed for virus neutralization in the U87-CD4-CXCR4 cell assay as described above. Hybridoma
- _ ⁇ supernatants were tested for the ability to neutralize two representative HTV clade B isolates (ACH168.10 (168P) and 92US657). Neutralization is expressed as the number of foci in the presence of hybridoma supernatant relative to the number in the presence of medium alone. Each point represents a selected hybridoma. As shown in Fig. 8, a majority of hybridomas were found to neutralize both PI viruses to 60-80% (0.4 - 0.2 fraction
- the FRMS immunogens of the invention are capable of eliciting neutralizing monoclonal antibodies against primary isolate virus.
- this example illustrates that an appropriately-presented clade B envelope protein can elicit potent neutralization against most PI viruses from multiple HTV clades and suggest that broad vaccine protection may not require an unlimited number HTV serotypes.
- the static forms of the envelope protein does not function as an effective immunogen
- the present invention recognizes that the critical fusion-related epitopes are sufficiently represented on the static protein to allow binding.
- the FRMS immunogens of the invention provide broad protection against a wide variety of viral serotypes and clades.
- three classes of mutations of the envelope protein include, but are not limited to: (1) mutations that abrogate proteolytic cleavage of the gpl60 precursor protein; (2) mutations that affect the N-terminal gp41 fusion peptide
- Mutations that abrogated proteolytic cleavage of the gpl60 precursor proteins include certain mutations that altered the highly conserved K/R-X-K/R-R site at the C-terminus of gpl20 prevent proteolytic cleavage of the gpl60 polyprotein and also 90 abrogate fusogenicity (Freed, E.O., et al., 1989, J. Virol. 63:4670-5; Guo H.G., et al., 1990, Virology, 174:217-24; Bosch V., et al., 1990, J. Virol. 1990:2337-44; Dubay, J.W., et al., 1995, J. Virol., 69:4675-82).
- Site-directed mutagenesis was used to introduce a documented cleavage site mutation into the 168P envelope protein (REKR to REKT).
- the cleavage-defective 168P envelope protein gpl60 was expressed on the cell surface but was unable to mediate cell-cell fusion or infection by pseudotyped HIV virions.
- Mutations that affect the N-terminal gp41 fusion peptide domain include mutation of the hydrophobic N-terminal region of gp41 which region is believed to mediate fusion by inserting into the cell membrane and destablizing the lipid bilayer. Certain amino - n acids changes within this region render the envelope protein nonfusogenics synthetic peptides.
- Mutations that alter the coiled-coil regions include mutations that are within the highly conserved coiled-coil motif is found in fusion proteins of many virus families (Weissenhorn, W., et al., 1998, Proc. Natl. Acad. Sci. USA 95:6032-6), for example, V570R and Y586E of HIV.
- changes are introduced into the 168P envelope gene and tested for expression and fusogenicity in cell assays before being
- n transferred to pAbT4587-168Penv for generation of the cognate recombinant vaccinia virus.
- Fusion-competent immunogens were prepared by co-cultivating calcium phosphate-transfected COS-7 (American Type Culture Collection, Rockville, MD) cells
- expressing the molecularly cloned HIV 168P envelope protein from ACH168.10 a syncytium-inducing primary isolate that utilized both CCR5 and CXCR4) (LaCasse, R.A., et al., 1998, Science 72:2491) with an equal number of cell fusion partners.
- the cell fusion partners were U87-CD4 cells expressing CCR5 (U87-CD4-CCR5) (Dan Liftman, Skirball Institute of Biomolecular Medicine, NYU Medical Center). Cultures were terminated at the
- Virus neutralization assays using the homologous PI virus 168P were performed using both U87-CD4-CCR5 and -CXCR4 cells, and in some cases sera were analyzed after the second as well as the third immunization. In all cases, results were concordant and are averaged in the data presented in Fig 9. Potent neutralization of the homologous 168P PI virus was consistently observed; neutralization levels of ⁇ 80% were typically obtained at a 1:100
- ACH320.2A.1.2 (320SI) (Amsterdam Cohort) is a molecularly cloned
- T-lymphocytropic PI virus which is particularly refractory to neutralization by HIV-IG, CD4-Ig, and IgG r bl2.
- Residual sera remaining in Example 3 were used to test the sensitivity of this PI virus to fusion-competent and fusion-incompetent immunogens. Although limited to a starting 1:100 dilution of fusion-competent serum, this heterologous
- . _ virus neutralization elicited by fusion-competent immunogens the sensitivity of a panel of representative PI viruses was examined from five prevalent and geographically-diverse phylogenetic clades.
- Neutralization assays in U87-CD4-co-receptor cells are as described above and in Example 1. Fusion-competent sera elicited by a functioning clade B envelope protein were able to neutralize 23 of 24 PI viruses tested-monocytropic/NSI and
- the FRMS of the subject invention were tagged using a commercially available system to facilitate their isolation and purification.
- CD4 molecules were purified either by 1) immunoprecipitation using MAb T4 (Coulter Co ⁇ ., Hialeah, FL) and rabbit- ⁇ -mouse Ig antibody-loaded Protein A agarose, or 2) affinity purification using S-protein agarose. Proteins were released by boiling in sodium dodecyl sulfate (SDS) sample buffer, resolved by 10% polyacrylamide-SDS gel electrophoresis, and transfe ⁇ ed to nitrocellulose. Biotinylated proteins were detected by SDS sample buffer, resolved by 10% polyacrylamide-SDS gel electrophoresis, and transfe ⁇ ed to nitrocellulose. Biotinylated proteins were detected by SDS sample buffer, resolved by 10% polyacrylamide-SDS gel electrophoresis, and transfe ⁇ ed to nitrocellulose. Biotinylated proteins were detected by SDS sample buffer, resolved by 10% polyacrylamide-SDS gel electrophoresis, and transfe ⁇ ed to nitrocellulose. Biotinyl
- COS-7 cells were transfected with CD4-Spep ( ⁇ CXCR4) and were _ subsequently harvested in 0.5% Triton X-100 lysis buffer (control), or subjected to ice-cold formaldehyde fixation prior to lysis.
- CD4-Spep protein was purified by S-protein agarose, and analyzed by Western blot using S-protein HRP (Novagen, Inc.). Minimal loss of S-peptide binding (and/or recovery) was seen with 0.2% formaldehyde fixation for 1 hr; some recovery was lost with 2% formaldehyde in 1 hr and more at 3 hr.
- reaction conditions e.g. 0.2% formaldehyde for 1-2 hr
- DTSSP is a homobifunctional N-hydroxysuccinimidyl ester that
- DTSSP reacts, as formaldehyde, with primary amines. Because of its negative charge, DTSSP cannot penetrate or cross the cell membrane and thus, in contrast to formaldehyde, is expected to spare the cytoplasmic S-peptide tag. Also unlike formaldehyde, DTSSP cross-linking is reversible; an internal disulfide linkage can be cleaved to release cross-linked components.
- COS-7 cells were transfected with 168P envelope or CD4-Spep plasmids; envelope expressing cells as well as mock transfected cells were metabolically labeled in a
- Isolated complexes can be used to define the molecular structure associated with the progression of envelope-mediated membrane fusion.
- Envelope-expressing cells were co-cultured with fusion partners (a-c) for 5 hr and complexes were solublized using 1% Brij-97 detergent (Lapham et al., 1996) and isolated using S-protein affinity chromatography. Co-purified envelope protein isolated using Brij-97 was detected by Western blot analysis using the HIV V3-directed mAb 50.1 (Fig 11). As anticipated, a large amount of 168P envelope protein was isolable following co-culture with cells expressing CD4-Spep. A lesser, but significant, amount of envelope protein was also isolable following co-culture with cells expressing CD4 and CCR5-Spep.
- ⁇ _ Contaminating envelope protein was not detectable from co-cultures with mock-transfected cells. Complexes were isolated from formaldehyde cross-linked fusion-competent co-cultures in similar experiments.
- S-protein purified molecules and complexes can be used as "split virus” or subunit vaccine immunogens.
- Example 7 Recombinant Vaccinia Viruses That Generate Fusion-Competent FRMS in Cell Culture or at the Site of Vaccination.
- rV-168Penv cells, rV-CD4/CCR5 cells, or cross-linked cocultured rV-168Penv and rV-CD4/CCR5 cells were used as FRMS to vaccinate transgenic mice, as described above.
- - Sera obtained from mice are vaccinated with cross-linked cocultured rV-168Penv and rV-CD4/CCR5 cells are capable of virus neutralization across a variety of clades of HIV indicating that the viral vectors may successfully be used in the methods of the invention in the formation of the FRMS.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Virology (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Molecular Biology (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- Engineering & Computer Science (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Biotechnology (AREA)
- Hematology (AREA)
- AIDS & HIV (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Microbiology (AREA)
- Pulmonology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
- Preparation Of Compounds By Using Micro-Organisms (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicinal Preparation (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Priority Applications (7)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2000563676A JP2002522448A (ja) | 1998-08-03 | 1999-08-03 | ウイルス疾患の予防および治療 |
BR9912732-6A BR9912732A (pt) | 1998-08-03 | 1999-08-03 | Estrutura molecular isolada, vìrus envelopadorecombinante, formilação de vacina, anticorpomonoclonal, anti-soro policlonal, geléia, espuma,creme ou unguento contraceptivo, amostras desangue de mamìfero e de humano, kit, linhagemcelular, processos de tratamento ou deprevenção de infecção por um vìrus em sujeito, detratamento ou de prevenção de infecção por hivem um humano, de inibição de infecção por um vìrusem uma amostra de sangue, de inibição de infecçãopor hiv em uma amostra de sangue humano, dedescontaminação de ferramentas cirúrgicas oudentais, de monitoração de produto de anticorpopara a estrutura molecular, de produção de umimunógeno para uso em uma vacina para otratamento ou a prevenção de infecção por umvìrus, e de produção de um imunólogeno para usoem uma vacina para o tratamento ou a prevençãode infecção por hiv, estrutura reticulada,anticorpo monoclonal para a estrutura, dedescontaminação de ferramentas cirúrgicas oudentais, processo de seleção de uma estruturamolecular para eficácia de vacina, mamìfero,complexo de proteìna isolado, formulação devacina, processos de imunização de um animal paraum vìrus, e de preparação de um complexo deproteìna, células fixadas, processos depurificação de um complexo de proteìna e detratamento de um hospedeiro que tem sidoexposto a um vìrus, ou de prevenção de infecçãode um hospedeiro pelo citado vìrus, e composição. |
EP99945011A EP1102788A4 (fr) | 1998-08-03 | 1999-08-03 | Prevention et traitement de pathologie virale |
IL14121199A IL141211A0 (en) | 1998-08-03 | 1999-08-03 | Prevention and treatment of viral disease |
CA002338983A CA2338983A1 (fr) | 1998-08-03 | 1999-08-03 | Prevention et traitement de pathologie virale |
KR1020017001483A KR20010085326A (ko) | 1998-08-03 | 1999-08-03 | 바이러스 질환의 예방과 치료 |
AU57715/99A AU5771599A (en) | 1998-08-03 | 1999-08-03 | Prevention and treatment of viral disease |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US9510598P | 1998-08-03 | 1998-08-03 | |
US60/095,105 | 1998-08-03 | ||
US14180699P | 1999-06-29 | 1999-06-29 | |
US60/141,806 | 1999-06-29 |
Publications (3)
Publication Number | Publication Date |
---|---|
WO2000008043A2 true WO2000008043A2 (fr) | 2000-02-17 |
WO2000008043A3 WO2000008043A3 (fr) | 2000-05-11 |
WO2000008043A9 WO2000008043A9 (fr) | 2001-12-13 |
Family
ID=26789760
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US1999/017487 WO2000008043A2 (fr) | 1998-08-03 | 1999-08-03 | Prevention et traitement de pathologie virale |
Country Status (9)
Country | Link |
---|---|
EP (1) | EP1102788A4 (fr) |
JP (1) | JP2002522448A (fr) |
KR (1) | KR20010085326A (fr) |
CN (1) | CN1321165A (fr) |
AU (1) | AU5771599A (fr) |
BR (1) | BR9912732A (fr) |
CA (1) | CA2338983A1 (fr) |
IL (1) | IL141211A0 (fr) |
WO (1) | WO2000008043A2 (fr) |
Cited By (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP1130089A1 (fr) * | 2000-02-17 | 2001-09-05 | Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts | Composition comprenant des particules sous-virales cibles et fusions de virus enveloppés, et vaccin la contenant |
WO2002053587A2 (fr) * | 2001-01-05 | 2002-07-11 | Aventis Pasteur | Polypeptide induisant des anticorps neutralisant le vih |
FR2819256A1 (fr) * | 2001-01-05 | 2002-07-12 | Aventis Pasteur | Polypeptide induisant des anticorps neutralisant le vih |
EP1331942A2 (fr) * | 2000-09-22 | 2003-08-06 | Duke University | Immunogene contenant la proteine d'enveloppe du vih liee a un ligand |
US7033593B2 (en) | 2000-09-22 | 2006-04-25 | Duke University | Immunogen comprising an HIV envelope protein, a ligand and H2 peptide |
US7943375B2 (en) | 1998-12-31 | 2011-05-17 | Novartis Vaccines & Diagnostics, Inc | Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof |
US8133494B2 (en) | 2001-07-05 | 2012-03-13 | Novartis Vaccine & Diagnostics Inc | Expression cassettes endcoding HIV-1 south african subtype C modified ENV proteins with deletions in V1 and V2 |
US10912824B2 (en) | 2014-07-14 | 2021-02-09 | Ospedale San Raffaele S.R.L. | Vector production |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
FR2789590B1 (fr) * | 1999-02-15 | 2003-01-17 | Inst Rech Developpement Ird | Compositions immunogenes utilisables comme vaccins |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1993005147A1 (fr) * | 1991-08-30 | 1993-03-18 | The United States Of America, Represented By The Secretary, Department Of Health And Human Services | Particules defectives interferentes du vih avec un cd-env chimerique |
US5714316A (en) * | 1991-06-21 | 1998-02-03 | The Wistar Institute Of Anatomy & Biology | Chimeric envelope proteins for viral targeting |
-
1999
- 1999-08-03 WO PCT/US1999/017487 patent/WO2000008043A2/fr not_active Application Discontinuation
- 1999-08-03 CA CA002338983A patent/CA2338983A1/fr not_active Abandoned
- 1999-08-03 EP EP99945011A patent/EP1102788A4/fr not_active Withdrawn
- 1999-08-03 BR BR9912732-6A patent/BR9912732A/pt not_active IP Right Cessation
- 1999-08-03 IL IL14121199A patent/IL141211A0/xx unknown
- 1999-08-03 AU AU57715/99A patent/AU5771599A/en not_active Abandoned
- 1999-08-03 CN CN99811641A patent/CN1321165A/zh active Pending
- 1999-08-03 JP JP2000563676A patent/JP2002522448A/ja active Pending
- 1999-08-03 KR KR1020017001483A patent/KR20010085326A/ko not_active Application Discontinuation
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5714316A (en) * | 1991-06-21 | 1998-02-03 | The Wistar Institute Of Anatomy & Biology | Chimeric envelope proteins for viral targeting |
WO1993005147A1 (fr) * | 1991-08-30 | 1993-03-18 | The United States Of America, Represented By The Secretary, Department Of Health And Human Services | Particules defectives interferentes du vih avec un cd-env chimerique |
Non-Patent Citations (1)
Title |
---|
See also references of EP1102788A2 * |
Cited By (12)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US7943375B2 (en) | 1998-12-31 | 2011-05-17 | Novartis Vaccines & Diagnostics, Inc | Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof |
EP1130089A1 (fr) * | 2000-02-17 | 2001-09-05 | Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts | Composition comprenant des particules sous-virales cibles et fusions de virus enveloppés, et vaccin la contenant |
EP1331942A2 (fr) * | 2000-09-22 | 2003-08-06 | Duke University | Immunogene contenant la proteine d'enveloppe du vih liee a un ligand |
EP1331942A4 (fr) * | 2000-09-22 | 2005-03-16 | Univ Duke | Immunogene contenant la proteine d'enveloppe du vih liee a un ligand |
US7033593B2 (en) | 2000-09-22 | 2006-04-25 | Duke University | Immunogen comprising an HIV envelope protein, a ligand and H2 peptide |
US7070787B2 (en) | 2000-09-22 | 2006-07-04 | Duke University | Method of inducing the production of antibodies to HIV |
WO2002053587A2 (fr) * | 2001-01-05 | 2002-07-11 | Aventis Pasteur | Polypeptide induisant des anticorps neutralisant le vih |
FR2819256A1 (fr) * | 2001-01-05 | 2002-07-12 | Aventis Pasteur | Polypeptide induisant des anticorps neutralisant le vih |
WO2002053587A3 (fr) * | 2001-01-05 | 2003-09-25 | Aventis Pasteur | Polypeptide induisant des anticorps neutralisant le vih |
US8133494B2 (en) | 2001-07-05 | 2012-03-13 | Novartis Vaccine & Diagnostics Inc | Expression cassettes endcoding HIV-1 south african subtype C modified ENV proteins with deletions in V1 and V2 |
US10912824B2 (en) | 2014-07-14 | 2021-02-09 | Ospedale San Raffaele S.R.L. | Vector production |
US11957747B2 (en) | 2014-07-14 | 2024-04-16 | Ospedale San Raffaele S.R.L. | Vector production |
Also Published As
Publication number | Publication date |
---|---|
AU5771599A (en) | 2000-02-28 |
EP1102788A4 (fr) | 2002-08-28 |
JP2002522448A (ja) | 2002-07-23 |
WO2000008043A3 (fr) | 2000-05-11 |
KR20010085326A (ko) | 2001-09-07 |
EP1102788A2 (fr) | 2001-05-30 |
CN1321165A (zh) | 2001-11-07 |
CA2338983A1 (fr) | 2000-02-17 |
BR9912732A (pt) | 2001-11-27 |
IL141211A0 (en) | 2002-02-10 |
WO2000008043A9 (fr) | 2001-12-13 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AP1282A (en) | HIV envelope polypeptides and vaccine. | |
US7939083B2 (en) | Soluble, stabilized, proteolytically cleaved, trimeric HIV-1 gp140 proteins comprising modifications in the N-terminus of the gp41 ectodomain | |
KR100547049B1 (ko) | 바이러스 감염증 치료를 위한 조성물 및 방법 | |
NO324047B1 (no) | Hoyaffinitetshumane monoklonale antistoff spesifikke for RSV F-protein samt anvendelse derav | |
JP2000505299A (ja) | 合成hiv遺伝子 | |
JP2000516445A (ja) | 合成遺伝子を含むワクチン | |
JP2009511623A (ja) | レンチウイルスベクターベースのワクチン | |
JP2005503147A (ja) | ウイルスコートタンパク質/受容体キメラおよび使用方法 | |
NL8701950A (nl) | Monoclonale antilichamen en peptiden, geschikt voor de behandeling en diagnose van hiv infecties. | |
US6585979B1 (en) | HIV envelope polypeptides and immunogenic composition | |
WO2000008043A2 (fr) | Prevention et traitement de pathologie virale | |
Hunter et al. | Induction of mucosal and systemic antibody responses against the HIV coreceptor CCR5 upon intramuscular immunization and aerosol delivery of a virus-like particle based vaccine | |
WO1998041536A1 (fr) | Glycoproteines enveloppes de vih et vis a glycosylation deficiente | |
WO1998041536A9 (fr) | Glycoproteines enveloppes de vih et vis a glycosylation deficiente | |
JP2004121266A (ja) | コンビナトリアルポリペプチド抗原 | |
JP2003520786A (ja) | コドン最適化hiv−1polおよび修飾hiv−1polを発現するポリヌクレオチドワクチン | |
JP2004525604A (ja) | Hivアクセサリータンパク質をコードするdnaワクチン | |
US7537769B2 (en) | Webbed immunogens comprising recombinant human immunodeficiency virus (HIV) envelope glycoproteins and the M9 scorpion toxin | |
MXPA01001264A (en) | Prevention and treatment of viral disease | |
AU2004201321A1 (en) | Compositions and methods for treating viral infections | |
MXPA99003380A (en) | Compositions and methods for treating viral infections |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
WWE | Wipo information: entry into national phase |
Ref document number: 99811641.6 Country of ref document: CN |
|
AK | Designated states |
Kind code of ref document: A2 Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A2 Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
AK | Designated states |
Kind code of ref document: A3 Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A3 Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG |
|
DFPE | Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101) | ||
ENP | Entry into the national phase |
Ref document number: 2338983 Country of ref document: CA Ref document number: 2338983 Country of ref document: CA Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 141211 Country of ref document: IL |
|
WWE | Wipo information: entry into national phase |
Ref document number: PA/a/2001/001264 Country of ref document: MX |
|
WWE | Wipo information: entry into national phase |
Ref document number: 1020017001483 Country of ref document: KR |
|
WWE | Wipo information: entry into national phase |
Ref document number: 57715/99 Country of ref document: AU |
|
WWE | Wipo information: entry into national phase |
Ref document number: 1999945011 Country of ref document: EP |
|
WWP | Wipo information: published in national office |
Ref document number: 1999945011 Country of ref document: EP |
|
REG | Reference to national code |
Ref country code: DE Ref legal event code: 8642 |
|
WWP | Wipo information: published in national office |
Ref document number: 1020017001483 Country of ref document: KR |
|
AK | Designated states |
Kind code of ref document: C2 Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: C2 Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG |
|
COP | Corrected version of pamphlet |
Free format text: PAGES 1-92, DESCRIPTION, REPLACED BY NEW PAGES 1-92; PAGES 93-104, CLAIMS, REPLACED BY NEW PAGES 93-104; PAGES 1/12-12/12, DRAWINGS, REPLACED BY NEW PAGES 1/23-23/23; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE |
|
WWW | Wipo information: withdrawn in national office |
Ref document number: 1999945011 Country of ref document: EP |
|
WWW | Wipo information: withdrawn in national office |
Ref document number: 1020017001483 Country of ref document: KR |