WO2020243166A1 - Th1-polarizing adjuvants for enhancing immunogenicity of hiv antigens - Google Patents
Th1-polarizing adjuvants for enhancing immunogenicity of hiv antigens Download PDFInfo
- Publication number
- WO2020243166A1 WO2020243166A1 PCT/US2020/034688 US2020034688W WO2020243166A1 WO 2020243166 A1 WO2020243166 A1 WO 2020243166A1 US 2020034688 W US2020034688 W US 2020034688W WO 2020243166 A1 WO2020243166 A1 WO 2020243166A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- thl
- hiv
- cells
- vaccine
- adjuvant
- Prior art date
Links
- 239000002671 adjuvant Substances 0.000 title claims abstract description 191
- 230000002708 enhancing effect Effects 0.000 title claims abstract description 17
- 239000000427 antigen Substances 0.000 title abstract description 76
- 108091007433 antigens Proteins 0.000 title abstract description 76
- 102000036639 antigens Human genes 0.000 title abstract description 76
- 230000005847 immunogenicity Effects 0.000 title description 13
- 238000000034 method Methods 0.000 claims abstract description 98
- 239000000203 mixture Substances 0.000 claims abstract description 74
- 230000001965 increasing effect Effects 0.000 claims abstract description 46
- 238000002255 vaccination Methods 0.000 claims abstract description 44
- 108090000623 proteins and genes Proteins 0.000 claims description 256
- 102000004169 proteins and genes Human genes 0.000 claims description 228
- 241000725303 Human immunodeficiency virus Species 0.000 claims description 194
- 210000004027 cell Anatomy 0.000 claims description 164
- 210000001280 germinal center Anatomy 0.000 claims description 120
- 230000004044 response Effects 0.000 claims description 117
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 98
- 210000002966 serum Anatomy 0.000 claims description 90
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 81
- 102000040430 polynucleotide Human genes 0.000 claims description 75
- 108091033319 polynucleotide Proteins 0.000 claims description 75
- 239000002157 polynucleotide Substances 0.000 claims description 75
- 229940021995 DNA vaccine Drugs 0.000 claims description 74
- 108010041986 DNA Vaccines Proteins 0.000 claims description 73
- 229920001184 polypeptide Polymers 0.000 claims description 73
- 241000713772 Human immunodeficiency virus 1 Species 0.000 claims description 52
- 238000003556 assay Methods 0.000 claims description 49
- 230000005875 antibody response Effects 0.000 claims description 46
- 230000000694 effects Effects 0.000 claims description 41
- 239000008194 pharmaceutical composition Substances 0.000 claims description 38
- 229940035032 monophosphoryl lipid a Drugs 0.000 claims description 34
- 239000013598 vector Substances 0.000 claims description 31
- 210000004369 blood Anatomy 0.000 claims description 30
- 239000008280 blood Substances 0.000 claims description 30
- 238000006386 neutralization reaction Methods 0.000 claims description 30
- 239000002502 liposome Substances 0.000 claims description 27
- 210000004877 mucosa Anatomy 0.000 claims description 25
- 230000001419 dependent effect Effects 0.000 claims description 21
- 238000009472 formulation Methods 0.000 claims description 21
- 150000002632 lipids Chemical class 0.000 claims description 20
- 210000004180 plasmocyte Anatomy 0.000 claims description 19
- 102000014150 Interferons Human genes 0.000 claims description 17
- 108010050904 Interferons Proteins 0.000 claims description 17
- 239000003937 drug carrier Substances 0.000 claims description 17
- 238000004520 electroporation Methods 0.000 claims description 16
- 229940079322 interferon Drugs 0.000 claims description 16
- 206010057249 Phagocytosis Diseases 0.000 claims description 15
- 230000008782 phagocytosis Effects 0.000 claims description 15
- 101710149951 Protein Tat Proteins 0.000 claims description 14
- 108091005804 Peptidases Proteins 0.000 claims description 11
- 239000004365 Protease Substances 0.000 claims description 11
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 claims description 11
- 230000002093 peripheral effect Effects 0.000 claims description 7
- 230000007541 cellular toxicity Effects 0.000 claims description 4
- 230000037317 transdermal delivery Effects 0.000 claims description 3
- 102100034343 Integrase Human genes 0.000 claims 2
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 claims 2
- 210000001744 T-lymphocyte Anatomy 0.000 abstract description 72
- 230000028993 immune response Effects 0.000 abstract description 31
- 230000010287 polarization Effects 0.000 abstract description 28
- 230000008105 immune reaction Effects 0.000 abstract 1
- 229960005486 vaccine Drugs 0.000 description 231
- 108020004414 DNA Proteins 0.000 description 112
- 102000053602 DNA Human genes 0.000 description 112
- 230000003053 immunization Effects 0.000 description 77
- 238000002649 immunization Methods 0.000 description 76
- 241001465754 Metazoa Species 0.000 description 67
- 101000916050 Homo sapiens C-X-C chemokine receptor type 3 Proteins 0.000 description 65
- 102100028990 C-X-C chemokine receptor type 3 Human genes 0.000 description 64
- 230000014509 gene expression Effects 0.000 description 62
- 230000006698 induction Effects 0.000 description 62
- 210000003719 b-lymphocyte Anatomy 0.000 description 43
- 210000001165 lymph node Anatomy 0.000 description 39
- 238000013459 approach Methods 0.000 description 38
- 102100031658 C-X-C chemokine receptor type 5 Human genes 0.000 description 36
- 101000922405 Homo sapiens C-X-C chemokine receptor type 5 Proteins 0.000 description 36
- 102100025248 C-X-C motif chemokine 10 Human genes 0.000 description 34
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 33
- 230000027455 binding Effects 0.000 description 33
- 102000004127 Cytokines Human genes 0.000 description 29
- 108090000695 Cytokines Proteins 0.000 description 29
- 241000700605 Viruses Species 0.000 description 29
- 239000013612 plasmid Substances 0.000 description 29
- 230000000638 stimulation Effects 0.000 description 28
- 229940033330 HIV vaccine Drugs 0.000 description 27
- 208000015181 infectious disease Diseases 0.000 description 27
- 101710098275 C-X-C motif chemokine 10 Proteins 0.000 description 26
- 241000282560 Macaca mulatta Species 0.000 description 25
- -1 4-Hydroxy-3-nitrophenylacetyl Chemical group 0.000 description 24
- 238000002965 ELISA Methods 0.000 description 24
- 241000580858 Simian-Human immunodeficiency virus Species 0.000 description 23
- 241000713311 Simian immunodeficiency virus Species 0.000 description 22
- 230000036039 immunity Effects 0.000 description 22
- 150000007523 nucleic acids Chemical group 0.000 description 22
- 102100039341 Atrial natriuretic peptide receptor 2 Human genes 0.000 description 21
- 101710102159 Atrial natriuretic peptide receptor 2 Proteins 0.000 description 21
- 230000000770 proinflammatory effect Effects 0.000 description 21
- 201000010099 disease Diseases 0.000 description 20
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 20
- 230000003325 follicular Effects 0.000 description 20
- 238000004458 analytical method Methods 0.000 description 19
- 239000012636 effector Substances 0.000 description 19
- 210000002074 inflammatory monocyte Anatomy 0.000 description 19
- 102100023990 60S ribosomal protein L17 Human genes 0.000 description 18
- 208000031886 HIV Infections Diseases 0.000 description 18
- 239000003550 marker Substances 0.000 description 18
- 230000037452 priming Effects 0.000 description 18
- 239000000523 sample Substances 0.000 description 18
- 239000003795 chemical substances by application Substances 0.000 description 17
- 230000004727 humoral immunity Effects 0.000 description 17
- 230000028327 secretion Effects 0.000 description 17
- 230000009885 systemic effect Effects 0.000 description 17
- 238000011225 antiretroviral therapy Methods 0.000 description 16
- 230000002163 immunogen Effects 0.000 description 16
- 238000004519 manufacturing process Methods 0.000 description 16
- 229940037003 alum Drugs 0.000 description 15
- 230000004069 differentiation Effects 0.000 description 15
- 210000002443 helper t lymphocyte Anatomy 0.000 description 15
- 230000003472 neutralizing effect Effects 0.000 description 15
- 208000030507 AIDS Diseases 0.000 description 14
- 108010048209 Human Immunodeficiency Virus Proteins Proteins 0.000 description 14
- 101100381525 Mus musculus Bcl6 gene Proteins 0.000 description 13
- 102000019034 Chemokines Human genes 0.000 description 12
- 108010012236 Chemokines Proteins 0.000 description 12
- 208000037357 HIV infectious disease Diseases 0.000 description 12
- 241000282412 Homo Species 0.000 description 12
- 108090001005 Interleukin-6 Proteins 0.000 description 12
- 102000004889 Interleukin-6 Human genes 0.000 description 12
- 150000001875 compounds Chemical class 0.000 description 12
- 239000003814 drug Substances 0.000 description 12
- 108010078428 env Gene Products Proteins 0.000 description 12
- 208000033519 human immunodeficiency virus infectious disease Diseases 0.000 description 12
- 239000007924 injection Substances 0.000 description 12
- 238000002347 injection Methods 0.000 description 12
- 102000039446 nucleic acids Human genes 0.000 description 12
- 108020004707 nucleic acids Proteins 0.000 description 12
- 230000003612 virological effect Effects 0.000 description 12
- 229940033332 HIV-1 vaccine Drugs 0.000 description 11
- 101000858088 Homo sapiens C-X-C motif chemokine 10 Proteins 0.000 description 11
- 102100034349 Integrase Human genes 0.000 description 11
- 102100034353 Integrase Human genes 0.000 description 11
- 108010074328 Interferon-gamma Proteins 0.000 description 11
- 102000008070 Interferon-gamma Human genes 0.000 description 11
- 230000024245 cell differentiation Effects 0.000 description 11
- 238000013461 design Methods 0.000 description 11
- 239000012634 fragment Substances 0.000 description 11
- 230000006870 function Effects 0.000 description 11
- 230000001939 inductive effect Effects 0.000 description 11
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 11
- 230000001225 therapeutic effect Effects 0.000 description 11
- 230000001052 transient effect Effects 0.000 description 11
- 102000015696 Interleukins Human genes 0.000 description 10
- 108010063738 Interleukins Proteins 0.000 description 10
- 108091028043 Nucleic acid sequence Proteins 0.000 description 10
- 230000005867 T cell response Effects 0.000 description 10
- 239000011324 bead Substances 0.000 description 10
- 238000010790 dilution Methods 0.000 description 10
- 239000012895 dilution Substances 0.000 description 10
- 238000000684 flow cytometry Methods 0.000 description 10
- 229960003130 interferon gamma Drugs 0.000 description 10
- 230000003834 intracellular effect Effects 0.000 description 10
- 238000007837 multiplex assay Methods 0.000 description 10
- 230000002265 prevention Effects 0.000 description 10
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 10
- CITHEXJVPOWHKC-UUWRZZSWSA-N 1,2-di-O-myristoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCCCCCCC CITHEXJVPOWHKC-UUWRZZSWSA-N 0.000 description 9
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 description 9
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 description 9
- 102100025074 C-C chemokine receptor-like 2 Human genes 0.000 description 9
- 108010029697 CD40 Ligand Proteins 0.000 description 9
- 102100032937 CD40 ligand Human genes 0.000 description 9
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 description 9
- 101000716068 Homo sapiens C-C chemokine receptor type 6 Proteins 0.000 description 9
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 9
- 230000008901 benefit Effects 0.000 description 9
- 238000011161 development Methods 0.000 description 9
- 230000018109 developmental process Effects 0.000 description 9
- 229960003724 dimyristoylphosphatidylcholine Drugs 0.000 description 9
- 229960005160 dimyristoylphosphatidylglycerol Drugs 0.000 description 9
- BPHQZTVXXXJVHI-AJQTZOPKSA-N ditetradecanoyl phosphatidylglycerol Chemical compound CCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@@H](O)CO)OC(=O)CCCCCCCCCCCCC BPHQZTVXXXJVHI-AJQTZOPKSA-N 0.000 description 9
- 230000001404 mediated effect Effects 0.000 description 9
- 239000012071 phase Substances 0.000 description 9
- 239000002953 phosphate buffered saline Substances 0.000 description 9
- 230000001681 protective effect Effects 0.000 description 9
- 230000002103 transcriptional effect Effects 0.000 description 9
- 102100030703 Interleukin-22 Human genes 0.000 description 8
- 230000004913 activation Effects 0.000 description 8
- 210000000612 antigen-presenting cell Anatomy 0.000 description 8
- 239000002552 dosage form Substances 0.000 description 8
- 108010074108 interleukin-21 Proteins 0.000 description 8
- 239000003446 ligand Substances 0.000 description 8
- 239000002773 nucleotide Substances 0.000 description 8
- 125000003729 nucleotide group Chemical group 0.000 description 8
- 238000003752 polymerase chain reaction Methods 0.000 description 8
- 230000001105 regulatory effect Effects 0.000 description 8
- VGTPCRGMBIAPIM-UHFFFAOYSA-M sodium thiocyanate Chemical compound [Na+].[S-]C#N VGTPCRGMBIAPIM-UHFFFAOYSA-M 0.000 description 8
- 208000024891 symptom Diseases 0.000 description 8
- 238000013518 transcription Methods 0.000 description 8
- 230000035897 transcription Effects 0.000 description 8
- 101710149863 C-C chemokine receptor type 4 Proteins 0.000 description 7
- 102100032976 CCR4-NOT transcription complex subunit 6 Human genes 0.000 description 7
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 7
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 7
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 7
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 7
- 101000713602 Homo sapiens T-box transcription factor TBX21 Proteins 0.000 description 7
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 7
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 7
- 241000282553 Macaca Species 0.000 description 7
- 101800001690 Transmembrane protein gp41 Proteins 0.000 description 7
- 239000000872 buffer Substances 0.000 description 7
- 230000036755 cellular response Effects 0.000 description 7
- 230000002596 correlated effect Effects 0.000 description 7
- 230000000875 corresponding effect Effects 0.000 description 7
- 238000001727 in vivo Methods 0.000 description 7
- 230000028709 inflammatory response Effects 0.000 description 7
- 230000015788 innate immune response Effects 0.000 description 7
- 230000003389 potentiating effect Effects 0.000 description 7
- 230000000069 prophylactic effect Effects 0.000 description 7
- 238000012360 testing method Methods 0.000 description 7
- 108020004684 Internal Ribosome Entry Sites Proteins 0.000 description 6
- 238000000585 Mann–Whitney U test Methods 0.000 description 6
- 241000714177 Murine leukemia virus Species 0.000 description 6
- 102100036840 T-box transcription factor TBX21 Human genes 0.000 description 6
- 150000001413 amino acids Chemical class 0.000 description 6
- 238000001574 biopsy Methods 0.000 description 6
- 230000001413 cellular effect Effects 0.000 description 6
- 230000008859 change Effects 0.000 description 6
- 239000003153 chemical reaction reagent Substances 0.000 description 6
- 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 6
- 210000004392 genitalia Anatomy 0.000 description 6
- 230000001900 immune effect Effects 0.000 description 6
- 230000007774 longterm Effects 0.000 description 6
- 230000004048 modification Effects 0.000 description 6
- 238000012986 modification Methods 0.000 description 6
- 210000001616 monocyte Anatomy 0.000 description 6
- 244000052769 pathogen Species 0.000 description 6
- 230000002688 persistence Effects 0.000 description 6
- 229920000642 polymer Polymers 0.000 description 6
- 239000001509 sodium citrate Substances 0.000 description 6
- NLJMYIDDQXHKNR-UHFFFAOYSA-K sodium citrate Chemical compound O.O.[Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O NLJMYIDDQXHKNR-UHFFFAOYSA-K 0.000 description 6
- YYGNTYWPHWGJRM-AAJYLUCBSA-N squalene Chemical class CC(C)=CCC\C(C)=C\CC\C(C)=C\CC\C=C(/C)CC\C=C(/C)CCC=C(C)C YYGNTYWPHWGJRM-AAJYLUCBSA-N 0.000 description 6
- 238000010186 staining Methods 0.000 description 6
- 230000004936 stimulating effect Effects 0.000 description 6
- 239000006228 supernatant Substances 0.000 description 6
- 229940023859 AIDSVAX Drugs 0.000 description 5
- 208000035473 Communicable disease Diseases 0.000 description 5
- 108010000227 GP 140 Proteins 0.000 description 5
- 101000611023 Homo sapiens Tumor necrosis factor receptor superfamily member 6 Proteins 0.000 description 5
- 101710102916 Ichor Proteins 0.000 description 5
- 108010002350 Interleukin-2 Proteins 0.000 description 5
- 102000000588 Interleukin-2 Human genes 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- 241000288906 Primates Species 0.000 description 5
- 102100040403 Tumor necrosis factor receptor superfamily member 6 Human genes 0.000 description 5
- 206010046865 Vaccinia virus infection Diseases 0.000 description 5
- 230000001154 acute effect Effects 0.000 description 5
- 230000003115 biocidal effect Effects 0.000 description 5
- 229940023860 canarypox virus HIV vaccine Drugs 0.000 description 5
- 230000022131 cell cycle Effects 0.000 description 5
- 230000008614 cellular interaction Effects 0.000 description 5
- 238000012512 characterization method Methods 0.000 description 5
- 230000004186 co-expression Effects 0.000 description 5
- 239000002299 complementary DNA Substances 0.000 description 5
- 238000011156 evaluation Methods 0.000 description 5
- 230000008348 humoral response Effects 0.000 description 5
- 230000001976 improved effect Effects 0.000 description 5
- 238000007918 intramuscular administration Methods 0.000 description 5
- 230000000670 limiting effect Effects 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 210000001806 memory b lymphocyte Anatomy 0.000 description 5
- 108020004999 messenger RNA Proteins 0.000 description 5
- 238000002360 preparation method Methods 0.000 description 5
- 239000000047 product Substances 0.000 description 5
- 229920002477 rna polymer Polymers 0.000 description 5
- 230000004083 survival effect Effects 0.000 description 5
- 230000002459 sustained effect Effects 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 238000011282 treatment Methods 0.000 description 5
- 208000007089 vaccinia Diseases 0.000 description 5
- 102100025279 C-X-C motif chemokine 11 Human genes 0.000 description 4
- 102100036170 C-X-C motif chemokine 9 Human genes 0.000 description 4
- 241000282693 Cercopithecidae Species 0.000 description 4
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 4
- 108091026890 Coding region Proteins 0.000 description 4
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 4
- 108010065805 Interleukin-12 Proteins 0.000 description 4
- 102000013462 Interleukin-12 Human genes 0.000 description 4
- 102000013691 Interleukin-17 Human genes 0.000 description 4
- 108050003558 Interleukin-17 Proteins 0.000 description 4
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 4
- 238000003559 RNA-seq method Methods 0.000 description 4
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 4
- 208000036142 Viral infection Diseases 0.000 description 4
- 238000009825 accumulation Methods 0.000 description 4
- 230000002411 adverse Effects 0.000 description 4
- 230000032683 aging Effects 0.000 description 4
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 4
- 230000005540 biological transmission Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 230000000295 complement effect Effects 0.000 description 4
- 238000010276 construction Methods 0.000 description 4
- 210000004443 dendritic cell Anatomy 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 238000010494 dissociation reaction Methods 0.000 description 4
- 230000005593 dissociations Effects 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 102000046438 human CXCL10 Human genes 0.000 description 4
- 230000028996 humoral immune response Effects 0.000 description 4
- 230000002757 inflammatory effect Effects 0.000 description 4
- 230000003993 interaction Effects 0.000 description 4
- 230000017306 interleukin-6 production Effects 0.000 description 4
- 239000002245 particle Substances 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 239000011780 sodium chloride Substances 0.000 description 4
- 238000007619 statistical method Methods 0.000 description 4
- 238000003860 storage Methods 0.000 description 4
- 238000007920 subcutaneous administration Methods 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 102000003390 tumor necrosis factor Human genes 0.000 description 4
- 210000000689 upper leg Anatomy 0.000 description 4
- 230000009385 viral infection Effects 0.000 description 4
- 239000013603 viral vector Substances 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 3
- 229940124718 AIDS vaccine Drugs 0.000 description 3
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 3
- 241000283707 Capra Species 0.000 description 3
- 108020004705 Codon Proteins 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 208000007212 Foot-and-Mouth Disease Diseases 0.000 description 3
- 241000710198 Foot-and-mouth disease virus Species 0.000 description 3
- 101710177291 Gag polyprotein Proteins 0.000 description 3
- 241000560067 HIV-1 group M Species 0.000 description 3
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 3
- 241000713340 Human immunodeficiency virus 2 Species 0.000 description 3
- 206010061218 Inflammation Diseases 0.000 description 3
- 108091092195 Intron Proteins 0.000 description 3
- 101710125418 Major capsid protein Proteins 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 3
- 241000699670 Mus sp. Species 0.000 description 3
- 108091034117 Oligonucleotide Proteins 0.000 description 3
- 229920001213 Polysorbate 20 Polymers 0.000 description 3
- 102100040247 Tumor necrosis factor Human genes 0.000 description 3
- 206010058874 Viraemia Diseases 0.000 description 3
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 3
- 238000002835 absorbance Methods 0.000 description 3
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 3
- 229910052782 aluminium Inorganic materials 0.000 description 3
- 229940024545 aluminum hydroxide Drugs 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 210000000628 antibody-producing cell Anatomy 0.000 description 3
- 230000004888 barrier function Effects 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 210000001185 bone marrow Anatomy 0.000 description 3
- 125000002091 cationic group Chemical group 0.000 description 3
- 230000007969 cellular immunity Effects 0.000 description 3
- 210000003756 cervix mucus Anatomy 0.000 description 3
- 235000012000 cholesterol Nutrition 0.000 description 3
- 125000004122 cyclic group Chemical group 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 238000006073 displacement reaction Methods 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 239000003623 enhancer Substances 0.000 description 3
- 239000000796 flavoring agent Substances 0.000 description 3
- 238000002825 functional assay Methods 0.000 description 3
- 230000004927 fusion Effects 0.000 description 3
- 230000016784 immunoglobulin production Effects 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 238000011835 investigation Methods 0.000 description 3
- 238000012933 kinetic analysis Methods 0.000 description 3
- 210000002809 long lived plasma cell Anatomy 0.000 description 3
- 230000005923 long-lasting effect Effects 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 238000013507 mapping Methods 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 210000000822 natural killer cell Anatomy 0.000 description 3
- 230000001717 pathogenic effect Effects 0.000 description 3
- 239000008188 pellet Substances 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 3
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 238000011321 prophylaxis Methods 0.000 description 3
- 230000010076 replication Effects 0.000 description 3
- 210000005000 reproductive tract Anatomy 0.000 description 3
- 238000005070 sampling Methods 0.000 description 3
- 238000012174 single-cell RNA sequencing Methods 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 238000005406 washing Methods 0.000 description 3
- 230000003442 weekly effect Effects 0.000 description 3
- YYGNTYWPHWGJRM-UHFFFAOYSA-N (6E,10E,14E,18E)-2,6,10,15,19,23-hexamethyltetracosa-2,6,10,14,18,22-hexaene Chemical compound CC(C)=CCCC(C)=CCCC(C)=CCCC=C(C)CCC=C(C)CCC=C(C)C YYGNTYWPHWGJRM-UHFFFAOYSA-N 0.000 description 2
- FPQQSJJWHUJYPU-UHFFFAOYSA-N 3-(dimethylamino)propyliminomethylidene-ethylazanium;chloride Chemical compound Cl.CCN=C=NCCCN(C)C FPQQSJJWHUJYPU-UHFFFAOYSA-N 0.000 description 2
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 2
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 108090001008 Avidin Proteins 0.000 description 2
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- 101710117545 C protein Proteins 0.000 description 2
- 101710098272 C-X-C motif chemokine 11 Proteins 0.000 description 2
- 101710085500 C-X-C motif chemokine 9 Proteins 0.000 description 2
- 101150013553 CD40 gene Proteins 0.000 description 2
- 101150004010 CXCR3 gene Proteins 0.000 description 2
- BQENDLAVTKRQMS-SBBGFIFASA-L Carbenoxolone sodium Chemical compound [Na+].[Na+].C([C@H]1C2=CC(=O)[C@H]34)[C@@](C)(C([O-])=O)CC[C@]1(C)CC[C@@]2(C)[C@]4(C)CC[C@@H]1[C@]3(C)CC[C@H](OC(=O)CCC([O-])=O)C1(C)C BQENDLAVTKRQMS-SBBGFIFASA-L 0.000 description 2
- 108010008978 Chemokine CXCL10 Proteins 0.000 description 2
- 102000009410 Chemokine receptor Human genes 0.000 description 2
- 108050000299 Chemokine receptor Proteins 0.000 description 2
- 241000606161 Chlamydia Species 0.000 description 2
- 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 2
- 101800001467 Envelope glycoprotein E2 Proteins 0.000 description 2
- 101710091045 Envelope protein Proteins 0.000 description 2
- 108010087819 Fc receptors Proteins 0.000 description 2
- 102000009109 Fc receptors Human genes 0.000 description 2
- 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 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- NYHBQMYGNKIUIF-UUOKFMHZSA-N Guanosine Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O NYHBQMYGNKIUIF-UUOKFMHZSA-N 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- 102000006354 HLA-DR Antigens Human genes 0.000 description 2
- 108010058597 HLA-DR Antigens Proteins 0.000 description 2
- 102100026122 High affinity immunoglobulin gamma Fc receptor I Human genes 0.000 description 2
- 102100037907 High mobility group protein B1 Human genes 0.000 description 2
- 101000858060 Homo sapiens C-X-C motif chemokine 11 Proteins 0.000 description 2
- 101000947172 Homo sapiens C-X-C motif chemokine 9 Proteins 0.000 description 2
- 101000913074 Homo sapiens High affinity immunoglobulin gamma Fc receptor I Proteins 0.000 description 2
- 101001025337 Homo sapiens High mobility group protein B1 Proteins 0.000 description 2
- 101000959820 Homo sapiens Interferon alpha-1/13 Proteins 0.000 description 2
- 101001033249 Homo sapiens Interleukin-1 beta Proteins 0.000 description 2
- 101000917826 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-a Proteins 0.000 description 2
- 101000917824 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-b Proteins 0.000 description 2
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 2
- 102100039065 Interleukin-1 beta Human genes 0.000 description 2
- 108090000978 Interleukin-4 Proteins 0.000 description 2
- 108090001007 Interleukin-8 Proteins 0.000 description 2
- 102100029204 Low affinity immunoglobulin gamma Fc region receptor II-a Human genes 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 108010009474 Macrophage Inflammatory Proteins Proteins 0.000 description 2
- 102000009571 Macrophage Inflammatory Proteins Human genes 0.000 description 2
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 108091005461 Nucleic proteins Proteins 0.000 description 2
- 108010058846 Ovalbumin Proteins 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 2
- 101710188315 Protein X Proteins 0.000 description 2
- 241001454523 Quillaja saponaria Species 0.000 description 2
- 235000009001 Quillaja saponaria Nutrition 0.000 description 2
- 102100022502 Receptor-interacting serine/threonine-protein kinase 2 Human genes 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 108091027544 Subgenomic mRNA Proteins 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- 101800001271 Surface protein Proteins 0.000 description 2
- 230000037453 T cell priming Effects 0.000 description 2
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 2
- BHEOSNUKNHRBNM-UHFFFAOYSA-N Tetramethylsqualene Natural products CC(=C)C(C)CCC(=C)C(C)CCC(C)=CCCC=C(C)CCC(C)C(=C)CCC(C)C(C)=C BHEOSNUKNHRBNM-UHFFFAOYSA-N 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 102000009618 Transforming Growth Factors Human genes 0.000 description 2
- 108010009583 Transforming Growth Factors Proteins 0.000 description 2
- 102000004142 Trypsin Human genes 0.000 description 2
- 108090000631 Trypsin Proteins 0.000 description 2
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 2
- 108020005202 Viral DNA Proteins 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 230000036436 anti-hiv Effects 0.000 description 2
- 230000000845 anti-microbial effect Effects 0.000 description 2
- 230000000840 anti-viral effect Effects 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 230000003190 augmentative effect Effects 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 239000000090 biomarker Substances 0.000 description 2
- 238000009395 breeding Methods 0.000 description 2
- 230000001488 breeding effect Effects 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 230000003915 cell function Effects 0.000 description 2
- 239000002738 chelating agent Substances 0.000 description 2
- 239000002975 chemoattractant Substances 0.000 description 2
- 230000003399 chemotactic effect Effects 0.000 description 2
- 238000000546 chi-square test Methods 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 229960001701 chloroform Drugs 0.000 description 2
- 239000003086 colorant Substances 0.000 description 2
- 230000001461 cytolytic effect Effects 0.000 description 2
- 238000007822 cytometric assay Methods 0.000 description 2
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 2
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 230000003111 delayed effect Effects 0.000 description 2
- 238000000151 deposition Methods 0.000 description 2
- 235000005911 diet Nutrition 0.000 description 2
- OGQYPPBGSLZBEG-UHFFFAOYSA-N dimethyl(dioctadecyl)azanium Chemical compound CCCCCCCCCCCCCCCCCC[N+](C)(C)CCCCCCCCCCCCCCCCCC OGQYPPBGSLZBEG-UHFFFAOYSA-N 0.000 description 2
- PRAKJMSDJKAYCZ-UHFFFAOYSA-N dodecahydrosqualene Natural products CC(C)CCCC(C)CCCC(C)CCCCC(C)CCCC(C)CCCC(C)C PRAKJMSDJKAYCZ-UHFFFAOYSA-N 0.000 description 2
- 239000013604 expression vector Substances 0.000 description 2
- 210000005002 female reproductive tract Anatomy 0.000 description 2
- 238000001914 filtration Methods 0.000 description 2
- 235000019634 flavors Nutrition 0.000 description 2
- 235000003599 food sweetener Nutrition 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 210000001102 germinal center b cell Anatomy 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- 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 2
- 230000036541 health Effects 0.000 description 2
- 230000003054 hormonal effect Effects 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000036737 immune function Effects 0.000 description 2
- 210000004201 immune sera Anatomy 0.000 description 2
- 229940042743 immune sera Drugs 0.000 description 2
- 229940088592 immunologic factor Drugs 0.000 description 2
- 239000000367 immunologic factor Substances 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 239000000411 inducer Substances 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 229940047122 interleukins Drugs 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 230000002147 killing effect Effects 0.000 description 2
- 238000001325 log-rank test Methods 0.000 description 2
- 230000014759 maintenance of location Effects 0.000 description 2
- 201000004792 malaria Diseases 0.000 description 2
- 230000035800 maturation Effects 0.000 description 2
- 229960002985 medroxyprogesterone acetate Drugs 0.000 description 2
- PSGAAPLEWMOORI-PEINSRQWSA-N medroxyprogesterone acetate Chemical compound C([C@@]12C)CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2CC[C@]2(C)[C@@](OC(C)=O)(C(C)=O)CC[C@H]21 PSGAAPLEWMOORI-PEINSRQWSA-N 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 230000003278 mimic effect Effects 0.000 description 2
- 238000001823 molecular biology technique Methods 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 150000003904 phospholipids Chemical class 0.000 description 2
- 210000003720 plasmablast Anatomy 0.000 description 2
- 230000002516 postimmunization Effects 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 229940023867 prime-boost vaccine Drugs 0.000 description 2
- 238000000513 principal component analysis Methods 0.000 description 2
- 230000001566 pro-viral effect Effects 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 238000012207 quantitative assay Methods 0.000 description 2
- 210000000664 rectum Anatomy 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 238000012552 review Methods 0.000 description 2
- 229930182490 saponin Natural products 0.000 description 2
- 235000017709 saponins Nutrition 0.000 description 2
- 229920006395 saturated elastomer Polymers 0.000 description 2
- 210000005212 secondary lymphoid organ Anatomy 0.000 description 2
- 238000013207 serial dilution Methods 0.000 description 2
- 230000011664 signaling Effects 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 229940031439 squalene Drugs 0.000 description 2
- TUHBEKDERLKLEC-UHFFFAOYSA-N squalene Natural products CC(=CCCC(=CCCC(=CCCC=C(/C)CCC=C(/C)CC=C(C)C)C)C)C TUHBEKDERLKLEC-UHFFFAOYSA-N 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 230000008093 supporting effect Effects 0.000 description 2
- 239000000829 suppository Substances 0.000 description 2
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 239000003765 sweetening agent Substances 0.000 description 2
- 230000002195 synergetic effect Effects 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 101150050362 thl gene Proteins 0.000 description 2
- 230000005945 translocation Effects 0.000 description 2
- 230000001960 triggered effect Effects 0.000 description 2
- 239000012588 trypsin Substances 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 229940124931 vaccine adjuvant Drugs 0.000 description 2
- 239000012646 vaccine adjuvant Substances 0.000 description 2
- 230000029812 viral genome replication Effects 0.000 description 2
- OZFAFGSSMRRTDW-UHFFFAOYSA-N (2,4-dichlorophenyl) benzenesulfonate Chemical compound ClC1=CC(Cl)=CC=C1OS(=O)(=O)C1=CC=CC=C1 OZFAFGSSMRRTDW-UHFFFAOYSA-N 0.000 description 1
- HBZBAMXERPYTFS-SECBINFHSA-N (4S)-2-(6,7-dihydro-5H-pyrrolo[3,2-f][1,3]benzothiazol-2-yl)-4,5-dihydro-1,3-thiazole-4-carboxylic acid Chemical compound OC(=O)[C@H]1CSC(=N1)c1nc2cc3CCNc3cc2s1 HBZBAMXERPYTFS-SECBINFHSA-N 0.000 description 1
- WHTVZRBIWZFKQO-AWEZNQCLSA-N (S)-chloroquine Chemical compound ClC1=CC=C2C(N[C@@H](C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-AWEZNQCLSA-N 0.000 description 1
- 101150072531 10 gene Proteins 0.000 description 1
- VGONTNSXDCQUGY-RRKCRQDMSA-N 2'-deoxyinosine Chemical group C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC2=O)=C2N=C1 VGONTNSXDCQUGY-RRKCRQDMSA-N 0.000 description 1
- KSXTUUUQYQYKCR-LQDDAWAPSA-M 2,3-bis[[(z)-octadec-9-enoyl]oxy]propyl-trimethylazanium;chloride Chemical compound [Cl-].CCCCCCCC\C=C/CCCCCCCC(=O)OCC(C[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC KSXTUUUQYQYKCR-LQDDAWAPSA-M 0.000 description 1
- PMUNIMVZCACZBB-UHFFFAOYSA-N 2-hydroxyethylazanium;chloride Chemical compound Cl.NCCO PMUNIMVZCACZBB-UHFFFAOYSA-N 0.000 description 1
- UAIUNKRWKOVEES-UHFFFAOYSA-N 3,3',5,5'-tetramethylbenzidine Chemical compound CC1=C(N)C(C)=CC(C=2C=C(C)C(N)=C(C)C=2)=C1 UAIUNKRWKOVEES-UHFFFAOYSA-N 0.000 description 1
- HRPVXLWXLXDGHG-UHFFFAOYSA-N Acrylamide Chemical compound NC(=O)C=C HRPVXLWXLXDGHG-UHFFFAOYSA-N 0.000 description 1
- 102100033647 Activity-regulated cytoskeleton-associated protein Human genes 0.000 description 1
- 241000710929 Alphavirus Species 0.000 description 1
- 108091006061 AviTagged proteins Proteins 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 241001446316 Bohle iridovirus Species 0.000 description 1
- 239000004255 Butylated hydroxyanisole Substances 0.000 description 1
- 239000004322 Butylated hydroxytoluene Substances 0.000 description 1
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 1
- 102100036848 C-C motif chemokine 20 Human genes 0.000 description 1
- 102100032367 C-C motif chemokine 5 Human genes 0.000 description 1
- 108091028026 C-DNA Proteins 0.000 description 1
- 108010075254 C-Peptide Proteins 0.000 description 1
- 108050006947 CXC Chemokine Proteins 0.000 description 1
- 102000019388 CXC chemokine Human genes 0.000 description 1
- 101100113692 Caenorhabditis elegans clk-2 gene Proteins 0.000 description 1
- 241000178270 Canarypox virus Species 0.000 description 1
- 241000008374 Capirona Species 0.000 description 1
- 241000713756 Caprine arthritis encephalitis virus Species 0.000 description 1
- 102000006579 Chemokine CXCL10 Human genes 0.000 description 1
- 241000606153 Chlamydia trachomatis Species 0.000 description 1
- 206010061041 Chlamydial infection Diseases 0.000 description 1
- VYZAMTAEIAYCRO-UHFFFAOYSA-N Chromium Chemical compound [Cr] VYZAMTAEIAYCRO-UHFFFAOYSA-N 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- MIKUYHXYGGJMLM-GIMIYPNGSA-N Crotonoside Natural products C1=NC2=C(N)NC(=O)N=C2N1[C@H]1O[C@@H](CO)[C@H](O)[C@@H]1O MIKUYHXYGGJMLM-GIMIYPNGSA-N 0.000 description 1
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 1
- 108010036949 Cyclosporine Proteins 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- NYHBQMYGNKIUIF-UHFFFAOYSA-N D-guanosine Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(CO)C(O)C1O NYHBQMYGNKIUIF-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 238000011238 DNA vaccination Methods 0.000 description 1
- 101100522258 Danio rerio ptmaa gene Proteins 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 239000012591 Dulbecco’s Phosphate Buffered Saline Substances 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 238000011510 Elispot assay Methods 0.000 description 1
- 241000792859 Enema Species 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000713730 Equine infectious anemia virus Species 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 101710127949 Extracellular matrix protein 1 Proteins 0.000 description 1
- 102100031758 Extracellular matrix protein 1 Human genes 0.000 description 1
- 241000713800 Feline immunodeficiency virus Species 0.000 description 1
- 241000710831 Flavivirus Species 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 208000009329 Graft vs Host Disease Diseases 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 241000556773 HIV-1 group N Species 0.000 description 1
- 241000560056 HIV-1 group O Species 0.000 description 1
- 229920000209 Hexadimethrine bromide Polymers 0.000 description 1
- 101000903742 Homo sapiens Basic leucine zipper transcriptional factor ATF-like Proteins 0.000 description 1
- 101000713099 Homo sapiens C-C motif chemokine 20 Proteins 0.000 description 1
- 101000797762 Homo sapiens C-C motif chemokine 5 Proteins 0.000 description 1
- 101000994375 Homo sapiens Integrin alpha-4 Proteins 0.000 description 1
- 101001109137 Homo sapiens Receptor-interacting serine/threonine-protein kinase 2 Proteins 0.000 description 1
- 101000650820 Homo sapiens Semaphorin-4A Proteins 0.000 description 1
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 1
- 241001135301 Hypleurochilus fissicornis Species 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 102100032818 Integrin alpha-4 Human genes 0.000 description 1
- 108010047761 Interferon-alpha Proteins 0.000 description 1
- 102000006992 Interferon-alpha Human genes 0.000 description 1
- 102000000589 Interleukin-1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 101150053046 MYD88 gene Proteins 0.000 description 1
- 101000858090 Macaca mulatta C-X-C motif chemokine 10 Proteins 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 241000736262 Microbiota Species 0.000 description 1
- 241001183012 Modified Vaccinia Ankara virus Species 0.000 description 1
- 101710151833 Movement protein TGBp3 Proteins 0.000 description 1
- 241000187479 Mycobacterium tuberculosis Species 0.000 description 1
- 102100024134 Myeloid differentiation primary response protein MyD88 Human genes 0.000 description 1
- NQTADLQHYWFPDB-UHFFFAOYSA-N N-Hydroxysuccinimide Chemical compound ON1C(=O)CCC1=O NQTADLQHYWFPDB-UHFFFAOYSA-N 0.000 description 1
- 108091061960 Naked DNA Proteins 0.000 description 1
- 108700001237 Nucleic Acid-Based Vaccines Proteins 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 229930040373 Paraformaldehyde Natural products 0.000 description 1
- 108010033276 Peptide Fragments Proteins 0.000 description 1
- 102000007079 Peptide Fragments Human genes 0.000 description 1
- 208000037581 Persistent Infection Diseases 0.000 description 1
- 108010004729 Phycoerythrin Proteins 0.000 description 1
- 108010076039 Polyproteins Proteins 0.000 description 1
- YDQIRODFTJGGMP-UHFFFAOYSA-N Prosapogenin Chemical group C12CC(C)(C)CCC2(C(=O)OC2C(C(O)C(O)C(CO)O2)O)CCC(C2(CCC34)C)(CO)C1=CCC2C3(C)CC(O)C(O)C4(C)C(=O)OC1OC(CO)C(O)C(O)C1O YDQIRODFTJGGMP-UHFFFAOYSA-N 0.000 description 1
- 108010015499 Protein Kinase C-theta Proteins 0.000 description 1
- 102100021566 Protein kinase C theta type Human genes 0.000 description 1
- 241001112090 Pseudovirus Species 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- MQUFAARYGOUYEV-UWEXFCAOSA-N Quillaic acid Natural products CC1(C)CC[C@@]2([C@H](O)C[C@]3(C)C(=CC[C@H]4[C@@]5(C)CC[C@H](O)[C@](C)(C=O)[C@H]5CC[C@@]34C)[C@H]2C1)C(=O)O MQUFAARYGOUYEV-UWEXFCAOSA-N 0.000 description 1
- 239000013614 RNA sample Substances 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 108010079933 Receptor-Interacting Protein Serine-Threonine Kinase 2 Proteins 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 102000014105 Semaphorin Human genes 0.000 description 1
- 108050003978 Semaphorin Proteins 0.000 description 1
- 102100027718 Semaphorin-4A Human genes 0.000 description 1
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 1
- 108010060825 Toll-Like Receptor 7 Proteins 0.000 description 1
- 108010060752 Toll-Like Receptor 8 Proteins 0.000 description 1
- 102100039390 Toll-like receptor 7 Human genes 0.000 description 1
- 102100033110 Toll-like receptor 8 Human genes 0.000 description 1
- 102100023935 Transmembrane glycoprotein NMB Human genes 0.000 description 1
- 101710090322 Truncated surface protein Proteins 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- FHICGHSMIPIAPL-HDYAAECPSA-N [2-[3-[6-[3-[(5R,6aS,6bR,12aR)-10-[6-[2-[2-[4,5-dihydroxy-3-(3,4,5-trihydroxyoxan-2-yl)oxyoxan-2-yl]ethoxy]ethyl]-3,4,5-trihydroxyoxan-2-yl]oxy-5-hydroxy-2,2,6a,6b,9,9,12a-heptamethyl-1,3,4,5,6,6a,7,8,8a,10,11,12,13,14b-tetradecahydropicene-4a-carbonyl]peroxypropyl]-5-[[5-[8-[3,5-dihydroxy-4-(3,4,5-trihydroxyoxan-2-yl)oxyoxan-2-yl]octoxy]-3,4-dihydroxy-6-methyloxan-2-yl]methoxy]-3,4-dihydroxyoxan-2-yl]propoxymethyl]-5-hydroxy-3-[(6S)-6-hydroxy-2,6-dimethylocta-2,7-dienoyl]oxy-6-methyloxan-4-yl] (2E,6S)-6-hydroxy-2-(hydroxymethyl)-6-methylocta-2,7-dienoate Chemical compound C=C[C@@](C)(O)CCC=C(C)C(=O)OC1C(OC(=O)C(\CO)=C\CC[C@](C)(O)C=C)C(O)C(C)OC1COCCCC1C(O)C(O)C(OCC2C(C(O)C(OCCCCCCCCC3C(C(OC4C(C(O)C(O)CO4)O)C(O)CO3)O)C(C)O2)O)C(CCCOOC(=O)C23C(CC(C)(C)CC2)C=2[C@@]([C@]4(C)CCC5C(C)(C)C(OC6C(C(O)C(O)C(CCOCCC7C(C(O)C(O)CO7)OC7C(C(O)C(O)CO7)O)O6)O)CC[C@]5(C)C4CC=2)(C)C[C@H]3O)O1 FHICGHSMIPIAPL-HDYAAECPSA-N 0.000 description 1
- 239000003070 absorption delaying agent Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 125000002252 acyl group Chemical group 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 230000009824 affinity maturation Effects 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- AZDRQVAHHNSJOQ-UHFFFAOYSA-N alumane Chemical class [AlH3] AZDRQVAHHNSJOQ-UHFFFAOYSA-N 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 238000005571 anion exchange chromatography Methods 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 230000014102 antigen processing and presentation of exogenous peptide antigen via MHC class I Effects 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 230000003305 autocrine Effects 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 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 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000008236 biological pathway Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 238000003738 britelite plus Methods 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 235000019282 butylated hydroxyanisole Nutrition 0.000 description 1
- 229940043253 butylated hydroxyanisole Drugs 0.000 description 1
- CZBZUDVBLSSABA-UHFFFAOYSA-N butylated hydroxyanisole Chemical compound COC1=CC=C(O)C(C(C)(C)C)=C1.COC1=CC=C(O)C=C1C(C)(C)C CZBZUDVBLSSABA-UHFFFAOYSA-N 0.000 description 1
- 235000010354 butylated hydroxytoluene Nutrition 0.000 description 1
- 229940095259 butylated hydroxytoluene Drugs 0.000 description 1
- BPKIGYQJPYCAOW-FFJTTWKXSA-I calcium;potassium;disodium;(2s)-2-hydroxypropanoate;dichloride;dihydroxide;hydrate Chemical compound O.[OH-].[OH-].[Na+].[Na+].[Cl-].[Cl-].[K+].[Ca+2].C[C@H](O)C([O-])=O BPKIGYQJPYCAOW-FFJTTWKXSA-I 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 239000011545 carbonate/bicarbonate buffer Substances 0.000 description 1
- 239000006143 cell culture medium Substances 0.000 description 1
- 230000011712 cell development Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000019522 cellular metabolic process Effects 0.000 description 1
- 230000003196 chaotropic effect Effects 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- 229960003677 chloroquine Drugs 0.000 description 1
- WHTVZRBIWZFKQO-UHFFFAOYSA-N chloroquine Natural products ClC1=CC=C2C(NC(C)CCCN(CC)CC)=CC=NC2=C1 WHTVZRBIWZFKQO-UHFFFAOYSA-N 0.000 description 1
- 229960001265 ciclosporin Drugs 0.000 description 1
- 238000012411 cloning technique Methods 0.000 description 1
- 238000003501 co-culture Methods 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 238000010835 comparative analysis Methods 0.000 description 1
- 230000002153 concerted effect Effects 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 239000012059 conventional drug carrier Substances 0.000 description 1
- 238000010219 correlation analysis Methods 0.000 description 1
- 230000000139 costimulatory effect Effects 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 239000006071 cream Substances 0.000 description 1
- 229930182912 cyclosporin Natural products 0.000 description 1
- 229940104302 cytosine Drugs 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000009849 deactivation Effects 0.000 description 1
- 238000000432 density-gradient centrifugation Methods 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 230000000378 dietary effect Effects 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 239000012470 diluted sample Substances 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 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 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000007920 enema Substances 0.000 description 1
- 229940079360 enema for constipation Drugs 0.000 description 1
- 108700004025 env Genes Proteins 0.000 description 1
- 101150030339 env gene Proteins 0.000 description 1
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 238000013401 experimental design Methods 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 235000013861 fat-free Nutrition 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 102000037865 fusion proteins Human genes 0.000 description 1
- 108020001507 fusion proteins Proteins 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 238000010199 gene set enrichment analysis Methods 0.000 description 1
- 230000014101 glucose homeostasis Effects 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 208000024908 graft versus host disease Diseases 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 229940029575 guanosine Drugs 0.000 description 1
- 210000004837 gut-associated lymphoid tissue Anatomy 0.000 description 1
- 208000002672 hepatitis B Diseases 0.000 description 1
- 102000055771 human CXCR3 Human genes 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 210000000985 iga plasma cell Anatomy 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 230000005934 immune activation Effects 0.000 description 1
- 230000007124 immune defense Effects 0.000 description 1
- 230000008076 immune mechanism Effects 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 239000003547 immunosorbent Substances 0.000 description 1
- 230000003116 impacting effect Effects 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000001524 infective effect Effects 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 150000002484 inorganic compounds Chemical class 0.000 description 1
- 229910010272 inorganic material Inorganic materials 0.000 description 1
- 239000012212 insulator Substances 0.000 description 1
- 108010021315 integrin beta7 Proteins 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108040002014 interleukin-18 receptor activity proteins Proteins 0.000 description 1
- 102000008625 interleukin-18 receptor activity proteins Human genes 0.000 description 1
- 108040006852 interleukin-4 receptor activity proteins Proteins 0.000 description 1
- 229940100601 interleukin-6 Drugs 0.000 description 1
- 230000000968 intestinal effect Effects 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 238000007917 intracranial administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 230000002601 intratumoral effect Effects 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 230000002427 irreversible effect Effects 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000021633 leukocyte mediated immunity Effects 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 239000008297 liquid dosage form Substances 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 238000003468 luciferase reporter gene assay Methods 0.000 description 1
- 210000005004 lymphoid follicle Anatomy 0.000 description 1
- 210000003563 lymphoid tissue Anatomy 0.000 description 1
- 239000008176 lyophilized powder Substances 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 210000003071 memory t lymphocyte Anatomy 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 238000002493 microarray Methods 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 239000003094 microcapsule Substances 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 244000309715 mini pig Species 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 210000003097 mucus Anatomy 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 239000007908 nanoemulsion Substances 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 229940023146 nucleic acid vaccine Drugs 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 229940046166 oligodeoxynucleotide Drugs 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 150000002894 organic compounds Chemical class 0.000 description 1
- 229940092253 ovalbumin Drugs 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 230000027758 ovulation cycle Effects 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 230000000242 pagocytic effect Effects 0.000 description 1
- 210000002741 palatine tonsil Anatomy 0.000 description 1
- 229920002866 paraformaldehyde Polymers 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 102000013415 peroxidase activity proteins Human genes 0.000 description 1
- 108040007629 peroxidase activity proteins Proteins 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 229940124531 pharmaceutical excipient Drugs 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 230000037081 physical activity Effects 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 239000000902 placebo Substances 0.000 description 1
- 229940068196 placebo Drugs 0.000 description 1
- 210000005134 plasmacytoid dendritic cell Anatomy 0.000 description 1
- 239000013600 plasmid vector Substances 0.000 description 1
- 229940031960 pneumococcal polysaccharide vaccine Drugs 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 238000003825 pressing Methods 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 239000000583 progesterone congener Substances 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 229940023143 protein vaccine Drugs 0.000 description 1
- 101150054513 ptmA gene Proteins 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 238000000746 purification Methods 0.000 description 1
- 239000012673 purified plant extract Substances 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 239000001397 quillaja saponaria molina bark Substances 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 102000037983 regulatory factors Human genes 0.000 description 1
- 108091008025 regulatory factors Proteins 0.000 description 1
- 238000009877 rendering Methods 0.000 description 1
- 238000012827 research and development Methods 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 238000002390 rotary evaporation Methods 0.000 description 1
- 239000012146 running buffer Substances 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 239000012898 sample dilution Substances 0.000 description 1
- 150000007949 saponins Chemical class 0.000 description 1
- 238000005204 segregation Methods 0.000 description 1
- 230000001568 sexual effect Effects 0.000 description 1
- 238000007493 shaping process Methods 0.000 description 1
- 239000001632 sodium acetate Substances 0.000 description 1
- 235000017281 sodium acetate Nutrition 0.000 description 1
- HRZFUMHJMZEROT-UHFFFAOYSA-L sodium disulfite Chemical compound [Na+].[Na+].[O-]S(=O)S([O-])(=O)=O HRZFUMHJMZEROT-UHFFFAOYSA-L 0.000 description 1
- 229940001584 sodium metabisulfite Drugs 0.000 description 1
- 235000010262 sodium metabisulphite Nutrition 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- CCEKAJIANROZEO-UHFFFAOYSA-N sulfluramid Chemical group CCNS(=O)(=O)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)F CCEKAJIANROZEO-UHFFFAOYSA-N 0.000 description 1
- 235000011149 sulphuric acid Nutrition 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 230000001360 synchronised effect Effects 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- 150000004044 tetrasaccharides Chemical class 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 230000002110 toxicologic effect Effects 0.000 description 1
- 231100000759 toxicological effect Toxicity 0.000 description 1
- 238000012085 transcriptional profiling Methods 0.000 description 1
- 108091006107 transcriptional repressors Proteins 0.000 description 1
- 108091007466 transmembrane glycoproteins Proteins 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- XETCRXVKJHBPMK-MJSODCSWSA-N trehalose 6,6'-dimycolate Chemical class C([C@@H]1[C@H]([C@H](O)[C@@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](COC(=O)C(CCCCCCCCCCC3C(C3)CCCCCCCCCCCCCCCCCC)C(O)CCCCCCCCCCCCCCCCCCCCCCCCC)O2)O)O1)O)OC(=O)C(C(O)CCCCCCCCCCCCCCCCCCCCCCCCC)CCCCCCCCCCC1CC1CCCCCCCCCCCCCCCCCC XETCRXVKJHBPMK-MJSODCSWSA-N 0.000 description 1
- 239000013638 trimer Substances 0.000 description 1
- 150000004043 trisaccharides Chemical class 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 201000008827 tuberculosis Diseases 0.000 description 1
- 230000006433 tumor necrosis factor production Effects 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 210000002845 virion Anatomy 0.000 description 1
- 239000011534 wash buffer Substances 0.000 description 1
Classifications
-
- 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
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- 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
-
- 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
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
-
- 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
- A61K2039/55505—Inorganic adjuvants
-
- 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
- A61K2039/55572—Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
-
- 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
- A61K2039/55577—Saponins; Quil A; QS21; ISCOMS
-
- 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
- A61K2039/57—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
-
- 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
- A61K2039/57—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
- A61K2039/575—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
-
- 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
- C12N2740/16034—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
-
- 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
-
- 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
- C12N2740/16211—Human Immunodeficiency Virus, HIV concerning HIV gagpol
- C12N2740/16234—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- ART Antiretroviral therapy
- CD4 T follicular helper cells are a specialized subset of CD4 T cells that migrate to germinal centers (GC) within secondary lymphoid organs and provide growth and differentiation signals to GC B cells within a few days of immunization.
- GCs are populated by antigen-activated, rapidly proliferating B cell clones, which rely on cytokines and co stimulatory signals from Tfh cells to undergo immunoglobulin affinity maturation, class- switch recombination, and differentiation to memory B cells and plasma cells. The maturation of GC B cells to plasma cells and the resulting long-lived humoral immunity hinges on effective Tfh help.
- Tfh cells are heterogeneous and, depending on inflammatory signals during T cell priming, differentiate into Thl, Th2, Thl7-type Tfh cells. Th polarization of a Tfh cell influences cytokine profile and co-stimulatory molecule expression.
- a method of enhancing the anti- Env antibody response during HIV vaccination in a subject, the method comprising (i) administering a first Thl -polarizing adjuvant to the subject together with a DNA vaccine comprising a polynucleotide encoding a first HIV Env polypeptide, wherein the first adjuvant comprises a polynucleotide encoding interferon-induced protein (IP)-10; and (ii) administering a second Thl -polarizing adjuvant to the subject together with a booster comprising a second HIV Env polypeptide, wherein the second adjuvant comprises QS-21.
- IP interferon-induced protein
- the second HIV Env polypeptide is a gpl40 polypeptide.
- the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
- the first adjuvant and the DNA vaccine are administered transdermally with electroporation.
- the HIV Env polypeptide, HIV gpl40 polypeptide, and/or one or more additional HIV polypeptides are from HIV-1.
- the HIV-1 is of clade C origin.
- the booster further comprises a lipid and/or liposomal adjuvant.
- the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
- the lipid adjuvant comprises monophosphoryl lipid A (MPLA).
- the first Thl- polarizing adjuvant and DNA vaccine are administered to the subject 1, 2 or 3 times prior to the administration of the booster.
- the first Thl -polarizing adjuvant and DNA vaccine are administered to the subject at 0, 8, and 16 weeks.
- the booster is administered to the subject 1 or 2 times.
- peripheral and germinal center (GC) Tfh cells isolated from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster display higher proportions of anti-Env Thl cells than do peripheral and germinal center (GC) Tfh cells taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
- blood taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster shows higher anti-gpl40 extrafollicular and/or plasma cell-derived titers than does blood from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
- serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show a broader cross-clade anti-Env response and/or increased specificity for gpl20 V1V2 loops than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
- serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster display elevated anti-Env titers that persist longer than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
- serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show higher avidity against gpl40 than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
- serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater neutralization activity against HIV-1 than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl- polarizing adjuvant.
- the neutralization of HIV-1 is assessed using the TZM-bl assay.
- serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater antibody- dependent cellular toxicity (ADCC) and/or antibody-dependent phagocytosis (ADP) activity against HIV-infected cells than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
- the HIV-infected cells are infected with HIV-1.
- the HIV-1 is of clade C origin.
- IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show higher anti-gpl40 titers than do IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
- the present disclosure provides a pharmaceutical composition for vaccinating a subject against HIV, the composition comprising a DNA vaccine comprising a polynucleotide encoding an HIV Env polypeptide, a polynucleotide encoding interferon- induced protein (IP)- 10, and a pharmaceutically acceptable carrier.
- the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
- the HIV Env polypeptide and/or one or more additional HIV polypeptides are from HIV-1.
- the HIV-1 is of clade C origin.
- the polynucleotide encoding the HIV Env polypeptide and the polynucleotide encoding IP- 10 are present within a single DNA vector.
- the composition is formulated for transdermal delivery with electroporation.
- the present disclosure provides a pharmaceutical composition comprising an HIV Env polypeptide, QS-21, and a pharmaceutically acceptable carrier.
- the HIV Env polypeptide is a gpl40 polypeptide.
- the composition further comprises a lipid and/or liposomal adjuvant.
- the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
- the lipid adjuvant comprises monophosphoryl lipid A (MPLA).
- the HIV Env polypeptide is from HIV-1.
- the HIV-1 is of clade C origin.
- FIGS. 1A-1D Immunization schedule for subtype C HIV-1 Envelope DNA prime and protein boost vaccine regimen.
- FIG. 1A Flow cytometric plots illustrate expression of HIV Env, SIV Gag, and IP-10 by 293-T cells transfected with DNA and DNAIP-10 plasmids. Grey overlay shows expression in non-transfected cells.
- FIG. IB Bar graph shows surface expression of HIV Env based on detection with a panel of monoclonal antibodies as indicated.
- FIG. 1C IP-10 titers in supernatants of transfected 293T cells show accumulation of IP- 10 following transfection with DNAIP-10.
- FIG. ID shows immunization schedule and sampling.
- FIGS. 2A-2G DIP-lOProteinALFQ vaccine induces robust anti -Env antibody titers with cross-clade breadth.
- FIG. 2A Antibody kinetics against C. 1086 Env gpl40 in serum at weeks 0, 2, and 8 following each protein boost assessed by binding antibody multiplex assay (BAMA); right panel shows scatter plot values for each animal at weeks 0, 2, and 8 post 2nd protein boost.
- FIG. 2B Kinetics of C. 1086 specific anti -Env titers after 2nd protein boost measured by ELISA; right panel shows titers for each individual animal.
- BAMA assay was used to measure responses against CH505 (FIG. 2C), Con C (FIG. 2D), Con S (FIG.
- FIG. 2G shows fold change in antibody titers at indicated time points after 2nd protein boost relative to the first.
- Animals receiving the DProALFA vaccine are represented by blue circles and animals receiving the DIP-lOProALFQ vaccine by red circles.
- Kinetic data show geometric means. Vertical dotted lines show immunization time points. In dot plots, geometric means are indicated as horizontal lines. Statistical significance across vaccine regimens was tested using unpaired, two-tailed Mann-Whitney U test; *p ⁇ 0.05, **p ⁇ 0.01, *** p ⁇ 0.001, **** p ⁇ 0.0001.
- FIGS. 3A-3M DIP-10 ProteinALFQ vaccine elicits high avidity anti-Env antibody with ADCC and ADP activities.
- FIG. 3A Surface Plasmon Resonance (SPR) was used to determine the avidity index (AI) in serum at 2 weeks after final DNA and each protein boost using C. 1086 gpl40 protein immobilized onto sensor chips.
- FIG. 3B shows SPR-based AI values in the two vaccine regimens over time.
- FIG. 3C shows significantly higher avidity in DIP-10 ProteinALFQ. at 2 weeks post 2nd protein boost. AI measured against C.1086 gpl40 using 2 M sodium thiocyanate (FIG. 3D) and 0.1 M sodium citrate (FIG.
- FIG. 3E Serum-neutralizing antibody responses were assessed against tier 1 A (MW965.26) pseudovirus and the 50% infective dose (ID50) was determined.
- FIG. 31 shows ADCC activity against SHIV CH505 infected target cells.
- FIG. 3J Antibody-dependent phagocytosis using gpl20-coated beads was measured using sera from week 8 post 2nd protein boost at serum dilutions ranging from 1 : 100 to 1 :2500.
- FIG. 3K shows individual ADP scores at 1:500 serum dilution.
- FIG. 3L C.1086 gpl20 subclass analysis was performed on week 8 post 2nd protein boost to measure IgGl, IgG2, IgG3, and IgG4.
- FIG. 3M shows IgGl/IgG4 ratio across vaccine groups at week 8 post 2nd protein boost.
- Statistical significance across vaccine regimens was tested using unpaired, two-tailed Mann-Whitney U test and within group differences over time were tested using Wilcoxon matched-pairs signed rank test; *p ⁇ 0.05, **p ⁇ 0.01, *** p ⁇ 0.001, **** p ⁇ 0.0001.
- FIGS. 4A-4E DNAIP-10 ProteinALFQ vaccine elicits robust anti-Env antibody in vaginal and rectal mucosal secretions. Vaginal and colorectal secretions were evaluated for C.1086 concentrations.
- FIGS. 4A-4B show kinetics of C.1086 IgA responses in vaginal and rectal secretions.
- FIGS. 4C-4D show IgA and IgG titers in rectal secretions.
- FIG. 4E shows gpl40 IgA kinetics in sera. Horizontal broken lines represent the assay limit of detection.
- Kinetic data show geometric means. Vertical dotted lines show immunization time points. In dot plots, geometric means are indicated as horizontal lines. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U test; *p ⁇ 0.05, **p ⁇ 0.01, *** p ⁇ 0.001, **** p ⁇ 0.0001.
- FIGS. 5A-5D DIP-10 ProteinALFQ vaccine induces Env-specific Tfh cells in peripheral blood.
- FIG. 5A Flow cytometric gating of CXCR5+ cells in PBMCs to identify 0X40+ CD25+ activated CD4 T subsets after stimulation with C.1086 protein and Con C peptide pools.
- FIG. 5B shows frequency of Env-specific CD4 T cells at week 1 post 1st protein boost which strongly correlate with responses against the C.ZA.1197MB boosting immunogen (FIG. 5C).
- FIG. 5D shows robust induction of IP- 10 in sera after 1st protein immunization. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U test. Spearman coefficient of correlation values were computed to determine associations; * p ⁇ 0.05, **p ⁇ 0.01, *** p ⁇ 0.001.
- FIGS. 6A-6L DIP-10 ProteinALFQ vaccine induces GC Tfh cells with distinctive Thl signatures.
- FIG. 6A Gating strategy to identify GC Tfh cells and GC B cells in lymph node at 2 weeks post 1st protein boost. Histograms show higher relative expression of Bcl-6 and ICOS in GC Tfh cells.
- FIG. 6B Ex vivo co-culture experiments with sorted GC Tfh cells demonstrates B helper capacity.
- FIG. 6C shows kinetics of GC Tfh responses in lymph node at specified time points.
- FIG. 6D Dot plot shows higher relative frequency of GC B cells in Thl vaccine group, and correlation of GC B cells with GC Tfh cells.
- FIG. 6E Histogram shows relative CXCR3 expression in GC Tfh cells and GC B cells and dot plot shows significantly higher CXCR3 expression on GC Tfh cells and GC B cells in Thl vaccine regimen.
- FIG. 6F Flow plot illustrates higher expression of CXCR3 on T-bet+ memory B cells.
- FIG. 6G Antibody titers at week 18 post 2nd protein predicted by frequency GC Tfh cells and proportion of CXCR3 -expressing GC Tfh cells at 2 weeks post 1st protein. Antibody avidity at week 8 post 2nd protein predicted by GC Tfh cells at 1st protein.
- FIG. 6H shows animals selected (triangles) for RNA-seq and Principal component analysis of RNA-Seq data (FIG. 61); colors represent populations as indicated in FIG. 6A.
- FIG. 6J Heatmap shows expression of genes differentially expressed in Tfh relative to naive across four sorted CD4 subsets indicated in FIG. 61.
- FIG. 6K Volcano plot of differentially expressed transcripts (adjusted p ⁇ 0.05 in red) for Tfh cells versus naive cells.
- FIG. 6L Log fold change values of key Tfh and Thl genes in Tfh (red) and memory Tfh (blue) cells in lymph node Thl vaccinated animals. Statistical significance was tested using unpaired, two-tailed Mann- Whitney U test. Spearman coefficient of correlation values were computed to determine associations; * p ⁇ 0.05, **** p ⁇ 0.0001.
- FIGS. 7A-7G DNAIP-10 immunization induces systemic expansion of pro- inflammatory monocytes and enhances GC Tfh responses.
- FIG. 7 A shows gating strategy to identify activated CXCR5+ cells in blood at day 0 and day 14 following DNA3 immunization.
- Kinetic data show transient accumulation of ICOS+ PD-1+ CXCR5+ cells in blood at day 14 following DNA3 when expressed as relative frequencies (left) and absolute counts (right).
- FIG. 7B Flow cytometric gating to identify inflammatory CD14+CD16+ monocytes which increase at day 3 following DNA immunization (FIG. 7C).
- FIG. 7D Higher relative increase in pro-inflammatory monocytes in DNA-PMO primed animals.
- FIG. 7 A shows gating strategy to identify activated CXCR5+ cells in blood at day 0 and day 14 following DNA3 immunization.
- Kinetic data show transient accumulation of ICOS+ PD-1+ CXCR5+ cells in
- FIG. 7E shows induction of IL-lb in sera 7 days following DNA immunization.
- FIG. 7F Fine needle aspirates of draining lymph nodes show increased frequencies of GC Tfh cells in DNA-IP-10 primed animals at day 14 post DNA.
- FIG. 7G shows that the frequency of pro- inflammatory monocytes predicts C.1086C gpl40 antibody titers at week 8 post 2nd protein boost. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U test. Spearman coefficient of correlation values were computed to determine associations; * p ⁇ 0.05, ***, p ⁇ 0.001, **** p ⁇ 0.0001.
- FIGS. 8A-8D Thl DNA prime enhances frequency of pro-inflammatory monocytes and GC Tfh cells.
- FIG. 8A Increase in net frequency of pro-inflammatory monocytes in Thl vaccine group.
- FIG. 8B Increase in IP- 10, IL-IB, IL-6 and IL-21 after DNA prime.
- FIG. 8C Increase in GC Tfh cells with Thl vaccine.
- FIG. 8D Frequency of pro-inflammatory monocytes after DNA prime predicts antibody response at memory.
- FIGS. 9A-9D Thl DNA prime enhances frequency of Env-specific Tfh cells.
- FIG. 9A shows time points of blood collection for T cell kinetic analysis.
- FIG. 9B Flow plot illustrates gating strategy to identify activated CD4 Tfh cells based on coexpression of ICOS and PD-1. Kinetics of relative frequencies and absolute counts of ICOS+PD-1+ Tfh cells.
- FIG. 9C shows gating strategy to identify Env-specific Tfh cells based on co-expression of 0X40 and CD25.
- FIG. 9D shows higher Env-specific Tfh cells in Thl regimen.
- FIGS. 10A-10B Schematic shows study design with three DNA primes (SHIV 1086C, 4 mg given I.D with E.P) followed by two protein boosts with Clade C gpl40 Envelope, HIV-1 isolate C.ZA.1197MB, 100 pg delivered subcutaneously (SQ).
- SHIV 1086C 4 mg given I.D with E.P
- Clade C gpl40 Envelope HIV-1 isolate C.ZA.1197MB
- SQ subcutaneously
- FIG. 10A Kinetics of antibody responses following 2 nd protein boost.
- FIG. 10B Scatter plot showing antibody titers in all animals within the experimental groups at weeks 0, 2, 8, and 18 following 2nd protein boost with antibody fold-change in Thl relative to Thl+2 group.
- FIGS. 11A-11D After Thl vaccine regimen, antibodies demonstrate increased breadth, avidity, and broader specificity. Antibody breadth (FIG. 11A), Avidity index (FIG. 11B), and specificity (FIG. 11C) and neutralization (FIG. 11D) titers at indicated time points after 2nd protein boost.
- FIG. 13. Experimental design of vaccine trial 1 (SI). All groups are immunized intradermally using a Bioject electroporation device with plasmid DNA-SHIV construct expressing 1086 Clade C Env, tat, and rev; and SIV239 Gag (3 mg/dose, week 0 and 4). Groups 2 and 3 receive a similar dose of IP-10 adjuvanted DNA-SHIV construct. The MVA- SHIV boost is intramuscular (10 L 8 PFU/dose, week 16,24).
- Animals are co-immunized with gpl40 protein (Clade C Env, 100 pg), in the thigh opposite to that of the MVA boost, formulated either in Alum (500 pg, Groups 1, 2) or AS01B (50 pg MPL+50 pg Q21) Group 3). Animals in Groups 1 and 3 are compared for vaccine efficacy and all groups are compared for immunogenicity.
- FIGS. 14A-14F DNA/MVA vaccine induces robust Env antibody.
- Monkeys were immunized with DNA (wks 0, 8) and MVA (wks 16, 32).
- FIGS. 15A-15E show a transient accumulation of CXCR5+ Tfh cells expressing the cell-cycle marker Ki-67 at the peak CD4 effector response following the MVA boost (FIGS. 15A-15B, *p ⁇ 0.05).
- IL Tfh cytokine interleukin
- FIG. 15D Ki- 67+ pTFH expressed CXCR3, a marker of Thl polarity
- the proportion of CXCR3+Ki-67+ pTFH cells markedly increased following MVA immunization relative to baseline (FIG. 15E, ***p ⁇ 0.001).
- FIGS. 16A-16F show aspects of the GC response following the 2nd MVA boost and how Pro+Alum modulated this response.
- GC Tfh were examined by high expression of programmed death (PD)1 and CXCR5, and Tfh cells based on the CXCR5+PD-1+ phenotype (FIG. 16A).
- M+Pro vaccine increased Thl-polarized Tfh cells (FIG. 16B) even in the presence of Alum.
- the proportion of CXCR3+ GC Tfh correlated with frequency of GC B cells (FIG. 16C), antibody avidity (FIG. 16D), durability (FIG. 16E), and neutralization (FIG. 16F).
- FIGS. 17A-17C IP-10 induces CXCR3+ Tfh cells.
- Flow plot (FIG. 17A) and histogram (FIG. 17B) show CXCR3 expression on Tfh cells at increasing IP- 10 doses.
- FIG. 17C Increase in CXCR3+ Tfh cells with IP- 10.
- FIGS. 18A-18C Construction of PMO-DNASHIV vaccine.
- FIG. 18A The DNA vaccine includes IP- 10 as a fusion to IRES or as a fusion to the 2 A peptide downstream of Env.
- FIG. 18B Characterization of CD40L-DNA vaccine.
- FIG. 18C Significantly higher magnitude and proportion (% responders indicated in grey area) vaccine-elicited CD4s after 1st prime in CD40L-DNA (blue) relative to DNA (clear); ****, p ⁇ 0.0001.
- FIGS. 19A-19C Identifying vaccine-elicited Thl and Th2 Tfh cells.
- the cell-cycle marker Ki-67 was utilized to interrogate CXCR5+ CD4 T cells at peak effector time points following each immunization for expression of CXCR3 and CCR4, as outlined in FIG. 19A. This is complemented by intracellular cytokine staining (ICS)-based assays designed to examine vaccine-specific responses (at peak and memory) after stimulation with relevant Gag and Env peptide pools. After stimulation, Ag-specific Tfh cells are identified based on cytokine positivity and CXCR5 expression, as shown in FIGS. 19B-19C.
- ICS cytokine staining
- FIG. 20 CD40L+ Tfh are Env specific. Vaccine-specific TFH cells are identified by translocation of CD40L after 5 hour stimulation with Env peptides in the presence of co stimulatory molecules.
- FIGS. 21A-21B The frequency of Env-specific IFNG+IL-21+ cells at 1 week post 2nd MV A, and the frequency of CXCR3+ GC Tfh cells at 2 weeks post 2nd MV A, correlate with antibody durability measured at 20 weeks post 2 nd MVA.
- FIGS. 22A-22B Assays to measure humoral immune responses.
- FIG. 22A ELISPOT assay captures increase in gpl40 ASCs with Pro+MVA relative to MVA alone.
- FIG. 22B ADCC increases following a 3rd protein boost, while no increase was observed in ADP.
- FIGS. 23A-23B CXCR3+ Tfh cells express CCR5.
- FIG. 23A CXCR3+ Tfh cells comprise higher frequency of CCR5 cells.
- FIG. 23B Higher levels of pro-viral DNA in X3+ Tfh cells in lymph node; ***, p ⁇ 0.001; *p ⁇ 0.05.
- FIGS. 24A-24B Thl vaccine regimen induces robust and durable serum HIV anti- Env antibody titers.
- Schematic shows study design with 3 DNA primes (SHIV 1086C, 4 mg given I.D with E.P) followed by two protein boosts with Clade C gpl40 Envelope, HIV-1 isolate C.ZA.1197MB, 100 pg delivered subcutaneously (SQ).
- DNA was adjuvanted with IP-10 and protein with ALFQ and the Thl+2 group got DNA alone with ALFA-adjuvanted protein boost (FIG. 24A).
- Kinetics of antibody responses following 2 nd protein boost (FIG. 24B). Scatter plot showing antibody titers in all animals within the experimental groups at week 0, 2, 8, and 18 following 2nd protein boost with antibody fold- change in Thl relative to Thl+2 group.
- FIGS. 25A-25D After Thl vaccine regimen antibodies demonstrate increased breadth, avidity, and broader specificity. Antibody breadth (FIG. 25A), Avidity index (FIG. 25B), and specificity (FIG. 25C) and neutralization (FIG. 25D) titers at indicated time points after 2nd protein boost.
- FIGS. 26A-26B Robust induction of anti-Env-specific IgA (FIG. 26A) and anti- Env-specific IgG (FIG. 26B) in vaginal and rectal mucosa with Thl vaccine regimen. Data are post 2nd protein boost.
- FIGS. 27A-27D Thl DNA prime induces higher magnitude Env-specific Tfh cells.
- FIG. 27A shows time points of blood collection for T cell kinetic analysis.
- FIG. 27B Flow plot illustrates gating strategy to identify activated CD4 Tfh cells based on co-expression of ICOS and PD-1. Kinetics of relative frequencies and absolute counts of ICOS+PD-1+ Tfh cells.
- FIG. 27C shows gating strategy to identify Env-specific Tfh cells based on co expression of 0X40 and CD25.
- FIG. 27D shows higher Env-specific Tfh cells in Thl regimen.
- FIGS. 28A-28G Induction of polyfunctional Thl/Thl7 CD4 Tfh cells in Thl vaccine regimen.
- FIG. 28A shows transient induction of pro-inflammatory Thl cytokine IP- 10, Thl7 cytokine IL-17A and IL-6 and IL-21.
- FIG. 28B shows higher frequencies of Env- specific Tfh cells in PBMCs and
- FIG. 28C illustrates a robust GC response in Thl vaccine regimen.
- FIG. 28D illustrates gating for GC Tfh cells with higher relative expression of Bcl- 6, Thl marker CXCR3 and Thl7 marker CCR6.
- FIG. 28E shows higher CD40L expression if IFNG+IL-17+ cells.
- FIG. 28G Phenotypic examination of GC subsets confirmed increase in proportion of CXCR3+CCR6+ GC Tfh cells in Thl vaccinated group.
- FIG. 29 Polyfunctional Thl/Thl7 CD4 T cells predict durability of anti-Env titers in sera and mucosa. The frequency of polyfunctional Thl/Thl7 GC TFH cells is directly predictive of binding titers as measured by ELISA and BAMA.
- FIGS. 30A-30D A single CAFOl adjuvanted protein immunization induces robust antibody responses.
- FIG. 30A Structure of the cationic liposome CAFOl. Single subcutaneous immunization with CAFOl -adjuvanted Chlamydia antigen (CTH 522, 5 pg; FIG.
- FIG. 30B shows robust induction of antibody responses at levels comparable or superior to that induced by the MF59 analog adjuvant, Addavax.
- FIG. 30D Notably, the frequency of OVA-bound cells at the site of injection was significantly higher at 24- and 48- hours following immunization, indicating formation of antigen depot at the site of immunization with CAFOl.
- FIG. 31 Robust protein expression of Env and Gag in DNA 1086C plasmid co expressing IP- 10 and IL-6.
- FIGS. 32A-32D Thl vaccine enhances frequency of pro-inflammatory monocytes and GC Tfh cells.
- FIG. 32A Increase in net frequency of pro-inflammatory monocytes in Thl vaccine group.
- FIG. 32B Increase in IP-10, IL-1B, IL-6 and IL-21 after DNA prime.
- FIG. 32C Increase in GC Tfh cells with Thl vaccine.
- FIG. 32D Frequency of pro- inflammatory monocytes after DNA prime predicts antibody response at memory.
- FIGS. 33A-33B Increased Env gpl40 1086.
- FIG. 33A shows higher avidity after sequential immunization in both vaccine groups.
- FIG. 33B shows increased avidity in Thl-Tfh vaccine regimen.
- FIGS. 34A-34B Higher levels of Env IgGl (FIG. 34A) and IgG3 (FIG. 34B) with Thl vaccine regimen. Titers were measured at week 8 post 2nd protein.
- the present disclosure relates to methods and compositions for enhancing the immune response in subjects undergoing HIV vaccination.
- the present methods and compositions can be used in conjunction with prime-boost vaccine regimens, e.g., using DNA vaccines against HIV, in order to enhance the Thl polarization of Tfh cells in a subject.
- the present disclosure is based in part on the discovery that Thl polarization in conjunction with a DNA prime substantially increases antigen-specific, e.g, Env-specific, Tfh cells, and that Thl polarization in conjunction with a protein boost, e.g., using gpl40, results in greater production of the IgGl subclass with enhanced longevity, breadth, avidity, ADCC, and ADP activities of the antigen-specific antibody.
- the present disclosure provides DNA constructs expressing both HIV (and/or SIV) genes and interferon protein 10 (IP- 10), as well as compositions comprising an HIV protein boost, e.g, gpl40, with a Thl -polarizing adjuvant such as QS-21.
- the present methods and compositions are effective at inducing, e.g, pro-inflammatory monocytes, Env-specific CD4 T follicular helper cells, and higher germinal center responses relative to DNA alone (i.e., in the absence of Thl polarization).
- the localized expression of IP- 10 using the present constructs enhances immunogenicity by concerted effects on antigen-presenting cells and T cells, and limits the potential for non-specific, systemic effects, thereby reducing side effects.
- the terms“about” and“approximately” as used herein shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Typically, exemplary degrees of error are within 20 percent (%), preferably within 10%, and more preferably within 5% of a given value or range of values.
- any reference to“about X” specifically indicates at least the values X, 0.8X, 0.81X, 0.82X, 0.83X, 0.84X, 0.85X, 0.86X, 0.87X, 0.88X, 0.89X, 0.9X, 0.91X, 0.92X, 0.93X, 0.94X, 0.95X, 0.96X, 0.97X, 0.98X, 0.99X, 1.01X, 1.02X, 1.03X, 1.04X, 1.05X, 1.06X, 1.07X, 1.08X, 1.09X, 1.1X, 1.11X, 1.12X, 1.13X, 1.14X, 1.15X, 1.16X, 1.17X, 1.18X, 1.19X, and 1.2X.
- “about X” is intended to teach and provide written description support for a claim limitation of, e.g.,“0.98X.”
- the term“antigen” refers to a molecule, or a portion thereof, that is capable of inducing an immune response (e.g ., in a subject). While in many instances an immune response involves the production of an antibody that targets or specifically binds to the antigen, as used herein the term“antigen” also refers to molecules that induce immune responses other than those that specifically involve the production of an antibody that targets the antigen, e.g., a cell-mediated immune response involving expansion of T cells that target antigen-derived peptides presented on the surface of target cells.
- an antigen refers to an HIV protein, or immunogenic fragment thereof, e.g, an HIV Env protein (e.g., gpl20, gpl40, gpl60).
- nucleic acid sequence encoding a peptide refers to a segment of DNA, which in some embodiments may be a gene or a portion thereof, that is involved in producing a peptide chain (e.g, an HIV protein or IP- 10).
- a gene will generally include regions preceding and following the coding region (leader and trailer) involved in the transcription/translation of the gene product and the regulation of the transcription/translation.
- a gene can also include intervening sequences (introns) between individual coding segments (exons).
- Leaders, trailers, and introns can include regulatory elements that are necessary during the transcription and the translation of a gene (e.g, promoters, terminators, translational regulatory sequences such as ribosome binding sites and internal ribosome entry sites, enhancers, silencers, insulators, boundary elements, replication origins, matrix attachment sites and locus control regions, etc.).
- A“gene product” can refer to either the mRNA or protein expressed from a particular gene.
- the terms“expression” and“expressed” refer to the production of a transcriptional and/or translational product, e.g., of a nucleic acid sequence encoding a protein (e.g, an HIV protein or IP- 10).
- the term refers to the production of a transcriptional and/or translational product encoded by a gene (e.g, a gene encoding an antigen) or a portion thereof.
- the level of expression of a DNA molecule in a cell may be assessed on the basis of either the amount of corresponding mRNA that is present within the cell or the amount of protein encoded by that DNA produced by the cell.
- recombinant when used with reference, e.g., to a polynucleotide, protein, vector, or cell, indicates that the polynucleotide, protein, vector, or cell has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified.
- recombinant polynucleotides contain nucleic acid sequences that are not found within the native (non-recombinant) form of the polynucleotide.
- immune response refers to any response that is induced (e.g, in a subject) by an antigen, including the induction of immunity against pathogens (e.g, viruses such as HIV).
- pathogens e.g, viruses such as HIV.
- Immune responses induced by systems, recombinant polynucleotides, compositions, and methods described herein are typically desired, intended, and/or protective immune responses.
- the term includes the production of antibodies against an antigen, as well as the development, maturation, differentiation, and activation of immune cells (e.g, B cells and T cells).
- an immune response comprises inducing pro-inflammatory monocytes, Env-specific CD4 T follicular helper cells, and germinal center responses.
- the term also includes increasing or decreasing the expression or activity of cytokines that are involved in regulating immune function (e.g, in a subject).
- Other markers of immune responses include, but are not limited to, Thl-Tfh responses, dendritic cell (DC)-T cell interactions, the magnitude of vaccine-specific T cells, Tfh differentiation, Thl polarization, IL-6 production in B cells, plasma cell differentiation, induction of CD80 and CD40 on DCs, the strength of cross-priming and magnitude of cytolytic responses, effects on APCs and T cells, and others.
- HIV refers to the human immunodeficiency virus, and can include any type, group, sub-group, clade, strain, variant, or isolate, e.g, HIV-1, HIV-2, HIV-1 Group M (including sub-groups or clades A, B, C, D, F, G, H, J), Group N, Group O, C.1086, Con C, Con S CH505, etc.
- HIV is also used to describe genes within the HIV genome (e.g, gag, pol, env, tat, rev, nef, vpr, vif, and vpu), or to proteins encoded by the genes, including Env (or gpl20, gpl40, gpl60, gp41), Gag (or MA, CA, SP1, NC, SP2, P6), Pol (or RT, RNAse H, IN, PR), Tat, Rev, Nef, Vpr, Vif, and Vpu.
- Env or gpl20, gpl40, gpl60, gp41
- Gag or MA, CA, SP1, NC, SP2, P6
- Pol or RT, RNAse H, IN, PR
- Tat Rev, Nef, Vpr, Vif, and Vpu.
- IP- 10 interferon-gamma-induced protein 10 (also known as CXCL10, or C-X- C motif chemokine 10, INP10, small-inducible cytokine B10, and other names) is an 8.7 kDa protein belonging to the CXC chemokine family.
- the gene encoding IP- 10 corresponds, e.g, to NCBI Gene ID No. 3627.
- IP-10 also encompasses polynucleotides comprising the human IP-10/CXCR3 mRNA of NCBI Reference Sequence NM_001565, or polynucleotides having a sequence comprising at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to the nucleotide sequence of NM_001565.
- IP-10 also encompasses polypeptides comprising (or polynucleotides encoding) the human IP- 10/CXCR3 protein of NCBI Reference Sequence NP_001556.2, or comprising (or polynucleotides encoding) a polypeptide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to NP_001556.2, as well as to derivatives, variants, and fragments thereof.
- Thl -polarization refers to the differentiation of CD4+ T cells into specific subsets in response to antigens presented by antigen presenting cells (APCs) and depending on, e.g., the particular cytokines present in the microenvironment. Polarization involves in part the modulation of particular transcription factors that results in the production of autocrine cytokines, producing a positive feedback loop that contributes to the polarization of the cells.
- CD4+ subsets include Thl, Th2, Th3, Th9, Thl7, Treg, Trl, and Tfh (see, e.g, Martinez- Sanchez et al., Front. Physiol. (2016); doi.org/10.3389/fphys.2018.00877).
- Thl polarization involves, e.g, the transcription factor T-bet and the cytokine IFNy; other markers of Thl polarization include, e.g., bcl-6, CXCR3, BCl-6, TBX21, SemA4A, and CCR5.
- Gpl40 is a soluble version of the Env protein that contains the entire ectodomain
- Gpl40 encompasses forms, variants, and derivatives of Gpl40, including monomeric, dimeric, trimeric, etc., forms, forms in which the gpl20 and gp41 components are linked, e.g, by a disulfide bond, mutated forms, e.g, with an I559P change within the gp41 component, and others.
- Gpl40 also comprises immunogenic fragments of full-length gpl40, e.g, comprising one or more portions of the gpl20 and/or gp41 components that are useful for eliciting immune responses in a subject.
- QS-21 is a purified plant extract containing water soluble triterpene glycoside saponins that is derived from the soap bark tree, Quillaja saponaria.
- QS-21 comprises a quillaic acid triterpene substituted with a branched trisaccharide and a linear tetrasaccharide, which is connected to an acyl chain via a hydrolytically labile ester.
- QS-21 is a 65:35 mixture of the apiose- and xylose-substituted variants of the above-described molecule.
- QS-21 can be natural or semi-synthetic, e.g, prepared by purifying the prosapogenin part of the molecule and synthetically adding the rest of the molecule. Fully synthetic forms of the molecule are encompassed as well. QS-21 can be obtained from commercial sources, e.g., from Desert King International (San Diego, CA).
- an“adjuvant” refers to a compound administered to a subject in conjunction with an antigen for enhancing an immune response to the antigen.
- Adjuvants can increase the immunogenicity of vaccines in any of a number of ways, and can include inorganic compounds such as salts, e.g, aluminum salts, as well as organic compounds and mixtures of compounds, including extracts and preparations, e.g, Freund’s incomplete adjuvant, squalene, MF59, monophosphoryl lipid A, QS-21.
- an adjuvant is a polypeptide such as IP- 10.
- an“adjuvant” as used herein refers to adjuvants that increase the Thl polarization of Tfh cells, e.g, IP-10 and/or QS-21.
- A“prime-boost” or“prime-booster” vaccine strategy refers to an immunization approach that is administered in two or more stages and in which the same antigen is presented to a subject in identical (in a homologous protocol) or different (in a heterologous protocol) forms.
- a polynucleotide e.g, DNA molecule encoding an immunogenic protein
- a polynucleotide is administered one or more times, wherein the protein encoded by the polynucleotide is expressed by cells in the body, and in a second stage the protein itself is administered.
- the form of the protein administered in the second stage need not be identical to that of the encoded protein; for example, in numerous embodiments of the present disclosure, a prime DNA vaccine is administered encoding the Env protein, and the gpl40 form of the Env protein is administered during the boost stage.
- a prime DNA vaccine is administered encoding the Env protein
- the gpl40 form of the Env protein is administered during the boost stage.
- the first, i.e., DNA prime, stage precedes the protein boost stage, but other sequences can be performed as well, including approaches in which the two stages overlap to some extent.
- Tfh cells are a subset of CD4+ T cells that help B cells produce antibodies against foreign pathogens (e.g, against HIV).
- Tfh cells are located both in circulation and in secondary lymphoid organs, e.g, tonsils, spleen, and lymph nodes, in particular in the B cell zones, where they interact with and stimulate B cells (e.g, through CD40 and by producing IL-21).
- Tfh cells are involved in the formation of the germinal centers (GCs), structures that form within the B cell zones during an immune response.
- GCs germinal centers
- Tfh cells are defined by, e.g, the expression of the transcription factor Bcl6 and cell surface markers such as CXCR5, PD1, and ICOS.
- the term“antigen-presenting cell” or“APC” refers to a cell that displays or presents an antigen, or a portion thereof, on the surface of the cell.
- antigens are displayed or presented with a major histocompatibility complex (MHC) molecule.
- MHC major histocompatibility complex
- APCs are found in a large number of different tissue types.
- Professional APCs such as dendritic cells, macrophages, and B cells, present antigens to T cells in a context that most efficiently leads to their activation and subsequent proliferation. Many cell types present antigens to cytotoxic T cells.
- polynucleotide refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof.
- the term includes, but is not limited to, single-, double-, or multi-stranded DNA or RNA, genomic DNA, cDNA, and DNA-RNA hybrids, as well as other polymers comprising purine and/or pyrimidine bases or other natural, chemically modified, biochemically modified, non-natural, synthetic, or derivatized nucleotide bases.
- nucleic acids containing known analogs of natural nucleotides that have similar binding properties as the reference nucleic acid Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g ., degenerate codon substitutions), homologs, and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al ., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al. , J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al. , Mol. Cell. Probes 8:91-98 (1994)).
- vector and“expression vector” refer to a nucleic acid construct, e.g., plasmid or viral vector, generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid sequence (e.g, encoding an HIV antigen and/or a protein having IP- 10-like activity) in a host cell or engineered cell.
- a vector includes a polynucleotide to be transcribed, operably linked to a promoter.
- a vector may contain those that enhance transcription (e.g, enhancers), those that terminate transcription (e.g, terminators), those that confer certain binding affinity or antigenicity to a protein (e.g, recombinant protein) produced from the vector, and those that enable replication of the vector and its packaging (e.g, into a viral particle).
- the vector is a viral vector ( i.e a viral genome or a portion thereof).
- a vector may contain nucleic acid sequences or mutations, for example, that increase tropism and/or modulate immune function.
- polypeptide “peptide,” and“protein” are used interchangeably herein to refer to a polymer of amino acid residues. All three terms apply to amino acid polymers in which one or more amino acid residues are an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. As used herein, the terms encompass amino acid chains of any length, including full-length proteins, wherein the amino acid residues are linked by covalent peptide bonds.
- the terms“subject,”“individual,” and“patient” are used interchangeably herein to refer to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, mice, rats, simians, humans, farm animals, sport animals, and pets. Tissues, cells and their progeny of a biological entity obtained in vivo or cultured in vitro are also encompassed.
- administering includes oral administration, topical contact, administration as a suppository, intravenous, intraperitoneal, intramuscular, intralesional, intratumoral, intrathecal, intranasal, intraosseous, or subcutaneous administration to a subject. Administration is by any route, including parenteral and transmucosal (e.g buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal). Parenteral administration includes, e.g., intravenous, intramuscular, intra arterial, intradermal, subcutaneous, intraperitoneal, intraventricular, intraosseous, and intracranial. Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.
- compositions may be administered to a subject at risk of developing a particular disease, condition, or symptom, or to a subject reporting one or more of the physiological symptoms of a disease, even though the disease, condition, or symptom may not yet be present.
- the term“therapeutically effective amount” or“sufficient amount” refers to the amount of a system, recombinant polynucleotide, or composition described herein that is sufficient to effect beneficial or desired results.
- the therapeutically effective amount may vary depending upon one or more of: the subject and disease condition being treated or prevented, the weight and age of the subject, the severity of the disease condition, the immune status of the subject, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
- the specific amount may vary depending on one or more of: the particular agent chosen, the target cell type, the location of the target cell in the subject, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, and the physical delivery system in which it is carried.
- an effective amount is determined by such considerations as may be known in the art.
- the amount must be effective to achieve the desired therapeutic effect in a subject suffering from, e.g., HIV infection, or to prevent an infection, e.g, by HIV.
- the desired therapeutic effect may include, for example, amelioration of undesired symptoms associated with the disease, prevention of the manifestation of such symptoms before they occur, slowing down the progression of symptoms associated with the disease, slowing down or limiting any irreversible damage caused by the disease, lessening the severity of or curing the disease, or improving the survival rate or providing more rapid recovery from the disease.
- the amount may also be effective to prevent the development of the disease or to prevent an infection (e.g, HIV infection).
- “pharmaceutically acceptable carrier” refers to a substance that aids the administration of an active agent to a cell, an organism, or a subject.“Pharmaceutically acceptable carrier” also refers to a carrier or excipient that can be included in the compositions described herein and that causes no significant adverse toxicological effect on the patient.
- Non-limiting examples of pharmaceutically acceptable carriers include water, sodium chloride (NaCl), normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors and colors, liposomes, dispersion media, microcapsules, cationic lipid carriers, isotonic and absorption delaying agents, and the like.
- the carrier may also comprise or consist of substances for providing the formulation with stability, sterility and isotonicity (e.g, antimicrobial preservatives, antioxidants, chelating agents and buffers), for preventing the action of microorganisms (e.g, antimicrobial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid and the like) or for providing the formulation with an edible flavor, etc.
- the carrier is an agent that facilitates the delivery of a polypeptide, fusion protein, or polynucleotide to a target cell or tissue.
- pharmaceutical carriers are useful in the present disclosure.
- the term“vaccine” refers to a biological composition that, when administered to a subject, has the ability to produce an acquired immunity to a particular pathogen or disease in the subject.
- a vaccine is used to produce an acquired immunity against HIV infection.
- one or more antigens, fragments of antigens, or polynucleotides encoding antigens or fragments of antigens that are associated with the pathogen or disease of interest are administered to the subject.
- cells are engineered to express proteins such that, when administered as a vaccine, they enhance the ability of a subject to acquire immunity to an organism that causes an infectious disease, e.g., HIV.
- a vaccine refers to an HIV (or SIV) vaccine, e.g, a DNA vaccine encoding one or more HIV (and/or SIV) proteins, e.g, Env, Gag, protease, reverse transcriptase, Rev, and combinations thereof, or a composition comprising an HIV protein, e.g, gpl40.
- the present disclosure provides methods and compositions for enhancing the anti- Env antibody response during HIV vaccination in a subject, e.g, during vaccination using a prime DNA vaccine encoding one or more HIV (or SIV) proteins, and/or during a boost step in which an HIV protein is administered.
- the methods and compositions involve the administration of one or more adjuvants, e.g, during the prime and/or boost step, in order to enhance the Thl-Tfh responses in the subject.
- Thl -polarization promoting compounds can be administered in coordination with a prime vaccination step, e.g, by administering to the subject a DNA vaccine encoding one or more HIV polypeptides and a Thl -polarizing protein.
- the adjuvant is a protein such as IP- 10, and a polynucleotide encoding IP- 10 is administered to the subject.
- a single DNA vector is administered one or more times to the subject that comprises polynucleotides encoding both the one or more HIV (or SIV) proteins and the adjuvant such as IP- 10.
- the Thl -polarizing protein administered in coordination with the DNA prime is interferon-induced protein 10 (IP- 10).
- IP- 10 is also known as, e.g., CXCL10 or INP10, and corresponds, e.g, to NCBI Gene ID No. 3627.
- a polynucleotide comprising the human IP-10/CXCL10 mRNA of NCBI Reference Sequence NM_001565, or of a polynucleotide sequence comprising at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to the nucleotide sequence of NM 001565, or encoding the polypeptide of NCBI Reference Sequence NP_001556.2, or encoding a polypeptide having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity to NCBI Reference Sequence NP_001556.2, is used.
- a polypeptide comprising the human IP-10/CXCL10 protein of NCBI Reference Sequence NP 001556.2, or comprising a polypeptide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to NP_001556.2, is used.
- a polynucleotide encoding IP-10 is used, and is administered to the patient on a DNA plasmid, e.g, a single DNA plasmid comprising polynucleotides encoding one or more HIV polypeptides and encoding IP- 10.
- Thl polarization can also be promoted by administering to the subject a Thl- polarizing compound, e.g, adjuvant, in conjunction with one or more protein boosts, e.g, administration of an HIV protein such as gpl40.
- a Thl- polarizing compound e.g, adjuvant
- one or more protein boosts e.g, administration of an HIV protein such as gpl40.
- the boost protein e.g, gpl40
- the adjuvant QS-21 is administered together with the adjuvant QS-21.
- QS-21 can be used in the present methods, including natural, semi-synthetic, and synthetic forms, and that QS-21 can be used at a variety of amounts, e.g, at 50 pg/500 m ⁇ of vaccine dose, or 40-60 pg/500 m ⁇ , 30-70 mg/500 m ⁇ , 20-80 mg/500 m ⁇ , 10-90 mg/500 m ⁇ , or 1-100 mg/500 m ⁇ of vaccine dose.
- a Thl -polarizing agent e.g, a polynucleotide encoding a Thl -polarizing protein such as IP-10, or an adjuvant such as QS-21
- an HIV antigen e.g, a DNA molecule encoding an HIV protein or a protein boost
- the agent and the antigen can be administered in any of a variety of ways with respect to one another.
- the agent can be co-administered with the antigen, e.g, co-formulated and administered together to a subject.
- a single DNA plasmid is administered that encodes one or more HIV (or SIV) proteins, e.g., Env, and also encodes IP-10.
- multiple plasmids or vectors e.g, one or more plasmids encoding one or more HIV (or SIV) proteins, and a separate plasmid encoding IP-10, are formulated together.
- a polypeptide boost such as gpl40 is co-formulated with a Thl-promoting adjuvant, e.g, QS-21.
- the Thl -polarizing agent is formulated independently from the HIV antigen, but they are administered at about the same time (e.g, within 10 minutes, 30 minutes, or 1 hour of each other, or during a single clinical visit, or on the same day). In other embodiments, the Thl -polarizing agent is formulated independently from the HIV antigen, and they are administered at different times (e.g, not during a single clinical visit, or not on the same day, or at least 1, 2, 3 or more days apart). In such embodiments, the Thl -polarizing agent can be administered before or after the administration of the HIV antigen.
- HIV vaccines described herein can take on any of a number of forms, including through the administration of proteins, peptides, and nucleic acids including RNA or DNA encoding one or more HIV (or SIV) proteins.
- the vaccine is a DNA vaccine, e.g, a DNA vector such as a DNA plasmid comprising one or more polynucleotides encoding one or more HIV proteins, operably linked to one or more promoters.
- the DNA vaccine comprises a polynucleotide encoding the Env protein, or an immunogenic fragment or derivative thereof.
- the DNA vaccine encodes multiple HIV proteins, e.g, any combination of Env, Gag, protease, reverse transcriptase, tat, and rev, or immunogenic fragments or derivatives thereof.
- the HIV proteins can be from HIV-1 or HIV- 2, from any group, e.g, HIV-1 group M, group N, group O, or group P, or HIV-2 group A, B, C, D, E, F, G, or H, and from any sub-group or clade, e.g, HIV-1 group M clade (sub-type) A, B, C, D, E, F, G, H, I, J, or K, or any strain, variant, or isolate from within any of such groups, sub-groups or clades.
- one or more of the proteins used for vaccination are from HIV-1 clade C.
- each encoded protein can be from one clade, strain or variant/isolate, one or more proteins can be from a second clade, strain, or variant/isolate, etc. Any of these proteins can be encoded as a full-length protein, or can be encoded as a fragment of the full-length protein, e.g., as a minimal peptide sequence capable of eliciting an immune response in a subject.
- Any protein encoded by the HIV genome e.g, gpl20 env, gpl40 env, gpl60 env, pi 8, gag, pol, vif, vpr, vpu, tat, ref, and nef, or any fragment, derivative, or combination of any of these proteins, can be used during the prime immunization phase (e.g, using polynucleotides encoding the protein, peptide, fragment, or derivative) and/or during the protein boost phase.
- polynucleotides e.g, DNA plasmids, encoding one or more HIV proteins and/or IP- 10
- the polynucleotides are typically present within one or more expression cassettes, i.e., the polynucleotides are operably linked to one or more promoters.
- Any promoter capable of driving expression of the polynucleotides in one or more cells of a subject can be used, including inducible and constitutive promoters.
- the CMV promoter is used.
- all of the one or more HIV proteins and IP- 10 are operably linked to a single promoter, e.g, a CMV promoter, that drives constitutive expression in cells, leading to the expression of a single mRNA from which the different proteins are expressed by subgenomic splicing and frameshifting.
- a single promoter e.g, a CMV promoter
- the DNA vaccine comprises a pGA2/JS2 plasmid, i.e., a DNA vector that encodes one or more of the HIV-1 Env, Tat, Rev, Gag, Protease, and reverse transcriptase proteins under the control of the CMV vector.
- a polynucleotide encoding IP- 10 is integrated into the vector, e.g, downstream of the env gene.
- Plasmid constructs can be produced, maintained, and cultured using standard molecular biology techniques, e.g, as taught in Sambrook el al. , (1989),“Molecular Cloning: A Laboratory Manual”, (2nd ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y.; and Ausubel et al. (Eds), (2000-2010),“Current Protocols in Molecular Biology”, John Wiley and Sons, Inc.
- the function of the constructs can be assessed, e.g, by transfection into human cells, e.g, 293 T cells, and then grown, harvested, and assessed with respect to the expression of HIV proteins using, e.g, flow cytometry using monoclonal antibodies.
- cellular and secreted expression of IP-10 or other Thl- polarizing proteins can be assessed, e.g., by intracellular cytokine signaling and/or ELISA.
- Recombinant polynucleotides can be prepared using standard molecular biology methods.
- Polynucleotides encoding protein antigens can be identified, e.g, by searching a human or other model organism DNA sequence database for any gene segment that has a certain percentage of sequence homology to a known nucleotide sequence, such as one encoding the antigen. Any DNA sequence so identified can be subsequently obtained by chemical synthesis and/or a polymerase chain reaction (PCR) technique such as the overlap extension method. For a short sequence, completely de novo synthesis may be sufficient; whereas further isolation of full length coding sequence from a human or other model organism cDNA or genomic library using a synthetic probe may be necessary to obtain a larger gene.
- PCR polymerase chain reaction
- a nucleic acid sequence can be isolated from a cDNA or genomic DNA library (e.g, human or rodent cDNA or human, rodent, bacterial, or viral genomic DNA library) using standard cloning techniques such as polymerase chain reaction (PCR), where homology-based primers can often be derived from a known nucleic acid sequence.
- PCR polymerase chain reaction
- degenerate oligonucleotides can be designed as primer sets and PCR can be performed under suitable conditions (see, e.g, White el al. , PCR Protocols: Current Methods and Applications, 1993; Griffin and Griffin, PCR Technology, CRC Press Inc. 1994) to amplify a segment of nucleotide sequence from a cDNA or genomic library. Using the amplified segment as a probe, the full-length nucleic acid encoding a protein of interest is obtained.
- Oligonucleotides used in the construction of the recombinant polynucleotides that are not commercially available can be chemically synthesized, e.g, according to the solid phase phosphoramidite triester method first described by Beaucage & Caruthers, Tetrahedron Lett. 22: 1859-1862 (1981), using an automated synthesizer, as described in Van Devanter et. al. , Nucleic Acids Res. 12: 6159-6168 (1984). Purification of oligonucleotides is performed using any art-recognized strategy, e.g, native acrylamide gel electrophoresis or anion-exchange HPLC as described in Pearson & Reanier, J. Chrom.
- the coding sequence can be further modified by a number of well-known techniques such as restriction endonuclease digestion, PCR, and PCR-related methods to generate coding sequences, including mutants and variants derived from the wild-type protein.
- the polynucleotide sequence encoding the desired polypeptide can then be subcloned into a vector, for instance, an expression vector, so that a recombinant polypeptide can be produced from the resulting construct. Further modifications to the coding sequence, e.g., nucleotide substitutions, may be subsequently made to alter the characteristics of the polypeptide.
- a variety of mutation-generating protocols are established and described in the art, and can be readily used to modify a polynucleotide sequence encoding a protein of interest. See, e.g., Zhang el al, Proc. Natl. Acad. Sci. USA, 94: 4504-4509 (1997); and Stemmer, Nature , 370: 389-391 (1994).
- the procedures can be used separately or in combination to produce variants of a set of nucleic acids, and hence variants of encoded polypeptides. Kits for mutagenesis, library construction, and other diversity -generating methods are commercially available.
- a recombinant polynucleotide or vector described herein contains a nucleic acid sequence that encodes a selectable marker.
- a selectable marker is useful, for example, when a polynucleotide described herein is being recombinantly modified. Taking antibiotic resistance genes as an example of a selectable marker, treating the cells that contain the recombinant polynucleotides with the antibiotic will identify which cells contain recombinant polynucleotides that have incorporated the antibiotic resistance gene (i.e., the cells that survive after antibiotic treatment must have incorporated the antibiotic resistance gene).
- the recombinant polynucleotides can be further screened (e.g, purified from the cells, amplified, and sequenced), in order to verify that the desired modification has been recombinantly introduced into the polynucleotide at the correct position.
- the subject will receive one or more boosts with HIV polypeptide antigens, e.g, gpl40.
- the boost can be administered any of a number of times following the immunization, e.g, 1, 2, 3, 4, 5 or more times, and at different intervals, e.g, every 1, 2, 3, 4, 5, 6, 7, 8, weeks or more.
- the booster is administered two times, e.g, at weeks 30 and 44 (with week 0 corresponding to the date of the first prime immunization).
- the boost can be administered at any of a variety of amounts, e.g., 100 pg protein/boost, or 10-50, 50-100, 100-150, or 150-200 pg protein/boost, or 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 200, 250, 300 or more pg protein, e.g, gpl40, per boost.
- amounts e.g., 100 pg protein/boost, or 10-50, 50-100, 100-150, or 150-200 pg protein/boost, or 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 200, 250, 300 or more pg protein, e.g, gpl40, per boost.
- the booster polypeptide can be administered with one or more other adjuvants.
- the booster can be administered with a liposomal and/or lipid adjuvant, e.g, Army Liposome Formulation (ALF) liposomes.
- ALF Army Liposome Formulation
- ALF liposomes comprise dimyristoyl phosphatidylcholine (DMPC), dimyristoyl phosphatidylglycerol (DMPG) saturated phospholipids, cholesterol (chol), and monophosphoryl lipid A (MPLA), e.g., synthetic MPLA (e.g., 3D-PHAD), each of which can be obtained commercial suppliers such as Avanti Polar Lipids (Alabaster, AL).
- DMPC dimyristoyl phosphatidylcholine
- DMPG dimyristoyl phosphatidylglycerol
- MPLA monophosphoryl lipid A
- Other liposomes that can be used comprise CAFOl, an adjuvant that promotes Thl polarization and which can therefore be used in the place of, or in addition to, QS-21.
- lipids can be mixed in a molar ratio of, e.g, 9: 1 : 12.2:0.36 (DMPC:DMPG:Chol:MPLA), dried, rehydrated with, e.g., Sorenson PBS (pH 6.2), followed by microfluidization and filtration.
- the HIV protein, e.g, gpl40 can then be mixed with ALFQ in a 1 : 1 volume ratio, and vaccine doses can comprise, e.g, 100 pg MPLA, 100 pg protein, and 50 pg QS-21 in a total volume of 500 pi.
- the present methods and compositions can be used for the immunization of any subject, e.g, a human, that could benefit from an enhanced immune response against HIV infection, e.g, an enhanced anti-Env antibody response during HIV vaccination.
- the subject is male.
- the subject is female.
- the subject is an adult (e.g, an adult male).
- the subject is an adolescent.
- the subject is a child.
- the subject has not been infected with HIV, e.g, and the methods and compositions are used to enhance the subject’s immune defenses against HIV in order to prevent future infection.
- the subject is infected with HIV, and the methods are used to enhance the subject’s immune response against HIV in order to slow or potentially reverse the original infection.
- the present methods and compositions provide for enhanced immune responses with vaccination against HIV (and/or SIV).
- the present methods and compositions provide for enhanced Thl polarization of Tfh cells.
- Thl polarization enhances the immune response by increasing the Env-specific Tfh cells when delivered with the DNA prime, and that Thl polarization with a protein boost results in greater production of the IgGl subclass with enhanced longevity, breadth, avidity, ADCC, and ADP activities of the anti-Env antibody.
- Tfh cells e.g., germinal center (GC) Tfh cells
- GC germinal center
- Suitable markers include, but are not limited to, CXCR3, T-bet, IFNy, CXCR3, BCl-6, TBX21, SemA4A, and CCR5.
- the administration of a Thl -polarizing adjuvant leads to an increase in or or more of the herein-described Thl polarization markers, e.g, to an increase relative to the level in the absence of the Thl -polarizing adjuvant.
- the efficacy of an Thl -polarizing adjuvant is assessed by detecting one or more aspects of the immune response in a subject to an HIV antigen, e.g, Env.
- an HIV antigen e.g, Env.
- the extrafollicular and/or plasma cell-derived titers e.g, vs. gpl40
- BAMA binding antibody multiplex assay
- ELISA enzyme-linked immunosorbent assay
- the administration of a Thl- polarizing agent in coordination with the administration of an HIV antigen leads to an increase in an extrafollicular and/or plasma cell-derived antibody response against an HIV protein, e.g, gpl40.
- the increase in antibody response can be, e.g, 1.25, 1.5, 1.75, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10 or more fold higher in subjects receiving a Thl -polarizing adjuvant than in subjects not receiving a Thl -polarizing adjuvant.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the breadth, e.g, cross-clade breadth, of serum IgG antibodies relative to the breadth in a subject not receiving a Thl -polarizing adjuvant.
- the breadth of the antibodies is assessed by determining the kinetics of the antibody response against different HIV antigens, e.g., titers against Env from different HIV clades.
- Antibody binding against different HIV proteins e.g, against HIV Env proteins from different Groups, Clades, or isolates, e.g, against Con S (group M consensus), Con C, or isolate Case A2 or CH505 proteins, can be assessed, e.g, using a binding antibody multiplex assay (BAMA) or ELISA.
- BAMA binding antibody multiplex assay
- binding to gpl20 V1V2 loops is assessed, e.g, scaffolded on murine leukemia virus (MLV) gpl70.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the longevity of the serum IgG antibody response that is greater than any increase in longevity that may be observed in a subject not receiving a Thl -polarizing adjuvant.
- the elevation of anti-Env titers observed following one or more HIV protein boosts can be detected over time and the duration of the elevated antibody response assessed.
- titers are assessed at, e.g, week 8 and week 18 following the second protein boost, and the elevation observed in subjects receiving the Thl -polarizing adjuvant at the second time point (e.g, at 18 weeks following the second protein boost) is at least as high as any elevation observed in subjects not receiving the Thl -polarizing adjuvant.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the avidity (e.g, expressed as dissociation constants, kd) of serum IgG antibodies relative to the avidity in a subject not receiving a Thl -polarizing adjuvant.
- Increases in avidity can be assessed, e.g, by Surface Plasmon Resonance (SPR) or by 2M sodium thiocyanate or 0.1 M sodium citrate displacement ELISA with, e.g, gpl40 protein or with Con C, Con S, or other HIV proteins from other groups, clades, variants or isolates.
- SPR Surface Plasmon Resonance
- 2M sodium thiocyanate or 0.1 M sodium citrate displacement ELISA with, e.g, gpl40 protein or with Con C, Con S, or other HIV proteins from other groups, clades, variants or isolates.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the capacity of immune sera to neutralize HIV, e.g, HIV-1, e.g, as assessed using a TZM-bl assay, in which neutralization titers are measured in TZM-bl cells infected with HIV vs. cells infected with a control virus such as MLV.
- titers can be assessed against, e.g, subtype C tier 1A variants, tier IB variants, and tier 2 isolates.
- the induction of tier 1A neutralizing antibodies in a subject receiving a Thl- polarizing adjuvant is at least as high as any induction observed in a subject not receiving a Thl -polarizing adjuvant, at one or more time points following one or more protein boosts.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in antibody- dependent cellular toxicity (ADCC) and/or antibody-dependent phagocytosis (ADP) triggered by engagement of Fc receptors on antibody -bound target cells by innate cells.
- ADCC can be assessed, e.g., by measuring killing of HIV-infected target cells, e.g, by NK cells ( e.g. , CD16+/FcyR3 NK cells) in the presence of immune serum.
- ADP can be assessed, e.g, by measuring the phagocytosis of gpl20-coated beads by CD32+ (FcyR2) and/or CD64+ (FcyRl) THP-1 monocytic cells.
- the increase in ADCC and/or ADP is at least as high in a subject receiving a Thl -polarizing adjuvant as it is in a subject not receiving a Thl -polarizing adjuvant.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in anti-gpl20 IgGl antibodies relative to IgG2, IgG3, or IgG4 antibodies, e.g, as measured by ELISA.
- Thl -polarizing adjuvants lead to a specific increase in anti-gpl20 IgGl antibodies in subjects, and in particular to an increase that is as high or higher than any increase observed in subjects not receiving a Thl -polarizing adjuvant.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to a specific increase in the IgGl/IgG4 ratio among anti-gpl20 antibodies, and in particular to an increase that is as high or higher than any increase in the IgGl/IgG4 ratio in subjects not receiving a Thl -polarizing adjuvant.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in mucosal anti- gpl20 IgG and IgA antibodies relative to IgG2, IgG3, or IgG4, and in particular to an increase that is as high or higher than any increase observed in subjects not receiving a Thl- polarizing adjuvant.
- Thl -polarizing adjuvants lead to a specific increase in anti-gpl40 IgG and/or IgA antibodies in vaginal and/or rectal secretions, wherein the increase is stronger and/or has greater longevity than any increase seen in subjects not receiving a Thl -polarizing adjuvant.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in Env-specific Tfh cells and GC Tfh cells with distinctive Thl signatures.
- PBMCs peripheral blood mononuclear cells
- overlapping peptide pools comprising one or more gpl40 proteins
- activation markers such as CD25 and 0X40 assessed by flow cytometry after stimulation.
- the frequency of Env-specific CD4 T cells is higher in subjects receiving a Thl -polarizing adjuvant than in subjects not receiving a Thl -polarizing adjuvant, as is the percentage of Env-specific CD95+ CD4 T cells.
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in germinal center (GC) Tfh cells (as assessed, e.g., by virtue of CXCR5+, PD-1+++ expression) and GC B cells (as assessed, e.g, by virtue of Ki-67+, Bcl-6+, CD20 expression), and in particular to an increase that is as high as or higher than any increase seen in subjects not receiving a Thl- polarizing adjuvant.
- GC germinal center
- subjects receiving Thl -polarizing adjuvants can show higher Env-specific Tfh cell frequencies in the lymph nodes (LN) and can show a correlation between Tfh cell frequencies in the LN and GC Tfh cells, but not with memory Tfh cells, indicating that GC Tfh cells are enriched for vaccine-induced follicular cells.
- LN lymph nodes
- the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to a systemic expansion of pro- inflammatory monocytes and an increase in GC Tfh responses, e.g, during the priming immunization phase.
- Thl -polarizing antibodies increase the relative frequencies and absolute counts of ICOS+ PD-1 + CXCR5+ CD4 T cells in blood, e.g., at day 14 following a priming immunization step, as well as an increase in CD 14+ CD 16+ HLA-DR+ cells in blood.
- Thl -polarizing adjuvants lead to an increase in IL-Ib levels, e.g, as determined using a flow-based Legendplex assay, and to an increase in GC Tfh cell frequencies within fine-needle aspirates of the draining LN, that is as high or higher than any increase observed in subjects not receiving a Thl -polarizing adjuvant.
- a Thl -polarizing adjuvant leads to an increase of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, or more in any of the herein-described parameters or effects relative to a control value (e.g, to the value observed in a subject not receiving a Thl -polarizing adjuvant).
- a Thl- polarizing adjuvant can refer to any agent that, when administered with either a DNA prime against HIV, or with a protein boost against HIV, can lead to an increase of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, or more in any of the herein-described parameters or effects relative to a control value (e.g ., the value observed in a subject not receiving a Thl -polarizing adjuvant), at any point of the immunization process (e.g., at any one or more points of the priming phase and/or booster phase).
- a control value e.g ., the value observed in a subject not receiving a Thl -polarizing adjuvant
- compositions comprising an immunogenic component (e.g, a DNA vaccine or an immunogenic polypeptide) capable of inducing immunity against a targeted agent (e.g, HIV Env protein), comprising a Thl -polarizing adjuvant (e.g, a polynucleotide encoding IP-10 or an agent such as QS-21), and potentially comprising one or more additional adjuvants or compounds such as Army liposome formulations.
- a targeted agent e.g, HIV Env protein
- a Thl -polarizing adjuvant e.g, a polynucleotide encoding IP-10 or an agent such as QS-21
- the compositions further comprise a pharmaceutically acceptable carrier.
- the present disclosure provides pharmaceutical compositions for inducing an anti -Env (or other HIV protein or antigen) in a subject.
- the composition comprises one or more polynucleotides encoding one or more HIV proteins, e.g, Env, and one or more polynucleotides encoding a Thl -polarizing polypeptide, e.g, IP- 10, and a pharmaceutically acceptable carrier.
- the composition comprises one or more HIV polypeptides, e.g, gpl40, one or more Thl-promoting adjuvants, e.g, QS-21, and a pharmaceutically acceptable carrier.
- compositions may be formulated for, e.g, injection, inhalation, or topical administration, e.g, facilitating direct exposure of host cells and tissues to the immunogenic component and Thl -polarizing adjuvant.
- the compositions e.g, prime DNA vaccines
- the compositions are formulated for transdermal injection, e.g, by electroporation.
- the compositions, e.g, protein boost are formulated for subcutaneous or intramuscular administration, e.g, delivery into the thigh.
- compositions e.g, protein boost
- liposomes e.g, Army liposome formulations (ALFA)
- AFA Army liposome formulations
- the compositions are formulated in nanoparticles or in dry powder form, e.g, suitable for delivery by particle bombardment.
- the compositions, e.g, DNA vaccine are formulated as naked DNA (see, e.g, US Patent Nos. 6,265,387, 6,972,013, and 7,922,709).
- the DNA vaccines are prepared as DNA vectors or plasmids, e.g, a pGA2 plasmid, or pGA2/JS2 plasmid, or derivative thereof.
- the compositions, e.g, DNA vaccine are prepared as recombinant viruses, e.g, by modifying a parent virus to incorporate exogenous genetic material, e.g, one or more polynucleotides encoding one or more HIV proteins or antigens and/or a polynucleotide encoding IP-10.
- lentiviruses e.g, HIV, HIV-1, HIV-2, FIV, BIV, EIAV, MW, CAEV, SIV
- adenoviruses and adeno-associated viruses e.g., alphaviruses, flaviviruses, and poxviruses.
- suitable viral vectors see, e.g, US Patent Nos.
- the viruses are typically recombination-competent (i.e., capable of reproducing in an infected host cell). Modification of such viruses and vectors or plasmids for the preparation of the present DNA vaccines can be achieved using standard molecular biology techniques, e.g, as taught in Sambrook el al. (1989)“Molecular Cloning: A Laboratory Manual” (2 nd ed. Cold Spring Harbor Press) and Ausubel et al. (Eds.) (2000-2010)“Current Protocols in Molecular Biology” (John Wiley and Sons).
- compositions described herein may comprise a pharmaceutically acceptable carrier.
- pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical compositions described herein (see, e.g, REMINGTON’S PHARMACEUTICAL SCIENCES, 18TH ED., Mack Publishing Co., Easton, PA (1990)).
- compositions described herein will often further comprise one or more buffers (e.g, neutral buffered saline or phosphate buffered saline), carbohydrates (e.g, glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants (e.g, ascorbic acid, sodium metabi sulfite, butylated hydroxytoluene, butylated hydroxyanisole, etc.), bacteriostats, chelating agents such as EDTA or glutathione, solutes that render the formulation isotonic, hypotonic or weakly hypertonic with the blood of a recipient, suspending agents, thickening agents, preservatives, flavoring agents, sweetening agents, and coloring compounds as appropriate.
- buffers e.g, neutral buffered saline or phosphate buffered saline
- carbohydrates e.g, glucose, mannose, sucrose or dextrans
- compositions described herein are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically or prophylactically effective.
- the quantity to be administered depends on a variety of factors including, e.g., the age, body weight, physical activity, hereditary characteristics, general health, sex, and diet of the individual, the condition or disease to be treated or prevented, and the stage or severity of the condition or disease.
- the size of the dose may also be determined by the existence, nature, and extent of any adverse side effects that accompany the administration of a therapeutic or prophylactic agent(s) in a particular individual.
- Other factors that can influence the specific dose level and frequency of dosage for any particular patient include the activity of the specific compound employed, the metabolic stability and length of action of that compound, the mode and time of administration, and the rate of excretion.
- an effective amount of a pharmaceutical composition described herein is an amount that is sufficient to obtain an enhanced immune response against an HIV vaccine, e.g, in view of any of the parameters or indices described herein, and/or a sufficient amount to enhance the immunity of a subject to HIV infection or to the propagation of an already-existing HIV infection in the subject.
- the dose may take the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as, for example, tablets, pills, pellets, capsules, powders, solutions, suspensions, emulsions, suppositories, retention enemas, creams, ointments, lotions, gels, aerosols, foams, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages.
- unit dosage form refers to physically discrete units suitable as unitary dosages for humans and other mammals, each unit containing a predetermined quantity of a therapeutic or prophylactic agent calculated to produce the desired onset, tolerability, and/or therapeutic or prophylactic effects, in association with a suitable pharmaceutical excipient (e.g, an ampoule).
- a suitable pharmaceutical excipient e.g, an ampoule
- more concentrated dosage forms may be prepared, from which the more dilute unit dosage forms may then be produced.
- the more concentrated dosage forms thus will contain substantially more than, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times the amount of the therapeutic or prophylactic compound.
- the dosage forms typically include a conventional pharmaceutical carrier or excipient and may additionally include other medicinal agents, carriers, adjuvants, diluents, tissue permeation enhancers, solubilizers, and the like.
- Appropriate excipients can be tailored to the particular dosage form and route of administration by methods well known in the art (see, e.g, REMINGTON’S PHARMACEUTICAL SCIENCES, supra).
- compositions described herein can be administered locally or systemically to the subject, e.g, intraperitoneally, intramuscularly, intra-arterially, orally, intravenously, intracranially, intrathecally, intraspinally, intralesionally, intranasally, subcutaneously, intracerebroventricularly, topically, and/or by inhalation.
- the compositions are administered by intradermal injection with electroporation (e.g, for the priming DNA vaccine) or subcutaneously (e.g, for the protein boost).
- the DNA vaccines and/or polynucleotides encoding IP-10 or other Thl -polarizing protein are administered, e.g, by intradermal injection with electroporation (e.g, using the ICHOR TriGrid Array; Ichor Medical Systems), and can be administered any of a number of times, e.g, 1, 2, 3, 4, 5 or more times, and following any of a number of vaccination regimens, e.g, every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more weeks.
- the immunization is performed 3 times, e.g, at weeks 0, 8, and 16.
- DNA vaccines can be administered at any of a number of levels, e.g, 4 mg of plasmid DNA vector per subject per vaccination, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mg of plasmid DNA vector per subject per vaccination, or 1-10, 1-20, 1-8, 1-7, 2-6, 3-5 mg or plasmid DNA vector per subject per vaccination.
- the protein boost and/or QS-21 or other Thl -polarizing adjuvant is administered by intradermal injection, e.g, into the thigh, and can be administered any of a number of times following the immunization, e.g, 1, 2, 3, 4, 5 or more times, and following any of a number of regimens, e.g, every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 weeks or more.
- the protein boost can be administered at any of a number of levels, e.g., 100 pg protein/boost, or 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 200, 250, 300 or more pg protein, e.g., gpl40, per boost.
- the protein boost e.g, gpl40
- vaccine doses comprise, e.g, 100 pg MPLA, 100 pg protein, and 50 pg QS-21 in a total volume of 500 pi.
- kits are provided herein.
- the kit comprises a DNA prime composition (e.g, a DNA vaccine encoding one or more HIV proteins, e.g, Env, and also encoding a Thl -polarizing protein, e.g, IP-10, and optionally a pharmaceutically acceptable carrier).
- the kit comprises a protein boost composition (e.g, an HIV protein, e.g, gpl40, a Thl -polarizing adjuvant, e.g, QS-21, optionally one or more additional adjuvants, e.g, ALF, and optionally a pharmaceutically acceptable carrier).
- the kit comprises one or more DNA prime compositions and one or more protein boost compositions.
- the kit is for inducing an enhanced (e.g, stronger, broader, longer) immune response against an antigen, e.g, HIV Env protein (e.g, in a subject).
- the kit is for preventing or treating a disease, e.g, HIV.
- kits described herein can be packaged in a way that allows for safe or convenient storage or use (e.g, in a box or other container having a lid).
- the kits described herein include one or more containers, each container storing a particular kit component such as a reagent, a control sample, and so on.
- the choice of container will depend on the particular form of its contents, e.g, a kit component that is in liquid form, powder form, etc.
- containers can be made of materials that are designed to maximize the shelf-life of the kit components.
- kit components that are light-sensitive can be stored in containers that are opaque.
- the kit contains one or more elements, e.g, syringe, useful for administering compositions (i.e., a pharmaceutical composition described herein) to a subject.
- the kit further comprises instructions for use, e.g, containing directions (i.e., protocols) for the practice of the methods described herein (e.g, instructions for using the kit for enhancing an immune response in a subject to an HIV protein).
- directions i.e., protocols
- the instructional materials typically comprise written or printed materials, they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this disclosure. Such media include, but are not limited to electronic storage media (e.g ., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials.
- Tfh follicular helper CD4 T cells
- GC germinal center
- Thl polarization of Tfh cells is an important process shaping the success of Tfh-GC B cell interactions by influencing co-stimulatory and cytokine-dependent Tfh help to B cells.
- Env HIV-1 vaccine-mediated antiviral envelope
- IP interferon-induced protein
- the Thl vaccine regimen increased serum antibody effector functions, induced higher IgGl and lower IgG4 antibodies, increased circulating Env-specific Tfh cells and generated greater anti-Env mucosal IgA responses.
- the frequency of GC Tfh cells predicted both the magnitude and avidity of anti-Env serum IgG response at memory time points.
- ID intradermally
- EP electroporation
- DIP-10 ProteinALFQ vaccine induces robust and durable anti-Env antibody titers with cross-clade breadth.
- BAMA binding antibody multiplex assay
- Env ALFQ boosted animals developed significantly higher responses against C.1086 gpl40; median AUC values in ALFA (Thl+2) and ALFQ (Thl) vaccine groups were: wk 0, 7496 and 20301, p ⁇ 0.01; wk 2, 46481 and 63469, p ⁇ 0.001; wk 8, 20714 and 36709, p ⁇ 0.0001 2nd protein boost.
- DIP-10 ProteinALFQ vaccine elicits high avidity anti-Env antibody with ADCC and ADP activities.
- IgG binding antibodies as disassociation constants, kd
- SPR Surface Plasmon Resonance
- C.1086 gpl40 protein 23
- the data showed that gp 140-specific antibodies reached higher avidity with each sequential immunization in both vaccine groups (p ⁇ 0.0001, FIGS. 3A and 3B).
- Increased avidities in the Thl vaccine group relative to the Thl+2 group was suggestive of more productive GC reactions in the Thl vaccine group (p ⁇ 0.05, FIG. 3C).
- ADCC antibody-dependent cellular cytotoxicity
- ADP antibody-dependent phagocytosis
- serum from Thl vaccinated animals demonstrated significantly greater ADCC activity at week 2 and week 8 after the 2nd protein boost when compared to Thl+2 immunized animals (p ⁇ 0.01).
- Serum collected from Thl vaccinated animals at week 8 post protein boost 2 also mediated significantly greater phagocytosis of C.1086 gpl20-coated beads by the CD32+ (FcyR2) and CD64+ (FcyRl) THP-1 monocytic cell line (FIGS. 3J and 3K).
- DNAIP-lOProteinALFQ vaccine elicits robust anti-Env antibody in vaginal and rectal mucosal compartments. Having established induction of higher serum IgG antibody titers in Thl vaccinated animals, we next sought to determine whether mucosal anti-Env antibodies were also correspondingly increased. To this end, we assayed rectal and vaginal secretions for C.1086 gpl40-specific IgG and IgA antibodies at baseline and longitudinally after each of the protein boosts.
- gp 140-specific IgG in secretions closely mimicked the kinetics of the serum IgG antibody response, with each protein boost increasing levels of Env-specific IgG antibodies in vaginal and rectal secretions (FIGS. 4A and 4B).
- the Thl vaccine regimen generated higher levels of specific IgG in secretions when compared to the Thl+2 vaccine.
- the gp 140-specific IgA in vaginal and rectal secretions was also increased to a greater extent by the Thl vaccine regimen (FIGS. 4C and 4D).
- vaginal IgA antibodies were still above the limit of detection in most Thl vaccinated animals but in only 2 of 10 Thl+2 vaccinated animals.
- Analysis of gp 140-specific IgA in serum revealed higher induction in the Thl group (FIG. 4E).
- the kinetics of the serum IgA response in Thl as well as Thl+2 animals differed strikingly from the mucosal IgA responses, especially in the reproductive tract (FIGS. 4C and D), suggesting a true mucosal (locally-derived) IgA response was generated in these animals.
- DIP- 10 ProteinALFQ vaccine induces Env-specific Tfti cells and GC Tfh cells with distinctive Thl signatures.
- the DIP-10 PALFQ Thl vaccine promoted anti-Env antibody longevity and functionality following the 1st protein boost. Based upon this finding, we wanted to determine whether this vaccine regimen also correspondingly enhanced Tfh responses in the periphery and LNs. To this end, we first assessed whether higher magnitude of Env-specific Tfh cells were induced 7 days after 1st boost, corresponding to the peak of the effector response. PBMCs were stimulated with overlapping peptide pools representing Con C gpl40 together with C.1086 gpl40 protein.
- the induction of the activation markers CD25 and 0X40 was assessed by flow cytometry after stimulation (FIG. 5A, flow plot) (30).
- the analysis revealed a higher frequency of Env-specific CD4 T cells in DIP-10 PALFQ animals.
- median frequencies of Env specific-CD4 T cells were on average 10-fold higher in Thl group, indicative of a higher magnitude Env-specific Tfh response (p ⁇ 0.001, FIG. 5B).
- the frequency of Env-specific CD4 T cells responding to the C.1086 gpl40 priming immunogen correlated positively with CD4 T cells responding to the C.ZA gpl40 boosting immunogen (p ⁇ 0.0001, FIG. 5C).
- IP- 10 levels in sera using a flow-based Legend plex assay at day 0 and 3 of 1st protein boost in the ALFQ-adjuvanted animals confirmed induction of IP- 10 following the Thl protein boost (p ⁇ 0.05, FIG. 5D).
- CD4 subsets from the LNs of 3 Thl group animals with highest antibody titers at week 8 post 1st protein boost. These subsets were naive cells (CD4+CD95- ), Tfh cells (CD95+CXCR5+PD-1 +/++), memory Tfh cells (CD95+CXCR5+ PD-1-), and memory non-Tfh cells (CD95+ CXCR5-PD-1-). Similar subsets were sorted from 2 high responder animals in the Thl+2 vaccine group (animal sorted identified by triangles, FIG. 6H).
- RNA samples meeting quality control checks were sequenced using a 3'-Tag-RNA- Seq library prep protocol on the Illumini HiSeq 4000 platform. Prior to analysis of sequenced reads, genes with fewer than 40 counts per million reads were filtered, leaving 7,086 genes. Differential expression analyses were conducted using the limma-voom Bioconductor pipeline. We performed significance analysis of microarrays using the Dseq package from R/Bioconductor to compare the transcriptome profiles. Principal component analysis of the top 500 differentially expressed genes showed that the majority of variation in the data was driven by CD4 subsets.
- Memory CD4 T cells (CXCR5+ and CXCR5-) clustered closely together relative to naive and Tfh subsets (FIG. 61). Segregation of Tfh cells between the Thl and Thl+2 groups indicated adjuvant dependent modulation of transcriptional activity within the germinal center, suggestive of qualitative differences in Tfh cells between vaccine groups.
- To extract information on biologically relevant gene-sets we performed gene set enrichment analysis with the goal of determining biological pathways that were enriched in Tfh cells in the Thl vaccine regimen. Genes regulating interleukin (IL)-12, tumor necrosis factor (TNFa), interferon gamma (IFNG), and IL-6 production were strongly enriched in Tfh cells. Consistent with metabolic activity of effector cells and functional capacity of Tfh cells, pathways regulating cellular metabolism, glucose homeostasis, and B cell proliferation were also enriched (FIG. 6J).
- IL interleukin
- TNFa tumor necrosis factor
- SEMA4A semaphorin protein
- Thl cells a co-stimulatory molecule expressed by Thl cells (32)
- HMGB1 high-mobility group box 1
- IL-18R inflammatory mediator regulating TNF and IL-6 production
- IL-18R receptor interacting serine/threonine kinase 2 (RIPK2), which drives IFNy in Thl cells and contributes to Thl differentiation
- RIPK2 receptor interacting serine/threonine kinase 2
- the corresponding downregulation of IL-4R in Tfh cells indicated enrichment of the Thl program within Tfh cells in DIP- 10 ProALFQ vaccinated animals. This, together with increased protein expression of CXCR3 within the GC, supports the conclusion that CD4 T cell help for humoral immunity was driven by Thl Tfh cells in the Thl vaccine regimen (FIG. 61).
- DNAIP-10 immunization induces systemic expansion of pro-inflammatory monocytes and enhances GC Tfh responses. Based on increased frequencies of Env-specific Tfh cells and evidence for induction of a Thl transcriptome program in Thl vaccinated animals following the 1st protein boost, we sought to assess Tfh responses during the priming immunization phase. First, we evaluated blood to quantify activated CXCR5+ CD4 T cells (FIG. 7A).
- the improved inflammatory response was associated with increased antibody longevity linking the innate immune response to effective induction of CD4 Tfh cells.
- potent priming of the immune response sets the stage for stronger boosting of cellular and humoral immunity in the setting of DNA prime, NYVAC boost and Ad5 prime, NYVAC boost vaccine regimens (25, 40).
- the effectiveness of priming is not limited to CD4 T cells and B cells; a DNA vaccine targeting conserved elements of SIV Gag robustly primes cytotoxic T cells which are effectively boosted following a long rest period (41, 42).
- the HVTN studies 070 and 080 employed the IL-12 DNA adjuvanted plasmid with the subtype B PENNVAX-B (PV) DNA plasmid and showed 80% response rates after the third DNA vaccination in PV+IL-12 recipients compared to a 44% response rate with the PV alone vaccine.
- a subsequent follow up study demonstrated robust recall of binding anti-Env antibody titers with ADCC activity following an MVA boost in PV+IL-12 recipients (43, 44). Because IL-12 is a classic innate mediator of Thl responses, the data suggest that an increase in Thl GC Tfh cells may underlie the observed effects.
- IP- 10 increases dendritic cell-T cell interactions, which could have favored Tfh differentiation (48). IP- 10 also increases IL-6 production in B cells which is known to support Tfh differentiation and enhance plasma cell differentiation (49). This, together with the potent immune stimulatory potential of MPL+QS-21 boost, may have synergized to enhance Tfh responses numerically and favored Thl differentiation program within Tfh cells (50).
- Thl-type Tfh cells GC Tfh cells induced following viral infections where Thl inflammatory responses predominate express Bcl-6, Tbx21, IFNG, and IL-21 consistent with the induction of Thl-type Tfh cells (51).
- Our transcriptomic analysis of Tfh cells following the 1st protein boost in the Thl vaccine regimen show coordinate expression of Thl regulated genes as evidenced by enrichment of pathways related to IFNG signaling.
- the higher relative expression of the Thl chemokine receptor CXCR3 in GC Tfh cells and GC B cells validate the gene expression data.
- Thl/Thl7 adjuvant cationic liposomal formulation suggests that induction of Thl/Thl7-type Tfh cells may be extremely important to drive robust mucosal IgA responses while correspondingly enhancing serum anti-Env IgG responses and functionality (52).
- Thl-DNA prime substantially increases the Env-specific Tfh cells relative to a Thl+Th2 vaccine regimen and that Thl-protein boost results in greater production of the IgGl subclass with enhanced longevity, breadth, avidity, ADCC, and ADP activities of anti-Env antibody.
- DNA immunizations were administered via intradermal injection with electroporation utilizing the ICHOR TriGrid Array (Ichor Medical Systems, San Diego) at weeks 0, 8, and 16.
- ICHOR TriGrid Array Ichor Medical Systems, San Diego
- two groups of 10 animals received 4 mg of the pGA2/JS2 plasmid DNA vector (53) encoding either SHIV C.1086 T/F Env + interferon-induced protein (IP)-10 (Group 1) or SHIV C.1086 T/F Env alone (Group 2). Details of the SHIV DNA construct have been described (54).
- Group 1 animals received boosts with 100 pg C.ZA 1197MB gpl40 protein (Immune Technology, New York, NY) adjuvanted with 100 pg MPLA +50 pg QS-21 (ALFQ) and Group 2 animals received 100 pg C.ZA 1197MB gpl40 adjuvanted with 100 pg MPLA + 600 pg Aluminum (ALFA).
- the protein formulation was delivered in a 250 pi volume (50 pg protein) subcutaneously in each thigh during both boosts.
- Adjuvants Dimyristoyl phosphatidylcholine (DMPC) and dimyristoyl phosphatidylglycerol (DMPG) saturated phospholipids, cholesterol (Choi), and synthetic monophosphoryl lipid A (MPLA, 3D-PHAD) were purchased from Avanti Polar Lipids (Alabaster, AL). DMPC and Choi were dissolved in chloroform, and DMPG and MPLA were dissolved in chloroform: methanol (9: 1). Alhydrogel ® , aluminum hydroxide (AH) in a gel suspension was purchased from Brenntag (Frederikssund, Denmark). Saponin, QS-21 were purchased from Desert King International (San Diego, CA) and was dissolved in Sorensen PBS, pH 5.6.
- DMPC dimyristoyl phosphatidylcholine
- DMPG dimyristoyl phosphatidylglycerol
- MPLA synthetic monophosphoryl lipid A
- DMPC :DMPG: Choi :MPLA DMPC :DMPG: Choi :MPLA
- Liposomes were formed by molecular biology grade water (Quality Biological, Gaithersburg, MD), microfluidized, and sterile filtered, followed by lyophilization. 100 pg of gpl40 protein was adsorbed to 600 pg of Alhydrogel in PBS, pH 7.4, and incubated on a tilted roller at room temperature (RT) for 1 h prior to adding to lyophilized ALF.
- RT room temperature
- lipids were mixed in a molar ratio of 9: 1 : 12.2:0.36 (DMPC:DMPG:Chol:MPLA), dried, rehydrated by adding Sorensen PBS, pH 6.2, followed by microfluidization and filtration.
- gpl40 was mixed with ALFQ in a 1 : 1 volume ratio.
- Each vaccine dose in 500 pi volume contained 100 pg MPLA (and 100 pg protein) and either 600 pg aluminum or 50 pg QS-21.
- Lymph node (LN) biopsies were obtained 2 weeks following each of the protein boosts and were manually processed by disassociation through IOOmM cell strainers and washing in complete media, as described previously (12). Two weeks after the 3rd DNA immunization, fine needle aspirates of LN were obtained using a 22 gauge needle, as previously described (55).
- PBMCs were isolated from whole blood collected in CPT vacutainer tubes by density gradient centrifugation as previously described (12). For serum, coagulated blood was centrifuged at 800g for 10 min to pellet clotted cells, followed by extraction of fluid and storage at -80° C. Rectal and vaginal secretions were collected using premoistened Week-Cel sponges and eluted as described (56).
- Env gpl40 were determined by ELISA. In brief, 96-well microtiter plates with high binding capacity (Thermo Fisher, MA) were coated overnight at 4° C with 1 pg/mL C.1086 Env gpl40C from the NIH AIDS Reagent Program (ARP) diluted in 0.1M carbonate-bicarbonate buffer, pH 9.2. Plates were washed with PBS containing 0.1% Tween-20 (PBST) and blocked with 5% w/v nonfat dry milk in PBS for 2h at RT followed by four washes with PBST.
- ARP NIH AIDS Reagent Program
- Sodium thiocyanate avidity assay C.1086 Env gpl40C-specific IgG antibody avidity was determined using a chaotropic displacement ELISA with NaSCN. Serum samples were incubated in duplicate at 6000 pg per well for 2h at RT. The plate was washed five times. For the dissociation step, one well of each sample was manually treated with 100 pL of 2 M NaSCN (Sigma-Aldrich) to dissociate antigen-antibody complexes and a second well of the same sample was treated with PBS as a control. The plate was incubated for 15 min at RT, followed by washing three times.
- 2 M NaSCN Sigma-Aldrich
- the plate was then developed as described above for the C.1086 gpl40C ELISA.
- antibody avidity was reported as an avidity index value (a percentage), which was calculated as the ratio of absorbance in the well treated with NaSCN to that in the well treated with PBS.
- Binding and avidity determination were conducted using Surface Plasmon Resonance (SPR) Biacore 4000 system.
- the immobilizations were performed in 10 mM HEPES and 150 mM NaCl pH 7.4 using a standard amine coupling kit, as previously described (23, 57).
- the CM5-S series chip surface was activated with a 1 : 1 mixture of 0.4 M 1 -ethyl-3 -(3 -dimethylaminopropyl) carbodimide hydrochloride (EDC) and 0.1 M N-hydroxysuccinimide (NHS) for 600s (GE Healthcare).
- cyclic biotinylated V2 C.1086 peptide 1 mM Streptavidin (Life Technologies) in 10 mM sodium acetate pH 4.5 (5,800 - 7,400 RU) was coupled for 720s. The immobilized surface was then deactivated with 1.0 M ethanolamine-HCl pH 8.5 for 600s. Spot 3 in each flow cell was left unmodified to serve as a reference. Following surface deactivation, 0.06 - 1.5 mM cyclic biotinylated V2 C.1086 peptide was captured, resulting in two range of densities; high density (1,900 - 2,300 RU) and low/medium density (340 - 580 RU).
- the bound surface was then enhanced with a 240 s injection of 30 pg/mL secondary antibody goat anti -monkey IgG.
- 175 mM HC1 was injected for 70s.
- 4 - 8 replicates were collected at a rate of 10 Hz, with an analysis temperature of 25° C. All sample injections were conducted at a flow rate of 10 pL/min.
- Data analysis was performed using Biacore 4000 Evaluation Software 4.1 with double subtractions for unmodified surface and buffer for blank. The fitting was conducted using the dissociation mode integrated with the Evaluation software 4.1.
- Binding Antibody Multiplex Assay (BAMA) and sodium citrate avidity assay.
- SIV-specific serum IgG BAMA was performed with a panel of Env and VI V2 antigens: C.1086 gpl40, CH505 TF gpl40, Con S (group M consensus) gpl40, and Con C (clade C consensus) gpl40, gp70-VlV2 Clade B/Case A2 scaffolded protein and C.1086 VI V2 avitagged protein.
- Samples were titrated in 5-fold serial dilutions starting at 1 :80 and binding magnitude is reported as AUC.
- Positivity criteria was as follows: (1) MFI >100; (2) MFI > Ag-specific cutoff (95th percentile of all baseline binding per antigen); (3) MFI 3 -fold > than that of the matched baseline before and after blank/MuLV subtraction. All BAMA and avidity assays were performed in a blinded fashion using magnetic beads. For avidity assays, samples were tested with and without sodium citrate (0.1 M, pH 3.0) at 2 dilutions for each antigen based on BAMA titration for maximum coverage of samples in the linear range of the assay.
- the dilutions were 1 :80 and 1 :400 for gp70-VlV2, 1 :400 for C.1086 VI V2, 1 :2000 for CH505TF gpl40, 1 :2000 for ConC gpl40, and 1 : 10000 for C.1086 gpl40 and ConS gpl40.
- Antibody avidity is reported as avidity index, which was calculated as 100 x (MFI in the citrate-treated well/MFI in the untreated well). Avidity index is reported for sample-antigen combinations that were (1) identified as positive responders in the IgG BAMA assay and (2) had an MFI within the linear range for the untreated sample.
- the pre set assay criteria for sample reporting were coefficient of variation per duplicate values of ⁇ 15% for each sample and >100 beads counted per sample.
- Neutralization Neutralization assays were performed as previously described (58) using TZM-bl cells. We measured neutralization activity against the tier 1 clade C pseuodvirus MW965.26 using MLV-pseudotyped virus as an indicator of non-HIV-specific activity in the assay. Neutralization titers were measured at week 2 and week 8 post 2nd protein boost and were considered to be positive for neutralizing antibody activity based on the criterion of signal >3x detected against the MLV negative control virus. The majority of positive titers detected were against the tier 1 virus MW965.26 with occasional very weak neutralization titers against the tier 2 viruses.
- Luciferase based ADCC assays were carried out as previously described with some modifications (59). Two million CEM.NKR-CCR5-sLTR-Luc target cells were spinoculated with SHIV.C.CH505.375H.dCT (38 ng p27) for 2 hours at 2,600 rpm at 30° C in the presence of 1 pg/mL polybrene. Subsequently, the target cell/virus mixture was incubated overnight at 37° C 5% CO2. The next day, virus was removed and cells were incubated for another 72 hours prior to the ADCC assay. For the ADCC assay, serum: effector cells: target cells were plated in a 1 : 1 : 1 volumetric ratio.
- Sera was heat inactivated and diluted (1 :50 dilution in RIO containing 10 U IL-2 per mL, with no CsA), mixed with PBS-washed, infected target cells (1 x 10 4 cells per well), and effector cells (5 x 10 4 cells per well). Assay plates were incubated overnight at 37°C and 5% CO2. Plates were then centrifuged at 1,800 rpm for 5 min at room temperature and 100 pL of the supernatant was removed. The cell pellets were resuspended and mixed with 50 pi of the luciferase substrate reagent BriteLite Plus (Perkin Elmer, MA).
- Luciferase activity was read in black 96-well plates according to the manufacturer’s instructions using a Synergy 2 micro plate luminometer (BioTek). Percent ADCC activity of each tested animal immune serum (week 2 and week 8 post 2nd protein) measured as reduction in RLUs, was calculated based on respective week 0 pre-immune serum (100% RLU). All samples were tested in triplicates and experiments were performed twice.
- IgG subclass antibodies Ten rows of a 96-well Immulon 4 microtiter plate (VWR) were coated overnight at 4° C with 50 ng per well of C.1086 gpl20 D7 K160N protein (61) in PBS. The remaining 2 rows were coated with duplicate 2-fold serial dilutions of rhesus IgGl, IgG2, IgG3 or IgG4 (Nonhuman Primate Reagent Program) starting at 25 ng/mL in PBS to generate a standard curve. Plates were washed with PBS containing 0.05% Tween 20 and blocked for 30 min at RT with reagent buffer (0.1% bovine serum albumin in wash buffer).
- the plate was then consecutively washed and treated with 100 ng/mL of biotinylated goat anti-mouse IgGl or IgG2a for 1 h at 37° C, neutralite-avidin peroxidase for 30 min at RT, and TMB (all from SouthernBiotech). Absorbance was recorded at 370 nm. SoftMax Pro software (Molecular Devices) was used to to construct a standard curve and determine concentrations of antibody. Preimmune serum samples had ⁇ 10 ng/mL of antibody in these assays.
- Activation induced Marker (AIM) assay Cells were stimulated with overlapping peptide pools of HIV consensus C and HIV-1 C.1086 Env gpl40C protein in AIM media as previously described (30). All antigens were used at a final concentration of 2 pg/mL in a stimulation cocktail made with using 0.2 pg of CD28 and 0.2 pg CD49d costimulatory antibodies per test. Unstimulated controls were treated with volume-controlled DMSO (Sigma-Aldrich). Tubes were placed in 5% CO2 incubator at 37° C and incubated overnight. Following an 18 hour stimulation, the cells were stained, fixed, and acquired the same day.
- DMSO volume-controlled DMSO
- Phenotype panel on lymph nodes and PBMCs was performed using standard flow cytometry assays.
- Legend Flex assay A Legendplex assay was performed to evaluate cytokines in rhesus macaque sera (Biolegend). The assay was performed according to the manufacturer’s protocol. Samples were acquired on a BD LSR Fortessa cell analyzer.
- Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med 192: 1545-1552. 2 MacLennan IC. 1994. Germinal centers. Annu Rev Immunol 12: 117-139.
- T-bet Transcription Factor Promotes Antibody-Secreting Cell Differentiation by Limiting the Inflammatory Effects of IFN-gamma on B Cells. Immunity 50: 1172-1187 el 177.
- IL-1 acts on antigen-presenting cells to enhance the in vivo proliferation of antigen-stimulated naive CD4 T cells via a CD28-dependent mechanism that does not involve increased expression of CD28 ligands. Eur J Immunol 34: 1085-1090.
- Singh S Ramirez-Salazar EG, Doueiri R, Valentin A, Rosati M, Hu X, Keele BF, Shen X, Tomaras GD, Ferrari G, LaBranche C, Montefiori DC, Das J, Alter G, Trinh HV, Hamlin C, Rao M, Dayton F, Bear J, Chowdhury B, Alicea C, Lifson JD, Broderick KE, Sardesai NY, Sivananthan SJ, Fox CB, Reed SG, Venzon DJ, Hirsch VM, Pavlakis GN, Felber BK. 2018.
- This Example focuses on achieving this goal using a recombinant DNA-prime, modified vaccinia Ankara (MVA)-boost vaccine platform in path for clinical development.
- MVA modified vaccinia Ankara
- Our data show that generation of Tfh cells expressing the chemokine receptor CXCR3 correlates strongly with multiple attributes of protective humoral immunity.
- CXCR3+ Tfh cells express higher levels of B cell helper factors relative to CXCR3- Tfh cells.
- HIV vaccines designed to induce high magnitude CXCR3+ Tfh cells increases antibody persistence, enhancing protection against a mucosal HIV challenge.
- Tfh cells Akin to CD4 T helper (h) cells, Tfh cells also encompass distinct Thl or Th2 subsets, and several recent studies have demonstrated that Th polarity of Tfh cells critically determines outcome of antibody responses to various vaccines/infections. Yet, it remains unclear whether Thl or Th2-type Tfh cells regulate HIV vaccine induced antibody. Bridging this gap will greatly advance vector-based vaccine design strategies and rational selection of adjuvants for protein boosts to extend the functional quality and durability of the vaccine- elicited antibody against HIV.
- This vaccine regimen is evaluated in a monkey study with three vaccine groups to determine whether increase in Thl-polarized Tfh cells during the prime with a DIP- lOMPro Alum vaccine and during both the prime and the boost with a DIP-lOMProASOlB vaccine enhances durable and protective HIV antibody relative to a reference DMProAlum platform.
- the first approach is to delineate immune determinants and characterize gene expression profiles of vaccine-specific Thl versus Th2-polarized Tfh cells.
- a novel IP-10-DNA-SHIV vaccine is constructed and characterized ex vivo.
- cytokine/chemokine determinants are delineated, and single-cell transcriptional profiling is performed of Env-specific Thl and Th2 Tfh cells in all three outlined vaccine groups.
- the second approach is to determine whether Thl-polarized Tfh cells enhance magnitude, quality and persistence of Env antibody titers. Specifically, it is determined whether the DIP- lOMPro Alum and DIP-lOMProASOlB vaccines increase Env binding titers in sera and mucosa and enhance neutralizing and non-neutralizing antibody titers.
- the third approach is to determine whether a Thl-polarized vaccine regimen enhances mucosal protection against a SHIV challenge and augments viral control. Specifically, it is determined whether the DIP-lOMProASOlB vaccine enhances protection against a vaginal heterologous SHIV challenge and potentiates viral control in infected animals. In addition, the susceptibility of CXCR3+ CD4 Tfh cells to SHIV infection is determined ex vivo.
- Tfh cells Similar to canonical CD4 T cells (11), Tfh cells also encompass characteristics of Thl, Th2, and Thl7-type cell subsets which can be identified by expression of specific chemokine receptors (12). Several recent studies have demonstrated that Thl and Th2 Tfh cells possess differential capacity for B cell help, and the induction of a specific Tfh subset critically determines outcome of the antibody response to vaccination/infection (13-15).
- Thl-polarized CXCR3 -expressing Tfh cells have been associated with durable antibody titers following influenza immunization (13, 16).
- MV A modified vaccinia Ankara
- Thl adjuvants such as MF59 and AS01B
- Th2 adjuvant Alum in Phase I/II HIV vaccine trials makes this a highly timely and clinically relevant question in HIV/AIDS vaccine research (18-20).
- Vaccine groups 1, 2, and 3 are used to investigate the outcome of graded increase in Thl immune response on antibody durability in systemic and mucosal compartments. Vaccine groups 1 and 3, at the opposite ends of the Thl response spectrum, are directly compared for efficacy against an intravaginal heterologous Clade C SHIV.
- DNA/MVA vaccine induces robust anti-Env antibody.
- intramuscular immunization with DNA/MVA vaccines elicits immunologically coordinated humoral immune responses in periphery and mucosa (27, 28).
- Determination of Env-specific binding titers by ELISA over the course of immunization shows a temporal increase in Env titers, which achieve robust peak after the 2nd MVA boost (FIG. 14A).
- DNA/MVA vaccine elicits Tfh responses, which are discernable in peripheral blood.
- Our data demonstrate a transient accumulation of CXCR5+ Tfh cells expressing the cell- cycle marker Ki-67 at the peak CD4 effector response following the MVA boost (FIGS. 15A and B, *p ⁇ 0.05).
- IP-10 increases induction of CXCR3+ Tfh cells.
- PBMC peripheral blood mononuclear cells
- first approach In order to effectively harness Tfh help for HIV vaccine design, understanding heterogeneity of the vaccine-elicited Tfh response and identifying factors contributing to this heterogeneity are critical.
- the goal of the first approach is to extensively define the cytokine/chemokine response to vaccination and how this influences Tfh cell magnitude and polarity.
- This approach is further designed to acquire comprehensive insights into division of labor within vaccine-elicited Tfh cells by interrogating the transcriptional signature of Env- specific CXCR3+ Thl and CCR4+ Th2 Tfh cells.
- IP-10-DNA-SHIV IRES and F2A constructs ex vivo using a variety of different approaches which include determining antigenicity of constructs by evaluating expression of HIV-1 Env (surface, clone PGT121), SIV Gag (intracellular, clone 2F12) and IP-10 (intracellular, clone J034D6) by flow cytometry on transfected 293-T cells.
- Cell supernatants from 293-T transfected cells are also be collected for measurement of secreted IP- 10 by ELISA and for functional activity of IP- 10 using cell supernatants to stimulate PBMCs.
- We select either the IRES or the 2A construct based on efficiency of Env and IP- 10 expression for in vivo experiments.
- Another objective is to understand how the in vivo chemokine/cytokine response immediately following immunization influences CD4 Tfh development and differentiation, which is directed by cues early in the immune response (36).
- Studies in a mouse model of acute viral infection show that IP-10 is rapidly induced in a type I IFN dependent manner (37), and studies in macaques show transient induction of pro-inflammatory factors including IP- 10 early after MVA immunization (29). Therefore, to optimally capture the early and transient induction of immune factors, we collect blood at 0, 12, 24, 48, and 76 hours post immunization to establish a comprehensive cytokine /chemokine profile temporally, and across vaccine platforms.
- cytokine, chemokine profile using a multiplex assay to measure soluble factors driving Thl polarization: interleukins (IL)-12, IL-Ib, IL-8, IL-2; macrophage inflammatory protein (MIP)la, MIRIb, MIP3a, MIR3b; macrophage chemotactic protein (MCP)-l; CXCR3 ligands; monokine induced by gamma (MIG/CXCL9), PMO/CXCLIO, interferon (IFN)- inducible T-cell alpha chemoattractant (I-TAC/CXCL11); tumor necrosis factor (TNF)a; IFNs PTMa,b,g.
- Th2/regulatory factors including IL-4, IL-10, IL- Ra, transforming growth factor
- CD3+CD4+Fas+CXCR5+PD-1+/++ (comprising Tfh and GC Tfh) cells are sorted using fluorescence-assisted cell sorting (FACS) into CXCR3+ and CCR4+ single positive subsets prior to stimulation.
- FACS fluorescence-assisted cell sorting
- RNA sequencing methodology allows us to capture the heterogeneity of the Tfh cell response to stimulation and give us the ability to resolve important associations between cell phenotype, stimulation status (by quantifying genes encoding Tbet (Thl) and IL-4 (Th2) that are rapidly up regulated upon TCR stimulation (41)), and cellular transcriptional program.
- Initial assays are done with the challenge SHIV, the immunogen SHIV, and clade-matched tier 1A, tier IB and tier 2 viruses, which include SHIV-1157ipEL-p and Cel086_B2. Further assays are performed against a multi-clade global virus panel described previously (47). To assess non-neutralizing antibody effector functions, we measure antibody-dependent phagocytosis (ADP) using the Ackerman flow cytometric assay (48) and antibody dependent cellular cytotoxicity (ADCC) using the Evans assay (49). Our data (FIG. 22B) show induction of antibodies that direct ADCC and ADP following a DNA/MVA immunization. We also measure antibody glycosylation (23).
- ADP antibody-dependent phagocytosis
- ADCC antibody dependent cellular cytotoxicity
- a significant impediment to enhancing vaccine efficacy is the waning of Env antibody titers following vaccination (5, 50). While booster protein immunizations are a means to augment antibody titers, they present the concern of inefficient CD8 T cell recall and potential generation of target cells, which together can create an environment that favors initial viral replication upon exposure (51). Consequently, vaccine strategies to establish durable humoral immunity while inducing robust cytolytic responses are highly desirable.
- IP- 10 IFN-gamma- inducible protein 10 (IP- 10; CXCL10)-deficient mice reveal a role for IP- 10 in effector T cell generation and trafficking. Journal of immunology. 2002; 168(7):3195-204. Epub 2002/03/22. PubMed PMID: 11907072.
- Anti-retroviral therapy has dramatically altered the HIV pandemic landscape by rendering the disease manageable - but still incurable. Significant barriers are associated with the global implementation of ART that limit its utility for sustainable prevention of HIV. Moreover, the intersection of aging-related conditions and the consequences of long-term ART will have a substantial impact on the healthcare system as well as on HIV patients. These challenges underscore the need to pursue strategies that will provide both sustainable HIV prevention and a functional HIV cure.
- the results of the RV144 trial indicate that vaccination may prevent HIV transmission in humans and that durability of anti-envelope (Env) HIV antibodies may be the key to this protection. Efforts to improve upon the RV144 trial have demonstrated that booster immunizations increase serum anti-Env antibody titers only transiently. This“anti-Env antibody persistence problem” impedes our efforts to develop an effective HIV vaccine.
- Antiretroviral therapy has a remarkable impact on the morbidity and mortality associated with HIV but significant barriers hamper its global implementation, limiting its full potential as a preventative tool. Moreover, the intersection of aging-related conditions and the consequences of long-term ART is predicted to adversely impact the healthcare system as well as the HIV patient. These challenges underscore the need to pursue strategies that will provide both sustainable HIV prevention and a functional HIV cure.
- the results of the RV144 trial indicate that vaccination may prevent HIV transmission in humans and that durability of anti-envelope (Env) HIV antibodies may be the key to this protection.
- Efforts to improve upon the RV144 trial have demonstrated that booster immunizations increase serum anti-Env titers but do so transiently. This short-lived impact demonstrates an ineffectiveness in the induction of long-lived Env-specific plasma cells, the source of the durable antibody. This“anti-Env antibody persistence problem” is impeding our progress in developing an effective HIV vaccine.
- Tfh CD4 T follicular helper cells
- Th CD4 helper
- Tfh cells are heterogeneous and encompass Thl, Th2, Thl7-type helper attributes.
- the proportion of each helper attribute determines the durability and functional quality of the antibody response.
- helper attributes are programmed during T-cell priming and are dictated by the initial inflammatory response following vaccination. This provides researchers a window of opportunity to manipulate the CD4 Tfh response in order to maximize the generation of durable anti-Env humoral immunity.
- Thl- DNA prime substantially increases the Env-specific Tfh cells relative to a Thl+Th2 vaccine regimen
- Thl -protein boost results in greater production of the IgGl and IgG3 subclasses with enhanced durability, breadth, specificity and avidity of anti-Env antibody
- Thl vaccine generation of polyfunctional Thl/Thl7 GC Tfh cells predicts the durability of anti- Env IgG titers in sera and anti-Env IgA titers in mucosa.
- the first approach is to demonstrate that a Thl/Thl7 DNA-prime is more effective at increasing the magnitude of Env-specific Tfh cells relative to a Thl vaccine regimen.
- we a) delineate innate determinants of Env-specific Tfh cell differentiation during the DNA prime; and b) delineate Env-specific Tfh cells induced in response to a Thl versus Thl/Thl7 DNA prime.
- the second approach is to demonstrate that synergistic boosting of Thl/Thl7 polarized Tfh cells elicits robust and durable anti-Env antibody titers in systemic and mucosal compartments.
- Thl vaccine regimen induces robust and durable serum HIV Env antibody titers.
- Detailed kinetic analysis revealed that HIV anti-Env titers peaked after the final boost with a median 5-fold boost in antibody titers at week 2 post-2nd protein relative to week 0.
- Thl vaccine regimen is more effective in generating durable binding titers in sera and mucosa, as well as HIV anti-Env antibodies with greater breadth, avidity, broader specificity, and neutralization potential relative to a mixed Thl+2 vaccine regimen.
- Thl vaccine induces higher frequencies of Env-specific CD4 T cells.
- Tfh frequencies were increased following the third DNA prime.
- a significant increase in frequencies and absolute counts of ICOS+PD-l+activated Tfh cells was observed (FIG. 27B).
- Env-specific Tfh cells were induced at significantly higher frequencies in the Thl group (FIGS. 27C and D).
- Sample Size There are 8 macaques in Group 1, and 12 macaques in Groups 2 and 3. We have worked with a statistician to ensure our studies are sufficiently powered to detect differences in immunogenicity. We evaluate individual innate, T cell, and antibody trajectories over time and compare mean trajectories across experimental groups. We then correlate individual trajectories of the innate inflammatory response with those of Tfh responses and humoral immunity. Our study design (sample sizes, repeated measures) provides confidence limits of about ⁇ 0.5s for the difference in rates of change of markers, compared to the variation s in individual trajectories (R package longpower, Edland 2009). We have 80% power to detect differences comparable to those found in our previous studies.
- sampling schedule is designed to optimally and rigorously capture immunological and biological dynamics to link innate immune parameters to Tfh responses and ultimately to antibody responses following vaccination. To this end, all animals are sampled at various time points at baseline and after each of the immunizations. We evaluate clinical chemistries immediately following and weeks after immunization to monitor safety (26).
- the improved inflammatory response was associated with increased levels of antibody durability linking the innate immune response to priming of effective CD4 Tfh help (FIG. 32D).
- FIG. 32D we compare induction of innate immune responses between Thl and Thl/Thl7 priming regimens. To optimally capture the early and transient induction of immune factors, we collect blood at 0, 48, and 72 hours post immunization to establish a comprehensive cytokine /chemokine profile temporally, and across vaccine platforms.
- Thl7 factors including IL-17, IL-6, TGF-b, IL-21, IL-23 and the CCR6 ligand CCL20.
- Thl7 factors including IL-17, IL-6, TGF-b, IL-21, IL-23 and the CCR6 ligand CCL20.
- Tfh cells After stimulation, Ag-specific Tfh cells are identified based on co-expression of AIM marker positivity and CXCR5 expression.
- CD3+CD4+Fas+CXCR5+PD-1+/++ (comprising Tfh and GC Tfh) are sorted using fluorescence-assisted cell sorting (FACS) into CXCR3+ and CCR6+ single positive subsets prior to stimulation.
- FACS fluorescence-assisted cell sorting
- Naive CD4 T cells (CD4+Fas-CXCR5-PD-1-CXCR3-) serve as a reference population.
- RNA sequencing methodology allows us to capture the heterogeneity of the Tfh cell response to stimulation and gives us the ability to resolve important associations between cell phenotype, stimulation status, and cellular transcriptional program.
- a method of enhancing the anti-Env antibody response during HIV vaccination in a subject comprising:
- DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
- liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
- ALF Army Liposome Formulation
- lipid adjuvant comprises monophosphoryl lipid A (MPLA).
- a pharmaceutical composition for vaccinating a subject against HIV comprising a DNA vaccine comprising a polynucleotide encoding an HIV Env polypeptide, a polynucleotide encoding interferon-induced protein (IP)- 10, and a pharmaceutically acceptable carrier.
- a DNA vaccine comprising a polynucleotide encoding an HIV Env polypeptide, a polynucleotide encoding interferon-induced protein (IP)- 10, and a pharmaceutically acceptable carrier.
- IP interferon-induced protein
- composition of embodiment 24, wherein the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
- a pharmaceutical composition for boosting vaccination against HIV comprising an HIV Env polypeptide, QS-21, and a pharmaceutically acceptable carrier.
- composition of embodiment 32, wherein the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
- ALF Army Liposome Formulation
- composition of embodiment 32 or 33, wherein the lipid adjuvant comprises monophosphoryl lipid A (MPLA).
- MPLA monophosphoryl lipid A
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Virology (AREA)
- Animal Behavior & Ethology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Communicable Diseases (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Oncology (AREA)
- Molecular Biology (AREA)
- Tropical Medicine & Parasitology (AREA)
- AIDS & HIV (AREA)
- Immunology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
The present disclosure provides methods and compositions for enhancing an immune response in a subject undergoing HIV vaccination. In particular, the present disclosure provides adjuvants and methods for increased Th1 polarization of T cells in a subject in order to enhance the immune reaction against an HIV antigen.
Description
TH1-POLARIZING ADJUVANTS FOR ENHANCING IMMUNOGENICITY OF HIV ANTIGENS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] The present application claims priority to U.S. Provisional Application No. 62/853,543, filed May 28, 2019, and U.S. Provisional Application No. 62/927,607, filed October 29, 2019, the disclosures of which are hereby incorporated by reference in their entirety for all purposes.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
[0002] This invention was made with Government support under Grant No. OD023034, awarded by the National Institutes of Health (NIH). The Government has certain rights in the invention.
BACKGROUND
[0003] Antiretroviral therapy (ART) has a remarkable impact on the morbidity and mortality associated with HIV but significant barriers hamper its global implementation, limiting its full potential as a preventative tool. Moreover, the intersection of aging-related conditions and the consequences of long-term ART is predicted to adversely impact the healthcare system as well as the HIV patient. These challenges underscore the need to pursue strategies that will provide both sustainable HIV prevention and a functional HIV cure.
[0004] The results of the RV144 trial demonstrated that vaccination may prevent HIV transmission in humans, and longevity of anti-Env antibodies may be the key to this protection. Efforts to improve upon the prime-boost vaccine regimen used in RV144 have indicated that booster immunizations increase serum anti-Env antibody titers, but only transiently. Poor antibody durability hampers efforts to develop an effective HIV-1 vaccine.
[0005] CD4 T follicular helper cells (Tfh) are a specialized subset of CD4 T cells that migrate to germinal centers (GC) within secondary lymphoid organs and provide growth and
differentiation signals to GC B cells within a few days of immunization. GCs are populated by antigen-activated, rapidly proliferating B cell clones, which rely on cytokines and co stimulatory signals from Tfh cells to undergo immunoglobulin affinity maturation, class- switch recombination, and differentiation to memory B cells and plasma cells. The maturation of GC B cells to plasma cells and the resulting long-lived humoral immunity hinges on effective Tfh help. Tfh cells are heterogeneous and, depending on inflammatory signals during T cell priming, differentiate into Thl, Th2, Thl7-type Tfh cells. Th polarization of a Tfh cell influences cytokine profile and co-stimulatory molecule expression.
[0006] There is a critical need to identify strategies that will augment vaccine-mediated humoral immunity for a successful HIV vaccine. The present disclosure satisfies this need and provides other advantages as well.
BRIEF SUMMARY
[0007] In one aspect of the present disclosure, a method is provided of enhancing the anti- Env antibody response during HIV vaccination in a subject, the method comprising (i) administering a first Thl -polarizing adjuvant to the subject together with a DNA vaccine comprising a polynucleotide encoding a first HIV Env polypeptide, wherein the first adjuvant comprises a polynucleotide encoding interferon-induced protein (IP)-10; and (ii) administering a second Thl -polarizing adjuvant to the subject together with a booster comprising a second HIV Env polypeptide, wherein the second adjuvant comprises QS-21.
[0008] In some embodiments of the method, the second HIV Env polypeptide is a gpl40 polypeptide. In some embodiments, the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof. In some embodiments, the first adjuvant and the DNA vaccine are administered transdermally with electroporation. In some embodiments, the HIV Env polypeptide, HIV gpl40 polypeptide, and/or one or more additional HIV polypeptides are from HIV-1. In some embodiments, the HIV-1 is of clade C origin. In some embodiments, the booster further comprises a lipid and/or liposomal adjuvant. In some embodiments, the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes. In some embodiments, the lipid adjuvant comprises monophosphoryl lipid A (MPLA). In some embodiments, the first Thl- polarizing adjuvant and DNA vaccine are administered to the subject 1, 2 or 3 times prior to
the administration of the booster. In some embodiments, the first Thl -polarizing adjuvant and DNA vaccine are administered to the subject at 0, 8, and 16 weeks. In some embodiments, the booster is administered to the subject 1 or 2 times.
[0009] In some embodiments of the method, peripheral and germinal center (GC) Tfh cells isolated from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster display higher proportions of anti-Env Thl cells than do peripheral and germinal center (GC) Tfh cells taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant. In some embodiments, blood taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster shows higher anti-gpl40 extrafollicular and/or plasma cell-derived titers than does blood from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant. In some embodiments, serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show a broader cross-clade anti-Env response and/or increased specificity for gpl20 V1V2 loops than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant. In some embodiments, serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster display elevated anti-Env titers that persist longer than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
[0010] In some embodiments of the method, serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show higher avidity against gpl40 than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant. In some embodiments, serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater neutralization activity against HIV-1 than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl- polarizing adjuvant. In some embodiments, the neutralization of HIV-1 is assessed using the TZM-bl assay. In some embodiments, serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater antibody- dependent cellular toxicity (ADCC) and/or antibody-dependent phagocytosis (ADP) activity
against HIV-infected cells than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant. In some embodiments, the HIV-infected cells are infected with HIV-1. In some embodiments, the HIV-1 is of clade C origin. In some embodiments, IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show higher anti-gpl40 titers than do IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
[0011] In another aspect, the present disclosure provides a pharmaceutical composition for vaccinating a subject against HIV, the composition comprising a DNA vaccine comprising a polynucleotide encoding an HIV Env polypeptide, a polynucleotide encoding interferon- induced protein (IP)- 10, and a pharmaceutically acceptable carrier. In some embodiments of the composition, the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof. In some embodiments, the HIV Env polypeptide and/or one or more additional HIV polypeptides are from HIV-1. In some embodiments, the HIV-1 is of clade C origin. In some embodiments, the polynucleotide encoding the HIV Env polypeptide and the polynucleotide encoding IP- 10 are present within a single DNA vector. In some embodiments, the composition is formulated for transdermal delivery with electroporation.
[0012] In another aspect, the present disclosure provides a pharmaceutical composition comprising an HIV Env polypeptide, QS-21, and a pharmaceutically acceptable carrier. In some embodiments, the HIV Env polypeptide is a gpl40 polypeptide. In some embodiments, the composition further comprises a lipid and/or liposomal adjuvant. In some embodiments, the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes. In some embodiments, the lipid adjuvant comprises monophosphoryl lipid A (MPLA). In some embodiments, the HIV Env polypeptide is from HIV-1. In some embodiments, the HIV-1 is of clade C origin.
[0013] Other objects, features, and advantages of the present invention will be apparent to one of skill in the art from the following detailed description and figures.
BRIEF DESCRIPTION OF THE DRAWINGS
[0014] FIGS. 1A-1D. Immunization schedule for subtype C HIV-1 Envelope DNA prime and protein boost vaccine regimen. FIG. 1A: Flow cytometric plots illustrate expression of HIV Env, SIV Gag, and IP-10 by 293-T cells transfected with DNA and DNAIP-10 plasmids. Grey overlay shows expression in non-transfected cells. FIG. IB: Bar graph shows surface expression of HIV Env based on detection with a panel of monoclonal antibodies as indicated. FIG. 1C: IP-10 titers in supernatants of transfected 293T cells show accumulation of IP- 10 following transfection with DNAIP-10. FIG. ID shows immunization schedule and sampling. Two groups of 10 rhesus macaques each were immunized three times with DNA followed by two immunizations with protein. DNA was delivered intradermally and three seconds later electrical pulses were delivered around the injection site using the ICHOR TriGrid Array. Group 1 animals (n=10) received DNA plasmid expressing IP-10 and an ALFQ-adjuvanted C.gpl40 boost (DIP-lOProALFQ). Group 2 (n=10) animals were immunized with DNA and boosted with ALFA-adjuvanted C.gpl40 protein (DProALFA).
[0015] FIGS. 2A-2G. DIP-lOProteinALFQ vaccine induces robust anti -Env antibody titers with cross-clade breadth. FIG. 2A: Antibody kinetics against C. 1086 Env gpl40 in serum at weeks 0, 2, and 8 following each protein boost assessed by binding antibody multiplex assay (BAMA); right panel shows scatter plot values for each animal at weeks 0, 2, and 8 post 2nd protein boost. FIG. 2B: Kinetics of C. 1086 specific anti -Env titers after 2nd protein boost measured by ELISA; right panel shows titers for each individual animal. BAMA assay was used to measure responses against CH505 (FIG. 2C), Con C (FIG. 2D), Con S (FIG. 2E), and gp70 VI V2 (FIG. 2F). FIG. 2G shows fold change in antibody titers at indicated time points after 2nd protein boost relative to the first. Animals receiving the DProALFA vaccine are represented by blue circles and animals receiving the DIP-lOProALFQ vaccine by red circles. Kinetic data show geometric means. Vertical dotted lines show immunization time points. In dot plots, geometric means are indicated as horizontal lines. Statistical significance across vaccine regimens was tested using unpaired, two-tailed Mann-Whitney U test; *p < 0.05, **p < 0.01, *** p < 0.001, **** p < 0.0001.
[0016] FIGS. 3A-3M. DIP-10 ProteinALFQ vaccine elicits high avidity anti-Env antibody with ADCC and ADP activities. FIG. 3A: Surface Plasmon Resonance (SPR) was used to determine the avidity index (AI) in serum at 2 weeks after final DNA and each protein boost using C. 1086 gpl40 protein immobilized onto sensor chips. FIG. 3B shows SPR-based AI
values in the two vaccine regimens over time. FIG. 3C shows significantly higher avidity in DIP-10 ProteinALFQ. at 2 weeks post 2nd protein boost. AI measured against C.1086 gpl40 using 2 M sodium thiocyanate (FIG. 3D) and 0.1 M sodium citrate (FIG. 3E) at week 8 post 2nd protein. AI against Con C (FIG. 3F) and Con S (FIG. 3G) measured using 0.1 M sodium citrate at week 8 post 2nd protein boost. FIG. 3H: Serum-neutralizing antibody responses were assessed against tier 1 A (MW965.26) pseudovirus and the 50% infective dose (ID50) was determined. FIG. 31 shows ADCC activity against SHIV CH505 infected target cells. FIG. 3J: Antibody-dependent phagocytosis using gpl20-coated beads was measured using sera from week 8 post 2nd protein boost at serum dilutions ranging from 1 : 100 to 1 :2500. FIG. 3K shows individual ADP scores at 1:500 serum dilution. FIG. 3L: C.1086 gpl20 subclass analysis was performed on week 8 post 2nd protein boost to measure IgGl, IgG2, IgG3, and IgG4. FIG. 3M shows IgGl/IgG4 ratio across vaccine groups at week 8 post 2nd protein boost. Statistical significance across vaccine regimens was tested using unpaired, two-tailed Mann-Whitney U test and within group differences over time were tested using Wilcoxon matched-pairs signed rank test; *p < 0.05, **p < 0.01, *** p < 0.001, **** p < 0.0001.
[0017] FIGS. 4A-4E. DNAIP-10 ProteinALFQ vaccine elicits robust anti-Env antibody in vaginal and rectal mucosal secretions. Vaginal and colorectal secretions were evaluated for C.1086 concentrations. FIGS. 4A-4B show kinetics of C.1086 IgA responses in vaginal and rectal secretions. FIGS. 4C-4D show IgA and IgG titers in rectal secretions. FIG. 4E shows gpl40 IgA kinetics in sera. Horizontal broken lines represent the assay limit of detection. Kinetic data show geometric means. Vertical dotted lines show immunization time points. In dot plots, geometric means are indicated as horizontal lines. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U test; *p < 0.05, **p < 0.01, *** p < 0.001, **** p < 0.0001.
[0018] FIGS. 5A-5D. DIP-10 ProteinALFQ vaccine induces Env-specific Tfh cells in peripheral blood. FIG. 5A: Flow cytometric gating of CXCR5+ cells in PBMCs to identify 0X40+ CD25+ activated CD4 T subsets after stimulation with C.1086 protein and Con C peptide pools. FIG. 5B shows frequency of Env-specific CD4 T cells at week 1 post 1st protein boost which strongly correlate with responses against the C.ZA.1197MB boosting immunogen (FIG. 5C). FIG. 5D shows robust induction of IP- 10 in sera after 1st protein immunization. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U
test. Spearman coefficient of correlation values were computed to determine associations; * p<0.05, **p < 0.01, *** p < 0.001.
[0019] FIGS. 6A-6L. DIP-10 ProteinALFQ vaccine induces GC Tfh cells with distinctive Thl signatures. FIG. 6A: Gating strategy to identify GC Tfh cells and GC B cells in lymph node at 2 weeks post 1st protein boost. Histograms show higher relative expression of Bcl-6 and ICOS in GC Tfh cells. FIG. 6B: Ex vivo co-culture experiments with sorted GC Tfh cells demonstrates B helper capacity. FIG. 6C shows kinetics of GC Tfh responses in lymph node at specified time points. FIG. 6D: Dot plot shows higher relative frequency of GC B cells in Thl vaccine group, and correlation of GC B cells with GC Tfh cells. Frequencies of Env- specific CD4 T cells in lymph node correlate with GC Tfh cells. FIG. 6E: Histogram shows relative CXCR3 expression in GC Tfh cells and GC B cells and dot plot shows significantly higher CXCR3 expression on GC Tfh cells and GC B cells in Thl vaccine regimen. FIG. 6F: Flow plot illustrates higher expression of CXCR3 on T-bet+ memory B cells. FIG. 6G: Antibody titers at week 18 post 2nd protein predicted by frequency GC Tfh cells and proportion of CXCR3 -expressing GC Tfh cells at 2 weeks post 1st protein. Antibody avidity at week 8 post 2nd protein predicted by GC Tfh cells at 1st protein. FIG. 6H shows animals selected (triangles) for RNA-seq and Principal component analysis of RNA-Seq data (FIG. 61); colors represent populations as indicated in FIG. 6A. FIG. 6J: Heatmap shows expression of genes differentially expressed in Tfh relative to naive across four sorted CD4 subsets indicated in FIG. 61. FIG. 6K: Volcano plot of differentially expressed transcripts (adjusted p < 0.05 in red) for Tfh cells versus naive cells. FIG. 6L: Log fold change values of key Tfh and Thl genes in Tfh (red) and memory Tfh (blue) cells in lymph node Thl vaccinated animals. Statistical significance was tested using unpaired, two-tailed Mann- Whitney U test. Spearman coefficient of correlation values were computed to determine associations; * p<0.05, **** p < 0.0001.
[0020] FIGS. 7A-7G. DNAIP-10 immunization induces systemic expansion of pro- inflammatory monocytes and enhances GC Tfh responses. FIG. 7 A shows gating strategy to identify activated CXCR5+ cells in blood at day 0 and day 14 following DNA3 immunization. Kinetic data show transient accumulation of ICOS+ PD-1+ CXCR5+ cells in blood at day 14 following DNA3 when expressed as relative frequencies (left) and absolute counts (right). FIG. 7B: Flow cytometric gating to identify inflammatory CD14+CD16+ monocytes which increase at day 3 following DNA immunization (FIG. 7C). FIG. 7D: Higher relative increase in pro-inflammatory monocytes in DNA-PMO primed animals. FIG.
7E shows induction of IL-lb in sera 7 days following DNA immunization. FIG. 7F: Fine needle aspirates of draining lymph nodes show increased frequencies of GC Tfh cells in DNA-IP-10 primed animals at day 14 post DNA. FIG. 7G shows that the frequency of pro- inflammatory monocytes predicts C.1086C gpl40 antibody titers at week 8 post 2nd protein boost. Statistical significance was tested using unpaired, two-tailed Mann-Whitney U test. Spearman coefficient of correlation values were computed to determine associations; * p< 0.05, ***, p < 0.001, **** p < 0.0001.
[0021] FIGS. 8A-8D. Thl DNA prime enhances frequency of pro-inflammatory monocytes and GC Tfh cells. FIG. 8A: Increase in net frequency of pro-inflammatory monocytes in Thl vaccine group. FIG. 8B: Increase in IP- 10, IL-IB, IL-6 and IL-21 after DNA prime. FIG. 8C: Increase in GC Tfh cells with Thl vaccine. FIG. 8D: Frequency of pro-inflammatory monocytes after DNA prime predicts antibody response at memory.
[0022] FIGS. 9A-9D. Thl DNA prime enhances frequency of Env-specific Tfh cells. FIG. 9A shows time points of blood collection for T cell kinetic analysis. FIG. 9B: Flow plot illustrates gating strategy to identify activated CD4 Tfh cells based on coexpression of ICOS and PD-1. Kinetics of relative frequencies and absolute counts of ICOS+PD-1+ Tfh cells. FIG. 9C shows gating strategy to identify Env-specific Tfh cells based on co-expression of 0X40 and CD25. FIG. 9D shows higher Env-specific Tfh cells in Thl regimen.
[0023] FIGS. 10A-10B. Schematic shows study design with three DNA primes (SHIV 1086C, 4 mg given I.D with E.P) followed by two protein boosts with Clade C gpl40 Envelope, HIV-1 isolate C.ZA.1197MB, 100 pg delivered subcutaneously (SQ). In the Thl group, DNA was adjuvanted with IP- 10 and protein with ALFQ and the Thl +2 group got DNA alone with ALFA-adjuvanted protein boost. FIG. 10A: Kinetics of antibody responses following 2nd protein boost. FIG. 10B: Scatter plot showing antibody titers in all animals within the experimental groups at weeks 0, 2, 8, and 18 following 2nd protein boost with antibody fold-change in Thl relative to Thl+2 group.
[0024] FIGS. 11A-11D. After Thl vaccine regimen, antibodies demonstrate increased breadth, avidity, and broader specificity. Antibody breadth (FIG. 11A), Avidity index (FIG. 11B), and specificity (FIG. 11C) and neutralization (FIG. 11D) titers at indicated time points after 2nd protein boost.
[0025] FIG. 12. Polyfunctional Thl/Thl7 CD4 T cells predict durability of anti-Env titers in sera and mucosa.
[0026] FIG. 13. Experimental design of vaccine trial 1 (SI). All groups are immunized intradermally using a Bioject electroporation device with plasmid DNA-SHIV construct expressing 1086 Clade C Env, tat, and rev; and SIV239 Gag (3 mg/dose, week 0 and 4). Groups 2 and 3 receive a similar dose of IP-10 adjuvanted DNA-SHIV construct. The MVA- SHIV boost is intramuscular (10L8 PFU/dose, week 16,24). Animals are co-immunized with gpl40 protein (Clade C Env, 100 pg), in the thigh opposite to that of the MVA boost, formulated either in Alum (500 pg, Groups 1, 2) or AS01B (50 pg MPL+50 pg Q21) Group 3). Animals in Groups 1 and 3 are compared for vaccine efficacy and all groups are compared for immunogenicity.
[0027] FIGS. 14A-14F. DNA/MVA vaccine induces robust Env antibody. Monkeys were immunized with DNA (wks 0, 8) and MVA (wks 16, 32). In FIG. 14B, some animals received gpl40 (Pro) +Alum with 2nd MVA ;**, p < 0.01 using a two-tailed t test; # p = 0.05.
[0028] FIGS. 15A-15E show a transient accumulation of CXCR5+ Tfh cells expressing the cell-cycle marker Ki-67 at the peak CD4 effector response following the MVA boost (FIGS. 15A-15B, *p < 0.05). Analogous to functionality of Gag and Env-specific CD4 T cells co expressing the Tfh cytokine interleukin (IL) 21 and the Thl cytokine IFNy (FIG. 15C); Ki- 67+ pTFH expressed CXCR3, a marker of Thl polarity (FIG. 15D). Notably, the proportion of CXCR3+Ki-67+ pTFH cells markedly increased following MVA immunization relative to baseline (FIG. 15E, ***p< 0.001).
[0029] FIGS. 16A-16F show aspects of the GC response following the 2nd MVA boost and how Pro+Alum modulated this response. GC Tfh were examined by high expression of programmed death (PD)1 and CXCR5, and Tfh cells based on the CXCR5+PD-1+ phenotype (FIG. 16A). M+Pro vaccine increased Thl-polarized Tfh cells (FIG. 16B) even in the presence of Alum. The proportion of CXCR3+ GC Tfh correlated with frequency of GC B cells (FIG. 16C), antibody avidity (FIG. 16D), durability (FIG. 16E), and neutralization (FIG. 16F).
[0030] FIGS. 17A-17C. IP-10 induces CXCR3+ Tfh cells. Flow plot (FIG. 17A) and histogram (FIG. 17B) show CXCR3 expression on Tfh cells at increasing IP- 10 doses. FIG. 17C: Increase in CXCR3+ Tfh cells with IP- 10.
[0031] FIGS. 18A-18C. Construction of PMO-DNASHIV vaccine. FIG. 18A: The DNA vaccine includes IP- 10 as a fusion to IRES or as a fusion to the 2 A peptide downstream of Env. FIG. 18B: Characterization of CD40L-DNA vaccine. FIG. 18C: Significantly higher magnitude and proportion (% responders indicated in grey area) vaccine-elicited CD4s after 1st prime in CD40L-DNA (blue) relative to DNA (clear); ****, p< 0.0001.
[0032] FIGS. 19A-19C. Identifying vaccine-elicited Thl and Th2 Tfh cells. The cell-cycle marker Ki-67 was utilized to interrogate CXCR5+ CD4 T cells at peak effector time points following each immunization for expression of CXCR3 and CCR4, as outlined in FIG. 19A. This is complemented by intracellular cytokine staining (ICS)-based assays designed to examine vaccine-specific responses (at peak and memory) after stimulation with relevant Gag and Env peptide pools. After stimulation, Ag-specific Tfh cells are identified based on cytokine positivity and CXCR5 expression, as shown in FIGS. 19B-19C.
[0033] FIG. 20. CD40L+ Tfh are Env specific. Vaccine-specific TFH cells are identified by translocation of CD40L after 5 hour stimulation with Env peptides in the presence of co stimulatory molecules.
[0034] FIGS. 21A-21B. The frequency of Env-specific IFNG+IL-21+ cells at 1 week post 2nd MV A, and the frequency of CXCR3+ GC Tfh cells at 2 weeks post 2nd MV A, correlate with antibody durability measured at 20 weeks post 2nd MVA.
[0035] FIGS. 22A-22B. Assays to measure humoral immune responses. FIG. 22A: ELISPOT assay captures increase in gpl40 ASCs with Pro+MVA relative to MVA alone. In FIG. 22B, ADCC increases following a 3rd protein boost, while no increase was observed in ADP.
[0036] FIGS. 23A-23B. CXCR3+ Tfh cells express CCR5. FIG. 23A: CXCR3+ Tfh cells comprise higher frequency of CCR5 cells. FIG. 23B: Higher levels of pro-viral DNA in X3+ Tfh cells in lymph node; ***, p< 0.001; *p < 0.05.
[0037] FIGS. 24A-24B. Thl vaccine regimen induces robust and durable serum HIV anti- Env antibody titers. Schematic shows study design with 3 DNA primes (SHIV 1086C, 4 mg given I.D with E.P) followed by two protein boosts with Clade C gpl40 Envelope, HIV-1 isolate C.ZA.1197MB, 100 pg delivered subcutaneously (SQ). In the Thl group, DNA was adjuvanted with IP-10 and protein with ALFQ and the Thl+2 group got DNA alone with ALFA-adjuvanted protein boost (FIG. 24A). Kinetics of antibody responses following 2nd
protein boost (FIG. 24B). Scatter plot showing antibody titers in all animals within the experimental groups at week 0, 2, 8, and 18 following 2nd protein boost with antibody fold- change in Thl relative to Thl+2 group.
[0038] FIGS. 25A-25D. After Thl vaccine regimen antibodies demonstrate increased breadth, avidity, and broader specificity. Antibody breadth (FIG. 25A), Avidity index (FIG. 25B), and specificity (FIG. 25C) and neutralization (FIG. 25D) titers at indicated time points after 2nd protein boost.
[0039] FIGS. 26A-26B. Robust induction of anti-Env-specific IgA (FIG. 26A) and anti- Env-specific IgG (FIG. 26B) in vaginal and rectal mucosa with Thl vaccine regimen. Data are post 2nd protein boost.
[0040] FIGS. 27A-27D. Thl DNA prime induces higher magnitude Env-specific Tfh cells. FIG. 27A shows time points of blood collection for T cell kinetic analysis. FIG. 27B: Flow plot illustrates gating strategy to identify activated CD4 Tfh cells based on co-expression of ICOS and PD-1. Kinetics of relative frequencies and absolute counts of ICOS+PD-1+ Tfh cells. FIG. 27C shows gating strategy to identify Env-specific Tfh cells based on co expression of 0X40 and CD25. FIG. 27D shows higher Env-specific Tfh cells in Thl regimen.
[0041] FIGS. 28A-28G. Induction of polyfunctional Thl/Thl7 CD4 Tfh cells in Thl vaccine regimen. FIG. 28A shows transient induction of pro-inflammatory Thl cytokine IP- 10, Thl7 cytokine IL-17A and IL-6 and IL-21. FIG. 28B shows higher frequencies of Env- specific Tfh cells in PBMCs and FIG. 28C illustrates a robust GC response in Thl vaccine regimen. FIG. 28D illustrates gating for GC Tfh cells with higher relative expression of Bcl- 6, Thl marker CXCR3 and Thl7 marker CCR6. FIG. 28E shows higher CD40L expression if IFNG+IL-17+ cells. FIG. 28F: RNA-sequencing analysis of GC Tfh cells in Thl vaccine treatment animals (n=3) reveals upregulation of Thl / Thl7 genes BATF, PRKCQ and MYD88. FIG. 28G: Phenotypic examination of GC subsets confirmed increase in proportion of CXCR3+CCR6+ GC Tfh cells in Thl vaccinated group.
[0042] FIG. 29. Polyfunctional Thl/Thl7 CD4 T cells predict durability of anti-Env titers in sera and mucosa. The frequency of polyfunctional Thl/Thl7 GC TFH cells is directly predictive of binding titers as measured by ELISA and BAMA.
[0043] FIGS. 30A-30D. A single CAFOl adjuvanted protein immunization induces robust antibody responses. FIG. 30A: Structure of the cationic liposome CAFOl. Single subcutaneous immunization with CAFOl -adjuvanted Chlamydia antigen (CTH 522, 5 pg; FIG. 30B) or 4-Hydroxy-3-nitrophenylacetyl hapten conjugated to ovalbumin (NP-OVA, 5 pg; FIG. 30C) in mice shows robust induction of antibody responses at levels comparable or superior to that induced by the MF59 analog adjuvant, Addavax. FIG. 30D: Notably, the frequency of OVA-bound cells at the site of injection was significantly higher at 24- and 48- hours following immunization, indicating formation of antigen depot at the site of immunization with CAFOl.
[0044] FIG. 31. Robust protein expression of Env and Gag in DNA 1086C plasmid co expressing IP- 10 and IL-6.
[0045] FIGS. 32A-32D. Thl vaccine enhances frequency of pro-inflammatory monocytes and GC Tfh cells. FIG. 32A: Increase in net frequency of pro-inflammatory monocytes in Thl vaccine group. FIG. 32B: Increase in IP-10, IL-1B, IL-6 and IL-21 after DNA prime. FIG. 32C: Increase in GC Tfh cells with Thl vaccine. FIG. 32D: Frequency of pro- inflammatory monocytes after DNA prime predicts antibody response at memory.
[0046] FIGS. 33A-33B. Increased Env gpl40 1086. C Antibody avidity (kd off-rate) in Thl vaccine group. Data represent quadruplicate samples for each animal. FIG. 33A shows higher avidity after sequential immunization in both vaccine groups. FIG. 33B shows increased avidity in Thl-Tfh vaccine regimen.
[0047] FIGS. 34A-34B. Higher levels of Env IgGl (FIG. 34A) and IgG3 (FIG. 34B) with Thl vaccine regimen. Titers were measured at week 8 post 2nd protein.
DETAILED DESCRIPTION
1. Introduction
[0048] The present disclosure relates to methods and compositions for enhancing the immune response in subjects undergoing HIV vaccination. In particular, the present methods and compositions can be used in conjunction with prime-boost vaccine regimens, e.g., using DNA vaccines against HIV, in order to enhance the Thl polarization of Tfh cells in a subject. The present disclosure is based in part on the discovery that Thl polarization in conjunction with a DNA prime substantially increases antigen-specific, e.g, Env-specific, Tfh cells, and
that Thl polarization in conjunction with a protein boost, e.g., using gpl40, results in greater production of the IgGl subclass with enhanced longevity, breadth, avidity, ADCC, and ADP activities of the antigen-specific antibody. In particular, the present disclosure provides DNA constructs expressing both HIV (and/or SIV) genes and interferon protein 10 (IP- 10), as well as compositions comprising an HIV protein boost, e.g, gpl40, with a Thl -polarizing adjuvant such as QS-21. The present methods and compositions are effective at inducing, e.g, pro-inflammatory monocytes, Env-specific CD4 T follicular helper cells, and higher germinal center responses relative to DNA alone (i.e., in the absence of Thl polarization). In addition, the localized expression of IP- 10 using the present constructs enhances immunogenicity by concerted effects on antigen-presenting cells and T cells, and limits the potential for non-specific, systemic effects, thereby reducing side effects.
2. Definitions
[0049] As used herein, the following terms have the meanings ascribed to them unless specified otherwise.
[0050] The terms“a,”“an,” or“the” as used herein not only include aspects with one member, but also include aspects with more than one member. For instance, the singular forms “a,” “an,” and“the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to“a cell” includes a plurality of such cells and reference to“the agent” includes reference to one or more agents known to those skilled in the art, and so forth.
[0051] The terms“about” and“approximately” as used herein shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Typically, exemplary degrees of error are within 20 percent (%), preferably within 10%, and more preferably within 5% of a given value or range of values. Any reference to“about X” specifically indicates at least the values X, 0.8X, 0.81X, 0.82X, 0.83X, 0.84X, 0.85X, 0.86X, 0.87X, 0.88X, 0.89X, 0.9X, 0.91X, 0.92X, 0.93X, 0.94X, 0.95X, 0.96X, 0.97X, 0.98X, 0.99X, 1.01X, 1.02X, 1.03X, 1.04X, 1.05X, 1.06X, 1.07X, 1.08X, 1.09X, 1.1X, 1.11X, 1.12X, 1.13X, 1.14X, 1.15X, 1.16X, 1.17X, 1.18X, 1.19X, and 1.2X. Thus,“about X” is intended to teach and provide written description support for a claim limitation of, e.g.,“0.98X.”
[0052] The term“antigen” refers to a molecule, or a portion thereof, that is capable of inducing an immune response ( e.g ., in a subject). While in many instances an immune response involves the production of an antibody that targets or specifically binds to the antigen, as used herein the term“antigen” also refers to molecules that induce immune responses other than those that specifically involve the production of an antibody that targets the antigen, e.g., a cell-mediated immune response involving expansion of T cells that target antigen-derived peptides presented on the surface of target cells. The term also refers to molecules that do not necessarily induce immune responses by themselves but can be made to induce an immune response when presented in the right context, and/or in combination with other molecules that facilitate immune responses. In particular embodiments of the present disclosure, an antigen refers to an HIV protein, or immunogenic fragment thereof, e.g, an HIV Env protein (e.g., gpl20, gpl40, gpl60).
[0053] The term“nucleic acid sequence encoding a peptide” refers to a segment of DNA, which in some embodiments may be a gene or a portion thereof, that is involved in producing a peptide chain (e.g, an HIV protein or IP- 10). A gene will generally include regions preceding and following the coding region (leader and trailer) involved in the transcription/translation of the gene product and the regulation of the transcription/translation. A gene can also include intervening sequences (introns) between individual coding segments (exons). Leaders, trailers, and introns can include regulatory elements that are necessary during the transcription and the translation of a gene (e.g, promoters, terminators, translational regulatory sequences such as ribosome binding sites and internal ribosome entry sites, enhancers, silencers, insulators, boundary elements, replication origins, matrix attachment sites and locus control regions, etc.). A“gene product” can refer to either the mRNA or protein expressed from a particular gene.
[0054] The terms“expression” and“expressed” refer to the production of a transcriptional and/or translational product, e.g., of a nucleic acid sequence encoding a protein (e.g, an HIV protein or IP- 10). In some embodiments, the term refers to the production of a transcriptional and/or translational product encoded by a gene (e.g, a gene encoding an antigen) or a portion thereof. The level of expression of a DNA molecule in a cell may be assessed on the basis of either the amount of corresponding mRNA that is present within the cell or the amount of protein encoded by that DNA produced by the cell.
[0055] The term “recombinant” when used with reference, e.g., to a polynucleotide, protein, vector, or cell, indicates that the polynucleotide, protein, vector, or cell has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. For example, recombinant polynucleotides contain nucleic acid sequences that are not found within the native (non-recombinant) form of the polynucleotide.
[0056] The term“immune response” refers to any response that is induced (e.g, in a subject) by an antigen, including the induction of immunity against pathogens (e.g, viruses such as HIV). Immune responses induced by systems, recombinant polynucleotides, compositions, and methods described herein are typically desired, intended, and/or protective immune responses. The term includes the production of antibodies against an antigen, as well as the development, maturation, differentiation, and activation of immune cells (e.g, B cells and T cells). In some instances, an immune response comprises inducing pro-inflammatory monocytes, Env-specific CD4 T follicular helper cells, and germinal center responses. The term also includes increasing or decreasing the expression or activity of cytokines that are involved in regulating immune function (e.g, in a subject). Other markers of immune responses include, but are not limited to, Thl-Tfh responses, dendritic cell (DC)-T cell interactions, the magnitude of vaccine-specific T cells, Tfh differentiation, Thl polarization, IL-6 production in B cells, plasma cell differentiation, induction of CD80 and CD40 on DCs, the strength of cross-priming and magnitude of cytolytic responses, effects on APCs and T cells, and others.
[0057] “HIV” refers to the human immunodeficiency virus, and can include any type, group, sub-group, clade, strain, variant, or isolate, e.g, HIV-1, HIV-2, HIV-1 Group M (including sub-groups or clades A, B, C, D, F, G, H, J), Group N, Group O, C.1086, Con C, Con S CH505, etc. HIV is also used to describe genes within the HIV genome (e.g, gag, pol, env, tat, rev, nef, vpr, vif, and vpu), or to proteins encoded by the genes, including Env (or gpl20, gpl40, gpl60, gp41), Gag (or MA, CA, SP1, NC, SP2, P6), Pol (or RT, RNAse H, IN, PR), Tat, Rev, Nef, Vpr, Vif, and Vpu.
[0058] “IP- 10”, or interferon-gamma-induced protein 10 (also known as CXCL10, or C-X- C motif chemokine 10, INP10, small-inducible cytokine B10, and other names) is an 8.7 kDa protein belonging to the CXC chemokine family. In humans, the gene encoding IP- 10 corresponds, e.g, to NCBI Gene ID No. 3627. IP-10 also encompasses polynucleotides
comprising the human IP-10/CXCR3 mRNA of NCBI Reference Sequence NM_001565, or polynucleotides having a sequence comprising at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to the nucleotide sequence of NM_001565. IP-10 also encompasses polypeptides comprising (or polynucleotides encoding) the human IP- 10/CXCR3 protein of NCBI Reference Sequence NP_001556.2, or comprising (or polynucleotides encoding) a polypeptide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to NP_001556.2, as well as to derivatives, variants, and fragments thereof.
[0059] “Thl -polarization” refers to the differentiation of CD4+ T cells into specific subsets in response to antigens presented by antigen presenting cells (APCs) and depending on, e.g., the particular cytokines present in the microenvironment. Polarization involves in part the modulation of particular transcription factors that results in the production of autocrine cytokines, producing a positive feedback loop that contributes to the polarization of the cells. CD4+ subsets include Thl, Th2, Th3, Th9, Thl7, Treg, Trl, and Tfh (see, e.g, Martinez- Sanchez et al., Front. Physiol. (2018); doi.org/10.3389/fphys.2018.00877). Thl polarization involves, e.g, the transcription factor T-bet and the cytokine IFNy; other markers of Thl polarization include, e.g., bcl-6, CXCR3, BCl-6, TBX21, SemA4A, and CCR5.
[0060] “Gpl40” is a soluble version of the Env protein that contains the entire ectodomain
(gpl20) and a portion of the transmembrane glycoprotein (gp41). As used herein, Gpl40 encompasses forms, variants, and derivatives of Gpl40, including monomeric, dimeric, trimeric, etc., forms, forms in which the gpl20 and gp41 components are linked, e.g, by a disulfide bond, mutated forms, e.g, with an I559P change within the gp41 component, and others. “Gpl40” also comprises immunogenic fragments of full-length gpl40, e.g, comprising one or more portions of the gpl20 and/or gp41 components that are useful for eliciting immune responses in a subject.
[0061] “QS-21” is a purified plant extract containing water soluble triterpene glycoside saponins that is derived from the soap bark tree, Quillaja saponaria. In particular embodiments, QS-21 comprises a quillaic acid triterpene substituted with a branched trisaccharide and a linear tetrasaccharide, which is connected to an acyl chain via a hydrolytically labile ester. In particular embodiments, QS-21 is a 65:35 mixture of the apiose- and xylose-substituted variants of the above-described molecule. As used herein, QS-21 can be natural or semi-synthetic, e.g, prepared by purifying the prosapogenin part of the
molecule and synthetically adding the rest of the molecule. Fully synthetic forms of the molecule are encompassed as well. QS-21 can be obtained from commercial sources, e.g., from Desert King International (San Diego, CA).
[0062] As used herein, an“adjuvant” refers to a compound administered to a subject in conjunction with an antigen for enhancing an immune response to the antigen. Adjuvants can increase the immunogenicity of vaccines in any of a number of ways, and can include inorganic compounds such as salts, e.g, aluminum salts, as well as organic compounds and mixtures of compounds, including extracts and preparations, e.g, Freund’s incomplete adjuvant, squalene, MF59, monophosphoryl lipid A, QS-21. In some embodiments, an adjuvant is a polypeptide such as IP- 10. In particular embodiments, an“adjuvant” as used herein refers to adjuvants that increase the Thl polarization of Tfh cells, e.g, IP-10 and/or QS-21.
[0063] A“prime-boost” or“prime-booster” vaccine strategy refers to an immunization approach that is administered in two or more stages and in which the same antigen is presented to a subject in identical (in a homologous protocol) or different (in a heterologous protocol) forms. For example, in one stage a polynucleotide, e.g, DNA molecule encoding an immunogenic protein, is administered one or more times, wherein the protein encoded by the polynucleotide is expressed by cells in the body, and in a second stage the protein itself is administered. The form of the protein administered in the second stage need not be identical to that of the encoded protein; for example, in numerous embodiments of the present disclosure, a prime DNA vaccine is administered encoding the Env protein, and the gpl40 form of the Env protein is administered during the boost stage. Generally the first, i.e., DNA prime, stage precedes the protein boost stage, but other sequences can be performed as well, including approaches in which the two stages overlap to some extent.
[0064] “Tfh” cells, or T follicular helper cells, are a subset of CD4+ T cells that help B cells produce antibodies against foreign pathogens (e.g, against HIV). Tfh cells are located both in circulation and in secondary lymphoid organs, e.g, tonsils, spleen, and lymph nodes, in particular in the B cell zones, where they interact with and stimulate B cells (e.g, through CD40 and by producing IL-21). Tfh cells are involved in the formation of the germinal centers (GCs), structures that form within the B cell zones during an immune response. Tfh cells are defined by, e.g, the expression of the transcription factor Bcl6 and cell surface markers such as CXCR5, PD1, and ICOS.
[0065] The term“antigen-presenting cell” or“APC” refers to a cell that displays or presents an antigen, or a portion thereof, on the surface of the cell. Typically, antigens are displayed or presented with a major histocompatibility complex (MHC) molecule. Almost all cell types can serve as APCs, and APCs are found in a large number of different tissue types. Professional APCs, such as dendritic cells, macrophages, and B cells, present antigens to T cells in a context that most efficiently leads to their activation and subsequent proliferation. Many cell types present antigens to cytotoxic T cells.
[0066] As used herein, the terms“polynucleotide,”“nucleic acid,” and“nucleotide,” refer to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof. The term includes, but is not limited to, single-, double-, or multi-stranded DNA or RNA, genomic DNA, cDNA, and DNA-RNA hybrids, as well as other polymers comprising purine and/or pyrimidine bases or other natural, chemically modified, biochemically modified, non-natural, synthetic, or derivatized nucleotide bases. Unless specifically limited, the term encompasses nucleic acids containing known analogs of natural nucleotides that have similar binding properties as the reference nucleic acid. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof ( e.g ., degenerate codon substitutions), homologs, and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al ., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al. , J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al. , Mol. Cell. Probes 8:91-98 (1994)).
[0067] The terms“vector” and“expression vector” refer to a nucleic acid construct, e.g., plasmid or viral vector, generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid sequence (e.g, encoding an HIV antigen and/or a protein having IP- 10-like activity) in a host cell or engineered cell. In some embodiments, a vector includes a polynucleotide to be transcribed, operably linked to a promoter. Other elements that may be present in a vector include those that enhance transcription (e.g, enhancers), those that terminate transcription (e.g, terminators), those that confer certain binding affinity or antigenicity to a protein (e.g, recombinant protein) produced from the vector, and those that enable replication of the vector and its packaging (e.g, into a viral particle). In some embodiments, the vector is a viral
vector ( i.e a viral genome or a portion thereof). A vector may contain nucleic acid sequences or mutations, for example, that increase tropism and/or modulate immune function.
[0068] The terms“polypeptide,”“peptide,” and“protein” are used interchangeably herein to refer to a polymer of amino acid residues. All three terms apply to amino acid polymers in which one or more amino acid residues are an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. As used herein, the terms encompass amino acid chains of any length, including full-length proteins, wherein the amino acid residues are linked by covalent peptide bonds.
[0069] The terms“subject,”“individual,” and“patient” are used interchangeably herein to refer to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, mice, rats, simians, humans, farm animals, sport animals, and pets. Tissues, cells and their progeny of a biological entity obtained in vivo or cultured in vitro are also encompassed.
[0070] As used herein, the term “administering” includes oral administration, topical contact, administration as a suppository, intravenous, intraperitoneal, intramuscular, intralesional, intratumoral, intrathecal, intranasal, intraosseous, or subcutaneous administration to a subject. Administration is by any route, including parenteral and transmucosal ( e.g buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal). Parenteral administration includes, e.g., intravenous, intramuscular, intra arterial, intradermal, subcutaneous, intraperitoneal, intraventricular, intraosseous, and intracranial. Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.
[0071] The term“treating” refers to an approach for obtaining beneficial or desired results including, but not limited to, a therapeutic benefit and/or a prophylactic benefit.“Therapeutic benefit” means any therapeutically relevant improvement in or effect on one or more diseases, conditions, or symptoms under treatment. Therapeutic benefit can also mean to effect a cure of one or more diseases, conditions, or symptoms under treatment. Furthermore, therapeutic benefit can also mean to increase survival. For prophylactic benefit, the compositions may be administered to a subject at risk of developing a particular disease, condition, or symptom, or to a subject reporting one or more of the physiological symptoms of a disease, even though the disease, condition, or symptom may not yet be present.
[0072] The term“therapeutically effective amount” or“sufficient amount” refers to the amount of a system, recombinant polynucleotide, or composition described herein that is sufficient to effect beneficial or desired results. The therapeutically effective amount may vary depending upon one or more of: the subject and disease condition being treated or prevented, the weight and age of the subject, the severity of the disease condition, the immune status of the subject, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The specific amount may vary depending on one or more of: the particular agent chosen, the target cell type, the location of the target cell in the subject, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, and the physical delivery system in which it is carried.
[0073] For the purposes herein, an effective amount is determined by such considerations as may be known in the art. The amount must be effective to achieve the desired therapeutic effect in a subject suffering from, e.g., HIV infection, or to prevent an infection, e.g, by HIV. The desired therapeutic effect may include, for example, amelioration of undesired symptoms associated with the disease, prevention of the manifestation of such symptoms before they occur, slowing down the progression of symptoms associated with the disease, slowing down or limiting any irreversible damage caused by the disease, lessening the severity of or curing the disease, or improving the survival rate or providing more rapid recovery from the disease. Further, in the context of prophylactic treatment the amount may also be effective to prevent the development of the disease or to prevent an infection (e.g, HIV infection).
[0074] The term“pharmaceutically acceptable carrier” refers to a substance that aids the administration of an active agent to a cell, an organism, or a subject.“Pharmaceutically acceptable carrier” also refers to a carrier or excipient that can be included in the compositions described herein and that causes no significant adverse toxicological effect on the patient. Non-limiting examples of pharmaceutically acceptable carriers include water, sodium chloride (NaCl), normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors and colors, liposomes, dispersion media, microcapsules, cationic lipid carriers, isotonic and absorption delaying agents, and the like. The carrier may also comprise or consist of substances for providing the formulation with stability, sterility and isotonicity (e.g, antimicrobial preservatives, antioxidants, chelating agents and buffers), for preventing the action of microorganisms (e.g, antimicrobial and antifungal agents, such as parabens, chlorobutanol,
phenol, sorbic acid and the like) or for providing the formulation with an edible flavor, etc. In some instances, the carrier is an agent that facilitates the delivery of a polypeptide, fusion protein, or polynucleotide to a target cell or tissue. One of skill in the art will recognize that other pharmaceutical carriers are useful in the present disclosure.
[0075] The term“vaccine” refers to a biological composition that, when administered to a subject, has the ability to produce an acquired immunity to a particular pathogen or disease in the subject. In particular embodiments of the present disclosure, a vaccine is used to produce an acquired immunity against HIV infection. Typically, one or more antigens, fragments of antigens, or polynucleotides encoding antigens or fragments of antigens that are associated with the pathogen or disease of interest are administered to the subject. In some instances, cells are engineered to express proteins such that, when administered as a vaccine, they enhance the ability of a subject to acquire immunity to an organism that causes an infectious disease, e.g., HIV. As used herein, the term“vaccine” includes, but is not limited to, systems and recombinant polynucleotides described herein, as well as viral particles, host cells, and pharmaceutical compositions that comprise systems or recombinant polynucleotides described herein. In particular embodiments, a vaccine refers to an HIV (or SIV) vaccine, e.g, a DNA vaccine encoding one or more HIV (and/or SIV) proteins, e.g, Env, Gag, protease, reverse transcriptase, Rev, and combinations thereof, or a composition comprising an HIV protein, e.g, gpl40.
3. Detailed Description of the Embodiments
[0076] The present disclosure provides methods and compositions for enhancing the anti- Env antibody response during HIV vaccination in a subject, e.g, during vaccination using a prime DNA vaccine encoding one or more HIV (or SIV) proteins, and/or during a boost step in which an HIV protein is administered. In particular, the methods and compositions involve the administration of one or more adjuvants, e.g, during the prime and/or boost step, in order to enhance the Thl-Tfh responses in the subject.
[0077] Any of a number of methods can be used to enhance Thl polarization in a subject. For example, one or more Thl -polarization promoting compounds can be administered in coordination with a prime vaccination step, e.g, by administering to the subject a DNA vaccine encoding one or more HIV polypeptides and a Thl -polarizing protein. In particular embodiments, the adjuvant is a protein such as IP- 10, and a polynucleotide encoding IP- 10 is
administered to the subject. In particular embodiments, a single DNA vector is administered one or more times to the subject that comprises polynucleotides encoding both the one or more HIV (or SIV) proteins and the adjuvant such as IP- 10.
[0078] In particular embodiments, the Thl -polarizing protein administered in coordination with the DNA prime is interferon-induced protein 10 (IP- 10). IP- 10 is also known as, e.g., CXCL10 or INP10, and corresponds, e.g, to NCBI Gene ID No. 3627. In some embodiments, a polynucleotide comprising the human IP-10/CXCL10 mRNA of NCBI Reference Sequence NM_001565, or of a polynucleotide sequence comprising at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to the nucleotide sequence of NM 001565, or encoding the polypeptide of NCBI Reference Sequence NP_001556.2, or encoding a polypeptide having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity to NCBI Reference Sequence NP_001556.2, is used. In some embodiments, a polypeptide comprising the human IP-10/CXCL10 protein of NCBI Reference Sequence NP 001556.2, or comprising a polypeptide sequence having at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or more identity to NP_001556.2, is used. In particular embodiments, a polynucleotide encoding IP-10 is used, and is administered to the patient on a DNA plasmid, e.g, a single DNA plasmid comprising polynucleotides encoding one or more HIV polypeptides and encoding IP- 10.
[0079] Thl polarization can also be promoted by administering to the subject a Thl- polarizing compound, e.g, adjuvant, in conjunction with one or more protein boosts, e.g, administration of an HIV protein such as gpl40. In particular embodiments, the boost protein, e.g, gpl40, is administered together with the adjuvant QS-21. Any form of QS-21 can be used in the present methods, including natural, semi-synthetic, and synthetic forms, and that QS-21 can be used at a variety of amounts, e.g, at 50 pg/500 mΐ of vaccine dose, or 40-60 pg/500 mΐ, 30-70 mg/500 mΐ, 20-80 mg/500 mΐ, 10-90 mg/500 mΐ, or 1-100 mg/500 mΐ of vaccine dose.
[0080] When a Thl -polarizing agent (e.g, a polynucleotide encoding a Thl -polarizing protein such as IP-10, or an adjuvant such as QS-21) is administered in coordination with an HIV antigen, e.g, a DNA molecule encoding an HIV protein or a protein boost, the agent and the antigen can be administered in any of a variety of ways with respect to one another. For example, the agent can be co-administered with the antigen, e.g, co-formulated and administered together to a subject. In some embodiments, as described elsewhere herein, a
single DNA plasmid is administered that encodes one or more HIV (or SIV) proteins, e.g., Env, and also encodes IP-10. In other embodiments, multiple plasmids or vectors, e.g, one or more plasmids encoding one or more HIV (or SIV) proteins, and a separate plasmid encoding IP-10, are formulated together. In some embodiments, a polypeptide boost such as gpl40 is co-formulated with a Thl-promoting adjuvant, e.g, QS-21.
[0081] In other embodiments, the Thl -polarizing agent is formulated independently from the HIV antigen, but they are administered at about the same time (e.g, within 10 minutes, 30 minutes, or 1 hour of each other, or during a single clinical visit, or on the same day). In other embodiments, the Thl -polarizing agent is formulated independently from the HIV antigen, and they are administered at different times (e.g, not during a single clinical visit, or not on the same day, or at least 1, 2, 3 or more days apart). In such embodiments, the Thl -polarizing agent can be administered before or after the administration of the HIV antigen.
Immunization
[0082] The HIV vaccines described herein can take on any of a number of forms, including through the administration of proteins, peptides, and nucleic acids including RNA or DNA encoding one or more HIV (or SIV) proteins.
Nucleic acid vaccines
[0083] In a particular embodiment, the vaccine is a DNA vaccine, e.g, a DNA vector such as a DNA plasmid comprising one or more polynucleotides encoding one or more HIV proteins, operably linked to one or more promoters. In particular embodiments, the DNA vaccine comprises a polynucleotide encoding the Env protein, or an immunogenic fragment or derivative thereof. In some embodiments, the DNA vaccine encodes multiple HIV proteins, e.g, any combination of Env, Gag, protease, reverse transcriptase, tat, and rev, or immunogenic fragments or derivatives thereof. The HIV proteins can be from HIV-1 or HIV- 2, from any group, e.g, HIV-1 group M, group N, group O, or group P, or HIV-2 group A, B, C, D, E, F, G, or H, and from any sub-group or clade, e.g, HIV-1 group M clade (sub-type) A, B, C, D, E, F, G, H, I, J, or K, or any strain, variant, or isolate from within any of such groups, sub-groups or clades. In particular embodiments, one or more of the proteins used for vaccination are from HIV-1 clade C. It will be appreciated that the precise HIV origin of each encoded protein is considered independently for the purposes of the present disclosure, e.g, one or more proteins can be from one clade, strain or variant/isolate, one or more proteins can be from a second clade, strain, or variant/isolate, etc. Any of these proteins can be encoded as
a full-length protein, or can be encoded as a fragment of the full-length protein, e.g., as a minimal peptide sequence capable of eliciting an immune response in a subject. Guidance to the selection of HIV proteins, protein fragments, peptides, variants or derivatives of HIV proteins, protein fragments, or peptides can be found, e.g, in Rappuioli and Bagnoli (eds.) (2011)“Vaccine Design: Innovative Approaches and Novel Strategies,” (Caister Academic Press, UK) or Agudelo and Patarroyo (2010),“Quantum chemical analysis of MHC-peptide interactions for vaccine design” Mini Rev Med Chem 10(8): 746-758, the entire disclosures of which are herein incorporated by reference. Any protein encoded by the HIV genome, e.g, gpl20 env, gpl40 env, gpl60 env, pi 8, gag, pol, vif, vpr, vpu, tat, ref, and nef, or any fragment, derivative, or combination of any of these proteins, can be used during the prime immunization phase (e.g, using polynucleotides encoding the protein, peptide, fragment, or derivative) and/or during the protein boost phase.
[0084] When polynucleotides, e.g, DNA plasmids, encoding one or more HIV proteins and/or IP- 10 are used, the polynucleotides are typically present within one or more expression cassettes, i.e., the polynucleotides are operably linked to one or more promoters. Any promoter capable of driving expression of the polynucleotides in one or more cells of a subject can be used, including inducible and constitutive promoters. In some embodiments, the CMV promoter is used. In some embodiments, all of the one or more HIV proteins and IP- 10 are operably linked to a single promoter, e.g, a CMV promoter, that drives constitutive expression in cells, leading to the expression of a single mRNA from which the different proteins are expressed by subgenomic splicing and frameshifting.
[0085] In particular embodiments, the DNA vaccine comprises a pGA2/JS2 plasmid, i.e., a DNA vector that encodes one or more of the HIV-1 Env, Tat, Rev, Gag, Protease, and reverse transcriptase proteins under the control of the CMV vector. In particular embodiments, a polynucleotide encoding IP- 10 is integrated into the vector, e.g, downstream of the env gene.
[0086] Plasmid constructs can be produced, maintained, and cultured using standard molecular biology techniques, e.g, as taught in Sambrook el al. , (1989),“Molecular Cloning: A Laboratory Manual”, (2nd ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y.; and Ausubel et al. (Eds), (2000-2010),“Current Protocols in Molecular Biology”, John Wiley and Sons, Inc. Before being used for vaccination, the function of the constructs can be assessed, e.g, by transfection into human cells, e.g, 293 T cells, and then grown, harvested, and assessed with respect to the expression of HIV proteins using, e.g, flow cytometry using
monoclonal antibodies. In addition, cellular and secreted expression of IP-10 or other Thl- polarizing proteins can be assessed, e.g., by intracellular cytokine signaling and/or ELISA.
[0087] Recombinant polynucleotides, (e.g, DNA plasmids, expression cassettes, polynucleotides encoding HIV antigens or IP- 10) can be prepared using standard molecular biology methods. Polynucleotides encoding protein antigens can be identified, e.g, by searching a human or other model organism DNA sequence database for any gene segment that has a certain percentage of sequence homology to a known nucleotide sequence, such as one encoding the antigen. Any DNA sequence so identified can be subsequently obtained by chemical synthesis and/or a polymerase chain reaction (PCR) technique such as the overlap extension method. For a short sequence, completely de novo synthesis may be sufficient; whereas further isolation of full length coding sequence from a human or other model organism cDNA or genomic library using a synthetic probe may be necessary to obtain a larger gene.
[0088] Alternatively, a nucleic acid sequence can be isolated from a cDNA or genomic DNA library (e.g, human or rodent cDNA or human, rodent, bacterial, or viral genomic DNA library) using standard cloning techniques such as polymerase chain reaction (PCR), where homology-based primers can often be derived from a known nucleic acid sequence. Commonly used techniques for this purpose are described in standard texts, e.g, Sambrook and Russell, supra.
[0089] Based on sequence homology, degenerate oligonucleotides can be designed as primer sets and PCR can be performed under suitable conditions (see, e.g, White el al. , PCR Protocols: Current Methods and Applications, 1993; Griffin and Griffin, PCR Technology, CRC Press Inc. 1994) to amplify a segment of nucleotide sequence from a cDNA or genomic library. Using the amplified segment as a probe, the full-length nucleic acid encoding a protein of interest is obtained. Oligonucleotides used in the construction of the recombinant polynucleotides that are not commercially available can be chemically synthesized, e.g, according to the solid phase phosphoramidite triester method first described by Beaucage & Caruthers, Tetrahedron Lett. 22: 1859-1862 (1981), using an automated synthesizer, as described in Van Devanter et. al. , Nucleic Acids Res. 12: 6159-6168 (1984). Purification of oligonucleotides is performed using any art-recognized strategy, e.g, native acrylamide gel electrophoresis or anion-exchange HPLC as described in Pearson & Reanier, J. Chrom. 255: 137-149 (1983).
[0090] Upon acquiring a nucleic acid sequence encoding a protein of interest, the coding sequence can be further modified by a number of well-known techniques such as restriction endonuclease digestion, PCR, and PCR-related methods to generate coding sequences, including mutants and variants derived from the wild-type protein. The polynucleotide sequence encoding the desired polypeptide can then be subcloned into a vector, for instance, an expression vector, so that a recombinant polypeptide can be produced from the resulting construct. Further modifications to the coding sequence, e.g., nucleotide substitutions, may be subsequently made to alter the characteristics of the polypeptide.
[0091] A variety of mutation-generating protocols are established and described in the art, and can be readily used to modify a polynucleotide sequence encoding a protein of interest. See, e.g., Zhang el al, Proc. Natl. Acad. Sci. USA, 94: 4504-4509 (1997); and Stemmer, Nature , 370: 389-391 (1994). The procedures can be used separately or in combination to produce variants of a set of nucleic acids, and hence variants of encoded polypeptides. Kits for mutagenesis, library construction, and other diversity -generating methods are commercially available.
[0092] In some embodiments, a recombinant polynucleotide or vector described herein contains a nucleic acid sequence that encodes a selectable marker. A selectable marker is useful, for example, when a polynucleotide described herein is being recombinantly modified. Taking antibiotic resistance genes as an example of a selectable marker, treating the cells that contain the recombinant polynucleotides with the antibiotic will identify which cells contain recombinant polynucleotides that have incorporated the antibiotic resistance gene (i.e., the cells that survive after antibiotic treatment must have incorporated the antibiotic resistance gene). If desired, the recombinant polynucleotides can be further screened (e.g, purified from the cells, amplified, and sequenced), in order to verify that the desired modification has been recombinantly introduced into the polynucleotide at the correct position.
Protein Booster
[0093] Following immunization using the prime vaccine, the subject will receive one or more boosts with HIV polypeptide antigens, e.g, gpl40. The boost can be administered any of a number of times following the immunization, e.g, 1, 2, 3, 4, 5 or more times, and at different intervals, e.g, every 1, 2, 3, 4, 5, 6, 7, 8, weeks or more. In one embodiment, the booster is administered two times, e.g, at weeks 30 and 44 (with week 0 corresponding to the
date of the first prime immunization). The boost can be administered at any of a variety of amounts, e.g., 100 pg protein/boost, or 10-50, 50-100, 100-150, or 150-200 pg protein/boost, or 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 200, 250, 300 or more pg protein, e.g, gpl40, per boost.
[0094] In addition to a Thl-promoting adjuvant such as QS-21, the booster polypeptide can be administered with one or more other adjuvants. For example, the booster can be administered with a liposomal and/or lipid adjuvant, e.g, Army Liposome Formulation (ALF) liposomes. ALF liposomes comprise dimyristoyl phosphatidylcholine (DMPC), dimyristoyl phosphatidylglycerol (DMPG) saturated phospholipids, cholesterol (chol), and monophosphoryl lipid A (MPLA), e.g., synthetic MPLA (e.g., 3D-PHAD), each of which can be obtained commercial suppliers such as Avanti Polar Lipids (Alabaster, AL). Other liposomes that can be used comprise CAFOl, an adjuvant that promotes Thl polarization and which can therefore be used in the place of, or in addition to, QS-21. For formulations adjuvanted with ALFQ (i.e., ALF + QS-21), lipids can be mixed in a molar ratio of, e.g, 9: 1 : 12.2:0.36 (DMPC:DMPG:Chol:MPLA), dried, rehydrated with, e.g., Sorenson PBS (pH 6.2), followed by microfluidization and filtration. The HIV protein, e.g, gpl40, can then be mixed with ALFQ in a 1 : 1 volume ratio, and vaccine doses can comprise, e.g, 100 pg MPLA, 100 pg protein, and 50 pg QS-21 in a total volume of 500 pi.
Subjects
[0095] The present methods and compositions can be used for the immunization of any subject, e.g, a human, that could benefit from an enhanced immune response against HIV infection, e.g, an enhanced anti-Env antibody response during HIV vaccination. In some embodiments, the subject is male. In some embodiments, the subject is female. In some embodiments, the subject is an adult (e.g, an adult male). In some embodiments, the subject is an adolescent. In some embodiments, the subject is a child.
[0096] In some embodiments, the subject has not been infected with HIV, e.g, and the methods and compositions are used to enhance the subject’s immune defenses against HIV in order to prevent future infection. In other embodiments, the subject is infected with HIV, and the methods are used to enhance the subject’s immune response against HIV in order to slow or potentially reverse the original infection.
Immune Response and Thl Polarization
[0097] The present methods and compositions provide for enhanced immune responses with vaccination against HIV (and/or SIV). In particular, the present methods and compositions provide for enhanced Thl polarization of Tfh cells. Without being bound to the following theory, it is believed that increased Thl polarization enhances the immune response by increasing the Env-specific Tfh cells when delivered with the DNA prime, and that Thl polarization with a protein boost results in greater production of the IgGl subclass with enhanced longevity, breadth, avidity, ADCC, and ADP activities of the anti-Env antibody.
[0098] The enhanced immune response and increased Thl polarization provided by the present methods and compositions can be detected, characterized, or quantified in any of a number of ways. For example, any of the assays described in any of the Examples to assess Thl polarization or the nature or strength of an immune response in a subject can be used. In some embodiments, Tfh cells, e.g., germinal center (GC) Tfh cells, are isolated from a subject, and the expression of one or more marker of Thl polarization assessed. Suitable markers include, but are not limited to, CXCR3, T-bet, IFNy, CXCR3, BCl-6, TBX21, SemA4A, and CCR5. In some embodiments, the administration of a Thl -polarizing adjuvant, e.g, with a DNA prime and/or with a protein boost, leads to an increase in or or more of the herein-described Thl polarization markers, e.g, to an increase relative to the level in the absence of the Thl -polarizing adjuvant.
[0099] In some embodiments, the efficacy of an Thl -polarizing adjuvant is assessed by detecting one or more aspects of the immune response in a subject to an HIV antigen, e.g, Env. For example, the extrafollicular and/or plasma cell-derived titers, e.g, vs. gpl40, can be evaluated using a binding antibody multiplex assay (BAMA) or an enzyme-linked immunosorbent assay (ELISA). In particular embodiments, the administration of a Thl- polarizing agent in coordination with the administration of an HIV antigen leads to an increase in an extrafollicular and/or plasma cell-derived antibody response against an HIV protein, e.g, gpl40. In some embodiments, the increase in antibody response can be, e.g, 1.25, 1.5, 1.75, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10 or more fold higher in subjects receiving a Thl -polarizing adjuvant than in subjects not receiving a Thl -polarizing adjuvant.
[0100] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the breadth, e.g, cross-clade breadth, of serum IgG antibodies relative to the breadth in a subject not
receiving a Thl -polarizing adjuvant. In some embodiments, the breadth of the antibodies is assessed by determining the kinetics of the antibody response against different HIV antigens, e.g., titers against Env from different HIV clades. Antibody binding against different HIV proteins, e.g, against HIV Env proteins from different Groups, Clades, or isolates, e.g, against Con S (group M consensus), Con C, or isolate Case A2 or CH505 proteins, can be assessed, e.g, using a binding antibody multiplex assay (BAMA) or ELISA. In some embodiments, binding to gpl20 V1V2 loops is assessed, e.g, scaffolded on murine leukemia virus (MLV) gpl70.
[0101] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the longevity of the serum IgG antibody response that is greater than any increase in longevity that may be observed in a subject not receiving a Thl -polarizing adjuvant. For example, the elevation of anti-Env titers observed following one or more HIV protein boosts can be detected over time and the duration of the elevated antibody response assessed. In one embodiment, titers are assessed at, e.g, week 8 and week 18 following the second protein boost, and the elevation observed in subjects receiving the Thl -polarizing adjuvant at the second time point (e.g, at 18 weeks following the second protein boost) is at least as high as any elevation observed in subjects not receiving the Thl -polarizing adjuvant.
[0102] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the avidity (e.g, expressed as dissociation constants, kd) of serum IgG antibodies relative to the avidity in a subject not receiving a Thl -polarizing adjuvant. Increases in avidity can be assessed, e.g, by Surface Plasmon Resonance (SPR) or by 2M sodium thiocyanate or 0.1 M sodium citrate displacement ELISA with, e.g, gpl40 protein or with Con C, Con S, or other HIV proteins from other groups, clades, variants or isolates.
[0103] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in the capacity of immune sera to neutralize HIV, e.g, HIV-1, e.g, as assessed using a TZM-bl assay, in which neutralization titers are measured in TZM-bl cells infected with HIV vs. cells infected with a control virus such as MLV. In such embodiments, for example, titers can be assessed against, e.g, subtype C tier 1A variants, tier IB variants, and tier 2 isolates. In particular embodiments, the induction of tier 1A neutralizing antibodies in a subject receiving a Thl-
polarizing adjuvant is at least as high as any induction observed in a subject not receiving a Thl -polarizing adjuvant, at one or more time points following one or more protein boosts.
[0104] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in antibody- dependent cellular toxicity (ADCC) and/or antibody-dependent phagocytosis (ADP) triggered by engagement of Fc receptors on antibody -bound target cells by innate cells. ADCC can be assessed, e.g., by measuring killing of HIV-infected target cells, e.g, by NK cells ( e.g. , CD16+/FcyR3 NK cells) in the presence of immune serum. ADP can be assessed, e.g, by measuring the phagocytosis of gpl20-coated beads by CD32+ (FcyR2) and/or CD64+ (FcyRl) THP-1 monocytic cells. In particular embodiments, the increase in ADCC and/or ADP is at least as high in a subject receiving a Thl -polarizing adjuvant as it is in a subject not receiving a Thl -polarizing adjuvant.
[0105] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in anti-gpl20 IgGl antibodies relative to IgG2, IgG3, or IgG4 antibodies, e.g, as measured by ELISA. In particular embodiments, Thl -polarizing adjuvants lead to a specific increase in anti-gpl20 IgGl antibodies in subjects, and in particular to an increase that is as high or higher than any increase observed in subjects not receiving a Thl -polarizing adjuvant. In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to a specific increase in the IgGl/IgG4 ratio among anti-gpl20 antibodies, and in particular to an increase that is as high or higher than any increase in the IgGl/IgG4 ratio in subjects not receiving a Thl -polarizing adjuvant.
[0106] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in mucosal anti- gpl20 IgG and IgA antibodies relative to IgG2, IgG3, or IgG4, and in particular to an increase that is as high or higher than any increase observed in subjects not receiving a Thl- polarizing adjuvant. For example, in some embodiments, Thl -polarizing adjuvants lead to a specific increase in anti-gpl40 IgG and/or IgA antibodies in vaginal and/or rectal secretions, wherein the increase is stronger and/or has greater longevity than any increase seen in subjects not receiving a Thl -polarizing adjuvant.
[0107] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in Env-specific
Tfh cells and GC Tfh cells with distinctive Thl signatures. For example, peripheral blood mononuclear cells (PBMCs) can be isolated and stimulated with overlapping peptide pools comprising one or more gpl40 proteins, and the induction of activation markers such as CD25 and 0X40 assessed by flow cytometry after stimulation. In some embodiments, the frequency of Env-specific CD4 T cells is higher in subjects receiving a Thl -polarizing adjuvant than in subjects not receiving a Thl -polarizing adjuvant, as is the percentage of Env-specific CD95+ CD4 T cells.
[0108] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to an increase in germinal center (GC) Tfh cells (as assessed, e.g., by virtue of CXCR5+, PD-1+++ expression) and GC B cells (as assessed, e.g, by virtue of Ki-67+, Bcl-6+, CD20 expression), and in particular to an increase that is as high as or higher than any increase seen in subjects not receiving a Thl- polarizing adjuvant. For example, subjects receiving Thl -polarizing adjuvants can show higher Env-specific Tfh cell frequencies in the lymph nodes (LN) and can show a correlation between Tfh cell frequencies in the LN and GC Tfh cells, but not with memory Tfh cells, indicating that GC Tfh cells are enriched for vaccine-induced follicular cells.
[0109] In some embodiments, the administration of a Thl -polarizing adjuvant in coordination with the administration of an HIV antigen leads to a systemic expansion of pro- inflammatory monocytes and an increase in GC Tfh responses, e.g, during the priming immunization phase. For example, Thl -polarizing antibodies increase the relative frequencies and absolute counts of ICOS+ PD-1 + CXCR5+ CD4 T cells in blood, e.g., at day 14 following a priming immunization step, as well as an increase in CD 14+ CD 16+ HLA-DR+ cells in blood. In some embodiments, Thl -polarizing adjuvants lead to an increase in IL-Ib levels, e.g, as determined using a flow-based Legendplex assay, and to an increase in GC Tfh cell frequencies within fine-needle aspirates of the draining LN, that is as high or higher than any increase observed in subjects not receiving a Thl -polarizing adjuvant.
[0110] Any of these parameters or effects, or any of the parameters or effects described in the Examples or elsewhere herein, can be used to assess the efficacy of a Thl -polarizing adjuvant. In some embodiments, a Thl -polarizing adjuvant leads to an increase of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, or more in any of the herein-described parameters or effects relative to a control value (e.g, to the value observed in a subject not receiving a Thl -polarizing adjuvant). As such, a Thl-
polarizing adjuvant can refer to any agent that, when administered with either a DNA prime against HIV, or with a protein boost against HIV, can lead to an increase of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 250%, 300%, or more in any of the herein-described parameters or effects relative to a control value ( e.g ., the value observed in a subject not receiving a Thl -polarizing adjuvant), at any point of the immunization process (e.g., at any one or more points of the priming phase and/or booster phase).
Pharmaceutical Compositions and Administration
[0111] The present disclosure provides compositions comprising an immunogenic component (e.g, a DNA vaccine or an immunogenic polypeptide) capable of inducing immunity against a targeted agent (e.g, HIV Env protein), comprising a Thl -polarizing adjuvant (e.g, a polynucleotide encoding IP-10 or an agent such as QS-21), and potentially comprising one or more additional adjuvants or compounds such as Army liposome formulations. In some embodiments, the compositions further comprise a pharmaceutically acceptable carrier. As such, the present disclosure provides pharmaceutical compositions for inducing an anti -Env (or other HIV protein or antigen) in a subject. In some embodiments, the composition comprises one or more polynucleotides encoding one or more HIV proteins, e.g, Env, and one or more polynucleotides encoding a Thl -polarizing polypeptide, e.g, IP- 10, and a pharmaceutically acceptable carrier. In some embodiments, the composition comprises one or more HIV polypeptides, e.g, gpl40, one or more Thl-promoting adjuvants, e.g, QS-21, and a pharmaceutically acceptable carrier.
[0112] The compositions may be formulated for, e.g, injection, inhalation, or topical administration, e.g, facilitating direct exposure of host cells and tissues to the immunogenic component and Thl -polarizing adjuvant. In particular embodiments, the compositions, e.g, prime DNA vaccines, are formulated for transdermal injection, e.g, by electroporation. In particular embodiments, the compositions, e.g, protein boost, are formulated for subcutaneous or intramuscular administration, e.g, delivery into the thigh. In particular embodiments, the compositions, e.g, protein boost, are formulated in liposomes, e.g, Army liposome formulations (ALFA), e.g, by the lipid deposition method. In other embodiments, the compositions are formulated in nanoparticles or in dry powder form, e.g, suitable for delivery by particle bombardment. In some embodiments, the compositions, e.g, DNA
vaccine, are formulated as naked DNA (see, e.g, US Patent Nos. 6,265,387, 6,972,013, and 7,922,709).
[0113] In particular embodiments, the DNA vaccines are prepared as DNA vectors or plasmids, e.g, a pGA2 plasmid, or pGA2/JS2 plasmid, or derivative thereof. In some embodiments, the compositions, e.g, DNA vaccine, are prepared as recombinant viruses, e.g, by modifying a parent virus to incorporate exogenous genetic material, e.g, one or more polynucleotides encoding one or more HIV proteins or antigens and/or a polynucleotide encoding IP-10. A non-limiting list of suitable viruses that can be used for the purposes of the present disclosure include lentiviruses (e.g, HIV, HIV-1, HIV-2, FIV, BIV, EIAV, MW, CAEV, SIV), adenoviruses and adeno-associated viruses, alphaviruses, flaviviruses, and poxviruses. For methods and examples concerning the use of suitable viral vectors, see, e.g, US Patent Nos. 5,219,740, 7,250,299, 7,608,273, 6,465,634, 7,811,812, 5,744,140, 8,124,398, 5,173,414, 7,022,519, 7,125,705, 6,905,862, 7,989,425, 6,468,711, 7,015,024,
7,338,662, 5,871,742, and 6,340,462. In such embodiments, the viruses are typically recombination-competent (i.e., capable of reproducing in an infected host cell). Modification of such viruses and vectors or plasmids for the preparation of the present DNA vaccines can be achieved using standard molecular biology techniques, e.g, as taught in Sambrook el al. (1989)“Molecular Cloning: A Laboratory Manual” (2nd ed. Cold Spring Harbor Press) and Ausubel et al. (Eds.) (2000-2010)“Current Protocols in Molecular Biology” (John Wiley and Sons).
[0114] The pharmaceutical compositions described herein may comprise a pharmaceutically acceptable carrier. In certain aspects, pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical compositions described herein (see, e.g, REMINGTON’S PHARMACEUTICAL SCIENCES, 18TH ED., Mack Publishing Co., Easton, PA (1990)).
[0115] The pharmaceutical compositions described herein will often further comprise one or more buffers (e.g, neutral buffered saline or phosphate buffered saline), carbohydrates (e.g, glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants (e.g, ascorbic acid, sodium metabi sulfite, butylated hydroxytoluene, butylated hydroxyanisole, etc.), bacteriostats, chelating agents such as
EDTA or glutathione, solutes that render the formulation isotonic, hypotonic or weakly hypertonic with the blood of a recipient, suspending agents, thickening agents, preservatives, flavoring agents, sweetening agents, and coloring compounds as appropriate.
[0116] The pharmaceutical compositions described herein are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically or prophylactically effective. The quantity to be administered depends on a variety of factors including, e.g., the age, body weight, physical activity, hereditary characteristics, general health, sex, and diet of the individual, the condition or disease to be treated or prevented, and the stage or severity of the condition or disease. In certain embodiments, the size of the dose may also be determined by the existence, nature, and extent of any adverse side effects that accompany the administration of a therapeutic or prophylactic agent(s) in a particular individual. Other factors that can influence the specific dose level and frequency of dosage for any particular patient include the activity of the specific compound employed, the metabolic stability and length of action of that compound, the mode and time of administration, and the rate of excretion.
[0117] Generally, for administering the compound (e.g, vaccine or adjuvant) for therapeutic or prophylactic (e.g, vaccination) purposes, the compound is given at a therpaeutically or prophylactically effective dose. In particular, an effective amount of a pharmaceutical composition described herein is an amount that is sufficient to obtain an enhanced immune response against an HIV vaccine, e.g, in view of any of the parameters or indices described herein, and/or a sufficient amount to enhance the immunity of a subject to HIV infection or to the propagation of an already-existing HIV infection in the subject.
[0118] In certain embodiments, the dose may take the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as, for example, tablets, pills, pellets, capsules, powders, solutions, suspensions, emulsions, suppositories, retention enemas, creams, ointments, lotions, gels, aerosols, foams, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages.
[0119] As used herein, the term“unit dosage form” refers to physically discrete units suitable as unitary dosages for humans and other mammals, each unit containing a predetermined quantity of a therapeutic or prophylactic agent calculated to produce the desired onset, tolerability, and/or therapeutic or prophylactic effects, in association with a suitable pharmaceutical excipient (e.g, an ampoule). In addition, more concentrated dosage
forms may be prepared, from which the more dilute unit dosage forms may then be produced. The more concentrated dosage forms thus will contain substantially more than, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more times the amount of the therapeutic or prophylactic compound.
[0120] Methods for preparing such dosage forms are known to those skilled in the art (see, e.g, REMINGTON’S PHARMACEUTICAL SCIENCES, supra). The dosage forms typically include a conventional pharmaceutical carrier or excipient and may additionally include other medicinal agents, carriers, adjuvants, diluents, tissue permeation enhancers, solubilizers, and the like. Appropriate excipients can be tailored to the particular dosage form and route of administration by methods well known in the art (see, e.g, REMINGTON’S PHARMACEUTICAL SCIENCES, supra).
[0121] The pharmaceutical compositions described herein can be administered locally or systemically to the subject, e.g, intraperitoneally, intramuscularly, intra-arterially, orally, intravenously, intracranially, intrathecally, intraspinally, intralesionally, intranasally, subcutaneously, intracerebroventricularly, topically, and/or by inhalation. In particular embodiments, the compositions are administered by intradermal injection with electroporation (e.g, for the priming DNA vaccine) or subcutaneously (e.g, for the protein boost).
[0122] In some embodiments, the DNA vaccines and/or polynucleotides encoding IP-10 or other Thl -polarizing protein are administered, e.g, by intradermal injection with electroporation (e.g, using the ICHOR TriGrid Array; Ichor Medical Systems), and can be administered any of a number of times, e.g, 1, 2, 3, 4, 5 or more times, and following any of a number of vaccination regimens, e.g, every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more weeks. In a particular embodiment, the immunization is performed 3 times, e.g, at weeks 0, 8, and 16. DNA vaccines can be administered at any of a number of levels, e.g, 4 mg of plasmid DNA vector per subject per vaccination, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more mg of plasmid DNA vector per subject per vaccination, or 1-10, 1-20, 1-8, 1-7, 2-6, 3-5 mg or plasmid DNA vector per subject per vaccination.
[0123] In some embodiments, the protein boost and/or QS-21 or other Thl -polarizing adjuvant is administered by intradermal injection, e.g, into the thigh, and can be administered any of a number of times following the immunization, e.g, 1, 2, 3, 4, 5 or more times, and following any of a number of regimens, e.g, every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14,
15, 16, 17, 18, 19, 20 weeks or more. The protein boost can be administered at any of a number of levels, e.g., 100 pg protein/boost, or 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 200, 250, 300 or more pg protein, e.g., gpl40, per boost. In some embodiments, the protein boost, e.g, gpl40, is mixed with ALFQ in a 1 : 1 volume ratio, and vaccine doses comprise, e.g, 100 pg MPLA, 100 pg protein, and 50 pg QS-21 in a total volume of 500 pi.
Kits
[0124] In another aspect, kits are provided herein. In some embodiments, the kit comprises a DNA prime composition (e.g, a DNA vaccine encoding one or more HIV proteins, e.g, Env, and also encoding a Thl -polarizing protein, e.g, IP-10, and optionally a pharmaceutically acceptable carrier). In some embodiments, the kit comprises a protein boost composition (e.g, an HIV protein, e.g, gpl40, a Thl -polarizing adjuvant, e.g, QS-21, optionally one or more additional adjuvants, e.g, ALF, and optionally a pharmaceutically acceptable carrier). In some embodiments, the kit comprises one or more DNA prime compositions and one or more protein boost compositions. In some embodiments, the kit is for inducing an enhanced (e.g, stronger, broader, longer) immune response against an antigen, e.g, HIV Env protein (e.g, in a subject). In other embodiments, the kit is for preventing or treating a disease, e.g, HIV.
[0125] The kits described herein can be packaged in a way that allows for safe or convenient storage or use (e.g, in a box or other container having a lid). Typically, the kits described herein include one or more containers, each container storing a particular kit component such as a reagent, a control sample, and so on. The choice of container will depend on the particular form of its contents, e.g, a kit component that is in liquid form, powder form, etc. Furthermore, containers can be made of materials that are designed to maximize the shelf-life of the kit components. As a non-limiting example, kit components that are light-sensitive can be stored in containers that are opaque.
[0126] In some embodiments, the kit contains one or more elements, e.g, syringe, useful for administering compositions (i.e., a pharmaceutical composition described herein) to a subject. In yet other embodiments, the kit further comprises instructions for use, e.g, containing directions (i.e., protocols) for the practice of the methods described herein (e.g, instructions for using the kit for enhancing an immune response in a subject to an HIV protein). While the instructional materials typically comprise written or printed materials,
they are not limited to such. Any medium capable of storing such instructions and communicating them to an end user is contemplated by this disclosure. Such media include, but are not limited to electronic storage media ( e.g ., magnetic discs, tapes, cartridges, chips), optical media (e.g., CD ROM), and the like. Such media may include addresses to internet sites that provide such instructional materials.
4. Examples
[0127] The following examples are offered to illustrate, but not to limit, the claimed invention.
Example 1. Adjuvant-Dependent Modulation of CD4 T Follicular Helper Cells Impacts Longevity and Functional Quality of Antibody Responses to a Subtype C HIV-1 Envelope Vaccine in Rhesus Macaques
Abstract
[0128] Enduring humoral immunity through vaccination is dependent upon effective interaction of follicular helper CD4 T cells (Tfh) with germinal center (GC) B cells. Thl polarization of Tfh cells is an important process shaping the success of Tfh-GC B cell interactions by influencing co-stimulatory and cytokine-dependent Tfh help to B cells. However, the question remains whether adjuvant-dependent modulation of Thl-Tfh cells enhances HIV-1 vaccine-mediated antiviral envelope (Env) antibody responses. In this study, we investigated whether an HIV-1 vaccine platform designed to increase the number of Thl- polarized Tfh cells enhances the functional quality and longevity of anti-Env antibodies. Utilizing a novel interferon-induced protein (IP)-lO-adjuvanted HIV subtype C DNA prime, followed by an MPLA+QS-21-adjuvanted subtype C Env protein boost in rhesus macaques (Thl group), we observed higher anti-Env serum IgG antibody titers with significantly increased cross-clade reactivity and heightened specificity to VI V2 loops when compared to macaques primed with DNA lacking IP- 10 and boosted with MPLA+alum-adjuvanted Env protein (Thl+2 group). The Thl vaccine regimen increased serum antibody effector functions, induced higher IgGl and lower IgG4 antibodies, increased circulating Env-specific Tfh cells and generated greater anti-Env mucosal IgA responses. Within lymph nodes, we observed augmented GC B cell responses and promotion of Thl gene expression profiles in GC Tfh cells. The frequency of GC Tfh cells predicted both the magnitude and avidity of anti-Env serum IgG response at memory time points. Together, these data demonstrate that
adjuvant-induced stimulation of Thl-Tfh cell production during the prime and the boost is an effective strategy to enhance the longevity and functional quality of the anti-Env antibody response.
Introduction
[0129] In this study, we investigated whether an HIV-1 vaccine platform designed to increase the number of Thl-polarized Tfh cells would enhance the functional quality and longevity of HIV-1 anti-Env antibodies. Our data show that a Thl prime followed by a Thl boost increases HIV-1 anti-Env specific binding antibody in serum and mucosal compartments. The Thl vaccine regimen augmented GC B cell responses and promoted Thl gene expression profiles in GC Tfh cells. The number of GC Tfh cells positively correlated with both longevity and avidity of anti-Env specific antibody responses. Together, these data show that by stimulating production of Thl-Tfh cells during the prime and boost regimen using an adjuvanted vaccine, we can enhance the longevity and function of the anti-HIV-1 - Env antibody response.
Results
[0130] Vaccination regimen. Twenty female rhesus macaques were assigned to one of two experimental groups: For Group 1 (n = 10), the Thl vaccine group, the Thl chemokine, interferon-induced protein (IP)- 10, a ligand for and an inducer of CXCR3, was used as a molecular adjuvant to a DNA vaccine, (DIP-10) to prime Thl-type Tfh cells. Group 2 (n=10) animals received the same DNA vaccine without adjuvant (FIG. ID). The DNA plasmid expressed SIVmac239 Gag, protease, reverse transcriptase, Tat, Rev, HIV C. 1086 Env, and the DIP-10 plasmid additionally expressed rhesus IP-10. The DNA was delivered intradermally (ID) with electroporation (EP) in both experimental groups.
[0131] Prior to immunizing animals, we evaluated plasmid constructs using 293 T cells. At 48 h following transfection, cells were harvested and expression of HIV proteins was assessed by flow cytometry using monoclonal antibodies (PG9, PG16, PGT121 for surface Env, 2F12 for intracellular SIV Gag, and J034D6 for intracellular IP-10). As illustrated in FIG. 1A flow plots, both constructs expressed comparable levels of Env and Gag proteins as determined by staining with PGT121 and 2F12, respectively. Cellular and secreted IP- 10 as determined by intracellular cytokine staining (FIG. IB) and ELISA (FIG. 1C), respectively, was specific to the DNA IP- 10 construct. Following DNA immunization, we used clade C C.ZA 1197MB gpl40 protein adjuvanted with Army Liposome Formulation (ALF)
liposomes, monophosphoryl lipid A (MPLA) and a detoxified saponin derivative, QS-21 (ALFQ) (20) to boost Thl primed responses (DIP-10 PALFQ; Thl group) (FIG. ID). Group 2 animals received an unadjuvanted ID, EP delivered DNA prime and protein adjuvanted with MPLA + aluminum-adsorbed ALF formulation (ALFA) (21), wherein the protein was adsorbed to aluminum hydroxide and then added to ALF (DPALFA;Thl+2 group). Blood was collected at weeks -8 and 0 of vaccination, and at weeks 1, 2, 4, 8, 18, and 20 following each vaccination, as indicated. Fine needle aspirates of lymph nodes (LN) or LN biopsies (draining) were collected to examine GC responses, and rectal and vaginal secretions were sampled to assess mucosal antibodies (FIG. ID).
[0132] DIP-10 ProteinALFQ vaccine induces robust and durable anti-Env antibody titers with cross-clade breadth. To ascertain whether induction of Thl versus mixed Thl+2 inflammatory responses elicited anti-Env antibody responses of different magnitudes, we first evaluated responses against C.1086 gpl40 Env using a binding antibody multiplex assay (BAMA)(22). We have previously shown that the transient extrafollicular plasmablast response contributes to peak serum IgG antibody titers following the boost, while titers at week 8 and beyond are mainly plasma cell derived (12). Therefore, we assessed antibody levels at weeks 0, 2, and 8 following each of the protein boosts to capture both extrafollicular (week 2) and plasma cell-derived (week 8 and beyond) titers. The data showed robust induction of anti-C.1086 Env responses following the 1st protein immunization in all 20 animals and potent recall of memory B cells following the 2nd protein immunization as evidenced by a robust boost in antibody responses (FIG. 2A). Strikingly, Env ALFQ boosted animals developed significantly higher responses against C.1086 gpl40; median AUC values in ALFA (Thl+2) and ALFQ (Thl) vaccine groups were: wk 0, 7496 and 20301, p < 0.01; wk 2, 46481 and 63469, p < 0.001; wk 8, 20714 and 36709, p < 0.0001 2nd protein boost.
[0133] We confirmed these findings by using an independent enzyme linked immunosorbent assay (ELISA) assay to explore C.1086 gpl40 anti-Env antibody kinetics after the 2nd protein boost (FIG. 2B). The assay revealed that anti-Env titers exhibited a median 5-fold increase at week 2 post-2nd protein immunization relative to week 0 indicating a successful booster response. In affirmation of the BAMA data, antibody titers were significantly higher in the Thl group in comparison to the Thl+2 group at week 0 (median ng/mL of anti-gpl40 IgG; Thl+2 = 257 and Thl = 1314; p < 0.01), at week 2 (Thl+2 = 9942 and Thl= 29104; p < 0.0001), week 8 (Thl+2=1830 and Thl = 7076; p < 0.001), and week 18 (Thl +2= 450 and Thl= 2431; pO.OOl).
[0134] We next assessed the breadth of the serum IgG antibody response and found that AUC values against CH505 subtype C Env were also significantly higher in the Thl group relative to the Thl+2 group (p< 0.01, FIG. 2C). Similarly, increased responses against the Con S (group M consensus) and Con C proteins at week 2 following the 2nd protein boost in the Thl group were sustained at week 8 demonstrating greater induction of antibodies with cross-clade breadth using the Thl vaccine regimen (p< 0.05, FIGS. 2D and 2E). We also assessed binding to gpl20 V1V2 loops from isolate Case A2, scaffolded on murine leukemia virus (MLV) gp70, at weeks 2 and 8 and found that significantly higher specificity to these important regions was induced in the Thl vaccine regimen following the second protein boost (p < 0.05, FIG. 2F).
[0135] Based on significantly elevated anti-Env antibody responses in the Thl vaccine regimen we sought to quantify antibody longevity. To this end, we calculated fold change in titers at week 8 and 18 following 2nd protein boost relative to titers at week 8 post 1st protein boost. Significantly higher titers at week 8 (mean 1.7-fold in Thl+2 versus 4.5-fold in Thl group; p< 0.05) and week 18 (mean 0.3-fold in Thl+2 versus 1.3-fold in Thl group; p< 0.01) post 2nd protein boost in Thl vaccinated animals demonstrated that the Thl vaccine regimen was effective at enhancing durability of anti-Env serum IgG titers (FIG. 2G). Together, the data show that the Thl group had higher induction of cross-clade breadth, elicited stronger binding to a gp70-VlV2 protein, and enhanced antibody longevity relative to the Thl+2 group.
[0136] DIP-10 ProteinALFQ vaccine elicits high avidity anti-Env antibody with ADCC and ADP activities. Next, we quantified avidity of IgG binding antibodies (as disassociation constants, kd) in sera collected at 2 weeks post final DNA prime and after each of the protein boosts using Surface Plasmon Resonance (SPR) to C.1086 gpl40 protein (23). The data showed that gp 140-specific antibodies reached higher avidity with each sequential immunization in both vaccine groups (p < 0.0001, FIGS. 3A and 3B). Increased avidities in the Thl vaccine group relative to the Thl+2 group was suggestive of more productive GC reactions in the Thl vaccine group (p< 0.05, FIG. 3C). To confirm that higher avidity antibodies in the Thl vaccine group were sustained, we determined avidity at 8 weeks following the 2nd protein boost using a 2M sodium thiocyanate displacement ELISA with C.1086C gpl40 antigen (12). The data showed sustained induction of higher avidity antibodies in the Thl group (p< 0.05, FIG. 3D), which was further corroborated with a 0.1 M sodium citrate ELISA (p< 0.01, FIG. 3E). Notably, higher avidity antibodies against Con C
and Con S gpl40 proteins were also induced in the Thl vaccine regimen (p< 0.01, FIGS. 3F and 3G).
[0137] After establishing induction of higher avidity antibodies in the Thl vaccine group, we next evaluated capacity of immune sera to neutralize HIV-1 using the classic TZM-bl assay (12). We detected robust activity against MW965.26, a subtype C tier 1 A variant (FIG. 3H) whereas neutralization of tier IB and tier 2 isolates was sporadic (24). The data showed higher induction of tier 1A neutralizing antibodies in the Thl vaccine group (ID50 range at week 2 post 2nd protein boost Thl+2: 37 -1126; Thl : 195-4977, p < 0.01). These titers dropped to an ID50 value of 20 in the Thl+2 group but were maintained between 24-1057 in the Thl group (p< 0.001). To assess generation of Fc-mediated antibody effector responses, we measured antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent phagocytosis (ADP) triggered by engagement of the Fc receptors on antibody-bound target cells by innate cells (25) (26, 27). ADCC was assessed by measuring killing of Clade C CH505 SHIV-infected CEM.NKR target cells by a rhesus CD16+ (FcyR3) NK cell line in the presence of immune serum. As shown in FIG. 31, serum from Thl vaccinated animals demonstrated significantly greater ADCC activity at week 2 and week 8 after the 2nd protein boost when compared to Thl+2 immunized animals (p< 0.01). Serum collected from Thl vaccinated animals at week 8 post protein boost 2 also mediated significantly greater phagocytosis of C.1086 gpl20-coated beads by the CD32+ (FcyR2) and CD64+ (FcyRl) THP-1 monocytic cell line (FIGS. 3J and 3K). To determine if the adjuvants given to animals in the Thl and Thl+2 groups generated different IgG subclass antibody repertoires, we quantified C.1086 gp 120-specific IgGl, IgG2, IgG3, and IgG4 by ELISA. We found that IgG2 and IgG3 antibodies were extremely low and did not differ between groups (FIG. 3L). However, Thl vaccinated animals had higher gpl20-specific IgGl (p< 0.0001, FIG. 3L) while Thl+2 vaccinated animals had higher gp 120-specific IgG4 resulting in markedly elevated IgGl/IgG4 ratio in the Thl vaccine group (p< 0.001, FIG. 3M). These results are consistent with the report that antibodies of the IgGl subclass are the most functional in rhesus macaques (28, 29).
[0138] DNAIP-lOProteinALFQ vaccine elicits robust anti-Env antibody in vaginal and rectal mucosal compartments. Having established induction of higher serum IgG antibody titers in Thl vaccinated animals, we next sought to determine whether mucosal anti-Env antibodies were also correspondingly increased. To this end, we assayed rectal and vaginal
secretions for C.1086 gpl40-specific IgG and IgA antibodies at baseline and longitudinally after each of the protein boosts.
[0139] The appearance of gp 140-specific IgG in secretions closely mimicked the kinetics of the serum IgG antibody response, with each protein boost increasing levels of Env-specific IgG antibodies in vaginal and rectal secretions (FIGS. 4A and 4B). As in serum, the Thl vaccine regimen generated higher levels of specific IgG in secretions when compared to the Thl+2 vaccine. The gp 140-specific IgA in vaginal and rectal secretions was also increased to a greater extent by the Thl vaccine regimen (FIGS. 4C and 4D). Notably, at week 16 post 2nd protein boost, vaginal IgA antibodies were still above the limit of detection in most Thl vaccinated animals but in only 2 of 10 Thl+2 vaccinated animals. Analysis of gp 140-specific IgA in serum revealed higher induction in the Thl group (FIG. 4E). However, the kinetics of the serum IgA response in Thl as well as Thl+2 animals differed strikingly from the mucosal IgA responses, especially in the reproductive tract (FIGS. 4C and D), suggesting a true mucosal (locally-derived) IgA response was generated in these animals. This was most evident after the 2nd protein boost, when vaginal IgA antibodies to gpl40 were found to be dramatically increased but serum IgA antibodies were reduced (FIGS. 4A and 4C). Together, these data demonstrate that the Thl vaccine regimen was more effective than the Thl+2 regimen for generating long-lived anti-Env binding antibodies in serum and secretions, as well as serum IgG antibodies with greater breadth, avidity, and function.
[0140] DIP- 10 ProteinALFQ vaccine induces Env-specific Tfti cells and GC Tfh cells with distinctive Thl signatures. The DIP-10 PALFQ Thl vaccine promoted anti-Env antibody longevity and functionality following the 1st protein boost. Based upon this finding, we wanted to determine whether this vaccine regimen also correspondingly enhanced Tfh responses in the periphery and LNs. To this end, we first assessed whether higher magnitude of Env-specific Tfh cells were induced 7 days after 1st boost, corresponding to the peak of the effector response. PBMCs were stimulated with overlapping peptide pools representing Con C gpl40 together with C.1086 gpl40 protein. The induction of the activation markers CD25 and 0X40 was assessed by flow cytometry after stimulation (FIG. 5A, flow plot) (30). The analysis revealed a higher frequency of Env-specific CD4 T cells in DIP-10 PALFQ animals. When expressed as a percentage of CD95+ CD4 T cells, median frequencies of Env specific-CD4 T cells were on average 10-fold higher in Thl group, indicative of a higher magnitude Env-specific Tfh response (p < 0.001, FIG. 5B). The frequency of Env-specific CD4 T cells responding to the C.1086 gpl40 priming immunogen correlated positively with
CD4 T cells responding to the C.ZA gpl40 boosting immunogen (p< 0.0001, FIG. 5C). Evaluation of IP- 10 levels in sera using a flow-based Legend plex assay at day 0 and 3 of 1st protein boost in the ALFQ-adjuvanted animals confirmed induction of IP- 10 following the Thl protein boost (p< 0.05, FIG. 5D).
[0141] Next, we assessed LN responses using biopsies collected at day 14 post 1st protein boost and identified GC Tfh cells as CXCR5+, PD-1+++ cells (red population, FIG. 6A) and GC B cells as Ki-67+, Bcl-6+ CD20 cells. As expected, Bcl-6 expression was highest on GC Tfh cells relative to PD1++ Tfh and PD1- memory Tfh populations. GC Tfh cells also expressed higher relative levels of ICOS compared to other CD4 subsets, and consistent with the functional ability of Tfh cells (12), our ex vivo analysis of sorted GC Tfh cells revealed their capacity to support IgG production from autologous B cells (FIG. 6B). Evaluation of GC TFH frequencies over the course of immunization revealed a significant induction of GC Tfh cells 2 weeks after protein boost 1, and significantly higher frequencies 2 weeks after protein boost 2 (FIG. 6C).
[0142] Following the 1st protein boost, we found that GC B cell frequencies were significantly higher in the Thl vaccine regimen (n=10 animals in each group; median Thl+2: 14.2% (of CD20+ cells) versus Thl : 25%, p < 0.05), and the frequency of GC Tfh cells strongly correlated with GC B cell responses. (FIG. 6D). Importantly, Env-specific Tfh cell frequencies in the LN directly correlated with GC Tfh cell frequencies but not memory Tfh cells, indicating that GC Tfh cells were enriched for vaccine-induced follicular cells (p<0.0001, FIG. 6D). Next we assessed expression of CXCR3, which is heterogeneously expressed by GC Tfh cells (histogram, FIG. 6E), and found higher relative expression of CXCR3 both within GC Tfh and GC B cells in the Thl vaccine group (p< 0.05, FIG. 6E). Notably, T-bet expression on B cells, a marker of memory B cells (31), corresponded with CXCR3 expression, suggesting a mechanistic basis for enhanced antibody responses in the Thl vaccine group. Consistent with this hypothesis, the magnitude of Tfh cells and the relative frequency of CXCR3+ Tfh cells within the GC was directly predictive of serum antibody titers at week 18 post 2nd protein boost. GC responses predicted antibody avidity at week 8 after the 2nd protein boost, supporting our hypothesis that induction of Thl-Tfh cells enhanced the quality of anti-Env antibody responses (p< 0.05, FIG. 6G).
[0143] To gain insights into the molecular mechanisms underlying successful antibody responses, we next determined transcriptional signature in GC Tfh cells. To achieve this goal,
we sorted four different CD4 subsets from the LNs of 3 Thl group animals with highest antibody titers at week 8 post 1st protein boost. These subsets were naive cells (CD4+CD95- ), Tfh cells (CD95+CXCR5+PD-1 +/++), memory Tfh cells (CD95+CXCR5+ PD-1-), and memory non-Tfh cells (CD95+ CXCR5-PD-1-). Similar subsets were sorted from 2 high responder animals in the Thl+2 vaccine group (animal sorted identified by triangles, FIG. 6H).
[0144] RNA samples meeting quality control checks were sequenced using a 3'-Tag-RNA- Seq library prep protocol on the Illumini HiSeq 4000 platform. Prior to analysis of sequenced reads, genes with fewer than 40 counts per million reads were filtered, leaving 7,086 genes. Differential expression analyses were conducted using the limma-voom Bioconductor pipeline. We performed significance analysis of microarrays using the Dseq package from R/Bioconductor to compare the transcriptome profiles. Principal component analysis of the top 500 differentially expressed genes showed that the majority of variation in the data was driven by CD4 subsets. Memory CD4 T cells (CXCR5+ and CXCR5-) clustered closely together relative to naive and Tfh subsets (FIG. 61). Segregation of Tfh cells between the Thl and Thl+2 groups indicated adjuvant dependent modulation of transcriptional activity within the germinal center, suggestive of qualitative differences in Tfh cells between vaccine groups. To extract information on biologically relevant gene-sets, we performed gene set enrichment analysis with the goal of determining biological pathways that were enriched in Tfh cells in the Thl vaccine regimen. Genes regulating interleukin (IL)-12, tumor necrosis factor (TNFa), interferon gamma (IFNG), and IL-6 production were strongly enriched in Tfh cells. Consistent with metabolic activity of effector cells and functional capacity of Tfh cells, pathways regulating cellular metabolism, glucose homeostasis, and B cell proliferation were also enriched (FIG. 6J).
[0145] To determine transcriptional activity of Tfh cells in the Thl vaccine group, we focused on differentially induced genes in Tfh cells relative to naive cells (n=89, adj. p < 0.05, FIG. 6K), of which induction of key Tfh transcripts including CXCR5, ICOS, and Bcl- 6 was common to both Tfh cells and memory Tfh cells (FIG. 6L). Consistent with representation of Thl genes in GSEA, Tfh cells showed higher expression of TBX21 and IFNG (FIG. 6L). The class IV semaphorin protein (SEMA4A), a co-stimulatory molecule expressed by Thl cells (32), was significantly induced, as was high-mobility group box 1 (HMGB1), an inflammatory mediator regulating TNF and IL-6 production (33). Induction of IL-18R suggested the capacity of IL-18 to drive IFNy production within the GC (34).
Likewise, we noted higher expression of receptor interacting serine/threonine kinase 2 (RIPK2), which drives IFNy in Thl cells and contributes to Thl differentiation (35). The corresponding downregulation of IL-4R in Tfh cells indicated enrichment of the Thl program within Tfh cells in DIP- 10 ProALFQ vaccinated animals. This, together with increased protein expression of CXCR3 within the GC, supports the conclusion that CD4 T cell help for humoral immunity was driven by Thl Tfh cells in the Thl vaccine regimen (FIG. 61).
[0146] DNAIP-10 immunization induces systemic expansion of pro-inflammatory monocytes and enhances GC Tfh responses. Based on increased frequencies of Env-specific Tfh cells and evidence for induction of a Thl transcriptome program in Thl vaccinated animals following the 1st protein boost, we sought to assess Tfh responses during the priming immunization phase. First, we evaluated blood to quantify activated CXCR5+ CD4 T cells (FIG. 7A). Based on co-expression of ICOS and PD-1, activation markers induced upon TCR stimulation, the data showed that DNA immunization significantly increased the relative frequencies and absolute counts of ICOS+ PD-1+ CXCR5+ CD4 T cells in blood at day 14 (n=20; median frequencies, day 0: 3.38%; day 14: 6.7%, p < 0.0001; n =20; absolute counts, day 0: 3.04; day 14, 8.7 day 14, p < 0.01, FIG. 7A). Based on the robust induction of activated CXCR5+ CD4 T cells, we next assessed whether a concomitant acute induction of pro-inflammatory monocytes (innate cells that drive Tfh responses) preceded the activated CXCR5+ response (36, 37). We quantified frequencies of CD14+ CD16+ HLA-DR+ (lineage-) cells in blood (FIG. 7B) and discovered rapid and robust expansion of pro- inflammatory monocytes with significantly higher induction in the Thl vaccine group (FIGS. 7C and 7D). Consistent with increased pro-inflammatory monocyte frequencies at day 3, IL- 1b levels, determined using a flow-based Legendplex assay, were elevated at day 7 following the 3rd DNA boost. Based on evidence showing that monocyte-derived IL-1B drives effective Tfh and CD4 T cell differentiation, we examined LN responses (FIG. 7E). Strikingly, the GC Tfh cell frequencies within the fine-needle aspirates of the draining LN were higher in the Thl vaccine group following the 3rd DNA prime (FIG. 7F). Notably, the greater inflammatory response was associated with increased levels of antibodies, linking the innate immune response to priming of effective CD4 Tfh help (FIG. 7G).
Discussion
[0147] The present study gives rise to three main conclusions; first, that an HIV-1 vaccine platform designed to promote Thl-polarized Tfh cells increases the number of circulating
Env-specific Tfh cells, enhances GC responses, increases anti-Env binding antibody titers in sera, stimulates serum antibody effector functions. Second, that a Thl vaccine regimen can elicit anti-Env vaginal and rectal IgA responses; and third that induction of high avidity antibodies, reflective of productive GC responses, are engendered by a Thl vaccine regimen. Collectively, the data demonstrate that adjuvant-induced stimulation of Thl-Tfh cell production during the vaccine prime and boost is an effective strategy to enhance the longevity and functionality of the anti-Env antibody response.
[0148] Productive T cell responses critically depend on cytokine signals during priming, and recent studies demonstrate that monocyte-derived IL-Ib drives effective CD4 T cell differentiation and Tfh responses (37-39). Here, investigation of the kinetics of pro- inflammatory monocytes - cellular innate biomarkers of adjuvanticity - revealed a transient increase in CD14+CD16+ monocytes in blood with a higher relative increase in the Thl vaccine group and a corresponding transient increase in IL-Ib. Strikingly, fine-needle aspirates of the draining LNs showed higher GC frequencies in the Thl vaccine group, indicating active/productive GC responses. Notably, the improved inflammatory response was associated with increased antibody longevity linking the innate immune response to effective induction of CD4 Tfh cells. Indeed, several recent studies show that potent priming of the immune response sets the stage for stronger boosting of cellular and humoral immunity in the setting of DNA prime, NYVAC boost and Ad5 prime, NYVAC boost vaccine regimens (25, 40). The effectiveness of priming is not limited to CD4 T cells and B cells; a DNA vaccine targeting conserved elements of SIV Gag robustly primes cytotoxic T cells which are effectively boosted following a long rest period (41, 42). These data open the possibility to a critical window of opportunity during the priming phase. This window can be exploited to prime for long-lasting, durable CD4, CD8 T cell, and antibody responses to HIV- 1 vaccination.
[0149] The HVTN studies 070 and 080 employed the IL-12 DNA adjuvanted plasmid with the subtype B PENNVAX-B (PV) DNA plasmid and showed 80% response rates after the third DNA vaccination in PV+IL-12 recipients compared to a 44% response rate with the PV alone vaccine. A subsequent follow up study demonstrated robust recall of binding anti-Env antibody titers with ADCC activity following an MVA boost in PV+IL-12 recipients (43, 44). Because IL-12 is a classic innate mediator of Thl responses, the data suggest that an increase in Thl GC Tfh cells may underlie the observed effects. Correspondingly, studies in rhesus macaques with an ALVAC prime, ALVAC + gpl20 protein boost using SIV immunogens
showed higher SIV Env titers with MF59 compared to alum adjuvanted protein boosts 2 weeks following the final immunization (45). While Tfh responses and memory antibody titers were not examined, a recent study in humans showed enhanced binding antibody titers 26 weeks after booster immunization with a Thl GLA-SE-adjuvanted malaria antigen relative to one formulated in aluminum (46). These studies in conjunction with the present study provide support to the immune potential of Thl-Tfh cells in fostering long-lived antibody titers. In contrast, a study using a homologous subtype C protein immunization reported induction of higher anti-Env antibody titers with aluminum-hydroxide Ahydrogel® relative to Addavax™, an MF59 analog, in rabbits (47). Collectively, these data indicate the importance of detailed studies to understand the context in which Thl responses are superior to Thl+2 responses and how viral versus DNA vectors and subunit proteins influence this paradigm.
[0150] Our findings raise the question of the mechanisms underlying the DIP-lOProALFQ vaccine-mediated enhancement in Tfh responses. A few possibilities can be explored; IP- 10 increases dendritic cell-T cell interactions, which could have favored Tfh differentiation (48). IP- 10 also increases IL-6 production in B cells which is known to support Tfh differentiation and enhance plasma cell differentiation (49). This, together with the potent immune stimulatory potential of MPL+QS-21 boost, may have synergized to enhance Tfh responses numerically and favored Thl differentiation program within Tfh cells (50). Indeed, GC Tfh cells induced following viral infections where Thl inflammatory responses predominate express Bcl-6, Tbx21, IFNG, and IL-21 consistent with the induction of Thl-type Tfh cells (51). Our transcriptomic analysis of Tfh cells following the 1st protein boost in the Thl vaccine regimen show coordinate expression of Thl regulated genes as evidenced by enrichment of pathways related to IFNG signaling. The higher relative expression of the Thl chemokine receptor CXCR3 in GC Tfh cells and GC B cells validate the gene expression data. Together, our transcriptomic and phenotypic data on Tfh cells indicate a role for adjuvant induced quantitative (increased Tfh numbers) and qualitative (increased proportion of Thl-Tfh cells) effects on antibody longevity. Mechanistic studies are needed to discern the respective contribution of increased Tfh numbers versus Thl skewing of Tfh cells on antibody responses as both these characteristics are inextricably linked in the current study.
[0151] In addition to adjuvant-dependent modulation of Tfh responses, we made the striking observation that the Thl vaccine regimen induced significantly higher anti-Env IgGl titers while the Thl+2 vaccine regimen induced relatively higher anti-Env IgG4 titers. These
data provide the strongest evidence for adjuvant impacting antibody subclass profile in rhesus macaques to an Clade C HIV vaccine regimen. Thus, the predominance of IgGl over IgG4 at week 8 following the last boost, a time point when majority of the circulating antibody is plasma cell derived, supports the conclusion that the Thl vaccine regimen induced qualitatively different GC responses relative to a Thl +2 vaccine regimen. Enhancement of antibody effector functions - both ADCC and ADP in the Thl vaccine group are consistent with report antibodies of the IgGl subclass are the most functional in rhesus macaques (28, 29) and corroborate our conclusions.
[0152] Another notable observation was induction of robust mucosal anti-Env IgA responses in the reproductive tract and in rectal secretions. Interestingly, unlike mucosal anti- Env IgA responses, which were boosted after the 2nd protein immunization, serum IgA responses were not correspondingly enhanced. This incongruity between mucosal and serum IgA responses suggests that a true mucosal (locally-derived) response was generated in vaccinated animals and that the Thl vaccine was more effective in inducing mucosal immunity. These findings raise the possibility that induction of Thl7-type Tfh cells within the mucosal draining lymph nodes could underlie mucosal IgA responses. Although our studies were not designed to evaluate this possibility, the effectiveness of the Thl/Thl7 adjuvant cationic liposomal formulation (CAFOl) suggests that induction of Thl/Thl7-type Tfh cells may be extremely important to drive robust mucosal IgA responses while correspondingly enhancing serum anti-Env IgG responses and functionality (52).
[0153] In summary, our findings demonstrate that Thl-DNA prime substantially increases the Env-specific Tfh cells relative to a Thl+Th2 vaccine regimen and that Thl-protein boost results in greater production of the IgGl subclass with enhanced longevity, breadth, avidity, ADCC, and ADP activities of anti-Env antibody.
Materials & Methods
[0154] Rhesus Macaques. Twenty adult female colony-bred rhesus macaques ( Macaco mulatto) were housed at the California National Primate Research Center and maintained in accordance with American Association for Accreditation of Laboratory Animal Care guidelines. All studies were approved by the University of California Davis Institutional Animal Care and Use Committee (IACUC). At study initiation, animals were 3.5 - 4.5 years of age with a median weight of 5.3 kg, were SIV- STLV- SRV-, had no history of dietary or pharmacological manipulation, and had intact ovaries.
[0155] Immunizations. DNA immunizations were administered via intradermal injection with electroporation utilizing the ICHOR TriGrid Array (Ichor Medical Systems, San Diego) at weeks 0, 8, and 16. For each DNA immunization, two groups of 10 animals received 4 mg of the pGA2/JS2 plasmid DNA vector (53) encoding either SHIV C.1086 T/F Env + interferon-induced protein (IP)-10 (Group 1) or SHIV C.1086 T/F Env alone (Group 2). Details of the SHIV DNA construct have been described (54). At weeks 30 and 44, Group 1 animals received boosts with 100 pg C.ZA 1197MB gpl40 protein (Immune Technology, New York, NY) adjuvanted with 100 pg MPLA +50 pg QS-21 (ALFQ) and Group 2 animals received 100 pg C.ZA 1197MB gpl40 adjuvanted with 100 pg MPLA + 600 pg Aluminum (ALFA). The protein formulation was delivered in a 250 pi volume (50 pg protein) subcutaneously in each thigh during both boosts.
[0156] Adjuvants. Dimyristoyl phosphatidylcholine (DMPC) and dimyristoyl phosphatidylglycerol (DMPG) saturated phospholipids, cholesterol (Choi), and synthetic monophosphoryl lipid A (MPLA, 3D-PHAD) were purchased from Avanti Polar Lipids (Alabaster, AL). DMPC and Choi were dissolved in chloroform, and DMPG and MPLA were dissolved in chloroform: methanol (9: 1). Alhydrogel®, aluminum hydroxide (AH) in a gel suspension was purchased from Brenntag (Frederikssund, Denmark). Saponin, QS-21 were purchased from Desert King International (San Diego, CA) and was dissolved in Sorensen PBS, pH 5.6.
[0157] Army liposome formulations (ALF) containing DMPC, DMPG, Choi, and MPLA were prepared by the lipid deposition method. For vaccine preparations adjuvanted with ALFA, dissolved lipids were mixed in a molar ratio of 9:1 :7.5:0.36
(DMPC :DMPG: Choi :MPLA) and dried by rotary evaporation followed by overnight desiccation. Liposomes were formed by molecular biology grade water (Quality Biological, Gaithersburg, MD), microfluidized, and sterile filtered, followed by lyophilization. 100 pg of gpl40 protein was adsorbed to 600 pg of Alhydrogel in PBS, pH 7.4, and incubated on a tilted roller at room temperature (RT) for 1 h prior to adding to lyophilized ALF. For vaccine preparations adjuvanted with ALFQ (ALF containing QS-21), lipids were mixed in a molar ratio of 9: 1 : 12.2:0.36 (DMPC:DMPG:Chol:MPLA), dried, rehydrated by adding Sorensen PBS, pH 6.2, followed by microfluidization and filtration. gpl40 was mixed with ALFQ in a 1 : 1 volume ratio. Each vaccine dose in 500 pi volume contained 100 pg MPLA (and 100 pg protein) and either 600 pg aluminum or 50 pg QS-21.
[0158] Specimen collection and processing. Lymph node (LN) biopsies were obtained 2 weeks following each of the protein boosts and were manually processed by disassociation through IOOmM cell strainers and washing in complete media, as described previously (12). Two weeks after the 3rd DNA immunization, fine needle aspirates of LN were obtained using a 22 gauge needle, as previously described (55). PBMCs were isolated from whole blood collected in CPT vacutainer tubes by density gradient centrifugation as previously described (12). For serum, coagulated blood was centrifuged at 800g for 10 min to pellet clotted cells, followed by extraction of fluid and storage at -80° C. Rectal and vaginal secretions were collected using premoistened Week-Cel sponges and eluted as described (56).
[0159] C.1086 gp 140-specific serum IgG ELISA. Serum IgG titers against HIV-1 C.1086
Env gpl40 were determined by ELISA. In brief, 96-well microtiter plates with high binding capacity (Thermo Fisher, MA) were coated overnight at 4° C with 1 pg/mL C.1086 Env gpl40C from the NIH AIDS Reagent Program (ARP) diluted in 0.1M carbonate-bicarbonate buffer, pH 9.2. Plates were washed with PBS containing 0.1% Tween-20 (PBST) and blocked with 5% w/v nonfat dry milk in PBS for 2h at RT followed by four washes with PBST. Standard (PG9 monoclonal antibody from the ARP) and serum samples were run at 3 dilutions/sample (1 :50-1 :450) in sample dilution buffer and incubated at RT for 2 h on a microplate shaker. After washing, the plate was incubated for 1 h with 1 : 10,000 HRP conjugated goat anti-monkey IgG (Nordic MUbio, Netherlands). The plates were washed and then developed with TMB substrate (Thermo Fisher) and the reaction was quenched with 2N H2SO4 (Sigma, St. Louis, MO). Absorbance was recorded at 450nm with a reference filter at 570 nm using a Spectramax 5 plate reader (Molecular Devices, Sunnyvale, CA). Baseline sera from each animal served as negative control and OD values 2-fold above baseline were considered positive and extrapolated to determine anti-Env antibody concentrations.
[0160] Sodium thiocyanate avidity assay. C.1086 Env gpl40C-specific IgG antibody avidity was determined using a chaotropic displacement ELISA with NaSCN. Serum samples were incubated in duplicate at 6000 pg per well for 2h at RT. The plate was washed five times. For the dissociation step, one well of each sample was manually treated with 100 pL of 2 M NaSCN (Sigma-Aldrich) to dissociate antigen-antibody complexes and a second well of the same sample was treated with PBS as a control. The plate was incubated for 15 min at RT, followed by washing three times. The plate was then developed as described above for the C.1086 gpl40C ELISA. For each sample, antibody avidity was reported as an avidity
index value (a percentage), which was calculated as the ratio of absorbance in the well treated with NaSCN to that in the well treated with PBS.
[0161] Biacore binding and avidity analysis. Binding and avidity determination were conducted using Surface Plasmon Resonance (SPR) Biacore 4000 system. The immobilizations were performed in 10 mM HEPES and 150 mM NaCl pH 7.4 using a standard amine coupling kit, as previously described (23, 57). The CM5-S series chip surface was activated with a 1 : 1 mixture of 0.4 M 1 -ethyl-3 -(3 -dimethylaminopropyl) carbodimide hydrochloride (EDC) and 0.1 M N-hydroxysuccinimide (NHS) for 600s (GE Healthcare). For the cyclic biotinylated V2 C.1086 peptide, 1 mM Streptavidin (Life Technologies) in 10 mM sodium acetate pH 4.5 (5,800 - 7,400 RU) was coupled for 720s. The immobilized surface was then deactivated with 1.0 M ethanolamine-HCl pH 8.5 for 600s. Spot 3 in each flow cell was left unmodified to serve as a reference. Following surface deactivation, 0.06 - 1.5 mM cyclic biotinylated V2 C.1086 peptide was captured, resulting in two range of densities; high density (1,900 - 2,300 RU) and low/medium density (340 - 580 RU). For C.1086 gpl40C, 0.56 - 15 pg/mL protein was immobilized directly on the sensor CM5 chip, resulting in four ranges of densities; very high density (9.800 - 10,100 RU); high density (3,400 - 4,100 RU); medium density (960 - 1,700 RU) and low density (240 - 670 RU). Following surface preparation, heat inactivated serum samples were diluted 1 :50 in the running buffer (10 mM Hepes, 300 mM NaCl and 0.005% Tween 20, pH 7.4). The diluted samples were injected onto the V2 peptide or gpl40 protein surface for 320 s followed by 1,800 s dissociation period. The bound surface was then enhanced with a 240 s injection of 30 pg/mL secondary antibody goat anti -monkey IgG. To regenerate the bound surface, 175 mM HC1 was injected for 70s. For each serum sample or controls, 4 - 8 replicates were collected at a rate of 10 Hz, with an analysis temperature of 25° C. All sample injections were conducted at a flow rate of 10 pL/min. Data analysis was performed using Biacore 4000 Evaluation Software 4.1 with double subtractions for unmodified surface and buffer for blank. The fitting was conducted using the dissociation mode integrated with the Evaluation software 4.1.
[0162] Binding Antibody Multiplex Assay (BAMA) and sodium citrate avidity assay. SIV- specific serum IgG BAMA was performed with a panel of Env and VI V2 antigens: C.1086 gpl40, CH505 TF gpl40, Con S (group M consensus) gpl40, and Con C (clade C consensus) gpl40, gp70-VlV2 Clade B/Case A2 scaffolded protein and C.1086 VI V2 avitagged protein. Samples were titrated in 5-fold serial dilutions starting at 1 :80 and binding magnitude is reported as AUC. Positivity criteria (determined at dilution 1 :80) was as follows: (1) MFI
>100; (2) MFI > Ag-specific cutoff (95th percentile of all baseline binding per antigen); (3) MFI 3 -fold > than that of the matched baseline before and after blank/MuLV subtraction. All BAMA and avidity assays were performed in a blinded fashion using magnetic beads. For avidity assays, samples were tested with and without sodium citrate (0.1 M, pH 3.0) at 2 dilutions for each antigen based on BAMA titration for maximum coverage of samples in the linear range of the assay. The dilutions were 1 :80 and 1 :400 for gp70-VlV2, 1 :400 for C.1086 VI V2, 1 :2000 for CH505TF gpl40, 1 :2000 for ConC gpl40, and 1 : 10000 for C.1086 gpl40 and ConS gpl40. Antibody avidity is reported as avidity index, which was calculated as 100 x (MFI in the citrate-treated well/MFI in the untreated well). Avidity index is reported for sample-antigen combinations that were (1) identified as positive responders in the IgG BAMA assay and (2) had an MFI within the linear range for the untreated sample. The pre set assay criteria for sample reporting were coefficient of variation per duplicate values of <15% for each sample and >100 beads counted per sample.
[0163] Neutralization. Neutralization assays were performed as previously described (58) using TZM-bl cells. We measured neutralization activity against the tier 1 clade C pseuodvirus MW965.26 using MLV-pseudotyped virus as an indicator of non-HIV-specific activity in the assay. Neutralization titers were measured at week 2 and week 8 post 2nd protein boost and were considered to be positive for neutralizing antibody activity based on the criterion of signal >3x detected against the MLV negative control virus. The majority of positive titers detected were against the tier 1 virus MW965.26 with occasional very weak neutralization titers against the tier 2 viruses.
[0164] Antibody-dependent cellular cytotoxicity. Engineered human cell lines CD 16+ KHYG-1 (effector cells) and CEM.NKR-CCR5-sLTR-Luc (target cells) provided by D. Evans were maintained in R10 cell culture medium consisting of RPMI medium supplemented with 10% fetal bovine serum, 25 mM HEPES, 2 mM 1-glutamine, and 0.1 mg/ml Primocin (59, 60). The growth medium of CD16+ KHYG-1 cells was additionally supplemented with cyclosporine (CsA) and interleukin-2 (IL-2) at a concentration of 1 pg/ml and 5U/mL, respectively.
[0165] Luciferase based ADCC assays were carried out as previously described with some modifications (59). Two million CEM.NKR-CCR5-sLTR-Luc target cells were spinoculated with SHIV.C.CH505.375H.dCT (38 ng p27) for 2 hours at 2,600 rpm at 30° C in the presence of 1 pg/mL polybrene. Subsequently, the target cell/virus mixture was incubated overnight at
37° C 5% CO2. The next day, virus was removed and cells were incubated for another 72 hours prior to the ADCC assay. For the ADCC assay, serum: effector cells: target cells were plated in a 1 : 1 : 1 volumetric ratio. Sera was heat inactivated and diluted (1 :50 dilution in RIO containing 10 U IL-2 per mL, with no CsA), mixed with PBS-washed, infected target cells (1 x 104 cells per well), and effector cells (5 x 104 cells per well). Assay plates were incubated overnight at 37°C and 5% CO2. Plates were then centrifuged at 1,800 rpm for 5 min at room temperature and 100 pL of the supernatant was removed. The cell pellets were resuspended and mixed with 50 pi of the luciferase substrate reagent BriteLite Plus (Perkin Elmer, MA). Luciferase activity was read in black 96-well plates according to the manufacturer’s instructions using a Synergy 2 micro plate luminometer (BioTek). Percent ADCC activity of each tested animal immune serum (week 2 and week 8 post 2nd protein) measured as reduction in RLUs, was calculated based on respective week 0 pre-immune serum (100% RLU). All samples were tested in triplicates and experiments were performed twice.
[0166] Antibody Dependent Phagocytosis. Serum antibodies were tested for ability to enhance phagocytosis of gpl20 expressing beads by THP-1 cells using methods similar to those previously described (58). Briefly, 5 pL of 1 pm avidin-coated Fluorospheres (Invitrogen) were labeled with 2 pg biotinylated anti-His tag antibody (Pierce), then 3.5 pg His-tagged Clade C gpl20 Dul51 protein (Immune Technologies) per plate. The gpl20 beads and triplicate 5-fold dilutions of heat-inactivated serum in a 50 pL volume were then pre-incubated at 37° C in V-bottom plates. After 1 h, 2 x 104 THP-1 cells in 50 pL were added to each well. After 5 h at 37°C in 5% CO2, the cells were washed in Ca+2 and Mg+2 - free DPBS and resuspended in 180 pL of warm 0.12% Trypsin/EDTA. After 5 min at 37° C, the trypsin was removed and the cells were resuspended in 1% paraformaldehyde. Fluorescence was evaluated using a FACS Canto (BD Biosciences) and Flo-jo software. Phagocytosis was measured by multiplying the % fluorescent cells by their median fluorescence intensity. The phagocytic score was then calculated by dividing phagocytosis of test samples by the average phagocytosis measured with preimmune serum.
[0167] IgG subclass antibodies. Ten rows of a 96-well Immulon 4 microtiter plate (VWR) were coated overnight at 4° C with 50 ng per well of C.1086 gpl20 D7 K160N protein (61) in PBS. The remaining 2 rows were coated with duplicate 2-fold serial dilutions of rhesus IgGl, IgG2, IgG3 or IgG4 (Nonhuman Primate Reagent Program) starting at 25 ng/mL in PBS to generate a standard curve. Plates were washed with PBS containing 0.05% Tween 20 and blocked for 30 min at RT with reagent buffer (0.1% bovine serum albumin in wash buffer).
Two- or three-fold dilutions of serum in reagent buffer were then added to the wells coated with gpl20. Reagent buffer was added to wells coated with standard. Following overnight storage at 4° C, the plate was washed and reacted for 1 h at 37° C with 1 pg/mL of the relevant monoclonal antibody from the Nonhuman Primate Reagent Program: anti -rhesus IgGl (mouse IgG2a clone 3C10), anti-rhesus IgG2 (mouse IgGl clone 3C10), anti -rhesus IgG3 (mouse IgGl clone 2G11) or anti-rhesus IgG4 (mouse IgGl clone 7A8). The plate was then consecutively washed and treated with 100 ng/mL of biotinylated goat anti-mouse IgGl or IgG2a for 1 h at 37° C, neutralite-avidin peroxidase for 30 min at RT, and TMB (all from SouthernBiotech). Absorbance was recorded at 370 nm. SoftMax Pro software (Molecular Devices) was used to to construct a standard curve and determine concentrations of antibody. Preimmune serum samples had < 10 ng/mL of antibody in these assays.
[0168] Mucosal antibodies and serum IgA. BAMA with C.1086 gpl40 K160N-labeled magnetic beads (MagPlex, BioRad) was used as previously described (61) to measure concentrations of antigen-specific IgG in secretions and IgA in both secretions and serum depleted of IgG. Briefly, beads reacted with dilutions of standard (62) and specimens at 1100 rpm and 4° C overnight were washed and developed with biotinylated anti-monkey IgG or - monkey IgA (Rockland) followed by Phycoerythrin-labeled Neutralite avidin (SouthernBiotech). Construction of standard curves and interpolation of antibody concentrations was done using Bioplex Manager software after measurement of fluorescence in a Bioplex 200 (BioRad). Concentrations of gp 120-specific IgG or IgA in secretions were divided by the total IgG or IgA measured in the sample by ELISA (63) to obtain the specific activity (ng IgG or IgA antibody per pg total IgG or IgA).
[0169] Activation induced Marker (AIM) assay. Cells were stimulated with overlapping peptide pools of HIV consensus C and HIV-1 C.1086 Env gpl40C protein in AIM media as previously described (30). All antigens were used at a final concentration of 2 pg/mL in a stimulation cocktail made with using 0.2 pg of CD28 and 0.2 pg CD49d costimulatory antibodies per test. Unstimulated controls were treated with volume-controlled DMSO (Sigma-Aldrich). Tubes were placed in 5% CO2 incubator at 37° C and incubated overnight. Following an 18 hour stimulation, the cells were stained, fixed, and acquired the same day. Phenotype panel on lymph nodes and PBMCs was performed using standard flow cytometry assays.
[0170] Legend Flex assay. A Legendplex assay was performed to evaluate cytokines in rhesus macaque sera (Biolegend). The assay was performed according to the manufacturer’s protocol. Samples were acquired on a BD LSR Fortessa cell analyzer.
Table 1.
[0171] Flow cytometry and cell sorting. Cells were acquired on a BD FACSymphony using FACS Diva version 8.0.1. Compensation, gating and analysis were performed using FlowJo (Versions 9 and 10). Cell sorting was performed using BD FACSAria III. Reagents used for flow cytometry are listed in Table 1.
[0172] Statistical analysis. Statistical analysis was performed using GraphPad Prism 7. Results for groups were compared using the two-tailed nonparametric Mann-Whitney rank sum test. For correlation analysis, the two-tailed Spearman rank correlation test was used.
References
1. Breitfeld D, Ohl L, Kremmer E, Ellwart J, Sallusto F, Lipp M, Forster R. 2000.
Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med 192: 1545-1552.
2 MacLennan IC. 1994. Germinal centers. Annu Rev Immunol 12: 117-139.
3. Crotty S. 2019. T Follicular Helper Cell Biology: A Decade of Discovery and Diseases. Immunity 50: 1132-1148.
4. Aljurayyan A, Puksuriwong S, Ahmed M, Sharma R, Krishnan M, Sood S, Davies K, Rajashekar D, Leong S, McNamara PS, Gordon S, Zhang Q. 2018. Activation and Induction of Antigen-Specific T Follicular Helper Cells Play a Critical Role in Live- Attenuated Influenza Vaccine-Induced Human Mucosal Anti-influenza Antibody Response. J Virol 92.
5. De Boer RJ, Perelson AS. 2017. How Germinal Centers Evolve Broadly Neutralizing Antibodies: the Breadth of the Follicular Helper T Cell Response. J Virol 91.
6. Bannard O, Cyster JG. 2017. Germinal centers: programmed for affinity maturation and antibody diversification. Curr Opin Immunol 45:21-30.
7. Song W, Craft J. 2019. T follicular helper cell heterogeneity: Time, space, and function. Immunol Rev 288:85-96.
8. Schmitt N, Ueno H. 2013. Blood Tfh cells come with colors. Immunity 39:629-630.
9. Schmitt N, Ueno H. 2015. Regulation of human helper T cell subset differentiation by cytokines. Curr Opin Immunol 34: 130-136.
10. Bentebibel SE, Khurana S, Schmitt N, Kurup P, Mueller C, Obermoser G, Palucka AK, Albrecht RA, Garcia-Sastre A, Golding H, Ueno H. 2016. ICOS(+)PD-l(+)CXCR3(+) T follicular helper cells contribute to the generation of high-avidity antibodies following influenza vaccination. Sci Rep 6:26494.
11. Baiyegunhi O, Ndlovu B, Ogunshola F, Ismail N, Walker BD, Ndung'u T, Ndhlovu ZM. 2018. Frequencies of Circulating Thl-Biased T Follicular Helper Cells in Acute HIV-1 Infection Correlate with the Development of HIV-Specific Antibody Responses and Lower Set Point Viral Load. J Virol 92.
12. Iyer SS, Gangadhara S, Victor B, Gomez R, Basu R, Hong JJ, Labranche C, Montefiori DC, Villinger F, Moss B, Amara RR. 2015. Codelivery of Envelope Protein in Alum with MVA Vaccine Induces CXCR3-Biased CXCR5+ and CXCR5- CD4 T Cell Responses in Rhesus Macaques. J Immunol 195:994-1005.
13. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Kamasuta C, Sutthent R, Liao HX, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. 2012. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 366: 1275-1286.
14. Kim JH, Excler JL, Michael NL. 2015. Lessons from the RV144 Thai phase III HIV-1 vaccine trial and the search for correlates of protection. Annu Rev Med 66:423-437.
15. Ding BB, Bi E, Chen H, Yu JJ, Ye BH. 2013. IL-21 and CD40L synergistically promote plasma cell differentiation through upregulation of Blimp- 1 in human B cells. J Immunol 190: 1827-1836.
16. Yusuf I, Kageyama R, Monticelli L, Johnston RJ, Ditoro D, Hansen K, Barnett B, Crotty S. 2010. Germinal center T follicular helper cell IL-4 production is dependent on signaling lymphocytic activation molecule receptor (CD150). J Immunol 185: 190-202.
17. Lin L, Finak G, Ushey K, Seshadri C, Hawn TR, Frahm N, Scriba TJ, Mahomed H, Hanekom W, Bart PA, Pantaleo G, Tomaras GD, Rerks-Ngarm S, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Michael NL, Kim JH, Robb ML, O'Connell RJ, Karasavvas N, Gilbert P, S CDR, McElrath MJ, Gottardo R. 2015. COMPASS identifies T-cell subsets correlated with clinical outcomes. Nat Biotechnol 33:610-616.
18. Pissani F, Schulte B, Eller MA, Schultz BT, Ratto-Kim S, Marovich M, Thongcharoen P, Sriplienchan S, Rerks-Ngarm S, Pitisuttithum P, Esser S, Alter G, Robb ML, Kim JH, Michael NL, Streeck H. 2018. Modulation of Vaccine-Induced CD4 T Cell Functional Profiles by Changes in Components of HIV Vaccine Regimens in Humans. J Virol 92.
19. Schultz BT, Teigler JE, Pissani F, Oster AF, Kranias G, Alter G, Marovich M, Eller MA, Dittmer U, Robb ML, Kim JH, Michael NL, Bolton D, Streeck H. 2016. Circulating HIV-Specific Interleukin-21(+)CD4(+) T Cells Represent Peripheral Tfh Cells with Antigen- Dependent Helper Functions. Immunity 44: 167-178.
20. Matyas GR, Mayorov AV, Rice KC, Jacobson AE, Cheng K, Iyer MR, Li F, Beck Z, Janda KD, Alving CR. 2013. Liposomes containing monophosphoryl lipid A: a potent adjuvant system for inducing antibodies to heroin hapten analogs. Vaccine 31 :2804-2810.
21. Beck Z, Torres OB, Matyas GR, Lanar DE, Alving CR. 2018. Immune response to antigen adsorbed to aluminum hydroxide particles: Effects of co-adsorption of ALF or ALFQ adjuvant to the aluminum-antigen complex. J Control Release 275: 12-19.
22. Pollara J, Jones DI, Huffman T, Edwards RW, Dennis M, Li SH, Jha S, Goodman D, Kumar A, LaBranche CC, Montefiori DC, Fouda GG, Hope TJ, Tomaras GD, Staats HF, Ferrari G, Permar SR. 2019. Bridging Vaccine-Induced HIV-1 Neutralizing and Effector Antibody Responses in Rabbit and Rhesus Macaque Animal Models. J Virol 93.
23. Wen Y, Trinh HV, Linton CE, Tani C, Norais N, Martinez-Guzman D, Ramesh P, Sun Y, Situ F, Karaca-Griffm S, Hamlin C, Onkar S, Tian S, Hilt S, Malyala P, Lodaya R, Li N, Otten G, Palladino G, Friedrich K, Aggarwal Y, LaBranche C, Duffy R, Shen X, Tomaras GD, Montefiori DC, Fulp W, Gottardo R, Burke B, Ulmer JB, Zolla-Pazner S, Liao HX, Haynes BF, Michael NL, Kim JH, Rao M, O'Connell RJ, Carfi A, Barnett SW. 2018. Generation and characterization of a bivalent protein boost for future clinical trials: HIV-1 subtypes CR01 AE and B gpl20 antigens with a potent adjuvant. PLoS One 13:e0194266.
24. Qualls ZM, Choudhary A, Honnen W, Prattipati R, Robinson JE, Pinter A. 2018. Identification of Novel Structural Determinants in MW965 Env That Regulate the Neutralization Phenotype and Conformational Masking Potential of Primary HIV-1 Isolates. J Virol 92.
25. Asbach B, Kibler KV, Kostler J, Perdiguero B, Yates NL, Stanfield-Oakley S, Tomaras GD, Kao SF, Foulds KE, Roederer M, Seaman MS, Montefiori DC, Parks R, Ferrari G, Forthal DN, Phogat S, Tartaglia J, Barnett SW, Self SG, Gottardo R, Cristillo AD, Weiss DE, Galmin L, Ding S, Heeney JL, Esteban M, Jacobs BL, Pantaleo G, Wagner R. 2019. Priming with a Potent HIV-1 DNA Vaccine Frames the Quality of Immune Responses prior to a Poxvirus and Protein Boost. J Virol 93.
26. Forthal DN, Finzi A. 2018. Antibody-dependent cellular cytotoxicity in HIV infection. AIDS 32:2439-2451.
27. Worley MJ, Fei K, Lopez-Denman AJ, Kelleher AD, Kent SJ, Chung AW. 2018. Neutrophils mediate HIV-specific antibody-dependent phagocytosis and ADCC. J Immunol Methods 457:41-52.
28. Huang Y, Ferrari G, Alter G, Forthal DN, Kappes JC, Lewis GK, Love JC, Borate B, Harris L, Greene K, Gao H, Phan TB, Landucci G, Goods BA, Dowell KG, Cheng HD, Bailey -Kellogg C, Montefiori DC, Ackerman ME. 2016. Diversity of Antiviral IgG Effector Activities Observed in HIV-Infected and Vaccinated Subjects. J Immunol 197:4603-4612.
29. Crowley AR, Ackerman ME. 2019. Mind the Gap: How Interspecies Variability in IgG and Its Receptors May Complicate Comparisons of Human and Non-human Primate Effector Function. Front Immunol 10:697.
30. Havenar-Daughton C, Reiss SM, Camathan DG, Wu JE, Kendric K, Torrents de la Pena A, Kasturi SP, Dan JM, Bothwell M, Sanders RW, Pulendran B, Silvestri G, Crotty S. 2016. Cytokine-Independent Detection of Antigen-Specific Germinal Center T Follicular Helper Cells in Immunized Nonhuman Primates Using a Live Cell Activation-Induced Marker Technique. J Immunol 197:994-1002.
31. Stone SL, Peel JN, Scharer CD, Risley CA, Chisolm DA, Schultz MD, Yu B, Ballesteros-Tato A, Wojciechowski W, Mousseau B, Misra RS, Hanidu A, Jiang H, Qi Z, Boss JM, Randall TD, Brodeur SR, Goldrath AW, Weinmann AS, Rosenberg AF, Lund FE. 2019. T-bet Transcription Factor Promotes Antibody-Secreting Cell Differentiation by Limiting the Inflammatory Effects of IFN-gamma on B Cells. Immunity 50: 1172-1187 el 177.
32. Kumanogoh A, Shikina T, Suzuki K, Uematsu S, Yukawa K, Kashiwamura S, Tsutsui H, Yamamoto M, Takamatsu H, Ko-Mitamura EP, Takegahara N, Marukawa S, Ishida I, Morishita H, Prasad DV, Tamura M, Mizui M, Toyofuku T, Akira S, Takeda K, Okabe M, Kikutani H. 2005. Nonredundant roles of Sema4A in the immune system: defective T cell priming and Thl/Th2 regulation in Sema4A-deficient mice. Immunity 22:305-316.
33. Li G, Liang X, Lotze MT. 2013. HMGBl : The Central Cytokine for All Lymphoid Cells. Front Immunol 4:68.
34. Dinarello CA. 1999. IL-18: A TH1 -inducing, proinflammatory cytokine and new member of the IL-1 family. J Allergy Clin Immunol 103: 11-24.
35. Chin AI, Dempsey PW, Bruhn K, Miller JF, Xu Y, Cheng G. 2002. Involvement of receptor-interacting protein 2 in innate and adaptive immune responses. Nature 416: 190-194.
36. Kwissa M, Nakaya HI, Oluoch H, Pulendran B. 2012. Distinct TLR adjuvants differentially stimulate systemic and local innate immune responses in nonhuman primates. Blood 119:2044-2055.
37. Barbet G, Sander LE, Geswell M, Leonardi I, Cerutti A, Iliev I, Blander JM. 2018. Sensing Microbial Viability through Bacterial RNA Augments T Follicular Helper Cell and Antibody Responses. Immunity 48:584-598 e585.
38. Ben-Sasson SZ, Hu-Li J, Quiel J, Cauchetaux S, Ratner M, Shapira I, Dinarello CA, Paul WE. 2009. IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation. Proc Natl Acad Sci U S A 106:7119-7124.
39. Khoruts A, Osness RE, Jenkins MK. 2004. IL-1 acts on antigen-presenting cells to enhance the in vivo proliferation of antigen-stimulated naive CD4 T cells via a CD28- dependent mechanism that does not involve increased expression of CD28 ligands. Eur J Immunol 34: 1085-1090.
40. Bart PA, Huang Y, Karuna ST, Chappuis S, Gaillard J, Kochar N, Shen X, Allen MA, Ding S, Hural J, Liao HX, Haynes BF, Graham BS, Gilbert PB, McElrath MJ, Montefiori DC, Tomaras GD, Pantaleo G, Frahm N. 2014. HIV-specific humoral responses benefit from stronger prime in phase lb clinical trial. J Clin Invest 124:4843-4856.
41. Hu X, Valentin A, Dayton F, Kulkarni V, Alicea C, Rosati M, Chowdhury B, Gautam R, Broderick KE, Sardesai NY, Martin MA, Mullins JI, Pavlakis GN, Felber BK. 2016. DNA Prime-Boost Vaccine Regimen To Increase Breadth, Magnitude, and Cytotoxicity of the Cellular Immune Responses to Subdominant Gag Epitopes of Simian Immunodeficiency Virus and HIV. J Immunol 197:3999-4013.
42. Hu X, Valentin A, Rosati M, Manocheewa S, Alicea C, Chowdhury B, Bear J, Broderick KE, Sardesai NY, Gall SL, Mullins JI, Pavlakis GN, Felber BK. 2017. HIV Env conserved element DNA vaccine alters immunodominance in macaques. Hum Vaccin Immunother 13:2859-2871.
43. Ake JA, Schuetz A, Pegu P, Wieczorek L, Eller MA, Kibuuka H, Sawe F, Maboko L, Polonis V, Karasavva N, Weiner D, Sekiziyivu A, Kosgei J, Missanga M, Kroidl A, Mann
P, Ratto-Kim S, Anne Eller L, Earl P, Moss B, Dorsey-Spitz J, Milazzo M, Laissa Ouedraogo G, Rizvi F, Yan J, Khan AS, Peel S, Sardesai NY, Michael NL, Ngauy V, Marovich M, Robb ML. 2017. Safety and Immunogenicity of PENNVAX-G DNA Prime Administered by Biojector 2000 or CELLECTRA Electroporation Device With Modified Vaccinia Ankara- CMDR Boost. J Infect Dis 216: 1080-1090.
44. Kalams SA, Parker SD, Elizaga M, Metch B, Edupuganti S, Hural J, De Rosa S, Carter DK, Rybczyk K, Frank I, Fuchs J, Koblin B, Kim DH, Joseph P, Keefer MC, Baden LR, Eldridge J, Boyer J, Sherwat A, Cardinali M, Allen M, Pensiero M, Butler C, Khan AS, Yan J, Sardesai NY, Kublin JG, Weiner DB, Network NHVT. 2013. Safety and comparative immunogenicity of an HIV-1 DNA vaccine in combination with plasmid interleukin 12 and impact of intramuscular electroporation for delivery. J Infect Dis 208:818-829.
45. Vaccari M, Gordon SN, Fourati S, Schifanella L, Liyanage NP, Cameron M, Keele BF, Shen X, Tomaras GD, Billings E, Rao M, Chung AW, Dowell KG, Bailey-Kellogg C, Brown EP, Ackerman ME, Vargas-Inchaustegui DA, Whitney S, Doster MN, Binello N, Pegu P, Montefiori DC, Foulds K, Quinn DS, Donaldson M, Liang F, Lore K, Roederer M, Koup RA, McDermott A, Ma ZM, Miller CJ, Phan TB, Forthal DN, Blackburn M, Caccuri F, Bissa M, Ferrari G, Kalyanaraman V, Ferrari MG, Thompson D, Robert-Guroff M, Ratto- Kim S, Kim JH, Michael NL, Phogat S, Barnett SW, Tartaglia J, Venzon D, Stablein DM, et al. 2016. Adjuvant-dependent innate and adaptive immune signatures of risk of SIVmac251 acquisition. Nat Med 22:762-770.
46. Hill DL, Pierson W, Bolland DJ, Mkindi C, Carr EJ, Wang J, Houard S, Wingett SW, Audran R, Wallin EF, Jongo SA, Kamaka K, Zand M, Spertini F, Daubenberger C, Corcoran AE, Linterman MA. 2019. The adjuvant GLA-SE promotes human Tfh cell expansion and emergence of public TCRbeta clonotypes. J Exp Med doi: 10.1084/jem.20190301.
47. van Diepen MT, Chapman R, Moore PL, Margolin E, Hermanus T, Morris L, Ximba P, Rybicki EP, Williamson AL. 2018. The adjuvant AlhydroGel elicits higher antibody titres than AddaVax when combined with HIV-1 subtype C gpl40 from CAP256. PLoS One 13:e0208310.
48. Kang TH, Bae HC, Kim SH, Seo SH, Son SW, Choi EY, Seong SY, Kim TW. 2009. Modification of dendritic cells with interferon-gamma-inducible protein-10 gene to enhance vaccine potency. J Gene Med 11 :889-898.
49. Xu W, Joo H, Clayton S, Dullaers M, Herve MC, Blankenship D, De La Morena MT, Balderas R, Picard C, Casanova JL, Pascual V, Oh S, Banchereau J. 2012. Macrophages induce differentiation of plasma cells through CXCLIO/IP-IO. J Exp Med 209: 1813-1823, S1811-1812.
50. Singh S, Ramirez-Salazar EG, Doueiri R, Valentin A, Rosati M, Hu X, Keele BF, Shen X, Tomaras GD, Ferrari G, LaBranche C, Montefiori DC, Das J, Alter G, Trinh HV, Hamlin C, Rao M, Dayton F, Bear J, Chowdhury B, Alicea C, Lifson JD, Broderick KE, Sardesai NY, Sivananthan SJ, Fox CB, Reed SG, Venzon DJ, Hirsch VM, Pavlakis GN, Felber BK. 2018. Control of Heterologous Simian Immunodeficiency Virus SIVsmE660 Infection by DNA and Protein Coimmunization Regimens Combined with Different Toll- Like-Receptor-4 -Based Adjuvants in Macaques. J Virol 92.
51. Iyer SS, Latner DR, Zilliox MJ, McCausland M, Akondy RS, Penaloza-Macmaster P, Hale JS, Ye L, Mohammed AU, Yamaguchi T, Sakaguchi S, Amara RR, Ahmed R. 2013. Identification of novel markers for mouse CD4(+) T follicular helper cells. Eur J Immunol 43:3219-3232.
52. Woodworth JS, Christensen D, Cassidy JP, Agger EM, Mortensen R, Andersen P. 2019. Mucosal boosting of H56:CAF01 immunization promotes lung-localized T cells and an accelerated pulmonary response to Mycobacterium tuberculosis infection without enhancing vaccine protection. Mucosal Immunol doi: 10.1038/s41385-019-0145-5.
53. Mulligan MJ, Russell ND, Celum C, Kahn J, Noonan E, Montefiori DC, Ferrari G, Weinhold KJ, Smith JM, Amara RR, Robinson HL, Network NNDHVT. 2006. Excellent safety and tolerability of the human immunodeficiency virus type 1 pGA2/JS2 plasmid DNA priming vector vaccine in HIV type 1 uninfected adults. AIDS Res Hum Retroviruses 22:678- 683.
54. Styles TM, Gangadhara S, Reddy PBJ, Hicks S, LaBranche C C, Montefiori DC, Derdeyn CA, Kozlowski PA, Velu V, Amara RR. HIV C.1086 Envelope Gpl40 Protein Boosts Following DNA/MVA Vaccination 1 Fail to Enhance Heterologous Anti-VlV2 Antibody Response and Protection 2 Against Clade C SHIV Challenge.
55. Xu Y, Fernandez C, Alcantara S, Bailey M, De Rose R, Kelleher AD, Zaunders J, Kent SJ. 2013. Serial study of lymph node cell subsets using fine needle aspiration in pigtail macaques. J Immunol Methods 394:73-83.
56. Kozlowski PA, Lynch RM, Patterson RR, Cu-Uvin S, Flanigan TP, Neutra MR. 2000. Modified wick method using Week-Cel sponges for collection of human rectal secretions and analysis of mucosal HIV antibody. J Acquir Immune Defic Syndr 24:297-309.
57. Pegu P, Vaccari M, Gordon S, Keele BF, Doster M, Guan Y, Ferrari G, Pal R, Ferrari MG, Whitney S, Hudacik L, Billings E, Rao M, Montefiori D, Tomaras G, Alam SM, Fenizia C, Lifson JD, Stablein D, Tartaglia J, Michael N, Kim J, Venzon D, Franchini G. 2013. Antibodies with high avidity to the gpl20 envelope protein in protection from simian immunodeficiency virus SIV(mac251) acquisition in an immunization regimen that mimics the RV-144 Thai trial. J Virol 87: 1708-1719.
58. Iyer SS, Gangadhara S, Victor B, Shen X, Chen X, Nabi R, Kasturi SP, Sabula MJ, Labranche CC, Reddy PB, Tomaras GD, Montefiori DC, Moss B, Spearman P, Pulendran B, Kozlowski PA, Amara RR. 2016. Virus-Like Particles Displaying Trimeric Simian Immunodeficiency Virus (SIV) Envelope gpl60 Enhance the Breadth of DNA/Modified Vaccinia Virus Ankara SIV Vaccine-Induced Antibody Responses in Rhesus Macaques. J Virol 90:8842-8854.
59. Alpert MD, Heyer LN, Williams DE, Harvey JD, Greenough T, Allhorn M, Evans DT. 2012. A novel assay for antibody-dependent cell-mediated cytotoxicity against HIV-1- or SIV-infected cells reveals incomplete overlap with antibodies measured by neutralization and binding assays. J Virol 86: 12039-12052.
60. von Bredow B, Arias JF, Heyer LN, Gardner MR, Farzan M, Rakasz EG, Evans DT. 2015. Envelope Glycoprotein Internalization Protects Human and Simian Immunodeficiency Virus-Infected Cells from Antibody-Dependent Cell-Mediated Cytotoxicity. J Virol 89: 10648-10655.
61. Phillips B, Fouda GG, Eudailey J, Pollara J, Curtis AD, 2nd, Kunz E, Dennis M, Shen X, Bay C, Hudgens M, Pickup D, Alam SM, Ardeshir A, Kozlowski PA, Van Rompay KKA, Ferrari G, Moody MA, Permar S, De Paris K. 2017. Impact of Poxvirus Vector Priming, Protein Coadministration, and Vaccine Intervals on HIV gpl20 Vaccine-Elicited Antibody Magnitude and Function in Infant Macaques. Clin Vaccine Immunol 24.
62. Petitdemange C, Kasturi SP, Kozlowski PA, Nabi R, Quamstrom CF, Reddy PBJ, Derdeyn CA, Spicer LM, Patel P, Legere T, Koval enkov YO, Labranche CC, Villinger F, Tomai M, Vasilakos J, Haynes B, Kang CY, Gibbs JS, Yewdell JW, Barouch D, Wrammert
J, Montefiori D, Hunter E, Amara RR, Masopust D, Pulendran B. 2019. Vaccine induction of antibodies and tissue-resident CD8+ T cells enhances protection against mucosal SHIV- infection in young macaques. JCI Insight 4.
63. Manrique M, Kozlowski PA, Cobo-Molinos A, Wang SW, Wilson RL, Montefiori DC, Mansfield KG, Carville A, Aldovini A. 2011. Long-term control of simian immunodeficiency virus mac251 viremia to undetectable levels in half of infected female rhesus macaques nasally vaccinated with simian immunodeficiency virus DNA/recombinant modified vaccinia virus Ankara. J Immunol 186:3581-3593.
Example 2. Targeting CD4 Tfh Cells to Enhance HIV Vaccine-Induced Humoral Immunity
Summary
[0173] With over 2.5 million new HIV infections per year, the majority in resource-poor countries with limited access to antiretroviral therapy, an effective HIV vaccine continues to remain among the most promising and safe strategies for preventing infection and reducing the burden of AIDS. While designing an effective HIV vaccine is a daunting goal, the modest efficacy of the RV144 Thai trial indicates that it is possible to prevent HIV infection in humans by vaccination, and that long-lasting antibody contributes significantly for this protection. Because vaccine-elicited CD4 T follicular helper cells (Tfh) are essential for establishing long-lived antibody, harnessing Tfh help is vital for an effective HIV vaccine. This Example focuses on achieving this goal using a recombinant DNA-prime, modified vaccinia Ankara (MVA)-boost vaccine platform in path for clinical development. Our data show that generation of Tfh cells expressing the chemokine receptor CXCR3 correlates strongly with multiple attributes of protective humoral immunity. Furthermore, CXCR3+ Tfh cells express higher levels of B cell helper factors relative to CXCR3- Tfh cells. Without being bound by the following theory, it is believed that HIV vaccines designed to induce high magnitude CXCR3+ Tfh cells increases antibody persistence, enhancing protection against a mucosal HIV challenge.
Approach
[0174] Long-lived plasma cells are primarily responsible for antibody durability, and the survival and differentiation of plasma cells within germinal centers (GC) depends on optimal
help from CD4 T follicular helper cells (Tfh). Therefore, it is imperative to design vaccines that can harness optimal Tfh cell help to establish durable anti-HIV antibodies.
[0175] Akin to CD4 T helper (h) cells, Tfh cells also encompass distinct Thl or Th2 subsets, and several recent studies have demonstrated that Th polarity of Tfh cells critically determines outcome of antibody responses to various vaccines/infections. Yet, it remains unclear whether Thl or Th2-type Tfh cells regulate HIV vaccine induced antibody. Bridging this gap will greatly advance vector-based vaccine design strategies and rational selection of adjuvants for protein boosts to extend the functional quality and durability of the vaccine- elicited antibody against HIV. Our recent studies using a DNA(D)-prime/modified vaccinia Ankara(MVA)+Alum-adjuvanted protein (Pro)- boost regimen showed that generation of CXCR3+ Thl-polarized Tfh cells within GCs associates with Env antibody durability, neutralization potential and avidity (Iyer SS, Amara R et ah, JI, 2015). Furthermore, CXCR3+ Tfh cells express higher levels of B cell helper co-stimulatory molecules and cytokines relative to CXCR3- Tfh cells.
[0176] We use the well-established DNA/MVA vaccine platform, which is in clinical development, for two integrated approaches; first, we employ the Thl chemokine, interferon induced protein (IP)-10, a ligand for and an inducer of CXCR3, as a molecular adjuvant to a DNA vaccine (DIP-10) to prime Thl-type Tfh cells. Second, we employ AS01B as a strong Thl adjuvant to the gpl40 protein (ProASOlB) to boost Thl-type Tfh cells. This vaccine regimen is evaluated in a monkey study with three vaccine groups to determine whether increase in Thl-polarized Tfh cells during the prime with a DIP- lOMPro Alum vaccine and during both the prime and the boost with a DIP-lOMProASOlB vaccine enhances durable and protective HIV antibody relative to a reference DMProAlum platform.
[0177] The first approach is to delineate immune determinants and characterize gene expression profiles of vaccine-specific Thl versus Th2-polarized Tfh cells. Specifically, a novel IP-10-DNA-SHIV vaccine is constructed and characterized ex vivo. In addition, cytokine/chemokine determinants are delineated, and single-cell transcriptional profiling is performed of Env-specific Thl and Th2 Tfh cells in all three outlined vaccine groups.
[0178] The second approach is to determine whether Thl-polarized Tfh cells enhance magnitude, quality and persistence of Env antibody titers. Specifically, it is determined whether the DIP- lOMPro Alum and DIP-lOMProASOlB vaccines increase Env binding titers in sera and mucosa and enhance neutralizing and non-neutralizing antibody titers.
[0179] The third approach is to determine whether a Thl-polarized vaccine regimen enhances mucosal protection against a SHIV challenge and augments viral control. Specifically, it is determined whether the DIP-lOMProASOlB vaccine enhances protection against a vaginal heterologous SHIV challenge and potentiates viral control in infected animals. In addition, the susceptibility of CXCR3+ CD4 Tfh cells to SHIV infection is determined ex vivo.
Introduction
[0180] The advent of highly effective antiretroviral therapy (ART) and its use in pre exposure prophylaxis (PrEP) has resulted in major strides in HIV/AIDS prevention (1). However, this has not diminished the need for a protective vaccine regimen, which, unlike PrEP, does not require conscious effort, continued adherence or frequent follow-up to be effective (2, 3). Therefore, development of an effective HIV/AIDS vaccine continues to be the most sustainable and cost-effective approach and a top priority for HIV/AIDS prevention. To date, there have been six HIV vaccine efficacy trials that have tested four different vaccine modalities. The first indication of efficacy was observed in the RV144 Thai trial, which tested a poxvirus vector prime (ALVAC, months 0,1) and ALVAC+gpl20 protein boost (months 3, 6) and demonstrated an estimated vaccine efficacy of 31% (4). Protection was linked with durability of anti-HIV Envelope (Env) antibody titers (5) emphasizing the need to enhance antibody persistence by induction of long-lived plasma cells (6, 7). Plasma cells are the product of germinal center (GC) reactions within lymphoid tissues, and the survival and differentiation of GC B cells to plasma cells critically depends on help from CD4 T follicular helper cells (Tfh) (8, 9). Therefore, it is imperative to design vaccines to harness optimal Tfh cell help to establish durable humoral immunity (10).
[0181] Similar to canonical CD4 T cells (11), Tfh cells also encompass characteristics of Thl, Th2, and Thl7-type cell subsets which can be identified by expression of specific chemokine receptors (12). Several recent studies have demonstrated that Thl and Th2 Tfh cells possess differential capacity for B cell help, and the induction of a specific Tfh subset critically determines outcome of the antibody response to vaccination/infection (13-15).
[0182] With respect to vaccination, Thl-polarized CXCR3 -expressing Tfh cells have been associated with durable antibody titers following influenza immunization (13, 16). In our HIV vaccine preclinical study, we recently showed that the magnitude of Thl-polarized CXCR3+ Tfh responses correlated with Env antibody durability, neutralization potential and
avidity following a modified vaccinia Ankara (MV A) boost (17). However, in a recent study in HIV+ individuals, CXCR3- Tfh cells were found to be associated with neutralizing titers (14) implying divergent functions of Thl-Tfh cells in vaccination versus chronic infection. These reports underscore a pressing need for unequivocally determining whether Thl or Th2 type Tfh cells regulate durable and protective antibody to viral vector and/ subunit protein HIV immunizations. Furthermore, the surge of interest in employing Thl adjuvants such as MF59 and AS01B over the Th2 adjuvant Alum in Phase I/II HIV vaccine trials makes this a highly timely and clinically relevant question in HIV/AIDS vaccine research (18-20).
Study Design
[0183] As shown in FIG. 13, all animals receive two DNA (D) primes and two MVA+gpl40 protein boosts (MPro). Vaccine groups 1, 2, and 3 are used to investigate the outcome of graded increase in Thl immune response on antibody durability in systemic and mucosal compartments. Vaccine groups 1 and 3, at the opposite ends of the Thl response spectrum, are directly compared for efficacy against an intravaginal heterologous Clade C SHIV.
Data
[0184] DNA/MVA vaccine induces robust anti-Env antibody. We have demonstrated that intramuscular immunization with DNA/MVA vaccines elicits immunologically coordinated humoral immune responses in periphery and mucosa (27, 28). Determination of Env-specific binding titers by ELISA over the course of immunization shows a temporal increase in Env titers, which achieve robust peak after the 2nd MVA boost (FIG. 14A).
[0185] Our data combining Pro+Alum with the 2nd MVA (FIG. 14B) demonstrate a modest but significant increase in peak titers in sera (FIG. 14C) and rectal mucosa (FIG. 14D). We also noted increase in heterologous tier 1 -neutralization titers in sera (FIG. 14E), and a trend for increase in antibody against cyclic VI V2 peptides in mucosa (FIG. 14F). These data demonstrate the feasibility of combining MVA with subunit protein immunogens to enhance humoral responses. Preliminary data from the HIV Vaccine Trials Network (HVTN) demonstrating high magnitude, durable anti-VlV2 antibodies in humans with AS01B relative to Alum as an adjuvant provide a strong rationale for utilizing AS01B to further enhance the magnitude and persistence of DNA/MVA+protein elicited humoral immunity.
[0186] DNA/MVA vaccine elicits Tfh responses, which are discernable in peripheral blood. Our data demonstrate a transient accumulation of CXCR5+ Tfh cells expressing the cell- cycle marker Ki-67 at the peak CD4 effector response following the MVA boost (FIGS. 15A and B, *p < 0.05). Analogous to functionality of Gag and Env-specific CD4 T cells co expressing the Tfh cytokine interleukin (IL) 21 and the Thl cytokine IFNG (FIG. 15C); Ki- 67+ pTfh expressed CXCR3, a marker of Thl polarity (FIG. 15D). Notably, the proportion of CXCR3+Ki-67+ pTfh cells markedly increased following MVA immunization relative to baseline (FIG. 15E, ***p< 0.001). These data demonstrate that DNA/MVA vaccination elicits Tfh cells of distinct Th polarity and establishes our ability to sensitively capture this response in peripheral blood (17).
[0187] Lymph node Tfti responses following DNA/MVA+Protein immunization. To capture salient aspects of the GC response following the 2nd MVA boost and how Pro+Alum modulated this response, we examined GC Tfh cells by high expression of programmed death (PD)1 and CXCR5, and Tfh cells based on the CXCR5+PD-1+ phenotype (FIG. 16A). Our data showed that M+Pro vaccine increased Thl -polarized Tfh cells (FIG. 16B) even in the presence of Alum.
[0188] Strikingly, the proportion of CXCR3+ GC Tfh correlated with frequency of GC B cells (FIG. 16C), antibody avidity (FIG. 16D), durability (FIG. 16E), and neutralization (FIG. 16F). These data provide a compelling incentive for determining whether a strong Thl DNA prime and a strong Thl MVA boost will enhance durable and protective antibody.
[0189] IP-10 increases induction of CXCR3+ Tfh cells. To confirm that utilizing IP-10 as a molecular adjuvant to a DNA vaccine induces Thl-type Tfh cells, we examined whether exogenous IP- 10 induces CXCR3 expression on Tfh cells ex vivo. Rhesus peripheral blood mononuclear cells (PBMC) were stimulated using CD3/CD28 beads in the absence or presence of increasing doses of IP- 10 for 48h. We utilized a dose of IP- 10 based on the range of serum concentrations reported following MVA immunization in macaques (29). Our data show that IP- 10 induces a distinct increase in proportion of Tfh cells expressing CXCR3 (FIGS. 17A and B) and importantly increases frequency of CXCR3+ Tfh cells (FIG. 17C).
Approaches
First approach
[0190] In order to effectively harness Tfh help for HIV vaccine design, understanding heterogeneity of the vaccine-elicited Tfh response and identifying factors contributing to this heterogeneity are critical. The goal of the first approach is to extensively define the cytokine/chemokine response to vaccination and how this influences Tfh cell magnitude and polarity. This approach is further designed to acquire comprehensive insights into division of labor within vaccine-elicited Tfh cells by interrogating the transcriptional signature of Env- specific CXCR3+ Thl and CCR4+ Th2 Tfh cells.
[0191] We utilize a DNA-SHIV construct expressing 1086 Clade C Env, tat and rev; and SIV239 Gag, protease, and reverse transcriptase using a CMV promoter. As previously described, the DNA vaccines express multiple HIV/SIV proteins from a single RNA by subgenomic splicing and frameshifting (28). We construct IP-10-DNA-SHIV vaccine by inserting the rhesus macaque IP- 10 sequence (297 base pairs) as a fusion to the (a) internal ribosome entry site (IRES) or (b) the peptide from foot and mouth disease (F2A) downstream of Env. Our rationale for testing two constructs ex vivo is to be able to select an optimal construct for in vivo vaccine experiments.
[0192] Based on ongoing studies using a CD40L-DNA-SHIV construct, we have observed robust expression of CD40L on 293-T transfected cells with the 2A DNA vaccine. Higher vaccine specific responses with the DNA-SHIV CD40L vaccine provide evidence for enhanced immunogenicity in vivo. These data provide an important proof-of-principle that the IP- 10 DNA vaccine can be designed and that enhanced immunogenicity can be quantified in vivo. We compare the IP-10-DNA-SHIV IRES and F2A constructs ex vivo using a variety of different approaches which include determining antigenicity of constructs by evaluating expression of HIV-1 Env (surface, clone PGT121), SIV Gag (intracellular, clone 2F12) and IP-10 (intracellular, clone J034D6) by flow cytometry on transfected 293-T cells. Cell supernatants from 293-T transfected cells are also be collected for measurement of secreted IP- 10 by ELISA and for functional activity of IP- 10 using cell supernatants to stimulate PBMCs. We compare supernatants for ability to activate DCs and induce CXCR3 expression on CD4 T cells. We select either the IRES or the 2A construct based on efficiency of Env and IP- 10 expression for in vivo experiments.
[0193] Another objective is to understand how the in vivo chemokine/cytokine response immediately following immunization influences CD4 Tfh development and differentiation, which is directed by cues early in the immune response (36). Studies in a mouse model of
acute viral infection show that IP-10 is rapidly induced in a type I IFN dependent manner (37), and studies in macaques show transient induction of pro-inflammatory factors including IP- 10 early after MVA immunization (29). Therefore, to optimally capture the early and transient induction of immune factors, we collect blood at 0, 12, 24, 48, and 76 hours post immunization to establish a comprehensive cytokine /chemokine profile temporally, and across vaccine platforms. In addition, we collect CVL at 0 and 24 hours after each immunization to evaluate the immune milieu in the genital mucosa. We determine (i) cytokine, chemokine profile using a multiplex assay to measure soluble factors driving Thl polarization: interleukins (IL)-12, IL-Ib, IL-8, IL-2; macrophage inflammatory protein (MIP)la, MIRIb, MIP3a, MIR3b; macrophage chemotactic protein (MCP)-l; CXCR3 ligands; monokine induced by gamma (MIG/CXCL9), PMO/CXCLIO, interferon (IFN)- inducible T-cell alpha chemoattractant (I-TAC/CXCL11); tumor necrosis factor (TNF)a; IFNs PTMa,b,g. We also examine a panel of Th2/regulatory factors including IL-4, IL-10, IL- Ra, transforming growth factor (TGF)a, TORb and additional CCR4 ligands including CCL5, CCL17, and CCL22.
[0194] To determine how the innate response influences (ii) vaccine-specific Tfh responses, we quantify vaccine-elicited Tfh cells in systemic, lymphoid, and mucosal compartments. We have demonstrated that the DNA/MVA vaccine induces robust frequencies of vaccine-specific CD4 T cells that are broadly distributed systemically (17, 27), within the spleen and lymph nodes, and in the rectal and genital mucosa. Due to evidence for CXCR3 in directing homing of T cells to the vaginal mucosa (38), we evaluate T cell responses in the genital tract. We examine blood and lymph node Tfh cells for a4b7 expression to understand how potentiating Thl responses impacts trafficking to the gut associated lymphoid tissue (39).
[0195] We study vaccine-specific Tfh cells utilizing two complementary approaches we have optimized and established (17). We utilize the cell-cycle marker Ki-67 to interrogate CXCR5+ CD4 T cells at peak effector time points following each immunization for expression of CXCR3 and CCR4 as outlined in FIG. 19A. This is complemented by intracellular cytokine staining (ICS)-based assays designed to examine vaccine-specific responses (at peak and memory) after stimulation with relevant Gag and Env peptide pools. After stimulation, Ag-specific Tfh cells are identified based on cytokine positivity and CXCR5 expression utilizing the panel as shown in FIGS. 19B and 19C. We evaluate GC
responses using flow cytometry and examine GC Tfh, Tfh and GC B cells for CXCR3 and CCR4 expression (similar to analysis shown above).
[0196] To gain a comprehensive understanding of molecular heterogeneity within vaccine elicited Tfh cells, we determine transcription profile of CXCR3+ vs. CCR4+ Tfh cells within the draining lymph node. Because robust antibody titers are established after the 2nd MVA boost, and active germinal centers are observed at 14 days post 2nd MVA (27), we collect biopsies from the draining lymph node at this time. Vaccine-specific Tfh cells are identified by translocation of CD40L after 5 hour stimulation with Env peptides in the presence of co stimulatory molecules (FIG. 20) (40). Samples from 5 animals within each of three vaccine groups are selected to understand heterogeneity within Tfh cell subsets and how it compares across vaccine regimens. Because changes in CXCR5, CXCR3, and PD-1 likely occur upon stimulation, CD3+CD4+Fas+CXCR5+PD-1+/++ (comprising Tfh and GC Tfh) cells are sorted using fluorescence-assisted cell sorting (FACS) into CXCR3+ and CCR4+ single positive subsets prior to stimulation. We also sort CXCR3- CCR4- double negative Tfh cells. Naive CD4 T cells (CD4+Fas- CXCR5-PD-1-CXCR3- ) serve as a reference population. After stimulation with Env peptides in the presence of co-stimulatory molecules using established protocols (17), cells are stained with surface CD40L and twenty-four CD40L+ cells from each of the two subsets are singly sorted into a 96-well plate. The single-cell RNA sequencing methodology allows us to capture the heterogeneity of the Tfh cell response to stimulation and give us the ability to resolve important associations between cell phenotype, stimulation status (by quantifying genes encoding Tbet (Thl) and IL-4 (Th2) that are rapidly up regulated upon TCR stimulation (41)), and cellular transcriptional program.
Second approach
[0197] Several recent studies have demonstrated functional heterogeneity within Tfh cells based on Th polarization, which is intertwined with chemokine receptor expression (12-15). Our data provide two strong pieces of evidence supporting a role of Thl-polarized Tfh cells in enhancing antibody durability. First, the frequency of Env-specific IFNG+IL-21+ cells at 1 week post 2nd MVA, and second, the frequency of CXCR3+ GC Tfh cells at 2 weeks post 2nd MVA correlate with antibody durability measured at 20 weeks post 2ndMVA (FIG. 20). In the second approach, we conduct a systematic and comprehensive assessment of antibody responses across three vaccine regimens designed to elicit graded increase in Thl-polarized Tfh cells.
[0198] We perform a broad repertoire of quantitative and functional assays to comprehensively assess the humoral response in the systemic and mucosal compartments. We perform Enzyme-linked immunosorbent spot (ELISPOT) assays using standard protocols (43, 44) to measure Env-specific antibody secreting cells (ASCs) using fresh PBMCs, at day 5 following 1st and 2nd MVA boost. We have established kinetics of gpl40-ASCs in our studies (17) (FIG. 22A). We quantitate plasma cells using bone marrow aspirates at memory post 2nd MVA. We measure Env-specific IgG and IgA binding titers and antibody IgG subclass (45) against gpl40 and ConAcaptured virus like particle trimeric gpl60 antigen by ELISA using established protocols (17, 27). We utilize the highly sensitive binding antibody multiplex assay (BAMA) to quantify Env responses against gpl20, gp41, V1V2, and V3 antigens in the mucosa and sera. We measure neutralization responses against tier 1 and tier 2 autologous and heterologous viruses using the TZM-bl assay after the 2nd MVA boost (46). Initial assays are done with the challenge SHIV, the immunogen SHIV, and clade-matched tier 1A, tier IB and tier 2 viruses, which include SHIV-1157ipEL-p and Cel086_B2. Further assays are performed against a multi-clade global virus panel described previously (47). To assess non-neutralizing antibody effector functions, we measure antibody-dependent phagocytosis (ADP) using the Ackerman flow cytometric assay (48) and antibody dependent cellular cytotoxicity (ADCC) using the Evans assay (49). Our data (FIG. 22B) show induction of antibodies that direct ADCC and ADP following a DNA/MVA immunization. We also measure antibody glycosylation (23).
Third approach
[0199] A significant impediment to enhancing vaccine efficacy is the waning of Env antibody titers following vaccination (5, 50). While booster protein immunizations are a means to augment antibody titers, they present the concern of inefficient CD8 T cell recall and potential generation of target cells, which together can create an environment that favors initial viral replication upon exposure (51). Consequently, vaccine strategies to establish durable humoral immunity while inducing robust cytolytic responses are highly desirable.
[0200] To determine vaccine efficacy, we determine protection against weekly intravaginal repeat low-dose challenge with heterologous clade C SHIV-1157ipd3N4 (35) for a maximum of 12 weeks (12 challenges). We utilize the stock titrated to infect about 30% of the unvaccinated animals at the first challenge. We determine whether DIP-lOMProASOlB
vaccine delayed rate of acquisition and decreased peak viremia in infected animals relative to DMProAlum.
[0201] Statistical Analyses. To arrive at sample sizes, we conducted power analyses using per-challenge infection rates computed from our prior trial using the SIV251 challenge virus. Our power analyses determined that we should achieve a minimum per-challenge infection rate of about 0.02 in group 3 to see a statistically significant (80% power at the alpha level of 0.05) difference between groups 1 and 3. We evaluate two main parameters to determine protection: 1) Status of Infection and 2) number of challenges to infection. Comparison of proportions of infected and uninfected animals between vaccine groups and controls is performed using Chi-square test. Comparisons of number of challenges until infection between groups is conducted using the two-sided log-rank test and visualized using Kaplan- Meier curves. Models of the protective effect of vaccination proposed by Hudgens and Gillbert are fitted, and the Akaike Information Criteria (AIC) is used to select the most parsimonious model (54). In addition, we determine peak viral load at 2 and 3 weeks post infection to determine whether DIP-lOMProASOlB vaccine enhanced viral control relative to DMProAlum vaccine.
[0202] The goal of this approach is to understand whether vaccine approaches that induce Thl polarized CD4 responses generate HIV target cells. Our data demonstrate enrichment of CCR5+ cells within the CXCR3+ Tfh cell subset (FIG. 23A). Furthermore, our data show significantly higher levels of pro-viral DNA in X3+ Tfh cells (FIG. 23B) suggestive of enhanced permissivity to infection. Therefore, we determine the frequency of CXCR3+ CD4 T cells in the context of CCR5 and CXCR5 in the vaginal mucosa, lymph node and blood prior to challenge and determine if this correlates with either acquisition or peak viremia. As an additional functional assay to quantitate vaccine-induced increase in susceptibility to infection (55), archived cells from vaginal biopsies at baseline, weeks 1 and 8 post 2nd MVA booster immunization are exposed to the SHIV challenge virus ex vivo for 18 hours; washed and cultured for 48 hours. We quantitate virus in the supernatant and infected cells are assessed by intracellular Gag staining. This assay yields information on whether vaccine- induced changes in the mucosal immune environment favor viral replication.
Bibliography and references cited
1. Dieffenbach CW, Fauci AS. Thirty years of HIV and AIDS: future challenges and opportunities. Annals of internal medicine. 2011 ; 154(11):766-71. Epub 2011/06/02. doi: 10.7326/0003-4819-154-11-201106070-00345. PubMed PMID: 21628350.
2. Baeten JM, Haberer JE, Liu AY, Sista N. Preexposure prophylaxis for HIV prevention: where have we been and where are we going? J Acquir Immune Defic Syndr. 2013;63 Suppl 2:S122-9. Epub 2013/06/21. doi: 10.1097/QAI.0b013e3182986f69. PubMed PMID: 23764623; PubMed Central PMCID: PMC3710117.
3. Fauci AS, Folkers GK, Marston HD. Ending the global HIV/AIDS pandemic: the critical role of an HIV vaccine. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2014;59 Suppl 2:S80-4. Epub 2014/08/26. doi: 10.1093/cid/ciu420. PubMed PMID: 25151483; PubMed Central PMCID: PMC4157695.
4. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Kamasuta C, Sutthent R, Liao HX, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. The New England journal of medicine. 2012;366(14): 1275-86. Epub 2012/04/06. doi: 10.1056/NEJMoal 113425. PubMed PMID: 22475592; PubMed Central PMCID: PMC3371689.
5. Corey L, Gilbert PB, Tomaras GD, Haynes BF, Pantaleo G, Fauci AS. Immune correlates of vaccine protection against HIV-1 acquisition. Science translational medicine. 015;7(310):310rv7. Epub 2015/10/23. doi: 10.1126/scitranslmed.aac7732. PubMed PMID: 26491081.
6. Kim JH, Excler JL, Michael NL. Lessons from the RV144 Thai phase III HIV-1 vaccine trial and the search for correlates of protection. Annual review of medicine. 2015;66:423-37. Epub 2014/10/24. doi: 10.1146/annurev-med-052912-123749. PubMed PMID: 25341006.
7. Slifka MK, Matloubian M, Ahmed R. Bone marrow is a major site of long-term antibody production after acute viral infection. Journal of virology. 1995;69(3): 1895-902. Epub 1995/03/01. PubMed PMID: 7853531; PubMed Central PMCID: PMC188803.
8. Cyster JG. Shining a light on germinal center B cells. Cell. 2010;143(4):503-5. Epub 2010/11/16. doi: 10.1016/j cell.2010.10.036. PubMed PMID: 21074042.
9. Crotty S. Follicular helper CD4 T cells (TFH). Annual review of immunology. 2011;29:621-63. Epub 2011/02/15. doi: 10.1146/annurev-immunol-031210-101400. PubMed PMID: 21314428.
10. Streeck H, D'Souza MP, Littman DR, Crotty S. Harnessing CD4(+) T cell responses in HIV vaccine development. Nature medicine. 2013; 19(2): 143-9. Epub 2013/02/08. doi: 10.1038/nm.3054. PubMed PMID: 23389614; PubMed Central PMCID: PMC3626561.
11. Swain SL. CD4 T cell development and cytokine polarization: an overview. Journal of leukocyte biology. 1995;57(5):795-8. Epub 1995/05/01. PubMed PMID: 7759960.
12. Morita R, Schmitt N, Bentebibel SE, Ranganathan R, Bourdery L, Zurawski G, Foucat E, Dullaers M, Oh S, Sabzghabaei N, Lavecchio EM, Punaro M, Pascual V, Banchereau J, Ueno H. Human blood CXCR5(+)CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity. 2011;34(1): 108-21. Epub 2011/01/11. doi: 10.1016/j.immuni.2010.12.012. PubMed PMID: 21215658; PubMed Central PMCID: PMC3046815.
13. Bentebibel SE, Lopez S, Obermoser G, Schmitt N, Mueller C, Harrod C, Flano E, Mejias A, Albrecht RA, Blankenship D, Xu H, Pascual V, Banchereau J, Garcia-Sastre A, Palucka AK, Ramilo O, Ueno H. Induction of ICOS+CXCR3+CXCR5+ TH cells correlates with antibody responses to influenza vaccination. Science translational medicine. 2013;5(176): 176ra32. Epub 2013/03/15. doi: 10.1126/scitranslmed.3005191. PubMed PMID: 23486778; PubMed Central PMCID: PMC3621097.
14. Locci M, Havenar-Daughton C, Landais E, Wu J, Kroenke MA, Arlehamn CL, Su LF, Cubas R, Davis MM, Sette A, Haddad EK, International AVIPCPI, Poignard P, Crotty S. Human circulating PD-1+CXCR3-CXCR5+ memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity. 2013;39(4):758-69. Epub 2013/09/17. doi: 10.1016/j.immuni.2013.08.031. PubMed PMID: 24035365; PubMed Central PMCID: PMC3996844.
15. Obeng-Adjei N, Portugal S, Tran TM, Yazew TB, Skinner J, Li S, Jain A, Feigner PL, Doumbo OK, Kayentao K, Ongoiba A, Traore B, Crompton PD. Circulating Thl-Cell- type Tfh Cells that Exhibit Impaired B Cell Help Are Preferentially Activated during Acute
Malaria in Children. Cell reports. 2015;13(2):425-39. Epub 2015/10/07. doi: 10.1016/j.celrep.2015.09.004. PubMed PMID: 26440897; PubMed Central PMCID: PMC4607674.
16. Obermoser G, Presnell S, Domico K, Xu H, Wang Y, Anguiano E, Thompson- Snipes L, Ranganathan R, Zeitner B, Bjork A, Anderson D, Speake C, Ruchaud E, Skinner J, Alsina L, Sharma M, Dutartre H, Cepika A, Israelsson E, Nguyen P, Nguyen QA, Harrod AC, Zurawski SM, Pascual V, Ueno H, Nepom GT, Quinn C, Blankenship D, Palucka K, Banchereau J, Chaussabel D. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity. 2013;38(4):831-44. Epub 2013/04/23. doi: 10.1016/j.immuni.2012.12.008. PubMed PMID: 23601689; PubMed Central PMCID: PMC3681204.
17. Iyer SS, Gangadhara S, Victor B, Gomez R, Basu R, Hong JJ, Labranche C, Montefiori DC, Villinger F, Moss B, Amara RR. Codelivery of Envelope Protein in Alum with MVA Vaccine Induces CXCR3-Biased CXCR5+ and CXCR5- CD4 T Cell Responses in Rhesus Macaques. Journal of immunology. 2015;195(3):994-1005. Epub 2015/06/28. doi: 10.4049/jimmunol.1500083. PubMed PMID: 26116502; PubMed Central PMCID: PMC4506863.
18. Alving CR, Peachman KK, Rao M, Reed SG. Adjuvants for human vaccines. Current opinion in immunology. 2012;24(3):310-5. Epub 2012/04/24. doi: 10.1016/j.coi.2012.03.008. PubMed PMID: 22521140; PubMed Central PMCID: PMC3383374.
19. Omosa-Manyonyi G, Mpendo J, Ruzagira E, Kilembe W, Chomba E, Roman F, Bourguignon P, Koutsoukos M, Collard A, Voss G, Laufer D, Stevens G, Hayes P, Clark L, Cormier E, Dally L, Barin B, Ackland J, Syvertsen K, Zachariah D, Anas K, Sayeed E, Lombardo A, Gilmour J, Cox J, Fast P, Priddy F. A Phase I Double Blind, Placebo- Controlled, Randomized Study of the Safety and Immunogenicity of an Adjuvanted HIV-1 Gag-Pol-Nef Fusion Protein and Adenovirus 35 Gag-RT-Int-Nef Vaccine in Healthy HIV Uninfected African Adults. PloS one. 2015;10(5):e0125954. Epub 2015/05/12. doi: 10.1371/journal. pone.0125954. PubMed PMID: 25961283; PubMed Central PMCID: PMC4427332.
20. Leroux-Roels G, Bourguignon P, Willekens J, Janssens M, Clement F, Didierlaurent AM, Fissette L, Roman F, Boutriau D. Immunogenicity and safety of a booster
dose of an investigational adjuvanted polyprotein HIV-1 vaccine in healthy adults and effect of administration of chloroquine. Clinical and vaccine immunology : CVI. 2014;21(3):302- 11. Epub 2014/01/07. doi: 10.1128/CVI.00617-13. PubMed PMID: 24391139; PubMed Central PMCID: PMC3957681.
21. Vandepapeliere P, Horsmans Y, Moris P, Van Mechelen M, Janssens M, Koutsoukos M, Van Belle P, Clement F, Hanon E, Wettendorff M, Garcon N, Leroux-Roels G. Vaccine adjuvant systems containing monophosphoryl lipid A and QS21 induce strong and persistent humoral and T cell responses against hepatitis B surface antigen in healthy adult volunteers. Vaccine. 2008;26(10): 1375-86. Epub 2008/02/15. doi: 10.1016/j.vaccine.2007.12.038. PubMed PMID: 18272264.
22. Lalvani A, Moris P, Voss G, Pathan AA, Kester KE, Brookes R, Lee E, Koutsoukos M, Plebanski M, Delchambre M, Flanagan KL, Carton C, Slaoui M, Van Hoecke C, Ballou WR, Hill AV, Cohen J. Potent induction of focused Thl-type cellular and humoral immune responses by RTS,S/SBAS2, a recombinant Plasmodium falciparum malaria vaccine. The Journal of infectious diseases. 1999; 180(5): 1656-64. Epub 1999/10/09. doi: 10.1086/315074. PubMed PMID: 10515829.
23. Barouch DH, Alter G, Broge T, Linde C, Ackerman ME, Brown EP, Borducchi EN,
Smith KM, Nkolola JP, Liu J, Shields J, Parenteau L, Whitney JB, Abbink P, Ng'ang'a DM, Seaman MS, Lavine CL, Perry JR, Li W, Colantonio AD, Lewis MG, Chen B, Wenschuh H, Reimer U, Piatak M, Lifson JD, Handley SA, Virgin HW, Koutsoukos M, Lorin C, Voss G, Weijtens M, Pau MG, Schuitemaker H. Protective efficacy of adenovirus/protein vaccines against SIV challenges in rhesus monkeys. Science. 2015;349(6245):320-4. Epub
2015/07/04. doi: 10.1126/science.aab3886. PubMed PMID: 26138104; PubMed Central PMCID: PMC4653134.
24. Duerr A, Huang Y, Buchbinder S, Coombs RW, Sanchez J, del Rio C, Casapia M,
Santiago S, Gilbert P, Corey L, Robertson MN. Extended follow-up confirms early vaccine- enhanced risk of HIV acquisition and demonstrates waning effect over time among participants in a randomized trial of recombinant adenovirus HIV vaccine (Step Study). The Journal of infectious diseases. 2012;206(2):258-66. Epub 2012/05/09. doi:
10.1093/infdis/jis342. PubMed PMID: 22561365; PubMed Central PMCID: PMC3490694.
25. Camathan DG, Wetzel KS, Yu J, Lee ST, Johnson BA, Paiardini M, Yan J, Morrow MP, Sardesai NY, Weiner DB, Ertl HC, Silvestri G. Activated CD4+CCR5+ T cells in the
rectum predict increased SIV acquisition in SIVGag/Tat-vaccinated rhesus macaques. Proceedings of the National Academy of Sciences of the United States of America. 2015; 112(2): 518-23. Epub 2015/01/01. doi: 10.1073/pnas.1407466112. PubMed PMID: 25550504; PubMed Central PMCID: PMC4299179.
26. Fouts TR, Bagley K, Prado IJ, Bobb KL, Schwartz JA, Xu R, Zagursky RJ, Egan MA, Eldridge JH, LaBranche CC, Montefiori DC, Le Buanec H, Zagury D, Pal R, Pavlakis GN, Felber BK, Franchini G, Gordon S, Vaccari M, Lewis GK, DeVico AL, Gallo RC. Balance of cellular and humoral immunity determines the level of protection by HIV vaccines in rhesus macaque models of HIV infection. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(9):E992-9. Epub 2015/02/15. doi: 10.1073/pnas.1423669112. PubMed PMID: 25681373; PubMed Central PMCID: PMC4352796.
27. Kwa S, Lai L, Gangadhara S, Siddiqui M, Pillai VB, Labranche C, Yu T, Moss B, Montefiori DC, Robinson HL, Kozlowski PA, Amara RR. CD40L-adjuvanted DNA/modified vaccinia virus Ankara simian immunodeficiency virus SIV239 vaccine enhances SIV-specific humoral and cellular immunity and improves protection against a heterologous SIVE660 mucosal challenge. Journal of virology. 2014;88(17):9579-89. Epub 2014/06/13. doi: 10.1128/JVI.00975-14. PubMed PMID: 24920805; PubMed Central PMCID: PMC4136340.
28. Lai L, Kwa S, Kozlowski PA, Montefiori DC, Ferrari G, Johnson WE, Hirsch V, Villinger F, Chennareddi L, Earl PL, Moss B, Amara RR, Robinson HL. Prevention of infection by a granulocyte-macrophage colony stimulating factor co-expressing DNA/modified vaccinia Ankara simian immunodeficiency virus vaccine. The Journal of infectious diseases. 2011;204(l): 164-73. Epub 2011/06/02. doi: 10.1093/infdis/jirl99. PubMed PMID: 21628671; PubMed Central PMCID: PMC3143670.
29. Teigler JE, Phogat S, Franchini G, Hirsch VM, Michael NL, Barouch DH. The canarypox virus vector ALVAC induces distinct cytokine responses compared to the vaccinia virus-based vectors MVA and NYVAC in rhesus monkeys. Journal of virology. 2014;88(3): 1809-14. Epub 2013/11/22. doi: 10.1128/JVI.02386-13. PubMed PMID: 24257612; PubMed Central PMCID: PMC3911591.
30. Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD. IFN-gamma- inducible protein 10 (IP- 10; CXCL10)-deficient mice reveal a role for IP- 10 in effector T cell
generation and trafficking. Journal of immunology. 2002; 168(7):3195-204. Epub 2002/03/22. PubMed PMID: 11907072.
31. Qin S, Rottman JB, Myers P, Kassam N, Weinblatt M, Loetscher M, Koch AE, Moser B, Mackay CR. The chemokine receptors CXCR3 and CCR5 mark subsets of T cells associated with certain inflammatory reactions. The Journal of clinical investigation. 1998;101(4):746-54. doi: 10.1172/jci 1422.
32. Groom JR, Richmond J, Murooka TT, Sorensen EW, Sung JH, Bankert K, von Andrian UH, Moon JJ, Mempel TR, Luster AD. CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation. Immunity. 2012;37(6): 1091-103. Epub 2012/11/06. doi: 10.1016/j.immuni.2012.08.016. PubMed PMID: 23123063; PubMed Central PMCID: PMC3525757.
33. Xu W, Joo H, Clayton S, Dullaers M, Herve MC, Blankenship D, De La Morena MT, Balderas R, Picard C, Casanova JL, Pascual V, Oh S, Banchereau J. Macrophages induce differentiation of plasma cells through CXCL 10/IP- 10. The Journal of experimental medicine. 2012;209(10): 1813-23, Sl-2. Epub 2012/09/19. doi: 10.1084/jem.20112142. PubMed PMID: 22987802; PubMed Central PMCID: PMC3457728.
34. Kang TH, Bae HC, Kim SH, Seo SH, Son SW, Choi EY, Seong SY, Kim TW. Modification of dendritic cells with interferon-gamma-inducible protein- 10 gene to enhance vaccine potency. The journal of gene medicine. 2009;l l(10):889-98. Epub 2009/07/21. doi: 10.1002/jgm. l371. PubMed PMID: 19618483.
35. Song RJ, Chenine AL, Rasmussen RA, Ruprecht CR, Mirshahidi S, Grisson RD, Xu W, Whitney JB, Goins LM, Ong H, Li PL, Shai-Kobiler E, Wang T, McCann CM, Zhang H, Wood C, Kankasa C, Secor WE, McClure HM, Strobert E, Else JG, Ruprecht RM. Molecularly cloned SHIV-1157ipd3N4: a highly replication competent, mucosally transmissible R5 simian-human immunodeficiency virus encoding HIV clade C Env. Journal of virology. 2006;80(17):8729-38. Epub 2006/08/17. doi: 10.1128/JVI.00558-06. PubMed PMID: 16912320; PubMed Central PMCID: PMC1563858.
36. Choi YS, Kageyama R, Eto D, Escobar TC, Johnston RJ, Monticelli L, Lao C, Crotty S. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity. 2011;34(6):932-46. Epub
2011/06/04. doi: 10.1016/j.immuni.2011.03.023. PubMed PMID: 21636296; PubMed Central PMC ID: PMC3124577.
37. Sung JH, Zhang H, Moseman EA, Alvarez D, Iannacone M, Henrickson SE, de la Torre JC, Groom JR, Luster AD, von Andrian UH. Chemokine guidance of central memory T cells is critical for antiviral recall responses in lymph nodes. Cell. 2012;150(6): 1249-63. Epub 2012/09/18. doi: 10.1016/j cell.2012.08.015. PubMed PMID: 22980984; PubMed Central PMCID: PMC3445043.
38. Olive AJ, Gondek DC, Stambach MN. CXCR3 and CCR5 are both required for T cell-mediated protection against C. trachomatis infection in the murine genital mucosa. Mucosal immunology. 2011;4(2):208-16. Epub 2010/09/17. doi: 10.1038/mi.2010.58. PubMed PMID: 20844481; PubMed Central PMCID: PMC3010299.
39. Petrovic A, Alpdogan O, Willis LM, Eng JM, Greenberg AS, Kappel BJ, Liu C, Murphy GJ, Heller G, van den Brink MR. LPAM (alpha 4 beta 7 integrin) is an important homing integrin on alloreactive T cells in the development of intestinal graft-versus-host disease. Blood. 2004;103(4): 1542-7. Epub 2003/10/18. doi: 10.1182/blood-2003-03-0957. PubMed PMID: 14563643.
40. Yamamoto T, Lynch RM, Gautam R, Matus-Nicodemos R, Schmidt SD, Boswell KL, Darko S, Wong P, Sheng Z, Petrovas C, McDermott AB, Seder RA, Keele BF, Shapiro L, Douek DC, Nishimura Y, Mascola JR, Martin MA, Koup RA. Quality and quantity of TFH cells are critical for broad antibody development in SHIVAD8 infection. Science translational medicine. 2015;7(298):298ral20. Epub 2015/08/01. doi: 10.1126/scitranslmed.aab3964. PubMed PMID: 26223303.
41. Hutchins AP, Poulain S, Fujii H, Miranda-Saavedra D. Discovery and characterization of new transcripts from RNA-seq data in mouse CD4(+) T cells. Genomics. 2012;100(5):303-13. Epub 2012/08/14. doi: 10.1016/j.ygeno.2012.07.014. PubMed PMID: 22884873.
42. Wu AR, Neff NF, Kalisky T, Dalerba P, Treutlein B, Rothenberg ME, Mburu FM, Mantalas GL, Sim S, Clarke MF, Quake SR. Quantitative assessment of single-cell RNA- sequencing methods. Nature methods. 2014; l l(l):41-6. Epub 2013/10/22. doi: 10.1038/nmeth.2694. PubMed PMID: 24141493; PubMed Central PMCID: PMC4022966.
43. Crotty S, Aubert RD, Glidewell J, Ahmed R. Tracking human antigen-specific memory B cells: a sensitive and generalized ELISPOT system. Journal of immunological methods. 2004;286(1-2): 111-22. Epub 2004/04/17. doi: 10.1016/j.jim.2003.12.015. PubMed PMID: 15087226.
44. Silveira EL, Kasturi SP, Kovalenkov Y, Rasheed AU, Yeiser P, Jinnah ZS, Legere TH, Pulendran B, Villinger F, Wrammert J. Vaccine-induced plasmablast responses in rhesus macaques: Phenotypic characterization and a source for generating antigen-specific monoclonal antibodies. Journal of immunological methods. 2014. Epub 2014/12/03. doi: 10.1016/j.jim.2014.11.003. PubMed PMID: 25445326.
45. Li H, Stephenson KE, Kang ZH, Lavine CL, Seaman MS, Barouch DH. Common features of mucosal and peripheral antibody responses elicited by candidate HIV-1 vaccines in rhesus monkeys. Journal of virology. 2014;88(22): 13510-5. Epub 2014/09/12. doi: 10.1128/JVI.02095-14. PubMed PMID: 25210178; PubMed Central PMCID: PMC4249108.
46. Montefiori DC. Evaluating neutralizing antibodies against HIV, SIV, and SHIV in luciferase reporter gene assays. Current protocols in immunology / edited by John E Coligan. 2005;Chapter 12:Unit 12 1. Epub 2008/04/25. doi: 10.1002/0471142735.iml211s64. PubMed PMID: 18432938.
47. deCamp A, Hraber P, Bailer RT, Seaman MS, Ochsenbauer C, Kappes J, Gottardo R, Edlefsen P, Self S, Tang H, Greene K, Gao H, Daniell X, Sarzotti-Kelsoe M, Gorny MK, Zolla-Pazner S, LaBranche CC, Mascola JR, Korber BT, Montefiori DC. Global panel of HIV-1 Env reference strains for standardized assessments of vaccine-elicited neutralizing antibodies. Journal of virology. 2014;88(5):2489-507. Epub 2013/12/20. doi: 10.1128/JVI.02853-13. PubMed PMID: 24352443; PubMed Central PMCID: PMC3958090.
48. Ackerman ME, Moldt B, Wyatt RT, Dugast AS, McAndrew E, Tsoukas S, Jost S, Berger CT, Sciaranghella G, Liu Q, Irvine DJ, Burton DR, Alter G. A robust, high- throughput assay to determine the phagocytic activity of clinical antibody samples. Journal of immunological methods. 201 l;366(l-2):8-19. Epub 2011/01/05. doi: 10.1016/j.jim.2010.12.016. PubMed PMID: 21192942; PubMed Central PMCID: PMC3050993.
49. Alpert MD, Heyer LN, Williams DE, Harvey JD, Greenough T, Allhorn M, Evans DT. A novel assay for antibody-dependent cell-mediated cytotoxicity against HIV-1- or SIV-
infected cells reveals incomplete overlap with antibodies measured by neutralization and binding assays. Journal of virology. 2012;86(22): 12039-52. Epub 2012/08/31. doi: 10.1128/JVI.01650-12. PubMed PMID: 22933282; PubMed Central PMCID: PMC3486484.
50. Gallo RC. Developing a Successful HIV Vaccine. The Journal of infectious diseases. 2015;212 Suppl 1 :S40-1. Epub 2015/06/28. doi: 10.1093/infdis/jiv069. PubMed PMID: 26116730; PubMed Central PMCID: PMC4574549.
51. Buge SL, Ma HL, Amara RR, Wyatt LS, Earl PL, Villinger F, Montefiori DC, Staprans SI, Xu Y, Carter E, O'Neil SP, Herndon JG, Hill E, Moss B, Robinson HL, McNicholl JM. Gpl20-alum boosting of a Gag-Pol-Env DNA/MVA AIDS vaccine: poorer control of a pathogenic viral challenge. AIDS research and human retroviruses. 2003; 19(10): 891-900. Epub 2003/10/31. doi: 10.1089/088922203322493067. PubMed PMID: 14585221.
52. Wille-Reece U, Flynn BJ, Lore K, Koup RA, Kedl RM, Mattapallil JJ, Weiss WR, Roederer M, Seder RA. HIV Gag protein conjugated to a Toll-like receptor 7/8 agonist improves the magnitude and quality of Thl and CD8+ T cell responses in nonhuman primates. Proceedings of the National Academy of Sciences of the United States of America. 2005; 102(42): 15190-4. Epub 2005/10/13. doi: 10.1073/pnas.0507484102. PubMed PMID: 16219698; PubMed Central PMCID: PMC1257732.
53. Tritel M, Stoddard AM, Flynn BJ, Darrah PA, Wu CY, Wille U, Shah JA, Huang Y, Xu L, Betts MR, Nabel GJ, Seder RA. Prime-boost vaccination with HIV-1 Gag protein and cytosine phosphate guanosine oligodeoxynucleotide, followed by adenovirus, induces sustained and robust humoral and cellular immune responses. Journal of immunology. 2003;171(5):2538-47. Epub 2003/08/21. PubMed PMID: 12928404.
54. Hudgens MG, Gilbert PB. Assessing vaccine effects in repeated low-dose challenge experiments. Biometrics. 2009;65(4): 1223-32. Epub 2009/04/29. doi: 10.111 l/j.1541- 0420.2009.01189.x. PubMed PMID: 19397589; PubMed Central PMCID: PMC2794923.
55. Goode D, Truong R, Villegas G, Calenda G, Guerra-Perez N, Piatak M, Lifson JD,
Blanchard J, Gettie A, Robbiani M, Martinelli E. HSV-2-driven increase in the expression of alpha4beta7 correlates with increased susceptibility to vaginal SHIV(SF162P3) infection. PLoS pathogens. 2014;10(12):el004567. Epub 2014/12/19. doi:
10.1371/journal.ppat.1004567. PubMed PMID: 25521298; PubMed Central PMCID: PMC4270786.
56. Camathan DG, Wetzel KS, Yu J, Lee ST, Johnson BA, Paiardini M, Yan J, Morrow MP, Sardesai NY, Weiner DB, Ertl HC, Silvestri G. Activated CD4+CCR5+ T cells in the rectum predict increased SIV acquisition in SIVGag/Tat-vaccinated rhesus macaques. Proceedings of the National Academy of Sciences of the United States of America. 2014. Epub 2015/01/01. doi: 10.1073/pnas.1407466112. PubMed PMID: 25550504.
Example 3 Targeting CD4 Follicular Helper T Cells for Enhancing HIV Vaccine-Induced
Humoral Immunity
Summary
[0203] Anti-retroviral therapy (ART) has dramatically altered the HIV pandemic landscape by rendering the disease manageable - but still incurable. Significant barriers are associated with the global implementation of ART that limit its utility for sustainable prevention of HIV. Moreover, the intersection of aging-related conditions and the consequences of long-term ART will have a substantial impact on the healthcare system as well as on HIV patients. These challenges underscore the need to pursue strategies that will provide both sustainable HIV prevention and a functional HIV cure. The results of the RV144 trial indicate that vaccination may prevent HIV transmission in humans and that durability of anti-envelope (Env) HIV antibodies may be the key to this protection. Efforts to improve upon the RV144 trial have demonstrated that booster immunizations increase serum anti-Env antibody titers only transiently. This“anti-Env antibody persistence problem” impedes our efforts to develop an effective HIV vaccine.
[0204] Here, we use a DNA-prime/Protein-boost vaccine regimen in rhesus macaques. In the first approach we focus on establishing how Thl versus Thl/Thl7 priming impacts innate inflammatory response, Env Tfh frequencies, phenotype, transcriptional profile, and B cell helper capacity, as well as systemic and mucosal anti-Env antibody titers. In the second approach we determine whether combining the state-of-the-art soluble Env mimic, an Env SOSIP turner protein immunogen with a highly potent and robust adjuvant (to induce Thl/Thl7 response, results in increased production of a high-quality, longlasting anti-Env antibodies relative to Thl vaccine regimen. We further investigate whether vaccine-mediated
induction of the polyfunctional Tfh response improves protection when faced with a mucosal simian HIV challenge.
Overview
[0205] Antiretroviral therapy (ART) has a remarkable impact on the morbidity and mortality associated with HIV but significant barriers hamper its global implementation, limiting its full potential as a preventative tool. Moreover, the intersection of aging-related conditions and the consequences of long-term ART is predicted to adversely impact the healthcare system as well as the HIV patient. These challenges underscore the need to pursue strategies that will provide both sustainable HIV prevention and a functional HIV cure. The results of the RV144 trial indicate that vaccination may prevent HIV transmission in humans and that durability of anti-envelope (Env) HIV antibodies may be the key to this protection. Efforts to improve upon the RV144 trial have demonstrated that booster immunizations increase serum anti-Env titers but do so transiently. This short-lived impact demonstrates an ineffectiveness in the induction of long-lived Env-specific plasma cells, the source of the durable antibody. This“anti-Env antibody persistence problem” is impeding our progress in developing an effective HIV vaccine.
[0206] The answer lies in the CD4 T follicular helper cells (Tfh), a subset of CD4 helper (Th) cells, vital for differentiation of short-lived germinal center B cells to long-lived antibody-producing plasma cells. Tfh cells are heterogeneous and encompass Thl, Th2, Thl7-type helper attributes. The proportion of each helper attribute determines the durability and functional quality of the antibody response. These helper attributes are programmed during T-cell priming and are dictated by the initial inflammatory response following vaccination. This provides researchers a window of opportunity to manipulate the CD4 Tfh response in order to maximize the generation of durable anti-Env humoral immunity. Because the characteristics that make up an optimal Tfh helper cell for induction of durable anti-Env antibody have not yet been clearly defined, our long-term goal is to determine how manipulating the Tfh helper profile impacts the longevity of anti-Env antibodies within the setting of priming and boosting immunizations.
[0207] Here, we establish whether improved durability and neutralization potential of anti- Env antibodies can be influenced by manipulating vaccine-induced generation of Thl / Thl7 Tfh cells. These investigations are supported by rigorous data demonstrating that: 1) a Thl- DNA prime substantially increases the Env-specific Tfh cells relative to a Thl+Th2 vaccine
regimen; 2) Thl -protein boost results in greater production of the IgGl and IgG3 subclasses with enhanced durability, breadth, specificity and avidity of anti-Env antibody; and 3) Thl vaccine generation of polyfunctional Thl/Thl7 GC Tfh cells predicts the durability of anti- Env IgG titers in sera and anti-Env IgA titers in mucosa.
[0208] Using a DNA/Protein vaccine platform, we describe two integrated approaches; 1) we employ interferon protein-10 and interleukin 6 as molecular adjuvants to a DNA (DIP-10- IL-6) vaccine to prime Thl/Thl7-type Tfh cells; 2) we use a cationic liposomal adjuvant, CAFOl, a strong Thl/Thl7 adjuvant to a state-of-the-art soluble Env mimic, an Env SOSIP trimer protein, to boost Thl/Thl7-type Tfh cells. Using monkeys, we evaluate this regimen with three vaccine groups to determine whether an increase in Thl/Thl7-type Tfh enhances the longevity of anti-Env, the protective HIV antibody, relative to a Thl platform only during the prime or during both the prime and the boost.
[0209] The first approach is to demonstrate that a Thl/Thl7 DNA-prime is more effective at increasing the magnitude of Env-specific Tfh cells relative to a Thl vaccine regimen. Here, we a) delineate innate determinants of Env-specific Tfh cell differentiation during the DNA prime; and b) delineate Env-specific Tfh cells induced in response to a Thl versus Thl/Thl7 DNA prime.
[0210] The second approach is to demonstrate that synergistic boosting of Thl/Thl7 polarized Tfh cells elicits robust and durable anti-Env antibody titers in systemic and mucosal compartments. Here, we a) determine anti-Env antibody quantity, quality, neutralization potential, and effector functions in systemic and mucosal compartments following vaccination; and b) determine whether specific immunity established by Thl/Thl7 regimen is more effective at preventing mucosal tier 2 SHIV acquisition.
Experimental approach and data.
[0211] Thl vaccine regimen induces robust and durable serum HIV Env antibody titers. Detailed kinetic analysis revealed that HIV anti-Env titers peaked after the final boost with a median 5-fold boost in antibody titers at week 2 post-2nd protein relative to week 0. Notably, antibody titers were significantly higher in the Thl regimen at week 0 (median gpl40 titers pg/ml; Thl +2 = 257.2 and Thl = 1314; p < 0.01), at week 2 (Thl+2 = 9942 and Thl=29104; p < 0.0001), week 8 (Thl+2=2067 and Thl = 6904; p < 0.001), and week 18 (Thl+2= 450 and Thl= 2431; p<0.001) (FIG. 24). We used binding antibody multiplex assay (BAMA) to assess the breadth and specificity of the serum antibody response. Responses to both 1086. C
and CH505 Clade C Env immunogens were significantly higher in the Thl group. Similarly, the increased titers against Con C and Con S were sustained at week 8 demonstrating induction of cross-clade breadth (FIG. 25A). Avidity index measured using 2M sodium thiocyanate revealed the presence of higher affinity antibodies in the Thl group (FIG. 25B). We also assessed binding to linear VI V2 and conformational epitopes at week 2 and 8 and found that significantly higher specificity to these important regions in the Thl vaccine regimen (FIG. 25C). Evaluation of HIV neutralization activity demonstrated higher induction of Tier 1A clade C isolate (ID50 range at week 2 post 2nd protein boost Thl+2: 37 -1126; Thl: 195-4977, p < 0.01) These titers dropped to an ID50 value of 20 in Thl+2 group and were maintained between 24-1057 in the Thl group (FIG. 25D).
[0212] We also measured secretions from vaginal and rectal sponges to assess anti-Env IgA responses. Similar to observed serum responses, the Thl vaccine initiated a significant increase in mucosal 1086. C gpl40-specific IgA and IgG titers in the vaginal and rectal mucosa relative to Thl+2 regimen (FIGS. 26A and B). Notably, at week 8 post 2nd protein boost, rectal IgA titers were below the limit of detection in all Thl+2 animals relative to detectable levels in 8/10 of Thl vaccinated animals. Together these data demonstrate that Thl vaccine regimen is more effective in generating durable binding titers in sera and mucosa, as well as HIV anti-Env antibodies with greater breadth, avidity, broader specificity, and neutralization potential relative to a mixed Thl+2 vaccine regimen. Thl vaccine induces higher frequencies of Env-specific CD4 T cells. To study Tfh responses in detail following the prime we examined whether Tfh frequencies were increased following the third DNA prime. A significant increase in frequencies and absolute counts of ICOS+PD-l+activated Tfh cells was observed (FIG. 27B). Consistent with this observation, Env-specific Tfh cells were induced at significantly higher frequencies in the Thl group (FIGS. 27C and D). These data establish the importance of a strong Thl DNA prime in eliciting strong Tfh cell responses.
[0213] Induction of polyfunctional Thl/Thl7 CD4 Tfh due to Thl vaccine regimen. Following the first protein boost, we characterized the innate inflammatory response and observed induction of cytokines important for Thl, Thl7, and Tfh programing (FIG. 28A). Following the boost, Env-specific Tfh cells were higher in the Thl vaccine regimen and were associated with increase in GC B cell frequencies (FIGS. 28B and C). GC Tfh cells expressed higher levels of Bcl-6, CXCR3, CCR6 and IFNG, IL-17+ cells expressed relatively higher levels of CD40L (FIG. 28E). Gene expression profiles revealed enrichment of Thl /
Thl7 and classical Tfh genes within GC Tfh cells in the Thl vaccine regimen (FIG. 28F). Correspondingly, we found that the frequency of Thl/Thl7 Tfh cells was higher in the Thl vaccine regimen suggesting the induction of polyfunctional CD4 Tfh cells (FIG. 28G).
[0214] Induction of polyfunctional Thl /Thl 7 CD4 Tfh cells predicts durability of anti-Env antibody titers in systemic and mucosal compartments. We performed correlational analysis to determine GC correlates of antibody titers, which showed the frequency of polyfunctional Thl/Thl7 GC Tfh cells to be directly predictive of binding titers measured by ELISA and BAMA (FIG. 29). Consistent with the role of Thl7 cells in inducing IgA responses, an association with mucosal Env IgA titers was also observed.
[0215] A single immunization with CAFOl-adjuvanted protein induces robust antibody responses. Our data strongly indicate that synergistic induction of polyfunctional Thl/Thl7 CD4 Tfh cells enhances HIV anti-Env antibody titers. Therefore, we sought to procure the CAFOl adjuvant - the strongest known adjuvant shown in multiple studies to induce Thl/Thl7 cells - for the studies described. Data in mice using CAFOl (44) (FIG. 30) adjuvanted to either a chlamydia (FIG. 30B) or ovalbumin protein (FIG. 30C) show gradual increase in Ag-specific titers in serum at levels comparable or superior to the squalene-based oil-in-water nano-emulsion adjuvant Addvax™, an MF59 analog. Quantification of antigen- bound cells at the site of immunization show higher antigen uptake with CAFOl suggestive that antigen depot induction at the immunization site (FIG. 30D) results in robust germinal center responses at the draining lymph node and subsequent enhancement in antibody titers. Together with CAFOl mediated skewing to a Thl and Thl 7 inflammatory response (39) and robust induction of IgA plasma cells in genital mucosa (41), these data provide a strong scientific rationale for determining whether an HIV vaccine platform designed to prime and reproducibly boost Thl/Thl7 CD4 Tfh cells employing a CAFOl adjuvant will engender high titer serum and mucosal HIV anti-Env responses.
Methods
[0216] Study design : The SI and S2 studies are designed to investigate the outcome of synchronous priming and boosting of polyfunctional Thl/Thl7 CD4 Tfh cells on antibody durability, breadth, specificity, and neutralization potential in both the systemic and mucosal compartments. All groups are directly compared for immunogenicity and for efficacy against intrarectal simian (S) HIV BG505 S375Y virus (weekly challenge with 1.4 x!07
virions/challenge). The DNA SHIV BG505-SOSIP.664 plasmid is used, into which we clone in IP- 10 and IL-6.
[0217] Animals. SIV- STLV- SRV-negative Indian origin rhesus macaques from the CNPRC breeding colony, between the ages of 3-6 years are selected for the study. Each group has equal males and females.
[0218] Sample Size : There are 8 macaques in Group 1, and 12 macaques in Groups 2 and 3. We have worked with a statistician to ensure our studies are sufficiently powered to detect differences in immunogenicity. We evaluate individual innate, T cell, and antibody trajectories over time and compare mean trajectories across experimental groups. We then correlate individual trajectories of the innate inflammatory response with those of Tfh responses and humoral immunity. Our study design (sample sizes, repeated measures) provides confidence limits of about ±0.5s for the difference in rates of change of markers, compared to the variation s in individual trajectories (R package longpower, Edland 2009). We have 80% power to detect differences comparable to those found in our previous studies.
[0219] Sampling and assays: The sampling schedule is designed to optimally and rigorously capture immunological and biological dynamics to link innate immune parameters to Tfh responses and ultimately to antibody responses following vaccination. To this end, all animals are sampled at various time points at baseline and after each of the immunizations. We evaluate clinical chemistries immediately following and weeks after immunization to monitor safety (26).
First approach
[0220] Part la) Delineate innate responses to a Thl versus Thl/Thl7 DNA-prime to define predictive correlates of Env-specific Tfh cells during the prime.
[0221] Experimental Approach: Productive T cell responses critically depend on cytokine responses during priming, and recent studies demonstrate that monocyte-derived IL-Ib drives effective Tfh and CD4 T cell differentiation (28, 60, 61). Detailed investigation of the kinetics of pro-inflammatory monocytes - cellular innate biomarkers of adjuvanticity - revealed a transient increase in CD14+CD16+ monocytes in blood with a higher relative increase in the Thl vaccine group (FIG. 32). Strikingly, the quantity of GC Tfh cells within the fine-needle aspirates of the draining lymph node showed higher GC frequencies in Thl vaccine group (FIG. 32C). Notably, the improved inflammatory response was associated
with increased levels of antibody durability linking the innate immune response to priming of effective CD4 Tfh help (FIG. 32D). Based on these data, we compare induction of innate immune responses between Thl and Thl/Thl7 priming regimens. To optimally capture the early and transient induction of immune factors, we collect blood at 0, 48, and 72 hours post immunization to establish a comprehensive cytokine /chemokine profile temporally, and across vaccine platforms. We determine (i) Cytokine, chemokine profile using a multiplex assay to measure soluble factors driving Thl polarization: interleukins (IL)-12, IL-Ib, IL-8, IL-2; macrophage inflammatory protein (MIP)la, MIRIb, MIP3a, MIR3b; macrophage chemotactic protein (MCP)-l; CXCR3 ligands; monokine induced by gamma (MIG/CXCL9), IP-10/CXCL10, interferon (IFN)-nducible T-cell alpha chemoattractant (I-TAC/CXCL11); tumor necrosis factor (TNF)a; IFNs IRNa,b,g. We examine a panel of Thl7 factors including IL-17, IL-6, TGF-b, IL-21, IL-23 and the CCR6 ligand CCL20. In addition, we capture dynamics of pro-inflammatory monocyte frequencies following immunization as shown in FIG. 32. Together, this approach provides a comprehensive in-depth overview of the inflammatory profile during priming.
[0222] Part lb): Delineate Env-specific Tfh responses to a Thl versus Thl/Thl7 DNA- prime.
[0223] Experimental Approach. We quantify vaccine-specific Tfh cells in systemic and lymphoid compartments. We have demonstrated that the DNA EP vaccine induces robust frequencies of vaccine-specific CD4 T cells in the blood and lymph node. We study vaccine- specific Tfh cells utilizing two complementary approaches we have optimized and established (17). We utilize the cell-cycle marker Ki-67 to interrogate CXCR5+ CD4 T cells at peak effector time points following each immunization for expression of CXCR3 and CCR6. This is complemented by activation induced marker (AIM) and standard intracellular cytokine staining (ICS)-based assays designed to examine vaccine-specific responses (at peak and memory) after stimulation with relevant Gag and Env peptide pools (62, 63). After stimulation, Ag-specific Tfh cells are identified based on co-expression of AIM marker positivity and CXCR5 expression. We evaluate GC responses using flow cytometry and examine GC Tfh, Tfh and GC B cells for CXCR3 and CCR6 expression.
[0224] To gain a comprehensive understanding of molecular heterogeneity within vaccine elicited Tfh cells, we determine transcription profiles of single-cell sorted CXCR3+ vs. CCR6+ Tfh cells within the draining lymph node. Since antibody titers are established after
the 3rd DNA boost in the majority of animals, and active germinal centers are observed at 14 days post 3rd DNA, we collect biopsies from the draining lymph node at this time. Vaccine- specific Tfh cells are identified by induction of 0X40 and CD25 after 5 hour stimulation with Env peptides in the presence of co-stimulatory molecules (40). Samples from 5 animals within each vaccine group are selected to understand heterogeneity within Tfh cell subsets and how it compares across vaccine regimens. Because changes in CXCR5, CXCR3, CCR6, and PD-1 likely occur upon stimulation, CD3+CD4+Fas+CXCR5+PD-1+/++ (comprising Tfh and GC Tfh) are sorted using fluorescence-assisted cell sorting (FACS) into CXCR3+ and CCR6+ single positive subsets prior to stimulation. We also sort CXCR3-CCR6- double negative Tfh cells. Naive CD4 T cells (CD4+Fas-CXCR5-PD-1-CXCR3-) serve as a reference population. After stimulation with Env peptide pools in the presence of co stimulatory molecules using established protocols (17), cells are stained with surface 0X40 and CD25 and twenty-four double positive cells from each of the two subsets are singly sorted into a 96-well plate. The single-cell RNA sequencing methodology allows us to capture the heterogeneity of the Tfh cell response to stimulation and gives us the ability to resolve important associations between cell phenotype, stimulation status, and cellular transcriptional program.
[0225] Part lc): Delineate anti -Env binding antibody titers elicited following the prime to Thl versus Thl/Thl7 DNA immunization.
[0226] Experimental Approach. We assess the humoral response in the systemic and mucosal compartments over the course of DNA immunization. We perform ELISA assays using standard protocols to measure Env-specific antibody in sera and mucosa.
Approach 2
[0227] Our data show marked potency of a Thl -Tfh vaccine regimen in eliciting humoral immunity and demonstrates that increasing the frequency of Thl/Thl7 GC Tfh cells within the draining lymph node may result in enhanced durability of antibody titers at the memory time point. These data together with published evidence in the literature provide a strong scientific rationale to utilize CAFOl adjuvanted trimeric Env protein immunogen BG505 SOSIP.664 boost to test the hypothesis that induction of polyfunctional Thl and Thl7 Tfh cells will enhance antibody titers in systemic and mucosal compartments.
[0228] Part 2a): Delineate innate responses and Env-specific Tfh responses to a Thl versus Thl/Thl7 boost. As outlined for Approach 1, we perform an in-depth and comprehensive interrogation of innate and Tfh responses following the boost.
[0229] Part 2b): Perform an in-depth characterization of systemic and mucosal titers following vaccination.
[0230] Experimental Approach. We perform a broad repertoire of quantitative and functional assays to comprehensively assess the humoral response in both the systemic and mucosal compartments. We measure anti-Env-specific IgG and IgA binding titers and antibody IgG subclass (45) against the BG505 SOSIP gpl40 by ELISA using established protocols. We utilize the highly sensitive binding antibody multiplex assay (BAMA) to quantify anti-Env responses against gpl20, gp41,VlV2, and V3 antigens in the mucosa and sera.
[0231] We measure neutralization responses against tier 1 and tier 2 autologous and heterologous viruses using the TZM-bl assay after the 2nd protein boost (64, 65). Initial assays are done with the challenge SHIV, the immunogen SHIV, and clade-matched tier 1 A, tier IB and tier 2 viruses. Encouraging tier 2 nAb responses stimulates further assays against a multi-clade global virus panel and bnAb mapping mutants for breadth and specificity of the Tier2 nAb response. In the event of absent or weak tier2 nAb, we assay viruses that can detect precursors to bnAbs that have been developed for V2 glycan and CD4bs lineages.
[0232] To assess non-neutralizing antibody effector functions, we measure antibody- dependent phagocytosis (ADP) using the Ackerman flow cytometric assay and antibody dependent cellular cytotoxicity (ADCC) using the Evans assay. We measure antibody avidity using surface plasmon resonance as described previously (66). Our data show generation of higher avidity gpl40 anti-Env antibodies with sequential immunization and significantly higher antibody avidity reached with Thl -Tfh group indicative of productive germinal center reaction (FIG. 33). Furthermore, Thl vaccine regimen results in improved IgGl and IgG3 subtype antibody responses (FIG. 34) prompting the question of whether this led to a corresponding increase in antibody-dependent cellular cytotoxicity (ADCC) and antibody- dependent cell-mediated virus inhibition (ADCVI) activities.
[0233] To follow up on these data, we measure ADCC and ADCVI functionalities using gp!20 coated and virus infected target cells (67). Given recent evidence of IgGl subclass
antibodies playing a role in neutralization, we also delineate the relationship between IgG subclasses and neutralization potential of sera (68).
[0234] Part 2c): Determine whether specific immunity established by Thl/Thl7 regimen is more effective at preventing mucosal tier 2 acquisition.
[0235] Experimental Approach. To determine vaccine efficacy, we determine protection against weekly Intrarectal repeat low-dose challenge with clade B BG505 N332 S375Y for a maximum of 12 weeks (12 challenges) (69). Age and sex-matched unvaccinated controls are derived from the CNRPC colony and we determine whether DIP-10/IL-6 SOSIPCAFOl vaccine delayed rate of acquisition and decreased peak viremia in infected animals relative to DIP-10/IL-6 SOSIPALFQ vaccine.
Statistical Analyses
[0236] To arrive at sample sizes, we conducted power analyses using per-challenge infection rates computed from our prior trials using the SIV251 challenge virus. Our power analyses determined that we should achieve a minimum per-challenge infection rate of about 0.02 in group 3 to see a statistically significant (80% power at the alpha level of 0.05) difference between groups 1 and 3.
[0237] We evaluate two main parameters to determine protection: 1) Status of Infection and 2) number of challenges to infection. Comparison of proportions of infected and uninfected animals between vaccine groups and controls are performed using Chi-square test. Comparisons of number of challenges until infection between groups are conducted using the two-sided log-rank test and visualized using Kaplan-Meier curves. Models of the protective effect of vaccination proposed by Hudgens and Gillbert are fitted, and the Akaike. Information Criteria (AIC) are used to select the most parsimonious model. In addition, we determine peak viral load at 2 and 3 weeks post-infection to determine whether DIP-10/IL-6 SOSIPCAFOl vaccine enhanced viral control relative to DIP-10/IL-6 SOSIPALFQ vaccine.
Bibliography and References
1. Latest statistics on the status of the AIDS epidemic. UNAIDS Fact Sheet www.unaids.org/en/resources/fact-sheet ed2018.
2. Cohen MS, Chen YQ, McCauley M, Gamble T, Hosseinipour MC, Kumarasamy N, Hakim JG, Kumwenda J, Grinsztejn B, Pilotto JH, Godbole SV, Mehendale S, Chariyalertsak
S, Santos BR, Mayer KH, Hoffman IF, Eshleman SH, Piwowar-Manning E, Wang L, Makhema J, Mills LA, de Bruyn G, Sanne I, Eron J, Gallant J, Havlir D, Swindells S, Ribaudo H, Elharrar V, Burns D, Taha TE, Nielsen-Saines K, Celentano D, Essex M, Fleming TR, Team HS. Prevention of HIV-1 infection with early antiretroviral therapy. N Engl J Med. 2011;365(6):493-505. Epub 2011/07/20. doi: 10.1056/NEJMoal 105243. PubMed PMID: 21767103; PMCID: PMC3200068.
3. Granich R, Williams B, Montaner J, Zuniga JM. 90-90-90 and ending AIDS: necessary and feasible. Lancet. 2017;390(10092):341-3. Epub 2017/07/27. doi: 10.1016/S0140-6736(17)31872-X. PubMed PMID: 28745591.
4. He T, Falwell E, Brocca-Cofano E, Pandrea I. Modeling aging in HIV infection in nonhuman primates to address an emerging challenge of the post-ART era. Curr Opin Virol. 2017;25:66-75. Epub 2017/08/15. doi: 10.1016/j.coviro.2017.07.012. PubMed PMID: 28803049; PMCID: PMC6392462.
5. Aepfelbacher JA, Balmaceda J, Purdy J, Mattingly A, Zambell K, Hawkins K, Chairez C, Curl KA, Dee N, Hadigan C. Increased Prevalence of Hepatic Steatosis in Young Adults with Life-long HIV. J Infect Dis. 2019. Epub 2019/03/11. doi: 10.1093/infdis/jiz096. PubMed PMID: 30852587.
6. Bakal DR, Coelho LE, Luz PM, Clark JL, De Boni RB, Cardoso SW, Veloso VG, Lake JE, Grinsztejn B. Obesity following ART initiation is common and influenced by both traditional and HIV-/ART-specific risk factors. J Antimicrob Chemother. 2018;73(8):2177- 85. Epub 2018/05/04. doi: 10.1093/jac/dkyl45. PubMed PMID: 29722811; PMCID: PMC6054231.
7. Fauci AS. An HIV Vaccine Is Essential for Ending the HIV/AIDS Pandemic. JAMA. 2017;318(16): 1535-6. Epub 2017/10/21. doi: 10.1001/jama.2017.13505. PubMed PMID: 29052689.
8. Fauci AS. An HIV Vaccine: Mapping Uncharted Territory. JAMA. 2016;316(2): 143-4. Epub 2016/07/13. doi: 10.1001/jama.2016.7538. PubMed PMID: 27404178.
9. Pitisuttithum P, Gilbert P, Gurwith M, Heyward W, Martin M, van Griensven F, Hu D, Tappero JW, Choopanya K, Bangkok Vaccine Evaluation G. Randomized, double-blind, placebo-controlled efficacy trial of a bivalent recombinant glycoprotein 120 HIV-1 vaccine
among injection drug users in Bangkok, Thailand. J Infect Dis. 2006; 194(12): 1661-71. Epub 2006/11/17. doi: 10.1086/508748. PubMed PMID: 17109337.
10. Adis International L. HIV gpl20 vaccine - VaxGen: AIDSVAX, AIDSVAX B/B, AIDSVAX B/E, HIV gpl20 vaccine - Genentech, HIV gpl20 vaccine AIDSVAX - VaxGen, HIV vaccine AIDSVAX - VaxGen. Drugs R D. 2003;4(4):249-53. Epub 2003/07/10. doi: 10.2165/00126839-200304040-00007. PubMed PMID: 12848591.
11. Hammer SM, Sobieszczyk ME, Janes H, Karuna ST, Mulligan MJ, Grove D, Koblin BA, Buchbinder SP, Keefer MC, Tomaras GD, Frahm N, Hural J, Anude C, Graham BS, Enama ME, Adams E, DeJesus E, Novak RM, Frank I, Bentley C, Ramirez S, Fu R, Koup RA, Mascola JR, Nabel GJ, Montefiori DC, Kublin J, McElrath MJ, Corey L, Gilbert PB, Team HS. Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. N Engl J Med. 2013;369(22):2083-92. Epub 2013/10/09. doi: 10.1056/NEJMoal310566. PubMed PMID: 24099601; PMCID: PMC4030634.
12. Rerks-Ngarm S, Pitisuttithum P, Excler JL, Nitayaphan S, Kaewkungwal J, Premsri N, Kunasol P, Karasavvas N, Schuetz A, Ngauy V, Sinangil F, Dawson P, deCamp AC, Phogat S, Garunathan S, Tartaglia J, DiazGranados C, Ratto-Kim S, Pegu P, Eller M, Karnasuta C, Montefiori DC, Sawant S, Vandergrift N, Wills S, Tomaras GD, Robb ML, Michael NL, Kim JH, Vasan S, O'Connell RJ, Team RVS. Randomized, Double-Blind Evaluation of Late Boost Strategies for HIV-Uninfected Vaccine Recipients in the RV144 HIV Vaccine Efficacy Trial. J Infect Dis. 2017;215(8): 1255-63. Epub 2017/03/23. doi: 10.1093/infdis/jix099. PubMed PMID: 28329190; PMCID: PMC5853427.
13. Butler AL, Fischinger S, Alter G. The Antibodiome-Mapping the Humoral Immune Response to HIV. Curr HIV/AIDS Rep. 2019; 16(2): 169-79. Epub 2019/03/25. doi: 10.1007/s 11904-019-00432-x. PubMed PMID: 30903381; PMCID: PMC6441398.
14. Saunders KO, Verkoczy LK, Jiang C, Zhang J, Parks R, Chen H, Housman M, Bouton-Verville H, Shen X, Trama AM, Scearce R, Sutherland L, Santra S, Newman A, Eaton A, Xu K, Georgiev IS, Joyce MG, Tomaras GD, Bonsignori M, Reed SG, Salazar A, Mascola JR, Moody MA, Cain DW, Centlivre M, Zurawski S, Zurawski G, Erickson HP, Kwong PD, Alam SM, Levy Y, Montefiori DC, Haynes BF. Vaccine Induction of Heterologous Tier 2 HIV-1 Neutralizing Antibodies in Animal Models. Cell Rep. 2017;21(13):3681-90. Epub 2017/12/28. doi: 10.1016/j.celrep.2017.12.028. PubMed PMID: 29281818; PMCID: PMC5777169.
15. Spearman P, Tomaras GD, Montefiori DC, Huang Y, Elizaga ML, Ferrari G, Alam SM, Isaacs A, Ahmed H, Hural J, McElrath MJ, Ouedraogo L, Pensiero M, Butler C, Kalams SA, Overton ET, Barnett SW, Team HP, Network NNHVT. Rapid Boosting of HIV-1 Neutralizing Antibody Responses in Humans Following a Prolonged Immunologic Rest. J Infect Dis. 2019. Epub 2019/01/08. doi: 10.1093/infdis/jiz008. PubMed PMID: 30615119.
16. Slifka MK, Matloubian M, Ahmed R. Bone marrow is a major site of long-term antibody production after acute viral infection. J Virol. 1995;69(3): 1895-902. Epub 1995/03/01. PubMed PMID: 7853531; PMCID: PMC188803.
17. Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immunity due to long-lived plasma cells. Immunity. 1998;8(3):363-72. Epub 1998/04/07. PubMed PMID: 9529153.
18. Crotty S. T follicular helper cell differentiation, function, and roles in disease. Immunity. 2014;41(4):529-42. Epub 2014/11/05. doi: 10.1016/j.immuni.2014.10.004. PubMed PMID: 25367570; PMCID: PMC4223692.
19. Havenar-Daughton C, Lee JH, Crotty S. Tfh cells and HIV bnAbs, an immunodominance model of the HIV neutralizing antibody generation problem. Immunol Rev. 2017;275(1):49-61. Epub 2017/01/31. doi: 10.1111/imr.12512. PubMed PMID: 28133798.
20. Jha V, Janoff EN. Complementary Role of CD4+ T Cells in Response to Pneumococcal Polysaccharide Vaccines in Humans. Vaccines (Basel). 2019;7(1). Epub 2019/02/14. doi: 10.3390/vaccines7010018. PubMed PMID: 30754689; PMCID: PMC6466080.
21. Cyster JG, Allen CDC. B Cell Responses: Cell Interaction Dynamics and Decisions. Cell. 2019;177(3):524-40. Epub 2019/04/20. doi: 10.1016/j .cell.2019.03.016. PubMed PMID: 31002794.
22. Read KA, Powell MD, Sreekumar BK, Oestreich KJ. In Vitro Differentiation of Effector CD4(+) T Helper Cell Subsets. Methods Mol Biol. 2019;1960:75-84. doi: 10.1007/978-l-4939-9167-9_6. PubMed PMID: 30798522.
23. Inoue T, Moran I, Shinnakasu R, Phan TG, Kurosaki T. Generation of memory B cells and their reactivation. Immunol Rev. 2018;283(1): 138-49. Epub 2018/04/18. doi: 10.1111/imr.12640. PubMed PMID: 29664566.
24. Excler JL, Michael NL. Lessons from HIV-1 vaccine efficacy trials. Curr Opin HIV AIDS. 2016;11(6):607-13. Epub 2016/08/19. doi: 10.1097/COH.0000000000000312. PubMed PMID: 27537673.
25. Morita R, Schmitt N, Bentebibel SE, Ranganathan R, Bourdery L, Zurawski G, Foucat E, Dullaers M, Oh S, Sabzghabaei N, Lavecchio EM, Punaro M, Pascual V, Banchereau J, Ueno H. Human blood CXCR5(+)CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity. 2011;34(1): 108-21. Epub 2011/01/11. doi: 10.1016/j.immuni.2010.12.012. PubMed PMID: 21215658; PMCID: PMC3046815.
26. Lee SY, Lee SH, Seo HB, Ryu JG, Jung K, Choi JW, Jhun J, Park JS, Kwon JY, Kwok SK, Youn J, Park SH, Cho ML. Inhibition of IL-17 ameliorates systemic lupus erythematosus in Roquin(san/san) mice through regulating the balance of TFH cells, GC B cells, Treg and Breg. Sci Rep. 2019;9(1):5227. doi: 10.1038/s41598-019-41534-l. PubMed PMID: 30914691; PMCID: PMC6435653.
27. Hirota K, Turner JE, Villa M, Duarte JH, Demengeot J, Steinmetz OM, Stockinger B. Plasticity of Thl7 cells in Peyer's patches is responsible for the induction of T cell- dependent IgA responses. Nat Immunol. 2013;14(4):372-9. doi: 10.1038/ni.2552. PubMed PMID: 23475182; PMCID: PMC3672955.
28. Barbet G, Sander LE, Geswell M, Leonardi I, Cerutti A, Iliev I, Blander JM.
Sensing Microbial Viability through Bacterial RNA Augments T Follicular Helper Cell and Antibody Responses. Immunity. 2018;48(3):584-98 e5. Epub 2018/03/20. doi:
10.1016/j.immuni.2018.02.015. PubMed PMID: 29548673; PMCID: PMC5924674.
29. Bentebibel SE, Khurana S, Schmitt N, Kurup P, Mueller C, Obermoser G, Palucka AK, Albrecht RA, Garcia-Sastre A, Golding H, Ueno H. ICOS(+)PD-l(+)CXCR3(+) T follicular helper cells contribute to the generation of high-avidity antibodies following influenza vaccination. Sci Rep. 2016;6:26494. Epub 2016/05/28. doi: 10.1038/srep26494. PubMed PMID: 27231124; PMCID: PMC4882544.
30. Iyer SS, Gangadhara S, Victor B, Gomez R, Basu R, Hong JJ, Labranche C, Montefiori DC, Villinger F, Moss B, Amara RR. Codelivery of Envelope Protein in Alum with MVA Vaccine Induces CXCR3-Biased CXCR5+ and CXCR5- CD4 T Cell Responses
in Rhesus Macaques. J Immunol. 2015;195(3):994-1005. Epub 2015/06/28. doi: 10.4049/jimmunol.1500083. PubMed PMID: 26116502; PMCID: PMC4506863.
31. Rao M, Onkar S, Peachman KK, White Y, Trinh HV, Jobe O, Zhou Y, Dawson P, Eller MA, Matyas GR, Alving CR. Liposome-Encapsulated Human Immunodeficiency Virus-1 gpl20 Induces Potent VlV2-Specific Antibodies in Humans. J Infect Dis. 2018;218(10): 1541-50. Epub 2018/06/13. doi: 10.1093/infdis/jiy348. PubMed PMID: 29893872.
32. Alving CR, Peachman KK, Rao M, Reed SG. Adjuvants for human vaccines. Curr Opin Immunol. 2012;24(3):310-5. Epub 2012/04/24. doi: 10.1016/j .coi.2012.03.008. PubMed PMID: 22521140; PMCID: PMC3383374.
33. He L, Gu W, Wang M, Chang X, Sun X, Zhang Y, Lin X, Yan C, Fan W, Su P, Wang Y, Yi C, Lin G, Li L, Jiang Y, Lu J, Dong C, Wang H, Sun B. Extracellular matrix protein 1 promotes follicular helper T cell differentiation and antibody production. Proc Natl Acad Sci U S A. 2018; 115(34):8621-6. Epub 2018/08/09. doi: 10.1073/pnas. l801196115. PubMed PMID: 30087185; PMCID: PMC6112729.
34. Spolski R, Leonard WJ. Cytokine mediators of Thl7 function. Eur J Immunol. 2009;39(3):658-61. doi: 10.1002/eji.200839066. PubMed PMID: 19283717; PMCID: PMC2735233.
35. Wichner K, Stauss D, Kampfrath B, Kruger K, Muller G, Rehm A, Lipp M, Hop ken UE. Dysregulated development of IL-17- and IL-21 -expressing follicular helper T cells and increased germinal center formation in the absence of RORgammat. FASEB J. 2016;30(2):761-74. doi: 10.1096/fj.15 -274001. PubMed PMID: 26499265.
36. Davidsen J, Rosenkrands I, Christensen D, Vangala A, Kirby D, Perrie Y, Agger EM, Andersen P. Characterization of cationic liposomes based on dimethyldioctadecylammonium and synthetic cord factor from M. tuberculosis (trehalose 6,6'-dibehenate)-a novel adjuvant inducing both strong CMI and antibody responses. Biochim Biophys Acta. 2005; 1718(1 -2):22-31. Epub 2005/12/03. doi: 10.1016/j.bbamem.2005.10.011. PubMed PMID: 16321607.
37. Martinez-Lopez M, Iborra S, Conde-Garrosa R, Mastrangelo A, Danne C, Mann ER, Reid DM, Gaboriau-Routhiau V, Chaparro M, Lorenzo MP, Minnerup L, Saz-Leal P, Slack E, Kemp B, Gisbert JP, Dzionek A, Robinson MJ, Ruperez FJ, Cerf-Bensussan N,
Brown GD, Bernardo D, LeibundGut-Landmann S, Sancho D. Microbiota Sensing by Mincle-Syk Axis in Dendritic Cells Regulates Interleukin- 17 and -22 Production and Promotes Intestinal Barrier Integrity. Immunity. 2019;50(2):446-61 e9. Epub 2019/02/03. doi: 10.1016/j.immuni.2018.12.020. PubMed PMID: 30709742; PMCID: PMC6382412.
38. Weminghaus K, Babiak A, Gross O, Holscher C, Dietrich H, Agger EM, Mages J, Mocsai A, Schoenen H, Finger K, Nimmerjahn F, Brown GD, Kirschning C, Heit A, Andersen P, Wagner H, Ruland J, Lang R. Adjuvanticity of a synthetic cord factor analogue for subunit Mycobacterium tuberculosis vaccination requires FcRgamma-Syk-Card9- dependent innate immune activation. J Exp Med. 2009;206(l):89-97. Epub 2009/01/14. doi: 10.1084/jem.20081445. PubMed PMID: 19139169; PMCID: PMC2626670.
39. Knudsen NP, Olsen A, Buonsanti C, Follmann F, Zhang Y, Coler RN, Fox CB, Meinke A, D'Oro U, Casini D, Bond A, Billeskov R, De Gregorio E, Rappuoli R, Harandi AM, Andersen P, Agger EM. Different human vaccine adjuvants promote distinct antigen- independent immunological signatures tailored to different pathogens. Sci Rep. 2016;6: 19570. Epub 2016/01/23. doi: 10.1038/srep 19570. PubMed PMID: 26791076; PMCID: PMC4726129.
40. Henriksen-Lacey M, Christensen D, Bramwell VW, Lindenstrom T, Agger EM,
Andersen P, Perrie Y. Comparison of the depot effect and immunogenicity of liposomes based on dimethyldioctadecylammonium (DDA), 3beta-[N-(N',N'-
Di methyl ami noethanejcarbomyl] cholesterol (DC-Chol), and l,2-Dioleoyl-3- trimethylammonium propane (DOTAP): prolonged liposome retention mediates stronger Thl responses. Mol Pharm. 2011;8(1): 153-61. Epub 2010/12/02. doi: 10.1021/mpl00208f. PubMed PMID: 21117621.
41. Emeholm K, Lorenzen E, Boje S, Olsen AW, Jungersen G, Jensen HE, Cassidy JP, Andersen P, Agerholm JS, Follmann F. Genital Infiltrations of CD4(+) and CD8(+) T Lymphocytes, IgA(+) and IgG(+) Plasma Cells and Intra-Mucosal Lymphoid Follicles Associate With Protection Against Genital Chlamydia trachomatis Infection in Minipigs Intramuscularly Immunized With UV-Inactivated Bacteria Adjuvanted With CAFOl. Front Microbiol. 2019;10: 197. Epub 2019/02/26. doi: 10.3389/fmicb.2019.00197. PubMed PMID: 30800114; PMCID: PMC6375829.
42. Jones AT, Shen X, Walter KL, LaBranche CC, Wyatt LS, Tomaras GD, Montefiori DC, Moss B, Barouch DH, Clements JD, Kozlowski PA, Varadarajan R, Amara RR. HIV-1
vaccination by needle-free oral injection induces strong mucosal immunity and protects against SHIV challenge. Nat Commun. 2019; 10(1):798. Epub 2019/02/20. doi: 10.1038/s41467-019-08739-4. PubMed PMID: 30778066; PMCID: PMC6379385.
43. Woodworth JS, Christensen D, Cassidy JP, Agger EM, Mortensen R, Andersen P. Mucosal boosting of H56:CAF01 immunization promotes lung-localized T cells and an accelerated pulmonary response to Mycobacterium tuberculosis infection without enhancing vaccine protection. Mucosal Immunol. 2019. Epub 2019/02/15. doi: 10.1038/s41385-019- 0145-5. PubMed PMID: 30760832.
44. Agger EM, Rosenkrands I, Hansen J, Brahimi K, Vandahl BS, Aagaard C, Werninghaus K, Kirschning C, Lang R, Christensen D, Theisen M, Follmann F, Andersen P. Cationic liposomes formulated with synthetic mycobacterial cordfactor (CAFOl): a versatile adjuvant for vaccines with different immunological requirements. PLoS One. 2008;3(9):e3116. Epub 2008/09/09. doi: 10.1371/joumal.pone.0003116. PubMed PMID: 18776936; PMCID: PMC2525815.
45. Uhlen M, Bandrowski A, Carr S, Edwards A, Ellenberg J, Lundberg E, Rimm DL, Rodriguez H, Hiltke T, Snyder M, Yamamoto T. A proposal for validation of antibodies. Nat Methods. 2016; 13(10):823-7. Epub 2016/09/07. doi: 10.1038/nmeth.3995. PubMed PMID: 27595404.
46. Fischinger S, Boudreau CM, Butler AL, Streeck H, Alter G. Sex differences in vaccine-induced humoral immunity. Semin Immunopathol. 2019;41(2):239-49. Epub 2018/12/14. doi: 10.1007/s00281-018-0726-5. PubMed PMID: 30547182; PMCID: PMC6373179.
47. Walker ML, Gordon TP, Wilson ME. Menstrual cycle characteristics of seasonally breeding rhesus monkeys. Biol Reprod. 1983 ;29(4): 841-8. Epub 1983/11/01. PubMed PMID: 6416317.
48. Carias AM, Allen SA, Fought AJ, Kotnik Halavaty K, Anderson MR, Jimenez ML, McRaven MD, Gioia CJ, Henning TR, Kersh EN, Smith JM, Pereira LE, Butler K, McNicholl SJ, Hendry RM, Kiser PF, Veazey RS, Hope TJ. Increases in Endogenous or Exogenous Progestins Promote Virus-Target Cell Interactions within the Non-human Primate Female Reproductive Tract. PLoS pathogens. 2016;12(9):el005885. doi: 10.1371/journal.ppat.1005885. PubMed PMID: 27658293; PMCID: PMC5033389.
49. Michel KG, Huijbregts RP, Gleason JL, Richter HE, Hel Z. Effect of hormonal contraception on the function of plasmacytoid dendritic cells and distribution of immune cell populations in the female reproductive tract. J Acquir Immune Defic Syndr. 2015;68(5):511- 8. Epub 2015/03/13. doi: 10.1097/QAI.0000000000000531. PubMed PMID: 25763784; PMC ID: PMC4874780.
50. Heffron R, Stalter R, Pyra M, Nanda K, Erikson DW, Hladik F, Blue SW, Davis NL, Mugo N, Kourtis AP, Lingappa JR, Baeten JM, Partners Pr EPS, Partners in Prevention HSVHIVTST. HIV risk associated with serum medroxyprogesterone acetate levels among women in East and southern Africa. AIDS 2019;33(4):735-44. Epub 2018/12/27. doi: 10.1097/QAD.0000000000002123. PubMed PMID: 30585845; PMCID: PMC6399047.
51. Huijbregts RP, Helton ES, Michel KG, Sabbaj S, Richter HE, Goepfert PA, Hel Z. Hormonal contraception and HIV-1 infection: medroxyprogesterone acetate suppresses innate and adaptive immune mechanisms. Endocrinology. 2013 ; 154(3): 1282-95. Epub 2013/01/29. doi: 10.1210/en.2012-1850. PubMed PMID: 23354099; PMCID: PMC3578997.
52. Hensley-McBain T, Wu MC, Manuzak JA, Cheu RK, Gustin A, Driscoll CB, Zevin AS, Miller CJ, Coronado E, Smith E, Chang J, Gale M, Jr., Somsouk M, Burgener AD, Hunt PW, Hope TJ, Collier AC, Klatt NR. Increased mucosal neutrophil survival is associated with altered microbiota in HIV infection. PLoS Pathog. 2019;15(4):el007672. Epub 2019/04/12. doi: 10.1371/joumal.ppat.1007672. PubMed PMID: 30973942.
53. Klatt NR, Manuzak JA. Microbial Effects on Immunity in HIV: Virus, Gender or Sexual Preference Induced? EBioMedicine. 2018;31 :7-8. Epub 2018/04/16. doi: 10.1016/j.ebiom.2018.04.008. PubMed PMID: 29655995; PMCID: PMC6014573.
54. Asbach B, Kibler KV, Kostler J, Perdiguero B, Yates NL, Stanfield-Oakley S, Tomaras GD, Kao SF, Foulds KE, Roederer M, Seaman MS, Montefiori DC, Parks R, Ferrari G, Forthal DN, Phogat S, Tartaglia J, Barnett SW, Self SG, Gottardo R, Cristillo AD, Weiss DE, Galmin L, Ding S, Heeney JL, Esteban M, Jacobs BL, Pantaleo G, Wagner R. Priming with a Potent HIV-1 DNA Vaccine Frames the Quality of Immune Responses prior to a Poxvirus and Protein Boost. J Virol. 2019;93(3). Epub 2018/11/16. doi: 10.1128/JVI.01529- 18. PubMed PMID: 30429343; PMCID: PMC6340047.
55. Kibler KV, Asbach B, Perdiguero B, Garcia- Arriaza J, Yates NL, Parks R, Stanfield-Oakley S, Ferrari G, Montefiori DC, Tomaras GD, Roederer M, Foulds KE, Forthal
DN, Seaman MS, Self S, Gottardo R, Phogat S, Tartaglia J, Barnett S, Cristillo AD, Weiss D, Galmin L, Ding S, Heeney JL, Esteban M, Wagner R, Pantaleo G, Jacobs BL. Replication- Competent NYVAC-KC Yields Improved Immunogenicity to HIV-1 Antigens in Rhesus Macaques Compared to Nonreplicating NYVAC. J Virol. 2019;93(3). Epub 2018/11/16. doi: 10.1128/JVI.01513-18. PubMed PMID: 30429340; PMCID: PMC6340019.
56. Ake JA, Schuetz A, Pegu P, Wieczorek L, Eller MA, Kibuuka H, Sawe F, Maboko L, Polonis V, Karasavva N, Weiner D, Sekiziyivu A, Kosgei J, Missanga M, Kroidl A, Mann P, Ratto-Kim S, Anne Eller L, Earl P, Moss B, Dorsey-Spitz J, Milazzo M, Laissa Ouedraogo G, Rizvi F, Yan J, Khan AS, Peel S, Sardesai NY, Michael NL, Ngauy V, Marovich M, Robb ML. Safety and Immunogenicity of PENNVAX-G DNA Prime Administered by Biojector 2000 or CELLECTRA Electroporation Device With Modified Vaccinia Ankara-CMDR Boost. J Infect Dis. 2017;216(9): 1080-90. Epub 2017/10/03. doi: 10.1093/infdis/jix456. PubMed PMID: 28968759; PMCID: PMC5853809.
57. Groom JR, Richmond J, Murooka TT, Sorensen EW, Sung JH, Bankert K, von Andrian UH, Moon JJ, Mempel TR, Luster AD. CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation. Immunity. 2012;37(6): 1091-103. Epub 2012/11/06. doi: 10.1016/j.immuni.2012.08.016. PubMed PMID: 23123063; PMCID: PMC3525757.
58. Groom JR, Luster AD. CXCR3 in T cell function. Exp Cell Res. 2011;317(5):620- 31. Epub 2011/03/08. doi: 10.1016/j.yexcr.2010.12.017. PubMed PMID: 21376175; PMCID: PMC3065205.
59. Mohty AM, Grob JJ, Mohty M, Richard MA, Olive D, Gaugler B. Induction of IP- 10/CXCL10 secretion as an immunomodulatory effect of low-dose adjuvant interferon-alpha during treatment of melanoma. Immunobiology. 2010;215(2): 113-23. Epub 2009/05/20. doi: 10.1016/j.imbio.2009.03.008. PubMed PMID: 19450896.
60. Ben-Sasson SZ, Hu-Li J, Quiel J, Cauchetaux S, Ratner M, Shapira I, Dinarello CA, Paul WE. IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation. Proc Natl Acad Sci U S A. 2009;106(17):7119-24. Epub 2009/04/11. doi: 10.1073/pnas.0902745106. PubMed PMID: 19359475; PMCID: PMC2678417.
61. Ben-Sasson SZ, Wang K, Cohen J, Paul WE. IL-lbeta strikingly enhances antigen- driven CD4 and CD8 T-cell responses. Cold Spring Harb Symp Quant Biol. 2013;78: 117-24. Epub 2013/10/05. doi: 10.1101/sqb.2013.78.021246. PubMed PMID: 24092469.
62. Dan JM, Lindestam Arlehamn CS, Weiskopf D, da Silva Antunes R, Havenar- Daughton C, Reiss SM, Brigger M, Bothwell M, Sette A, Crotty S. A Cytokine-Independent Approach To Identify Antigen- Specific Human Germinal Center T Follicular Helper Cells and Rare Antigen- Specific CD4+ T Cells in Blood. J Immunol. 2016;197(3):983-93. Epub 2016/06/28. doi: 10.4049/jimmunol.1600318. PubMed PMID: 27342848; PMCID: PMC4955771.
63. Reiss S, Baxter AE, Cirelli KM, Dan JM, Morou A, Daigneault A, Brassard N, Silvestri G, Routy JP, Havenar-Daughton C, Crotty S, Kaufmann DE. Comparative analysis of activation induced marker (AIM) assays for sensitive identification of antigen-specific CD4 T cells. PLoS One. 2017;12(10):e0186998. Epub 2017/10/25. doi: 10.1371/journal. pone.0186998. PubMed PMID: 29065175; PMCID: PMC5655442.
64. Petitdemange C, Kasturi SP, Kozlowski PA, Nabi R, Quamstrom CF, Reddy PBJ, Derdeyn CA, Spicer LM, Patel P, Legere T, Koval enkov YO, Labranche CC, Villinger F, Tomai M, Vasilakos J, Haynes B, Kang CY, Gibbs JS, Yewdell JW, Barouch D, Wrammert J, Montefiori D, Hunter E, Amara RR, Masopust D, Pulendran B. Vaccine induction of antibodies and tissue-resident CD8+ T cells enhances protection against mucosal SHIV infection in young macaques. JCI Insight. 2019;4(4). Epub 2019/03/05. doi: 10.1172/jci.insight.126047. PubMed PMID: 30830870; PMCID: PMC6478416.
65. Rosenberg YJ, Lewis GK, Montefiori DC, LaBranche CC, Lewis MG, Urban LA, Lees JP, Mao L, Jiang X. Introduction of the YTE mutation into the non-immunogenic HIV bnAb PGT121 induces anti-drug antibodies in macaques. PLoS One. 2019;14(2):e0212649. Epub 2019/02/21. doi: 10.1371/joumal.pone.0212649. PubMed PMID: 30785963; PMCID: PMC6437720.
66. Wen Y, Trinh HV, Linton CE, Tani C, Norais N, Martinez-Guzman D, Ramesh P, Sun Y, Situ F, Karaca-Griffm S, Hamlin C, Onkar S, Tian S, Hilt S, Malyala P, Lodaya R, Li N, Otten G, Palladino G, Friedrich K, Aggarwal Y, LaBranche C, Duffy R, Shen X, Tomaras GD, Montefiori DC, Fulp W, Gottardo R, Burke B, Ulmer JB, Zolla-Pazner S, Liao HX, Haynes BF, Michael NL, Kim JH, Rao M, O'Connell RJ, Carfi A, Barnett SW. Generation and characterization of a bivalent protein boost for future clinical trials: HIV-1 subtypes
CROI AE and B gpl20 antigens with a potent adjuvant. PLoS One. 2018;13(4):e0194266. Epub 2018/04/27. doi: 10.1371/joumal.pone.0194266. PubMed PMID: 29698406; PMCID: PMC5919662.
67. Forthal DN, Finzi A. Antibody-dependent cellular cytotoxicity in HIV infection. AIDS. 2018;32(17):2439-51. Epub 2018/09/21. doi: 10.1097/QAD.0000000000002011. PubMed PMID: 30234611.
68. Kadelka C, Liechti T, Ebner H, Schanz M, Rusert P, Friedrich N, Stiegeler E, Braun DL, Huber M, Scherrer AU, Weber J, Uhr T, Kuster H, Misselwitz B, Cavassini M, Bemasconi E, Hoffmann M, Calmy A, Battegay M, Rauch A, Yerly S, Aubert V, Klimkait T, Boni J, Kouyos RD, Gunthard HF, Trkola A, Swiss HIVCS. Distinct, IgGl-driven antibody response landscapes demarcate individuals with broadly HIV-1 neutralizing activity. J Exp Med. 2018;215(6): 1589-608. Epub 2018/05/26. doi: 10.1084/jem.20180246. PubMed PMID: 29794117; PMCID: PMC5987927.
69. Pauthner MG, Nkolola JP, Havenar-Daughton C, Murrell B, Reiss SM, Bastidas R, Prevost J, Nedellec R, von Bredow B, Abbink P, Cottrell CA, Kulp DW, Tokatlian T, Nogal B, Bianchi M, Li H, Lee JH, Butera ST, Evans DT, Hangartner L, Finzi A, Wilson IA, Wyatt RT, Irvine DJ, Schief WR, Ward AB, Sanders RW, Crotty S, Shaw GM, Barouch DH, Burton DR. Vaccine-Induced Protection from Homologous Tier 2 SHIV Challenge in Nonhuman Primates Depends on Serum-Neutralizing Antibody Titers. Immunity. 2019;50(l):241-52 e6. Epub 2018/12/16. doi: 10.1016/j.immuni.2018.11.011. PubMed PMID: 30552025; PMCID: PMC6335502.
70. Ackerman ME, Das J, Pittala S, Broge T, Linde C, Suscovich TJ, Brown EP, Bradley T, Natarajan H, Lin S, Sassic JK, O'Keefe S, Mehta N, Goodman D, Sips M, Weiner JA, Tomaras GD, Haynes BF, Lauffenburger DA, Bailey-Kellogg C, Roederer M, Alter G. Route of immunization defines multiple mechanisms of vaccine mediated protection against SIV. Nat Med. 2018;24(10): 1590-8. Epub 2018/09/05. doi: 10.1038/s41591 -018-0161 -0. PubMed PMID: 30177821; PMCID: PMC6482471.
71. Sui Y, Lewis GK, Wang Y, Berckmueller K, Frey B, Dzutsev A, Vargas- Inchaustegui D, Mohanram V, Musich T, Shen X, DeVico A, Fouts T, Venzon D, Kirk J, Waters RC, Talton J, Klinman D, Clements J, Tomaras GD, Franchini G, Robert-Guroff M, Trinchieri G, Gallo RC, Berzofsky JA. Mucosal vaccine efficacy against intrarectal SHIV is
independent of anti-Env antibody response. J Clin Invest. 2019; 129(3): 1314-28. Epub 2019/02/19. doi: 10.1172/JCI122110. PubMed PMID: 30776026; PMCID: PMC6391089.
72. Brody IB, Calcedo R, Connell MJ, Carnathan DG, Nason M, Lawson BO, Nega MT, Boyd S, Qin Q, Vanderford TH, Wilson JM, Wilson JM, Silvestri G, Betts MR. Susceptibility to SIV Infection After Adenoviral Vaccination in a Low Dose Rhesus Macaque Challenge Model. Pathog Immun. 2019;4(l): l-20. Epub 2019/04/18. doi: 10.2041 l/pai.v4i 1.241. PubMed PMID: 30993250; PMCID: PMC6457171.
73. Lai SH, Starke CE, Flynn JK, Vinton CL, Ortiz AM, Mudd JC, Brenchley JM. Simian Immunodeficiency Virus Infects Functionally Polarized Memory CD4 T Cells Equivalently In Vivo. J Virol. 2019;93(9). doi: 10.1128/JVI.02163-18. PubMed PMID: 30787150.
74. Christensen-Quick A, Lafferty M, Sun L, Marchionni L, DeVico A, Garzino-Demo A. Human Thl7 Cells Lack HIV-Inhibitory RNases and Are Highly Permissive to Productive HIV Infection. J Virol. 2016;90(17):7833-47. doi: 10.1128/JVI.02869-15. PubMed PMID: 27334595; PMCID: PMC4988157.
75. Stieh DJ, Matias E, Xu H, Fought AJ, Blanchard JL, Marx PA, Veazey RS, Hope TJ. Thl7 Cells Are Preferentially Infected Very Early after Vaginal Transmission of SIV in Macaques. Cell Host Microbe. 2016; 19(4): 529-40. Epub 2016/04/15. doi: 10.1016/j.chom.2016.03.005. PubMed PMID: 27078070; PMCID: PMC4841252.
5. Exemplary Embodiments
[0238] Exemplary embodiments provided in accordance with the presently disclosed subject matter include, but are not limited to, the claims and the following embodiments:
1. A method of enhancing the anti-Env antibody response during HIV vaccination in a subject, the method comprising:
(i) administering a first Thl -polarizing adjuvant to the subject together with a DNA vaccine comprising a polynucleotide encoding a first HIV Env polypeptide, wherein the first adjuvant comprises a polynucleotide encoding interferon-induced protein (IP)-10; and
(ii) administering a second Thl -polarizing adjuvant to the subject together with a booster comprising a second HIV Env polypeptide, wherein the second adjuvant comprises QS-21.
2. The method of embodiment 1, wherein the second HIV Env polypeptide is a gpl40 polypeptide.
3. The method of embodiment 1 or 2, wherein the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
4. The method of any one of embodiments 1 to 3, wherein the first adjuvant and the DNA vaccine are administered transdermally with electroporation.
5. The method of any one of embodiments 1 to 4, wherein the first HIV Env polypeptide, second HIV Env polypeptide, and/or one or more additional HIV polypeptides are from HIV-T
6. The method of embodiment 5, wherein the HIV-1 is of clade C origin.
7. The method of any one of embodiments 1 to 6, wherein the booster further comprises a lipid and/or liposomal adjuvant.
8. The method of embodiment 7, wherein the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
9. The method of embodiment 7 or 8, wherein the lipid adjuvant comprises monophosphoryl lipid A (MPLA).
10. The method of any one of embodiments 1 to 9, wherein the first Thl- polarizing adjuvant and DNA vaccine are administered to the subject 1, 2 or 3 times prior to the administration of the booster.
11. The method of embodiment 10, wherein the first Thl -polarizing adjuvant and DNA vaccine are administered to the subject at 0, 8, and 16 weeks.
12. The method of any one of embodiments 1 to 11, wherein the booster is administered to the subject 1 or 2 times.
13. The method of any one of embodiments 1 to 12, wherein peripheral and germinal center (GC) Tfh cells isolated from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster display higher proportions of anti-Env Thl cells than do peripheral and germinal center (GC) Tfh cells taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
14. The method of any one of embodiments 1 to 13, wherein blood taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster shows higher anti-gpl40 extrafollicular and/or plasma cell-derived titers than does blood from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
15. The method of any one of embodiments 1 to 14, wherein serum IgG antibodies taken from the subject subsequent to the administration of the second Thl- polarizing adjuvant and booster show a broader cross-clade anti-Env response and/or increased specificity for gpl20 V1V2 loops than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
16. The method of any one of embodiments 1 to 15, wherein serum IgG antibodies taken from the subject subsequent to the administration of the second Thl- polarizing adjuvant and booster display elevated anti-Env titers that persist longer than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
17. The method of any one of embodiments 1 to 16, wherein serum IgG antibodies taken from the subject subsequent to the administration of the second Thl- polarizing adjuvant and booster show higher avidity against gpl40 than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
18. The method of any one of embodiments 1 to 17, wherein serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater neutralization activity against HIV-1 than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
19. The method of embodiment 18, wherein the neutralization of HIV- 1 is assessed using a TZM-bl assay.
20. The method of any one of embodiments 1 to 19, wherein serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater antibody-dependent cellular toxicity (ADCC) and/or antibody-dependent phagocytosis (ADP) activity against HIV-infected cells than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
21. The method of embodiment 20, wherein the HIV-infected cells are infected with HIV-1.
22. The method of embodiment 21, wherein the HIV-1 is of clade C origin.
23. The method of any one of embodiments 1 to 22, wherein IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show higher anti-gpl40 titers than do IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
24. A pharmaceutical composition for vaccinating a subject against HIV, the composition comprising a DNA vaccine comprising a polynucleotide encoding an HIV Env polypeptide, a polynucleotide encoding interferon-induced protein (IP)- 10, and a pharmaceutically acceptable carrier.
25. The pharmaceutical composition of embodiment 24, wherein the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
26. The pharmaceutical composition of embodiment 24 or 25, wherein the HIV Env polypeptide and/or one or more additional HIV polypeptides are from HIV-1.
27. The pharmaceutical composition of embodiment 26, wherein the HIV- 1 is of clade C origin.
28. The pharmaceutical composition of any one of embodiments 24 to 27, wherein the polynucleotide encoding the HIV Env polypeptide and the polynucleotide encoding IP- 10 are present within a single DNA vector.
29. The pharmaceutical composition of any one of embodiments 24 to 28, wherein the composition is formulated for transdermal delivery with electroporation.
30. A pharmaceutical composition for boosting vaccination against HIV, the composition comprising an HIV Env polypeptide, QS-21, and a pharmaceutically acceptable carrier.
31. The pharmaceutical composition of embodiment 30, wherein the HIV Env polypeptide is a gpl40 polypeptide.
32. The pharmaceutical composition of embodiment 30 or 31, wherein the composition further comprises a lipid and/or liposomal adjuvant.
33. The pharmaceutical composition of embodiment 32, wherein the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
34. The pharmaceutical composition of embodiment 32 or 33, wherein the lipid adjuvant comprises monophosphoryl lipid A (MPLA).
35. The pharmaceutical composition of any one of embodiments 30 to 34, wherein the HIV Env polypeptide is from HIV-1.
36. The pharmaceutical composition of embodiment 35, wherein the HIV- 1 is of clade C origin.
[0239] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, one of skill in the art will appreciate that certain changes and modifications may be practiced within the scope of the appended claims. In addition, each reference provided herein is incorporated by reference in its entirety to the same extent as if each reference was individually incorporated by reference.
Claims
1. A method of enhancing the anti-Env antibody response during HIV vaccination in a subject, the method comprising:
(i) administering a first Thl -polarizing adjuvant to the subject together with a DNA vaccine comprising a polynucleotide encoding a first HIV Env polypeptide, wherein the first adjuvant comprises a polynucleotide encoding interferon-induced protein (IP)-10; and
(ii) administering a second Thl -polarizing adjuvant to the subject together with a booster comprising a second HIV Env polypeptide, wherein the second adjuvant comprises QS-21.
2. The method of claim 1, wherein the second HIV Env polypeptide is a gpl40 polypeptide.
3. The method of claim 1, wherein the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
4. The method of claim 1, wherein the first adjuvant and the DNA vaccine are administered transdermally with electroporation.
5. The method of claim 1 , wherein the first HIV Env polypeptide, second HIV Env polypeptide, and/or one or more additional HIV polypeptides are from HIV-1.
6. The method of claim 5, wherein the HIV-1 is of clade C origin.
7. The method of claim 1, wherein the booster further comprises a lipid and/or liposomal adjuvant.
8. The method of claim 7, wherein the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
9. The method of claim 7, wherein the lipid adjuvant comprises monophosphoryl lipid A (MPLA).
10. The method of claim 1, wherein the first Thl -polarizing adjuvant and DNA vaccine are administered to the subject 1, 2 or 3 times prior to the administration of the booster.
11. The method of claim 10, wherein the first Thl -polarizing adjuvant and DNA vaccine are administered to the subject at 0, 8, and 16 weeks.
12. The method of claim 1, wherein the booster is administered to the subject 1 or 2 times.
13. The method of claim 1, wherein peripheral and germinal center (GC) Tfh cells isolated from the subject subsequent to the administration of the second Thl- polarizing adjuvant and booster display higher proportions of anti-Env Thl cells than do peripheral and germinal center (GC) Tfh cells taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
14. The method of claim 1, wherein blood taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster shows higher anti-gpl40 extrafollicular and/or plasma cell-derived titers than does blood from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
15. The method of claim 1, wherein serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show a broader cross-clade anti-Env response and/or increased specificity for gpl20 V1V2 loops than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
16. The method of claim 1, wherein serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster display elevated anti-Env titers that persist longer than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
17. The method of claim 1, wherein serum IgG antibodies taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show higher avidity against gpl40 than do serum IgG antibodies taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
18. The method of claim 1, wherein serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater neutralization activity against HIV-1 than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
19. The method of claim 18, wherein the neutralization of HIV-1 is assessed using a TZM-bl assay.
20. The method of claim 1, wherein serum taken from the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster have greater antibody-dependent cellular toxicity (ADCC) and/or antibody-dependent phagocytosis (ADP) activity against HIV-infected cells than does serum taken from a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl -polarizing adjuvant.
21. The method of claim 20, wherein the HIV-infected cells are infected with HIV- 1.
22. The method of claim 21, wherein the HIV-1 is of clade C origin.
23. The method of claim 1, wherein IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of the subject subsequent to the administration of the second Thl -polarizing adjuvant and booster show higher anti-gpl40 titers than do IgG and/or IgA antibodies isolated from the rectal and/or vaginal mucosa of a second subject that has received the DNA vaccine and booster but that has not received the first and/or second Thl- polarizing adjuvant.
24. A pharmaceutical composition for vaccinating a subject against HIV, the composition comprising a DNA vaccine comprising a polynucleotide encoding an HIV
Env polypeptide, a polynucleotide encoding interferon-induced protein (IP)- 10, and a pharmaceutically acceptable carrier.
25. The pharmaceutical composition of claim 24, wherein the DNA vaccine further comprises one or more additional polynucleotides encoding one or more additional HIV polypeptides selected from the group consisting of Gag, protease, reverse transcriptase, Tat, Rev, and combinations thereof.
26. The pharmaceutical composition of claim 24, wherein the HIV Env polypeptide and/or one or more additional HIV polypeptides are from HIV-1.
27. The pharmaceutical composition of claim 26, wherein the HIV-1 is of clade C origin.
28. The pharmaceutical composition of claim 24, wherein the polynucleotide encoding the HIV Env polypeptide and the polynucleotide encoding IP- 10 are present within a single DNA vector.
29. The pharmaceutical composition of claim 24, wherein the composition is formulated for transdermal delivery with electroporation.
30. A pharmaceutical composition for boosting vaccination against HIV, the composition comprising an HIV Env polypeptide, QS-21, and a pharmaceutically acceptable carrier.
31. The pharmaceutical composition of claim 30, wherein the HIV Env polypeptide is a gpl40 polypeptide.
32. The pharmaceutical composition of claim 30, wherein the composition further comprises a lipid and/or liposomal adjuvant.
33. The pharmaceutical composition of claim 32, wherein the liposomal adjuvant comprises Army Liposome Formulation (ALF) liposomes.
34. The pharmaceutical composition of claim 32, wherein the lipid adjuvant comprises monophosphoryl lipid A (MPLA).
35. The pharmaceutical composition of claim 30, wherein the HIV Env polypeptide is from HIV-1.
36. The pharmaceutical composition of claim 35, wherein the HIV-1 is of clade C origin.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201962853543P | 2019-05-28 | 2019-05-28 | |
US62/853,543 | 2019-05-28 | ||
US201962927607P | 2019-10-29 | 2019-10-29 | |
US62/927,607 | 2019-10-29 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2020243166A1 true WO2020243166A1 (en) | 2020-12-03 |
Family
ID=73552941
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2020/034688 WO2020243166A1 (en) | 2019-05-28 | 2020-05-27 | Th1-polarizing adjuvants for enhancing immunogenicity of hiv antigens |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2020243166A1 (en) |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20050089526A1 (en) * | 2001-09-06 | 2005-04-28 | Moore John P. | Human immunodeficiency virus envelope clycoprotein mutants and uses thereof |
US20080199492A1 (en) * | 2004-06-08 | 2008-08-21 | Novartis Vaccines And Diagnostics Inc. | Env Polypeptide Complexes and Methods Of Use |
US20120177597A1 (en) * | 2011-01-12 | 2012-07-12 | Baylor Research Institute | Compositions and methods of alteration of autoimmune diseases |
US20160032245A1 (en) * | 2012-11-09 | 2016-02-04 | President And Fellows Of Harvard College | Compositions and Methods for Modulating an Immune Response |
US20170182152A1 (en) * | 2014-03-25 | 2017-06-29 | The Government Of The United States Of America As Represented By The Secretary Of The Army | Non-toxic adjuvant formulation comprising a monophosphoryl lipid a (mpla)-containing liposome composition and a saponin |
-
2020
- 2020-05-27 WO PCT/US2020/034688 patent/WO2020243166A1/en active Application Filing
Patent Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20050089526A1 (en) * | 2001-09-06 | 2005-04-28 | Moore John P. | Human immunodeficiency virus envelope clycoprotein mutants and uses thereof |
US20080199492A1 (en) * | 2004-06-08 | 2008-08-21 | Novartis Vaccines And Diagnostics Inc. | Env Polypeptide Complexes and Methods Of Use |
US20120177597A1 (en) * | 2011-01-12 | 2012-07-12 | Baylor Research Institute | Compositions and methods of alteration of autoimmune diseases |
US20160032245A1 (en) * | 2012-11-09 | 2016-02-04 | President And Fellows Of Harvard College | Compositions and Methods for Modulating an Immune Response |
US20170182152A1 (en) * | 2014-03-25 | 2017-06-29 | The Government Of The United States Of America As Represented By The Secretary Of The Army | Non-toxic adjuvant formulation comprising a monophosphoryl lipid a (mpla)-containing liposome composition and a saponin |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Verma et al. | Impact of Th1 CD4 follicular helper T cell skewing on antibody responses to an HIV-1 vaccine in rhesus macaques | |
Lai et al. | Prevention of infection by a granulocyte-macrophage colony-stimulating factor co-expressing DNA/modified vaccinia Ankara simian immunodeficiency virus vaccine | |
US12090200B2 (en) | Methods of producing cells resistant to HIV infection | |
US10036038B2 (en) | HIV pre-immunization and immunotherapy | |
Belyakov et al. | Impact of vaccine-induced mucosal high-avidity CD8+ CTLs in delay of AIDS viral dissemination from mucosa | |
US20240117017A1 (en) | Hiv vaccines and methods of making and using | |
US12053517B2 (en) | HIV vaccines and methods of using | |
Robinson et al. | Studies on GM-CSF DNA as an adjuvant for neutralizing Ab elicited by a DNA/MVA immunodeficiency virus vaccine | |
Styles et al. | Human immunodeficiency virus C. 1086 envelope gp140 protein boosts following DNA/modified vaccinia virus Ankara vaccination fail to enhance heterologous anti-V1V2 antibody response and protection against clade C simian-human immunodeficiency virus challenge | |
Zhan et al. | Multi-envelope HIV-1 vaccine devoid of SIV components controls disease in macaques challenged with heterologous pathogenic SHIV | |
EP2021356B1 (en) | Hiv vaccine | |
Lakhashe et al. | Multimodality vaccination against clade C SHIV: partial protection against mucosal challenges with a heterologous tier 2 virus | |
WO2020243166A1 (en) | Th1-polarizing adjuvants for enhancing immunogenicity of hiv antigens | |
Tramont et al. | Progress in the development of an HIV vaccine | |
Ruffin et al. | Rational design of HIV vaccines and microbicides: report of the EUROPRISE annual conference 2011 | |
Brinckmann et al. | Rational design of HIV vaccines and microbicides: report of the EUROPRISE network annual conference 2010 | |
Moskovljevic | INVESTIGATING HIV-1 LATENCY REVERSAL UPON COGNATE ANTIGEN RECOGNITION IN CD4+ T CELLS FROM PEOPLE ON ANTIRETROVIRAL THERAPY | |
Ruffin et al. | Journal of Translational Medicine | |
AU2015236147A1 (en) | Mosaic HIV-1 sequences and uses thereof | |
WO2024015741A1 (en) | Hiv immunogenic polypeptides and vaccines and uses thereof | |
Basu et al. | Strong, but Age-Dependent, Protection Elicited by a DNA/Modified Vaccinia Ankara Simian Immunodeficiency Virus Vaccine | |
Işık | HIV-1 envelope glycoproteins with costimulatory domains for vaccine applications | |
Wanga et al. | Chapter-3: The enhanced antibody responses elicited by a CpG adjuvant does not improve the protective effect of an AT-2 inactivated SIV therapeutic AIDS vaccine | |
Brinckmann et al. | King’s Research Portal |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 20813561 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 20813561 Country of ref document: EP Kind code of ref document: A1 |