WO2024040025A2 - Th2 vaccine-based prevention and treatment of inflammation in obesity - Google Patents
Th2 vaccine-based prevention and treatment of inflammation in obesity Download PDFInfo
- Publication number
- WO2024040025A2 WO2024040025A2 PCT/US2023/072164 US2023072164W WO2024040025A2 WO 2024040025 A2 WO2024040025 A2 WO 2024040025A2 US 2023072164 W US2023072164 W US 2023072164W WO 2024040025 A2 WO2024040025 A2 WO 2024040025A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- amino acids
- vector
- nucleic acid
- mice
- polypeptide
- Prior art date
Links
- 208000008589 Obesity Diseases 0.000 title claims abstract description 40
- 235000020824 obesity Nutrition 0.000 title claims abstract description 40
- 206010061218 Inflammation Diseases 0.000 title claims description 17
- 230000004054 inflammatory process Effects 0.000 title claims description 17
- 229960005486 vaccine Drugs 0.000 title abstract description 24
- 238000011282 treatment Methods 0.000 title abstract description 7
- 230000002265 prevention Effects 0.000 title abstract description 6
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 49
- 239000000203 mixture Substances 0.000 claims abstract description 39
- 238000000034 method Methods 0.000 claims abstract description 36
- 201000010099 disease Diseases 0.000 claims abstract description 29
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 29
- 206010006187 Breast cancer Diseases 0.000 claims abstract description 22
- 230000002503 metabolic effect Effects 0.000 claims abstract description 22
- 208000026310 Breast neoplasm Diseases 0.000 claims abstract description 20
- 201000011510 cancer Diseases 0.000 claims abstract description 18
- 208000008338 non-alcoholic fatty liver disease Diseases 0.000 claims abstract description 11
- 239000013598 vector Substances 0.000 claims description 106
- 150000001413 amino acids Chemical class 0.000 claims description 92
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 78
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 73
- 229920001184 polypeptide Polymers 0.000 claims description 62
- 150000007523 nucleic acids Chemical class 0.000 claims description 48
- 102000039446 nucleic acids Human genes 0.000 claims description 36
- 108020004707 nucleic acids Proteins 0.000 claims description 36
- 230000014509 gene expression Effects 0.000 claims description 33
- 239000013603 viral vector Substances 0.000 claims description 25
- 108020004999 messenger RNA Proteins 0.000 claims description 19
- 102100030431 Fatty acid-binding protein, adipocyte Human genes 0.000 claims description 18
- 101001062864 Homo sapiens Fatty acid-binding protein, adipocyte Proteins 0.000 claims description 18
- 102100039418 Plasminogen activator inhibitor 1 Human genes 0.000 claims description 18
- 101001129187 Homo sapiens Patatin-like phospholipase domain-containing protein 2 Proteins 0.000 claims description 17
- 102100031248 Patatin-like phospholipase domain-containing protein 2 Human genes 0.000 claims description 17
- 101000924017 Homo sapiens Dual specificity protein phosphatase 1 Proteins 0.000 claims description 16
- 101000881110 Homo sapiens Dual specificity protein phosphatase 12 Proteins 0.000 claims description 16
- 108010022233 Plasminogen Activator Inhibitor 1 Proteins 0.000 claims description 16
- 108020004414 DNA Proteins 0.000 claims description 15
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 15
- 239000002671 adjuvant Substances 0.000 claims description 13
- 239000013612 plasmid Substances 0.000 claims description 10
- 230000008488 polyadenylation Effects 0.000 claims description 6
- 206010018429 Glucose tolerance impaired Diseases 0.000 claims description 5
- 208000001280 Prediabetic State Diseases 0.000 claims description 5
- 201000009104 prediabetes syndrome Diseases 0.000 claims description 5
- 230000001105 regulatory effect Effects 0.000 claims description 4
- 239000003550 marker Substances 0.000 claims description 3
- 238000012545 processing Methods 0.000 claims description 3
- 230000002103 transcriptional effect Effects 0.000 claims description 3
- 230000029069 type 2 immune response Effects 0.000 claims description 3
- 102100034428 Dual specificity protein phosphatase 1 Human genes 0.000 claims 4
- 125000003275 alpha amino acid group Chemical group 0.000 claims 2
- 241000699670 Mus sp. Species 0.000 abstract description 85
- 108090000623 proteins and genes Proteins 0.000 abstract description 50
- 102000004169 proteins and genes Human genes 0.000 abstract description 42
- 210000000577 adipose tissue Anatomy 0.000 abstract description 33
- 210000001744 T-lymphocyte Anatomy 0.000 abstract description 28
- 210000001789 adipocyte Anatomy 0.000 abstract description 17
- 230000002757 inflammatory effect Effects 0.000 abstract description 16
- 235000005911 diet Nutrition 0.000 abstract description 9
- 230000037213 diet Effects 0.000 abstract description 9
- 230000002163 immunogen Effects 0.000 abstract description 4
- 229930006000 Sucrose Natural products 0.000 abstract description 3
- 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 abstract description 3
- 210000001596 intra-abdominal fat Anatomy 0.000 abstract description 3
- 239000005720 sucrose Substances 0.000 abstract description 3
- 108060008682 Tumor Necrosis Factor Proteins 0.000 abstract description 2
- 102100040247 Tumor necrosis factor Human genes 0.000 abstract description 2
- 210000004027 cell Anatomy 0.000 description 45
- 239000000427 antigen Substances 0.000 description 44
- 108091007433 antigens Proteins 0.000 description 40
- 102000036639 antigens Human genes 0.000 description 40
- 238000013116 obese mouse model Methods 0.000 description 30
- 102000040430 polynucleotide Human genes 0.000 description 25
- 108091033319 polynucleotide Proteins 0.000 description 25
- 239000002157 polynucleotide Substances 0.000 description 25
- 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 17
- 239000008103 glucose Substances 0.000 description 17
- 230000004044 response Effects 0.000 description 15
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 14
- YMWUJEATGCHHMB-UHFFFAOYSA-N Dichloromethane Chemical compound ClCCl YMWUJEATGCHHMB-UHFFFAOYSA-N 0.000 description 12
- 102100037573 Dual specificity protein phosphatase 12 Human genes 0.000 description 12
- 241000725303 Human immunodeficiency virus Species 0.000 description 11
- 241000699666 Mus <mouse, genus> Species 0.000 description 11
- 108091034117 Oligonucleotide Proteins 0.000 description 11
- 230000003053 immunization Effects 0.000 description 11
- 238000002649 immunization Methods 0.000 description 11
- 239000002207 metabolite Substances 0.000 description 11
- 230000001177 retroviral effect Effects 0.000 description 11
- 230000003612 virological effect Effects 0.000 description 11
- 239000003623 enhancer Substances 0.000 description 10
- 238000004806 packaging method and process Methods 0.000 description 10
- 210000001519 tissue Anatomy 0.000 description 10
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 10
- 238000002255 vaccination Methods 0.000 description 10
- 206010022489 Insulin Resistance Diseases 0.000 description 9
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 9
- 241000700605 Viruses Species 0.000 description 8
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 7
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 7
- 102000004877 Insulin Human genes 0.000 description 7
- 108090001061 Insulin Proteins 0.000 description 7
- 102100037850 Interferon gamma Human genes 0.000 description 7
- 108010074328 Interferon-gamma Proteins 0.000 description 7
- 238000004458 analytical method Methods 0.000 description 7
- 229940125396 insulin Drugs 0.000 description 7
- 150000002632 lipids Chemical class 0.000 description 7
- 241000702421 Dependoparvovirus Species 0.000 description 6
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 6
- 102000016267 Leptin Human genes 0.000 description 6
- 108010092277 Leptin Proteins 0.000 description 6
- 229940037003 alum Drugs 0.000 description 6
- 230000001413 cellular effect Effects 0.000 description 6
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 6
- 230000003247 decreasing effect Effects 0.000 description 6
- 239000012634 fragment Substances 0.000 description 6
- 238000009396 hybridization Methods 0.000 description 6
- NRYBAZVQPHGZNS-ZSOCWYAHSA-N leptin Chemical compound O=C([C@H](CO)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](N)CC(C)C)CCSC)N1CCC[C@H]1C(=O)NCC(=O)N[C@@H](CS)C(O)=O NRYBAZVQPHGZNS-ZSOCWYAHSA-N 0.000 description 6
- 229940039781 leptin Drugs 0.000 description 6
- 210000002966 serum Anatomy 0.000 description 6
- 210000000952 spleen Anatomy 0.000 description 6
- 208000024891 symptom Diseases 0.000 description 6
- 238000012360 testing method Methods 0.000 description 6
- -1 (8C) Glutamine Chemical compound 0.000 description 5
- 241000713869 Moloney murine leukemia virus Species 0.000 description 5
- 241000283973 Oryctolagus cuniculus Species 0.000 description 5
- 208000003721 Triple Negative Breast Neoplasms Diseases 0.000 description 5
- 210000004369 blood Anatomy 0.000 description 5
- 239000008280 blood Substances 0.000 description 5
- 239000000872 buffer Substances 0.000 description 5
- 230000000295 complement effect Effects 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 235000014113 dietary fatty acids Nutrition 0.000 description 5
- 229930195729 fatty acid Natural products 0.000 description 5
- 239000000194 fatty acid Substances 0.000 description 5
- 150000004665 fatty acids Chemical class 0.000 description 5
- 238000007446 glucose tolerance test Methods 0.000 description 5
- 230000034659 glycolysis Effects 0.000 description 5
- 238000011534 incubation Methods 0.000 description 5
- 125000003729 nucleotide group Chemical group 0.000 description 5
- 239000002245 particle Substances 0.000 description 5
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 5
- 230000003248 secreting effect Effects 0.000 description 5
- 208000022679 triple-negative breast carcinoma Diseases 0.000 description 5
- 230000004614 tumor growth Effects 0.000 description 5
- 238000001262 western blot Methods 0.000 description 5
- 239000013607 AAV vector Substances 0.000 description 4
- 241000702423 Adeno-associated virus - 2 Species 0.000 description 4
- 241001634120 Adeno-associated virus - 5 Species 0.000 description 4
- 241000713704 Bovine immunodeficiency virus Species 0.000 description 4
- 241000713756 Caprine arthritis encephalitis virus Species 0.000 description 4
- 108091026890 Coding region Proteins 0.000 description 4
- 102000004127 Cytokines Human genes 0.000 description 4
- 108090000695 Cytokines Proteins 0.000 description 4
- 241000713730 Equine infectious anemia virus Species 0.000 description 4
- 241000713800 Feline immunodeficiency virus Species 0.000 description 4
- 102100027581 Forkhead box protein P3 Human genes 0.000 description 4
- 241000713813 Gibbon ape leukemia virus Species 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 description 4
- 241000713311 Simian immunodeficiency virus Species 0.000 description 4
- 230000000692 anti-sense effect Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 108010078428 env Gene Products Proteins 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 239000013604 expression vector Substances 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 239000002773 nucleotide Substances 0.000 description 4
- 230000037361 pathway Effects 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 210000003289 regulatory T cell Anatomy 0.000 description 4
- 230000035945 sensitivity Effects 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 230000009885 systemic effect Effects 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- 241001655883 Adeno-associated virus - 1 Species 0.000 description 3
- 241000202702 Adeno-associated virus - 3 Species 0.000 description 3
- 241000580270 Adeno-associated virus - 4 Species 0.000 description 3
- 241000972680 Adeno-associated virus - 6 Species 0.000 description 3
- 241001164823 Adeno-associated virus - 7 Species 0.000 description 3
- 241001164825 Adeno-associated virus - 8 Species 0.000 description 3
- 102000053602 DNA Human genes 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- 241000701024 Human betaherpesvirus 5 Species 0.000 description 3
- 102100034353 Integrase Human genes 0.000 description 3
- 241000713666 Lentivirus Species 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 208000001145 Metabolic Syndrome Diseases 0.000 description 3
- 108091093037 Peptide nucleic acid Proteins 0.000 description 3
- 230000005867 T cell response Effects 0.000 description 3
- 201000000690 abdominal obesity-metabolic syndrome Diseases 0.000 description 3
- 230000003110 anti-inflammatory effect Effects 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 230000009286 beneficial effect Effects 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 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 3
- 239000002299 complementary DNA Substances 0.000 description 3
- 230000007423 decrease Effects 0.000 description 3
- 239000012636 effector Substances 0.000 description 3
- 230000000694 effects Effects 0.000 description 3
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 3
- 210000003527 eukaryotic cell Anatomy 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 235000009200 high fat diet Nutrition 0.000 description 3
- 150000002500 ions Chemical class 0.000 description 3
- 210000004698 lymphocyte Anatomy 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 238000012083 mass cytometry Methods 0.000 description 3
- 230000003818 metabolic dysfunction Effects 0.000 description 3
- 108091070501 miRNA Proteins 0.000 description 3
- 239000002679 microRNA Substances 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- BDERNNFJNOPAEC-UHFFFAOYSA-N propan-1-ol Chemical compound CCCO BDERNNFJNOPAEC-UHFFFAOYSA-N 0.000 description 3
- 230000010076 replication Effects 0.000 description 3
- 230000003393 splenic effect Effects 0.000 description 3
- 210000004988 splenocyte Anatomy 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 238000013518 transcription Methods 0.000 description 3
- 230000035897 transcription Effects 0.000 description 3
- 230000005748 tumor development Effects 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- 230000009278 visceral effect Effects 0.000 description 3
- 238000005406 washing Methods 0.000 description 3
- 229910001868 water Inorganic materials 0.000 description 3
- VRYALKFFQXWPIH-PBXRRBTRSA-N (3r,4s,5r)-3,4,5,6-tetrahydroxyhexanal Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)CC=O VRYALKFFQXWPIH-PBXRRBTRSA-N 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 101000805768 Banna virus (strain Indonesia/JKT-6423/1980) mRNA (guanine-N(7))-methyltransferase Proteins 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 238000011740 C57BL/6 mouse Methods 0.000 description 2
- 241000282465 Canis Species 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 101710132601 Capsid protein Proteins 0.000 description 2
- 101710197658 Capsid protein VP1 Proteins 0.000 description 2
- 108010051109 Cell-Penetrating Peptides Proteins 0.000 description 2
- 102000020313 Cell-Penetrating Peptides Human genes 0.000 description 2
- 241000282556 Cercocebus atys Species 0.000 description 2
- 101000686790 Chaetoceros protobacilladnavirus 2 Replication-associated protein Proteins 0.000 description 2
- 101000864475 Chlamydia phage 1 Internal scaffolding protein VP3 Proteins 0.000 description 2
- 241000282552 Chlorocebus aethiops Species 0.000 description 2
- 101710094648 Coat protein Proteins 0.000 description 2
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 2
- 101000803553 Eumenes pomiformis Venom peptide 3 Proteins 0.000 description 2
- 101000583961 Halorubrum pleomorphic virus 1 Matrix protein Proteins 0.000 description 2
- 101000609255 Homo sapiens Plasminogen activator inhibitor 1 Proteins 0.000 description 2
- 108090001005 Interleukin-6 Proteins 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 101710081079 Minor spike protein H Proteins 0.000 description 2
- 241000713862 Moloney murine sarcoma virus Species 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 101710118046 RNA-directed RNA polymerase Proteins 0.000 description 2
- 108091028664 Ribonucleotide Proteins 0.000 description 2
- YASAKCUCGLMORW-UHFFFAOYSA-N Rosiglitazone Chemical compound C=1C=CC=NC=1N(C)CCOC(C=C1)=CC=C1CC1SC(=O)NC1=O YASAKCUCGLMORW-UHFFFAOYSA-N 0.000 description 2
- 241000714474 Rous sarcoma virus Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 241000713880 Spleen focus-forming virus Species 0.000 description 2
- 108700012920 TNF Proteins 0.000 description 2
- 108700019146 Transgenes Proteins 0.000 description 2
- 102000004243 Tubulin Human genes 0.000 description 2
- 108090000704 Tubulin Proteins 0.000 description 2
- 101710108545 Viral protein 1 Proteins 0.000 description 2
- 241000713325 Visna/maedi virus Species 0.000 description 2
- 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 2
- 230000004913 activation Effects 0.000 description 2
- PMMURAAUARKVCB-UHFFFAOYSA-N alpha-D-ara-dHexp Natural products OCC1OC(O)CC(O)C1O PMMURAAUARKVCB-UHFFFAOYSA-N 0.000 description 2
- 210000000612 antigen-presenting cell Anatomy 0.000 description 2
- 230000000890 antigenic effect Effects 0.000 description 2
- 230000027455 binding Effects 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 235000014633 carbohydrates Nutrition 0.000 description 2
- 239000013592 cell lysate Substances 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 238000004163 cytometry Methods 0.000 description 2
- 238000000326 densiometry Methods 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- 108700004025 env Genes Proteins 0.000 description 2
- 238000013401 experimental design Methods 0.000 description 2
- 208000010706 fatty liver disease Diseases 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 229910052739 hydrogen Inorganic materials 0.000 description 2
- 239000001257 hydrogen Substances 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 230000028993 immune response Effects 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 230000010120 metabolic dysregulation Effects 0.000 description 2
- 230000007102 metabolic function Effects 0.000 description 2
- 230000004060 metabolic process Effects 0.000 description 2
- 238000002705 metabolomic analysis Methods 0.000 description 2
- 230000001431 metabolomic effect Effects 0.000 description 2
- 230000006540 mitochondrial respiration Effects 0.000 description 2
- 238000010172 mouse model Methods 0.000 description 2
- 238000002552 multiple reaction monitoring Methods 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000002018 overexpression Effects 0.000 description 2
- 239000008194 pharmaceutical composition Substances 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 239000013600 plasmid vector Substances 0.000 description 2
- 230000001124 posttranscriptional effect Effects 0.000 description 2
- 230000000770 proinflammatory effect Effects 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 238000003259 recombinant expression Methods 0.000 description 2
- 239000002336 ribonucleotide Substances 0.000 description 2
- 125000002652 ribonucleotide group Chemical group 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 230000004580 weight loss Effects 0.000 description 2
- MNULEGDCPYONBU-WMBHJXFZSA-N (1r,4s,5e,5'r,6'r,7e,10s,11r,12s,14r,15s,16s,18r,19s,20r,21e,25s,26r,27s,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trio Polymers O([C@@H]1CC[C@@H](/C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)[C@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)O[C@H]([C@H]2C)[C@H]1C)CC)[C@]12CC[C@@H](C)[C@@H](C[C@H](C)O)O1 MNULEGDCPYONBU-WMBHJXFZSA-N 0.000 description 1
- MNULEGDCPYONBU-DJRUDOHVSA-N (1s,4r,5z,5'r,6'r,7e,10s,11r,12s,14r,15s,18r,19r,20s,21e,26r,27s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers O([C@H]1CC[C@H](\C=C/C=C/C[C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@H](C)[C@@H](O)C(C)C(=O)[C@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)OC([C@H]2C)C1C)CC)[C@]12CC[C@@H](C)[C@@H](CC(C)O)O1 MNULEGDCPYONBU-DJRUDOHVSA-N 0.000 description 1
- DZLOHEOHWICNIL-QGZVFWFLSA-N (2R)-2-[6-(4-chlorophenoxy)hexyl]-2-oxiranecarboxylic acid ethyl ester Chemical compound C=1C=C(Cl)C=CC=1OCCCCCC[C@]1(C(=O)OCC)CO1 DZLOHEOHWICNIL-QGZVFWFLSA-N 0.000 description 1
- MNULEGDCPYONBU-YNZHUHFTSA-N (4Z,18Z,20Z)-22-ethyl-7,11,14,15-tetrahydroxy-6'-(2-hydroxypropyl)-5',6,8,10,12,14,16,28,29-nonamethylspiro[2,26-dioxabicyclo[23.3.1]nonacosa-4,18,20-triene-27,2'-oxane]-3,9,13-trione Polymers CC1C(C2C)OC(=O)\C=C/C(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)C\C=C/C=C\C(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-YNZHUHFTSA-N 0.000 description 1
- MNULEGDCPYONBU-VVXVDZGXSA-N (5e,5'r,7e,10s,11r,12s,14s,15r,16r,18r,19s,20r,21e,26r,29s)-4-ethyl-11,12,15,19-tetrahydroxy-6'-[(2s)-2-hydroxypropyl]-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers C([C@H](C)[C@@H](O)[C@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@H](C)[C@@H](O)[C@H](C)/C=C/C(=O)OC([C@H]1C)[C@H]2C)\C=C\C=C\C(CC)CCC2OC21CC[C@@H](C)C(C[C@H](C)O)O2 MNULEGDCPYONBU-VVXVDZGXSA-N 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- MNULEGDCPYONBU-UHFFFAOYSA-N 4-ethyl-11,12,15,19-tetrahydroxy-6'-(2-hydroxypropyl)-5',10,12,14,16,18,20,26,29-nonamethylspiro[24,28-dioxabicyclo[23.3.1]nonacosa-5,7,21-triene-27,2'-oxane]-13,17,23-trione Polymers CC1C(C2C)OC(=O)C=CC(C)C(O)C(C)C(=O)C(C)C(O)C(C)C(=O)C(C)(O)C(O)C(C)CC=CC=CC(CC)CCC2OC21CCC(C)C(CC(C)O)O2 MNULEGDCPYONBU-UHFFFAOYSA-N 0.000 description 1
- 229940126565 ATP-synthase inhibitor Drugs 0.000 description 1
- 208000021959 Abnormal metabolism Diseases 0.000 description 1
- 241000649045 Adeno-associated virus 10 Species 0.000 description 1
- 241000649046 Adeno-associated virus 11 Species 0.000 description 1
- 241000649047 Adeno-associated virus 12 Species 0.000 description 1
- 102000014777 Adipokines Human genes 0.000 description 1
- 108010078606 Adipokines Proteins 0.000 description 1
- 102000011690 Adiponectin Human genes 0.000 description 1
- 108010076365 Adiponectin Proteins 0.000 description 1
- USFZMSVCRYTOJT-UHFFFAOYSA-N Ammonium acetate Chemical compound N.CC(O)=O USFZMSVCRYTOJT-UHFFFAOYSA-N 0.000 description 1
- 239000005695 Ammonium acetate Substances 0.000 description 1
- UIFFUZWRFRDZJC-UHFFFAOYSA-N Antimycin A1 Natural products CC1OC(=O)C(CCCCCC)C(OC(=O)CC(C)C)C(C)OC(=O)C1NC(=O)C1=CC=CC(NC=O)=C1O UIFFUZWRFRDZJC-UHFFFAOYSA-N 0.000 description 1
- NQWZLRAORXLWDN-UHFFFAOYSA-N Antimycin-A Natural products CCCCCCC(=O)OC1C(C)OC(=O)C(NC(=O)c2ccc(NC=O)cc2O)C(C)OC(=O)C1CCCC NQWZLRAORXLWDN-UHFFFAOYSA-N 0.000 description 1
- 238000012169 CITE-Seq Methods 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 102000053642 Catalytic RNA Human genes 0.000 description 1
- 108090000994 Catalytic RNA Proteins 0.000 description 1
- NBSCHQHZLSJFNQ-GASJEMHNSA-N D-Glucose 6-phosphate Chemical compound OC1O[C@H](COP(O)(O)=O)[C@@H](O)[C@H](O)[C@H]1O NBSCHQHZLSJFNQ-GASJEMHNSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 1
- 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 1
- 238000008157 ELISA kit Methods 0.000 description 1
- 238000011510 Elispot assay Methods 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 108010087819 Fc receptors Proteins 0.000 description 1
- 102000009109 Fc receptors Human genes 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 208000033962 Fontaine progeroid syndrome Diseases 0.000 description 1
- 108091006027 G proteins Proteins 0.000 description 1
- 102000030782 GTP binding Human genes 0.000 description 1
- 108091000058 GTP-Binding Proteins 0.000 description 1
- 101710177291 Gag polyprotein Proteins 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 101100118916 Gibbon ape leukemia virus env gene Proteins 0.000 description 1
- VFRROHXSMXFLSN-UHFFFAOYSA-N Glc6P Natural products OP(=O)(O)OCC(O)C(O)C(O)C(O)C=O VFRROHXSMXFLSN-UHFFFAOYSA-N 0.000 description 1
- BCCRXDTUTZHDEU-VKHMYHEASA-N Gly-Ser Chemical group NCC(=O)N[C@@H](CO)C(O)=O BCCRXDTUTZHDEU-VKHMYHEASA-N 0.000 description 1
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 241001559542 Hippocampus hippocampus Species 0.000 description 1
- 101000976075 Homo sapiens Insulin Proteins 0.000 description 1
- 101001034652 Homo sapiens Insulin-like growth factor 1 receptor Proteins 0.000 description 1
- 101000650804 Homo sapiens Semaphorin-3E Proteins 0.000 description 1
- 241000598436 Human T-cell lymphotropic virus Species 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 102100025087 Insulin receptor substrate 1 Human genes 0.000 description 1
- 101710201824 Insulin receptor substrate 1 Proteins 0.000 description 1
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 1
- 102000004218 Insulin-Like Growth Factor I Human genes 0.000 description 1
- 102100039688 Insulin-like growth factor 1 receptor Human genes 0.000 description 1
- 108020004684 Internal Ribosome Entry Sites Proteins 0.000 description 1
- 208000037396 Intraductal Noninfiltrating Carcinoma Diseases 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 108010052014 Liberase Proteins 0.000 description 1
- 101710125418 Major capsid protein Proteins 0.000 description 1
- 206010064912 Malignant transformation Diseases 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 108700011259 MicroRNAs Proteins 0.000 description 1
- 241000714177 Murine leukemia virus Species 0.000 description 1
- 101100244211 Mus musculus Pnpla2 gene Proteins 0.000 description 1
- BAWFJGJZGIEFAR-NNYOXOHSSA-N NAD zwitterion Chemical compound NC(=O)C1=CC=C[N+]([C@H]2[C@@H]([C@H](O)[C@@H](COP([O-])(=O)OP(O)(=O)OC[C@@H]3[C@H]([C@@H](O)[C@@H](O3)N3C4=NC=NC(N)=C4N=C3)O)O2)O)=C1 BAWFJGJZGIEFAR-NNYOXOHSSA-N 0.000 description 1
- 241000772415 Neovison vison Species 0.000 description 1
- DFPAKSUCGFBDDF-UHFFFAOYSA-N Nicotinamide Chemical compound NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 1
- 208000035327 Oestrogen receptor positive breast cancer Diseases 0.000 description 1
- 240000007019 Oxalis corniculata Species 0.000 description 1
- 241000701945 Parvoviridae Species 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 101800001494 Protease 2A Proteins 0.000 description 1
- 101800001066 Protein 2A Proteins 0.000 description 1
- 108010076504 Protein Sorting Signals Proteins 0.000 description 1
- LCTONWCANYUPML-UHFFFAOYSA-M Pyruvate Chemical compound CC(=O)C([O-])=O LCTONWCANYUPML-UHFFFAOYSA-M 0.000 description 1
- 206010038802 Reticuloendothelial system stimulated Diseases 0.000 description 1
- 102100027752 Semaphorin-3E Human genes 0.000 description 1
- 102000054727 Serum Amyloid A Human genes 0.000 description 1
- 108700028909 Serum Amyloid A Proteins 0.000 description 1
- 101150043341 Socs3 gene Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 102000058015 Suppressor of Cytokine Signaling 3 Human genes 0.000 description 1
- 108700027337 Suppressor of Cytokine Signaling 3 Proteins 0.000 description 1
- 230000006044 T cell activation Effects 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 208000000389 T-cell leukemia Diseases 0.000 description 1
- 208000028530 T-cell lymphoblastic leukemia/lymphoma Diseases 0.000 description 1
- 210000000447 Th1 cell Anatomy 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 108700029229 Transcriptional Regulatory Elements Proteins 0.000 description 1
- 206010046865 Vaccinia virus infection Diseases 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- 108091093126 WHP Posttrascriptional Response Element Proteins 0.000 description 1
- 230000006682 Warburg effect Effects 0.000 description 1
- 230000001133 acceleration Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 230000033289 adaptive immune response Effects 0.000 description 1
- 239000000478 adipokine Substances 0.000 description 1
- 210000003486 adipose tissue brown Anatomy 0.000 description 1
- 230000006536 aerobic glycolysis Effects 0.000 description 1
- HXXFSFRBOHSIMQ-VFUOTHLCSA-N alpha-D-glucose 1-phosphate Chemical compound OC[C@H]1O[C@H](OP(O)(O)=O)[C@H](O)[C@@H](O)[C@@H]1O HXXFSFRBOHSIMQ-VFUOTHLCSA-N 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 235000019257 ammonium acetate Nutrition 0.000 description 1
- 229940043376 ammonium acetate Drugs 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- UIFFUZWRFRDZJC-SBOOETFBSA-N antimycin A Chemical compound C[C@H]1OC(=O)[C@H](CCCCCC)[C@@H](OC(=O)CC(C)C)[C@H](C)OC(=O)[C@H]1NC(=O)C1=CC=CC(NC=O)=C1O UIFFUZWRFRDZJC-SBOOETFBSA-N 0.000 description 1
- PVEVXUMVNWSNIG-UHFFFAOYSA-N antimycin A3 Natural products CC1OC(=O)C(CCCC)C(OC(=O)CC(C)C)C(C)OC(=O)C1NC(=O)C1=CC=CC(NC=O)=C1O PVEVXUMVNWSNIG-UHFFFAOYSA-N 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 239000006286 aqueous extract Substances 0.000 description 1
- 238000013473 artificial intelligence Methods 0.000 description 1
- 238000007681 bariatric surgery Methods 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 208000036815 beta tubulin Diseases 0.000 description 1
- 229920002988 biodegradable polymer Polymers 0.000 description 1
- 239000004621 biodegradable polymer Substances 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 210000000234 capsid Anatomy 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 210000004970 cd4 cell Anatomy 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 230000019522 cellular metabolic process Effects 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 235000013330 chicken meat Nutrition 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 239000007979 citrate buffer Substances 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 239000005515 coenzyme Substances 0.000 description 1
- 206010009887 colitis Diseases 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 208000029078 coronary artery disease Diseases 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 230000000139 costimulatory effect Effects 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 239000008367 deionised water Substances 0.000 description 1
- 229910021641 deionized water Inorganic materials 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 229960003957 dexamethasone Drugs 0.000 description 1
- 239000003085 diluting agent Substances 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
- 230000005750 disease progression Effects 0.000 description 1
- BFMYDTVEBKDAKJ-UHFFFAOYSA-L disodium;(2',7'-dibromo-3',6'-dioxido-3-oxospiro[2-benzofuran-1,9'-xanthene]-4'-yl)mercury;hydrate Chemical compound O.[Na+].[Na+].O1C(=O)C2=CC=CC=C2C21C1=CC(Br)=C([O-])C([Hg])=C1OC1=C2C=C(Br)C([O-])=C1 BFMYDTVEBKDAKJ-UHFFFAOYSA-L 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 208000028715 ductal breast carcinoma in situ Diseases 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 210000000750 endocrine system Anatomy 0.000 description 1
- 230000037149 energy metabolism Effects 0.000 description 1
- 101150030339 env gene Proteins 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 201000010063 epididymitis Diseases 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 201000007281 estrogen-receptor positive breast cancer Diseases 0.000 description 1
- 229950006213 etomoxir Drugs 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000006539 extracellular acidification Effects 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 230000002349 favourable effect Effects 0.000 description 1
- 230000004151 fermentation Effects 0.000 description 1
- 238000000855 fermentation Methods 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 238000004401 flow injection analysis Methods 0.000 description 1
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 1
- 230000004907 flux Effects 0.000 description 1
- 238000002290 gas chromatography-mass spectrometry Methods 0.000 description 1
- 238000001476 gene delivery Methods 0.000 description 1
- 108091006104 gene-regulatory proteins Proteins 0.000 description 1
- 102000034356 gene-regulatory proteins Human genes 0.000 description 1
- 230000004110 gluconeogenesis Effects 0.000 description 1
- 150000002303 glucose derivatives Chemical class 0.000 description 1
- 230000014101 glucose homeostasis Effects 0.000 description 1
- 229950010772 glucose-1-phosphate Drugs 0.000 description 1
- 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 1
- 210000003714 granulocyte Anatomy 0.000 description 1
- 230000002440 hepatic effect Effects 0.000 description 1
- 229920000140 heteropolymer Polymers 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 230000000910 hyperinsulinemic effect Effects 0.000 description 1
- 208000006575 hypertriglyceridemia Diseases 0.000 description 1
- 230000007954 hypoxia Effects 0.000 description 1
- 238000011493 immune profiling Methods 0.000 description 1
- 230000037451 immune surveillance Effects 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 238000002743 insertional mutagenesis Methods 0.000 description 1
- PBGKTOXHQIOBKM-FHFVDXKLSA-N insulin (human) Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@H](C(N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3C=CC(O)=CC=3)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=3NC=NC=3)NC(=O)[C@H](CO)NC(=O)CNC1=O)C(=O)NCC(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(=O)N[C@@H](CC(N)=O)C(O)=O)=O)CSSC[C@@H](C(N2)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C(C)C)NC(=O)[C@@H](NC(=O)CN)[C@@H](C)CC)[C@@H](C)CC)[C@@H](C)O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@@H](NC(=O)[C@@H](N)CC=1C=CC=CC=1)C(C)C)C1=CN=CN1 PBGKTOXHQIOBKM-FHFVDXKLSA-N 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000031261 interleukin-10 production Effects 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 235000007260 kalia Nutrition 0.000 description 1
- 230000008463 key metabolic pathway Effects 0.000 description 1
- 239000004310 lactic acid Substances 0.000 description 1
- 235000014655 lactic acid Nutrition 0.000 description 1
- 239000010410 layer Substances 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 239000002479 lipoplex Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 231100000053 low toxicity Toxicity 0.000 description 1
- 208000026534 luminal B breast carcinoma Diseases 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 230000036212 malign transformation Effects 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 210000003071 memory t lymphocyte Anatomy 0.000 description 1
- 230000009245 menopause Effects 0.000 description 1
- 230000006371 metabolic abnormality Effects 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 239000000693 micelle Substances 0.000 description 1
- 239000003607 modifier Substances 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 230000003387 muscular Effects 0.000 description 1
- 229950006238 nadide Drugs 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 229960003966 nicotinamide Drugs 0.000 description 1
- 235000005152 nicotinamide Nutrition 0.000 description 1
- 239000011570 nicotinamide Substances 0.000 description 1
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 230000009871 nonspecific binding Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 229930191479 oligomycin Natural products 0.000 description 1
- MNULEGDCPYONBU-AWJDAWNUSA-N oligomycin A Polymers O([C@H]1CC[C@H](/C=C/C=C/C[C@@H](C)[C@H](O)[C@@](C)(O)C(=O)[C@@H](C)[C@H](O)[C@@H](C)C(=O)[C@@H](C)[C@H](O)[C@@H](C)/C=C/C(=O)O[C@@H]([C@@H]2C)[C@@H]1C)CC)[C@@]12CC[C@H](C)[C@H](C[C@@H](C)O)O1 MNULEGDCPYONBU-AWJDAWNUSA-N 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 239000012044 organic layer Substances 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 230000010627 oxidative phosphorylation Effects 0.000 description 1
- 230000036284 oxygen consumption Effects 0.000 description 1
- 230000002023 papillomaviral effect Effects 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 239000000816 peptidomimetic Substances 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 238000000918 plasma mass spectrometry Methods 0.000 description 1
- 108010089520 pol Gene Products Proteins 0.000 description 1
- 229920000575 polymersome Polymers 0.000 description 1
- 238000013105 post hoc analysis Methods 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 210000000229 preadipocyte Anatomy 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 239000012925 reference material Substances 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000035806 respiratory chain Effects 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 108091092562 ribozyme Proteins 0.000 description 1
- 229960004586 rosiglitazone Drugs 0.000 description 1
- 229940080817 rotenone Drugs 0.000 description 1
- JUVIOZPCNVVQFO-UHFFFAOYSA-N rotenone Natural products O1C2=C3CC(C(C)=C)OC3=CC=C2C(=O)C2C1COC1=C2C=C(OC)C(OC)=C1 JUVIOZPCNVVQFO-UHFFFAOYSA-N 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 230000007781 signaling event Effects 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 230000009870 specific binding Effects 0.000 description 1
- 238000004611 spectroscopical analysis Methods 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- 235000013619 trace mineral Nutrition 0.000 description 1
- 239000011573 trace mineral Substances 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- 230000014621 translational initiation Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 150000003626 triacylglycerols Chemical class 0.000 description 1
- ZMOOZQVYWNRWRB-UHFFFAOYSA-N trifluoromethyl 1-cyano-n-phenylmethanehydrazonate Chemical compound FC(F)(F)OC(C#N)=NNC1=CC=CC=C1 ZMOOZQVYWNRWRB-UHFFFAOYSA-N 0.000 description 1
- UFTFJSFQGQCHQW-UHFFFAOYSA-N triformin Chemical compound O=COCC(OC=O)COC=O UFTFJSFQGQCHQW-UHFFFAOYSA-N 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 208000007089 vaccinia Diseases 0.000 description 1
- 238000010200 validation analysis Methods 0.000 description 1
- 210000005167 vascular cell Anatomy 0.000 description 1
- 238000003260 vortexing Methods 0.000 description 1
- 238000004260 weight control Methods 0.000 description 1
Classifications
-
- 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
- A61K39/0005—Vertebrate antigens
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- 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
- A61K2039/57—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
- A61K2039/70—Multivalent vaccine
Definitions
- Obesity is an important risk factor for breast cancer, especially in postmenopausal women. Obesity occurring after menopause increases the risk of developing estrogen receptor positive breast cancer by 20 -40% compared to normal weight women.
- BMI body mass index
- TNBC triple negative breast cancer
- metabolic obesity is used to describe individuals who are not obese on the basis of height and weight, but who, like people with overt obesity, are hyperinsulinemic, insulin-resistant, and predisposed to type 2 diabetes, hypertriglyceridemia, premature coronary heart disease+ and an increased risk of breast cancer.6-8 [0005] There remains a need to treat those with metabolic dysregulation that significantly increases their risk of breast cancer, as well as to effectively target obesity-related conditions, including pre-diabetes, cancers, and inflammation. SUMMARY [0006] The material described herein meets these needs and others by providing a vaccine that drives Type II T-cells to inflammatory adipose tissue.
- Identified herein are overexpressed proteins associated with inflammatory adipocytes, which are immunogenic in humans and mice.
- Newly identified antigens, DUSP1, FABP4, PAI-1, and ATGL, and epitopes thereof, can be used in such a vaccine, as well as epitopes of IGF-1R and HIF1a identified herein.
- nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising at least two epitopes selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing.
- the polypeptide is selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29- 43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing.
- the polypeptide comprises each of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108 and 158-172of PAI-1; (d) amino acids 213-227 and 374- 388 of ATGL; (e) amino acids 276-295 of HIF1a; and (f) amino acids 388-402 and 545-558 of IGF-1R.
- the amino acid sequences of these epitopes are listed in Table 1. [0007]
- the polypeptide is selected from amino acids 53-67 of FABP4, amino acids 29-43 of DUSP1, amino acids 276-295 of HIF1a, and a combination thereof.
- the polypeptide comprises amino acids 94-108 and/or 158- 172 of PAI-1, amino acids 213-227 and/or 374-388 of ATGL, amino acids 388-402 and/or 545-558 of IGF-1R, or a combination thereof. These epitopes are associated with significantly more antigen-specific IL-10 response.
- the polypeptide comprises one or more epitopes of (a), (b), (c), and (d) as listed above. In some embodiments, the polypeptide comprises at least three epitopes selected from (a), (b), (c), and (d).
- the polypeptide comprises at least one of each of (a), (b), (c), and (d). [0010] In some embodiments, the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f), and optionally further comprises one or more linker sequences disposed between the epitopes. In some embodiments, the linker sequence is GPGPG or GGGS. [0011] In some embodiments, the nucleic acid sequence comprises a promoter sequence. In some embodiments, the nucleic acid sequence comprises a heterologous sequence.
- the heterologous sequence encodes a promoter, a transcriptional start site, a translational start site, a detectable marker, a mRNA processing splice site, a polyadenylation sequence, and/or a regulatory element.
- a vector comprising the nucleic acid molecule as described herein, wherein the vector is capable of directing expression of the encoded polypeptide(s).
- the vector is a DNA plasmid, messenger RNA (mRNA), or viral vector.
- the viral vector is a lentiviral vector, an adenoviral vector, or a poxviral vector.
- the vector is mRNA.
- polypeptide comprising at least two epitopes selected from the group consisting of the epitopes listed in Table 1.
- the epitopes selected include an amino acid sequence having at least 90% identity with any one of the amino acid sequences listed in Table 1.
- the polypeptide comprises a combination of at least three, four, five, six, or more of the epitopes listed in Table 1.
- the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f).
- the polypeptide further comprises one or more linker sequences disposed between the epitopes.
- composition comprising the polypeptide described herein.
- the composition further comprises an adjuvant.
- described herein is a method for eliciting a Type 2 immune response directed at inflammation related to obesity, for preventing or treating inflammation in obesity, and/or for preventing or treating disease associated with metabolic obesity in a subject.
- the method comprises administering to the subject a composition comprising a nucleic acid molecule, polypeptide, or composition described herein.
- the disease associated with metabolic obesity is cancer.
- the cancer is breast cancer.
- the disease associated with metabolic obesity is non-alcoholic fatty liver disease (NAFLD), metabolic-associated fatty liver disease (MAFLD), or pre-diabetes.
- the subject is human.
- the subject is suspected of having, or has been diagnosed with obesity, a disease associated with metabolic obesity, cancer, such as pre-diabetes/insulin insensitivity, breast cancer, MAFLD, or NAFLD.
- BRIEF DESCRIPTION OF THE DRAWINGS [0017] FIGS.1A-1D.
- IL-10-selective epitopes can be identified from aberrantly expressed proteins in adipose tissue in obesity. Th ratio when stimulated with the indicated epitope from (1A) FABP4, (1B) DUSP1 (1C) ATGL or (1D) PAI1.
- FIGS.2A-2D Overexpressed antigens identified in human obese adipose tissue are also overexpressed in the adipose tissue of obese mice.
- FIGS.3A-3F Representative Western blot of cell lysate for the indicated antigen from mice fed a high fat diet (HFD) or normal chow diet (NCD). ⁇ / ⁇ Tubulin was used as a loading control.
- FIGS.3A-3F IL-10-secreting epitopes identified via in vitro screening generated a Th2-selective response after immunization with each single antigen.
- FIGS.6A-6D AdVac increased glucose sensitivity and reduced insulin resistance in obese mice.
- ITT Insulin tolerance test
- n 20 mice/group; **p ⁇ 0.01,***p ⁇ 0.001, ****p ⁇ 0.0001 (compared to control obese).
- FIGS.7A-7D AdVac reduced CD8+ T cell and increased T-regulatory cell levels in the adipose tissue of obese mice.
- Percent CD4+/FOXP3+ T-regulatory cells from (7C) adipose tissue or (7D) spleen from lean mice or obese mice immunized with Alum or AdVac.
- FIGS.8A-8F ADVac metabolically reprograms the adipose tissue microenvironment.
- FIGS.9A-9B Obese TgMMTV-neu develop a greater number of tumors with a faster growth rate than lean TgMMTV-neu mice.
- FIGS.10A-10C AdVac immunization decreases CD8+ T-cells in mammary tissue, lowers serum leptin levels, and results in significant tumor inhibition in obese Tg-MMTV-neu mice.
- 10A Percent CD8+ T-cells in viable lymphocytes derived from mammary fat from the indicated treatment group.
- the term “comprising” is intended to mean that the compositions and methods include the recited elements, but do not exclude others.
- the transitional phrase “consisting essentially of” (and grammatical variants) is to be interpreted as encompassing the recited materials or steps “and those that do not materially affect the basic and novel characteristic(s)” of the recited embodiment.
- the term “consisting essentially of” as used herein should not be interpreted as equivalent to “comprising.”
- Consisting of shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions disclosed herein. Aspects defined by each of these transition terms are within the scope of the disclosure herein.
- a linker refers to an amino acid sequence that connects two proteins, polypeptides, peptides, domains, regions, or motifs and may provide a spacer function compatible with interaction of the two sub-binding domains so that the resulting polypeptide retains a specific binding affinity to a target molecule or retains signaling activity.
- a linker is comprised of about two to about 35 amino acids, for instance, or about four to about 20 amino acids or about eight to about 15 amino acids or about 15 to about 25 amino acids.
- nucleic acid sequence or “polynucleotide” refers to nucleotides of any length which are deoxynucleotides (i.e. DNAs), or derivatives thereof; ribonucleotides (i.e. RNAs) or derivatives thereof; or peptide nucleic acids (PNAs) or derivatives thereof.
- the terms include, without limitation, single-stranded, double-stranded, or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, oligonucleotides (oligos), or other natural, synthetic, modified, mutated or non-natural forms of DNA or RNA.
- the term “vector” refers to, without limitation, a recombinant genetic construct or plasmid or expression construct or expression vector that retains the ability to infect and transduce non-dividing and/or slowly-dividing cells and integrate into the target cell’s genome.
- the vector may be derived from or based on a wild-type virus. Aspects of this disclosure relate to an adeno-associated virus vector, an adenovirus vector, and a lentivirus vector.
- expression control element refers to any sequence that regulates the expression of a coding sequence, such as a gene.
- Exemplary expression control elements include but are not limited to promoters, enhancers, microRNAs, post- transcriptional regulatory elements, polyadenylation signal sequences, and introns.
- Expression control elements may be, without limitation, constitutive, inducible, repressible, or tissue-specific.
- a “promoter” is a control sequence that is a region of a polynucleotide sequence at which initiation and rate of transcription are controlled. It may contain genetic elements at which regulatory proteins and molecules may bind such as RNA polymerase and other transcription factors.
- expression control by a promoter is tissue-specific.
- An “enhancer” is a region of DNA that can be bound by activating proteins to increase the likelihood or frequency of transcription.
- Non-limiting exemplary enhancers and posttranscriptional regulatory elements include the CMV enhancer and WPRE.
- multicistronic or “polycistronic” or “bicistronic” or tricistronic” refers to mRNA with multiple, i.e., double or triple coding areas or exons, and as such will have the capability to express from mRNA two or more, or three or more, or four or more, etc., proteins from a single construct. Multicistronic vectors simultaneously express two or more separate proteins from the same mRNA.
- the two strategies most widely used for constructing multicistronic configurations are through the use of 1) an IRES or 2) a 2A self- cleaving site.
- an “IRES” refers to an internal ribosome entry site or portion thereof of viral, prokaryotic, or eukaryotic origin which are used within polycistronic vector constructs.
- an IRES is an RNA element that allows for translation initiation in a cap- independent manner.
- self-cleaving peptides or “sequences encoding self- cleaving peptides” or “2A self-cleaving site” refer to linking sequences which are used within vector constructs to incorporate sites to promote ribosomal skipping and thus to generate two polypeptides from a single promoter, such self-cleaving peptides include without limitation, T2A, and P2A peptides or sequences encoding the self-cleaving peptides.
- “Homology” or “identity” or “similarity” refers to sequence similarity between two peptides or between two nucleic acid molecules.
- Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid, then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An “unrelated” or “non-homologous” sequence shares less than 40% identity, or alternatively less than 25% identity, with one of the sequences of disclosed herein. [0036] Percent similarity or percent complementary of any of the disclosed sequences may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG).
- UWGCG University of Wisconsin Genetics Computer Group
- the GAP program utilizes the alignment method of Needleman and Wunsch (1970). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) which are similar, divided by the total number of symbols in the shorter of the two sequences.
- the preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non- identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess (1986), (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
- Nucleotide sequence refers to a heteropolymer of deoxyribonucleotides, ribonucleotides, or peptide-nucleic acid sequences that may be assembled from smaller fragments, isolated from larger fragments, or chemically synthesized de novo or partially synthesized by combining shorter oligonucleotide linkers, or from a series of oligonucleotides.
- protein As used herein, the terms “protein”, “peptide”, and “polypeptide” refer to amino acid subunits, amino acid analogs, or peptidomimetics. The subunits may be linked by peptide bonds.
- the subunit may be linked by other bonds, e.g., ester, ether, etc.
- amino acid refers to either natural and/or unnatural or synthetic amino acids.
- recombinant expression system or “recombinant expression vector” refers to a genetic construct for the expression of certain genetic material formed by recombination.
- effective amount or “therapeutically effective amount” or “prophylactically effective amount”, refer to an amount of an active agent described herein that is effective to provide the desired/intended result and/or biological activity.
- an effective amount of a composition described herein is an amount that is effective to result in remission or slowing the progression of disease, and/or to improve or to ameliorate symptoms of and/or to treat disease.
- an equivalent or a biologically equivalent of such is intended within the scope of this disclosure.
- biological equivalent thereof is intended to be synonymous with “equivalent thereof” when referring to a reference small molecule, polypeptide, protein, polynucleotide, nucleic acid, oligonucleotide, antisense, or miRNA even those reference molecules having minimal homology while still maintaining desired structure or functionality. Unless specifically recited herein, it is contemplated that any nucleic acid, polynucleotide, oligonucleotide, antisense, miRNA, polypeptide, or protein mentioned herein also includes equivalents thereof.
- an equivalent intends at least about 70% homology or identity, or at least 80 % homology or identity and alternatively, or at least about 85 %, or alternatively at least about 90 %, or alternatively at least about 95 %, or alternatively 98 % percent homology or identity and exhibits substantially equivalent biological activity to the reference protein, polypeptide or nucleic acid.
- an equivalent thereof is a polynucleotide that hybridizes under stringent conditions to the reference polynucleotide or its complement.
- the polypeptide and/or polynucleotide sequences are provided herein for use in gene and protein transfer and expression techniques described herein.
- Such sequences provided herein can be used to provide the expression product as well as substantially identical sequences that produce a protein that has the same biological properties.
- These “biologically equivalent” or “biologically active” or “equivalent” polypeptides are encoded by equivalent polynucleotides as described herein. They may possess at least 60%, or alternatively, at least 65%, or alternatively, at least 70%, or alternatively, at least 75%, or alternatively, at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95% or alternatively at least 98%, identical primary amino acid sequence to the reference polypeptide when compared using sequence identity methods run under default conditions. Specific polynucleotide or polypeptide sequences are provided as examples of particular embodiments.
- an equivalent polynucleotide is one that hybridizes under stringent conditions to the reference polynucleotide or its complement or in reference to a polypeptide, a polypeptide encoded by a polynucleotide that hybridizes to the reference encoding polynucleotide under stringent conditions or its complementary strand.
- an equivalent polypeptide or protein is one that is expressed from an equivalent polynucleotide.
- “Hybridization” refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide residues.
- the hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner.
- the complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these.
- a hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PC reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme.
- Examples of stringent hybridization conditions include: incubation temperatures of about 25°C to about 37°C; hybridization buffer concentrations of about 6x SSC to about 10x SSC; formamide concentrations of about 0% to about 25%; and wash solutions from about 4x SSC to about 8x SSC.
- Examples of moderate hybridization conditions include: incubation temperatures of about 40°C to about 50°C; buffer concentrations of about 9x SSC to about 2x SSC; formamide concentrations of about 30% to about 50%; and wash solutions of about 5x SSC to about 2x SSC.
- high stringency conditions include: incubation temperatures of about 55°C to about 68°C; buffer concentrations of about lx SSC to about 0.1x SSC; formamide concentrations of about 55% to about 75%; and wash solutions of about lx SSC, 0.1x SSC, or deionized water.
- hybridization incubation times are from 5 minutes to 24 hours, with 1, 2, or more washing steps, and wash incubation times are about 1, 2, or 15 minutes.
- SSC is 0.15 M NaCl and 15 mM citrate buffer. It is understood that equivalents of SSC using other buffer systems can be employed.
- treating or “treatment” of a disease in a subject refers to (1) preventing the symptoms or disease from occurring in a subject that is predisposed or does not yet display symptoms of the disease; (2) inhibiting the disease or arresting its development; or (3) ameliorating or causing regression of the disease or the symptoms of the disease.
- treatment is an approach for obtaining beneficial or desired results, including clinical results.
- beneficial or desired results can include one or more, but are not limited to, alleviation or amelioration of one or more symptoms of disease associated with metabolic obesity, diminishment of same, stabilized (i.e., not worsening) state of the condition (including disease), delay or slowing of the condition (including disease), progression, amelioration or palliation of a condition (including disease), states of and remission of (whether partial or total) disease associated with metabolic obesity, whether detectable or undetectable.
- the term "isolated” means that a naturally occurring DNA fragment, DNA molecule, coding sequence, or oligonucleotide is removed from its natural environment, or is a synthetic molecule or cloned product.
- the DNA fragment, DNA molecule, coding sequence, or oligonucleotide is purified, i.e., essentially free from any other DNA fragment, DNA molecule, coding sequence, or oligonucleotide and associated cellular products or other impurities.
- the term “cell” as used herein refers to either a prokaryotic or eukaryotic cell, optionally obtained from a subject or a commercially available source.
- Cells treated, transfected, transformed, or otherwise in contact with compositions and/or nucleic acid molecules disclosed herein include without limitation, cells of a human, non-human animal, mammal, or non-human mammal, including without limitation, cells of murine, canine, or non-human primate species.
- Cells treated, transfected, transformed, or otherwise in contact with compositions and/or nucleic acid molecules disclosed herein are, without limitation, T cells, antigen-presenting cells, and other suitable host cells.
- the term "subject" includes any human or non-human animal.
- non-human animal includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, horses, sheep, dogs, cows, pigs, chickens, and other veterinary subjects.
- a or “an” means at least one, unless clearly indicated otherwise.
- to “prevent” or “protect against” a condition or disease means to hinder, reduce or delay the onset or progression of the condition or disease.
- encode refers to a polynucleotide which is said to “encode” a polypeptide, an mRNA, or an effector RNA if, in its native state or when manipulated by methods well known to those skilled in the art, can be transcribed and/or translated to produce the effector RNA, the mRNA, or an mRNA that can for the polypeptide and/or a fragment thereof.
- the antisense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom.
- expression refers to the process by which polynucleotides are transcribed into mRNA and/or the process by which the transcribed mRNA is subsequently translated into peptides, polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in a eukaryotic cell. The expression level of a gene may be determined by measuring the amount of mRNA or protein in a cell or tissue sample; further, the expression level of multiple genes can be determined to establish an expression profile for a particular sample.
- the term “functional” may be used to modify any molecule, biological, or cellular material to intend that it accomplishes a particular, specified effect.
- the term “about,” as used herein when referring to a measurable value such as an amount, level or concentration, for example and without limitation, is meant to encompass variations of 20%, 10%, 5%, 1 %, 0.5%, or even 0.1 % of the specified amount, or fold differences in levels of a quantifiable comparison with a standard or control or reference material, such as 1-fold, 2-fold, 3-fold, 4-fold...10-fold, 100-fold, etc. of the specified level of comparison.
- AAV adeno-associated virus
- Non-limiting exemplary serotypes useful in the methods disclosed herein include any of the 11 or 12 serotypes, e.g., AAV2, AAV5, and AAV8, or variant serotypes, e.g. AAV-DJ.
- the AAV structural particle is composed of 60 protein molecules made up of VP1, VP2, and VP3. Each particle contains approximately 5 VP1 proteins, 5 VP2 proteins and 50 VP3 proteins ordered into an icosahedral structure.
- compositions and/or nucleic acid molecules for preventing and treating inflammation associated with obesity, or a disease associated with metabolic obesity, in a subject.
- a nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide that includes one or more of the epitopes described herein.
- the polypeptide is selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing.
- the polypeptide comprises each of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108 and 158-172of PAI-1; (d) amino acids 213-227 and 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; and (f) amino acids 388-402 and 545- 558 of IGF-1R.
- the amino acid sequences of these epitopes are listed in Table 1.
- Table 1 Epitopes Included in ADVac (indicated) and Candidates for Inclusion in ADVac *The mature IGF1R protein is cleaved at 30 amino acids at the N-terminus.
- the nucleic acid sequence encodes a polypeptide that comprises a combination of one or more epitopes.
- the encoded polypeptide comprises one or more of the epitopes listed in Table 1.
- the polypeptide comprises the first nine epitopes listed in Table 1.
- the polypeptide comprises one or more of the epitopes listed in the latter portion of Table 1 as candidates for inclusion in an ADVac vaccine.
- the polypeptide comprises 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more of the epitopes listed in Table 1.
- the nucleic acid sequence optionally further comprises one or more linker sequences disposed between the epitopes.
- the linker sequence is glycine-serine.
- the nucleic acid sequence comprises a promoter sequence.
- the nucleic acid sequence comprises a heterologous sequence.
- the heterologous sequence encodes a promoter, a transcriptional start site, a translational start site, a detectable marker, a mRNA processing splice site, a polyadenylation sequence, and/or a regulatory element.
- Such nucleic acid molecules may be delivered by viral or non-viral means.
- viral delivery is adeno-associated virus (AAV).
- AAV adeno-associated virus
- retrovirus lentivirus
- baculovirus delivery
- CPP cell penetrating peptide
- Polynucleotide constructs may also be modified, such as through chemical modification, to improve their stability and/or suitability for delivery.
- the oligonucleotide is modified by locked nucleic acids and/or phosphorothioate linkages.
- a delivery system is selected for improved bioavailability, such as PEGylated liposomes, lipidoids, or biodegradable polymers, as examples.
- the vector disclosed herein is a viral vector.
- the vector is an adenoviral vector, an adeno-associated viral (AAV) vector, or a lentiviral vector.
- the vector is a retroviral vector, an adenoviral/retroviral chimera vector, a herpes simplex viral I or II vector, a parvoviral vector, a reticuloendotheliosis viral vector, a polioviral vector, a papillomaviral vector, a vaccinia viral vector, or any hybrid or chimeric vector incorporating favorable aspects of two or more viral vectors.
- the vector further comprises one or more expression control elements operably linked to the polynucleotide. In some embodiments, the vector further comprises one or more selectable markers. [0064] In some embodiments, the vector disclosed herein is an AAV vector with low toxicity. In some embodiments, the AAV vector does not incorporate into the host genome, thereby having a low probability of causing insertional mutagenesis. In some embodiments, the AAV vector can encode a range total polynucleotides from 4.5 kb to 4.75 kb.
- exemplary AAV vectors that may be used in any of the herein described compositions, systems, methods, and kits can include an AAV1 vector, a modified AAV1 vector, an AAV2 vector, a modified AAV2 vector, an AAV3 vector, a modified AAV3 vector, an AAV4 vector, a modified AAV4 vector, an AAV5 vector, a modified AAV5 vector, an AAV6 vector, a modified AAV6 vector, an AAV7 vector, a modified AAV7 vector, an AAV8 vector, an AAV9 vector, an AAV.rh10 vector, a modified AAV.rh10 vector, an AAV.rh32/33 vector, a modified AAV.rh32/33 vector, an AAV.rh43 vector, a modified AAV.rh43 vector, an AAV.rh64R1 vector, and a modified AAV.rh64R1 vector and any combinations or equivalents thereof.
- the vector disclosed herein is a lentiviral vector.
- the lentiviral vector is an integrase-competent lentiviral vector (ICLV).
- the lentiviral vector can refer to the transgene plasmid vector as well as the transgene plasmid vector in conjunction with related plasmids (e.g., a packaging plasmid, a rev expressing plasmid, an envelope plasmid) as well as a lentiviral-based particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism.
- Lentiviral vectors are well-known in the art.
- exemplary lentiviral vectors that may be used in relation to any of the herein described compositions, nucleic acid molecules and/or methods, and can include a human immunodeficiency virus (HIV) 1 vector, a modified human immunodeficiency virus (HIV) 1 vector, a human immunodeficiency virus (HIV) 2 vector, a modified human immunodeficiency virus (HIV) 2 vector, a sooty mangabey simian immunodeficiency virus (SIV SM ) vector, a modified sooty mangabey simian immunodeficiency virus (SIV SM ) vector, a African green monkey simian immunodeficiency virus (SIV AGM ) vector, a modified African green monkey simian immunodeficiency virus (SIV AGM ) vector, a equine infectious anemia virus (EIAV) vector, a modified equine infectious anemia virus (EIAV) vector, a feline immunode
- HAV
- a vector of the disclosure is a viral vector.
- the viral vector comprises a sequence isolated or derived from a retrovirus.
- the viral vector comprises a sequence isolated or derived from a lentivirus.
- the viral vector comprises a sequence isolated or derived from an adenovirus.
- the viral vector comprises a sequence isolated or derived from an adeno-associated virus (AAV).
- AAV adeno-associated virus
- the viral vector is replication incompetent.
- the viral vector is isolated or recombinant.
- the viral vector is self-complementary.
- the viral vector comprises a sequence isolated or derived from an adeno-associated virus (AAV).
- AAV adeno-associated virus
- the viral vector comprises an inverted terminal repeat sequence or a capsid sequence that is isolated or derived from an AAV of serotype AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, or AAV12, or the vector and/or components are derived from a synthetic AAV serotype, such as, without limitation, Anc80 AAV (an ancestor of AAV 1, 2, 6, 8 and 9).
- the viral vector is replication incompetent.
- the viral vector is isolated or recombinant (rAAV). In some embodiments, the viral vector is self-complementary (scAAV).
- a vector of the disclosure is a non-viral vector. In some embodiments, the vector comprises or consists of a nanoparticle, a micelle, a liposome or lipoplex, a polymersome, a polyplex or a dendrimer.
- expression vector or viral vector disclosed herein is used to transfect, transform, or come in contact with a cell which is a eukaryotic cell. In some embodiments, the cell is an animal cell.
- the cell is a mammalian cell. In some embodiments, the cell is a bovine, murine, feline, equine, porcine, canine, simian, or human cell.
- a cell is a packaging cell or a producer cell for production of a viral particle.
- viral particles comprising, consisting of, or consisting essentially of a vector comprising, consisting of, or consisting essentially of a polynucleotide sequence encoding one or more polypeptides as described herein.
- methods of packaging genetic material such as RNA or DNA into one or more vectors is well known in the art.
- the genetic material may be packaged using a packaging vector and cell lines and introduced via traditional recombinant methods.
- the packaging vector may include, but is not limited to retroviral vector, lentiviral vector, adenoviral vector, and adeno-associated viral vector.
- the packaging vector contains elements and sequences that facilitate the delivery of genetic materials into cells.
- the retroviral constructs are packaging plasmids comprising at least one retroviral helper DNA sequence derived from a replication-incompetent retroviral genome encoding in trans all virion proteins required to package a replication incompetent retroviral vector, and for producing virion proteins capable of packaging the replication-incompetent retroviral vector at high titer, without the production of replication-competent helper virus.
- the retroviral DNA sequence lacks the region encoding the native enhancer and/or promoter of the viral 5’ LTR of the virus, and lacks both the psi function sequence responsible for packaging helper genome and the 3’ LTR, but encodes a foreign polyadenylation site, for example the SV40 polyadenylation site, and a foreign enhancer and/or promoter which directs efficient transcription in a cell type where virus production is desired.
- the retrovirus is a leukemia virus such as a Moloney Murine Leukemia Virus (MMLV), the Human Immunodeficiency Virus (HIV), or the Gibbon Ape Leukemia virus (GALV).
- the foreign enhancer and promoter may be the human cytomegalovirus (HCMV) immediate early (IE) enhancer and promoter, the enhancer and promoter (U3 region) of the Moloney Murine Sarcoma Virus (MMSV), the U3 region of Rous Sarcoma Virus (RSV), the U3 region of Spleen Focus Forming Virus (SFFV), or the HCMV IE enhancer joined to the native Moloney Murine Leukemia Virus (MMLV) promoter.
- HCMV human cytomegalovirus
- IE immediate early
- IE Enhancr and promoter
- U3 region of the Moloney Murine Sarcoma Virus
- RSV Rous Sarcoma Virus
- SFFV Spleen Focus Forming Virus
- HCMV IE enhancer joined to the native Moloney Murine Leukemia Virus
- the retroviral packaging plasmid may consist of two retroviral helper DNA sequences encoded by plasmid-based expression vectors, for example where a first helper sequence contains a cDNA encoding the gag and pol proteins of ecotropic MMLV or GALV and a second helper sequence contains a cDNA encoding the env protein.
- the Env gene which determines the host range, may be derived from the genes encoding xenotropic, amphotropic, ecotropic, polytropic (mink focus forming) or 10A1 murine leukemia virus env proteins, or the Gibbon Ape Leukemia Virus (GALV env protein, the Human Immunodeficiency Virus env (gp160) protein, the Vesicular Stomatitus Virus (VSV) G protein, the Human T cell leukemia (HTLV) type I and II env gene products, chimeric envelope gene derived from combinations of one or more of the above env genes or chimeric envelope genes encoding the cytoplasmic and transmembrane of the above env gene products and a monoclonal antibody directed against a specific surface molecule on a desired target cell.
- GLV env protein Gibbon Ape Leukemia Virus
- gp160 Human Immunodeficiency Virus env
- VSV Vesicular Stomatitus
- Similar vector-based systems may employ other vectors such as sleeping beauty vectors or transposon elements.
- the resulting packaged expression systems may then be introduced via an appropriate route of administration, discussed in detail with respect to the method aspects disclosed herein.
- Polypeptides [0077] Also provided is a polypeptide that includes one or more epitopes of MCPyV.
- the polypeptide is selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing.
- the amino acid sequences of these epitopes are listed in Table 1.
- the epitopes selected include an amino acid sequence having at least 90% identity with any one of the amino acid sequences listed in Table 1.
- the polypeptide comprises a combination of at least three, four, five, six, or more of the epitopes listed in Table 1.
- the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f).
- the polypeptide further comprises one or more linker sequences disposed between the epitopes.
- compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
- buffers such as neutral buffered saline, phosphate buffered saline and the like
- carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol
- proteins polypeptides or amino acids such as glycine
- antioxidants chelating agents such as EDTA or glutathione
- adjuvants e.g., aluminum hydroxide
- preservatives e.g., aluminum hydroxide
- Compositions of the disclosure may be formulated for administration as a vaccine, for example, with the inclusion of an adjuvant.
- the composition comprises a nucleic acid molecule disclosed herein.
- Methods for eliciting a Type 2 immune response directed at inflammation related to obesity, for preventing and treating inflammation in obesity, and for preventing and treating disease associated with metabolic obesity in a subject. Also provided are methods for reducing insulin resistance, reducing systemic leptin levels, and reducing tumors associated with obesity.
- diseases associated with metabolic obesity include, but are not limited to, diabetes, pre-diabetes, non-alcoholic fatty liver disease (NAFLD), metabolic-associated fatty liver disease (MAFLD), colitis, and cancer, including, for example, breast cancer.
- the method comprises administering to a subject in need thereof a composition as described herein.
- a composition as described herein Such compositions are formulated to deliver a therapeutically effective amount of the epitopes, as described herein. Determination of amounts and appropriate means of administration are determined under the guidance of a treating physician, taking into account an individual patient’s condition.
- Administration and Dosage [0084] The compositions and/or nucleic acid molecules disclosed herein are administered in any suitable manner, often with pharmaceutically acceptable carriers.
- compositions, compounds, molecules, nucleic acids, and vectors in the context of the present invention are available, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route.
- the dose administered to a patient should be sufficient to result in a beneficial therapeutic response in the patient over time, or to inhibit disease progression.
- the composition is administered to a subject in an amount sufficient to elicit an effective response and/or to alleviate, reduce, cure or at least partially arrest symptoms and/or complications from the disease.
- a therapeutically effective dose An amount adequate to accomplish this is defined as a "therapeutically effective dose.”
- Routes, order and/or frequency of administration of the therapeutic compositions disclosed herein, as well as dosage, will vary from individual to individual, and may be readily established using standard techniques. In general, an appropriate dosage and treatment regimen provides the active compound(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit. Such a response can be monitored by establishing an improved clinical outcome in treated patients as compared to non-treated patients.
- EXAMPLES [0087] The following examples are presented to illustrate the present invention and to assist one of ordinary skill in making and using the same. The examples are not intended in any way to otherwise limit the scope of the invention.
- Example 1 Type I adipose inflammation reflects an adaptive immune response driving metabolic dysregulation.
- the inflammatory infiltrate found in adipose tissue of “metabolically obese” individuals is an adaptive Type I T-cell mediated response that drives metabolic dysfunction.
- 1 Infiltration of CD8 T-cells into fat is an early event.
- 2 Type I cytokines secreted by CD8 T-cells upregulate costimulatory molecules on enlarged adipocytes which, as antigen presenting cells, provide antigenic peptides to CD4 and CD8 T-cells, further stimulating Type I T-cell activation.
- 3-5 Thus, inflamed adipocytes vaccine against adipocyte associated proteins.
- Type I CD4 T-cells become essential to maintaining inflammation. 6,7
- the resulting effector and memory T-cells generated compete for glucose and fatty acids which leads to metabolic dysfunction in both the adipose tissue and the T-cells themselves.
- 8 T-cells are not able to maintain immune surveillance
- 9 and secretion of adipokines promotes epithelial to mesenchymal and malignant transformations.
- 10,11 Losing weight does not solve the problem.
- Immunologic memory prevents inflammation from resolving even if an individual becomes normal weight. Studies in formerly obese mice show that serum cytokine expression and proinflammatory genes in the mammary fat pad are similar to those in currently obese mice and much higher than in lean controls. 12,13 Further, T-cell metabolic defects are not fully corrected with weight loss.
- Example 2 Development of a vaccine that drives Type T-cells to inflammatory adipose tissue.
- This Example demonstrates the development of a vaccine that can drive Type II T-cells to inflammatory adipose tissue.
- Validation of the signature in an independent set of 85 cases and 170 matched controls resulted in an area under the curve (AUC) of 0.812 (CI 0.755-0.870).
- Antigen identification Antigens were identified by searching on key words: [(“obese” OR “fat” OR “obesity) AND (“inflammation” OR “inflammatory” OR “immune” OR “immunogenic”) AND (“overexpression”) NOT (“macrophage” OR “neutrophil” OR “innate” OR “monocyte” OR “granulocyte” OR “myeloid” OR “dendritic”) NOT (“nervous system” OR “endocrine system” OR “muscular system” OR “hepatic”) NOT (“expressed in brown adipose tissue”) in PubMed, retrieving 19 candidates.
- Targets were prioritized if they were associated with hypoxia and/or metabolic syndrome. The final 10 candidates identified are listed in Table 3.
- Human subjects Sample collection was approved by the University of Washington (UW) Human Subjects Division.
- PBMC Peripheral blood mononuclear cells
- Analysis of peptide-specific T-cell responses Peptides, predicted to promiscuously bind human MHCII, were selected using web based algorithms as previously described (Table 3) (2). The peptides were constructed and purified by high-performance liquid chromatography (>90% pure; CPC Scientific).
- Human PBMC were evaluated by ELISPOT for antigen-specific IFN-gamma ( ⁇ ) or IL-10 production as previously described (3) using 10 ⁇ g/ml peptides.
- Mouse splenic cells were evaluated by ELISPOT for antigen-specific cytokine secretion as published, with the following modifications; splenic cells were incubated with antigens for 72hr and spots were developed with the AEC substrate kit (BD Biosciences) (2). Positive responses were defined by a statistically significant difference (p ⁇ 0.05) between the mean number of spots in the experimental wells and the mean number from no antigen control wells, all performed with 4 replicates.
- mice were fasted 4h prior to the metabolic studies.
- mice were injected with 1g glucose per kg body weight and for the insulin tolerance test mice, mice were injected with 0.75 U per kg body weight human insulin (Eli Lilly).
- Glucose concentration was measured on a glucometer (Freestyle Freedom Lite) from blood collected pre-injection and at 30, 60 and 120 min after injection.
- Lipidomics Ten mg of frozen adipose samples were homogenized in 15% water, 60% methanol, and 25% dichloromethane. The mixture was vortexed for 5 s and 25 ⁇ L of the isotope labeled internal standards mixture were added to the tube.
- the lipid molecular species were measured using multiple reaction monitoring (MRM) and positive/negative polarity switching. Positive ion mode detected lipid classes SM/DAG/CE/CER/DCER/HCER/DCER/TAG and negative ion mode detected lipid classes LPE/LPC/PC/PE/FFA. A total of 1070 lipids and fatty acids were targeted in the analysis. Data were acquired and processed using Analyst 1.6.3 and Lipidomics Workflow Manager 1.0.5.0. [0119] Aqueous metabolomics. Ten mg of frozen adipose tissue was homogenized in 80% Methanol: 9% PBS: 11% H2O. After centrifugation, aqueous extracts were collected and dried at 30°C in a Speed-Vac.
- MRM multiple reaction monitoring
- Epididymal adipose tissue was collected, weighed and put in a C-tube (Miltenyi) containing 5ml DMEM supplemented with 10mg/mL BSA, 0.03mg/mL Liberase Blendzyme 3 (Worthington) and 50 U/mL DNase, then incubated in the gentleMACs dissociator using the manufacturer’s “mTDK1” program.
- the aqueous slurry was filtered and the red blood cells were lysed.
- the washed cells (1-2x10 6 ) were incubated with a live/dead stain (FVS450; ThermoFisher), washed and incubated with an FC-receptor blocking antibody to reduce non-specific binding.
- the cells were stained with fluorochrome-conjugated monoclonal antibodies for phenotyping analyses.
- the cells were surface-stained with CD3 PE-Cy7, CD4 BV605 and CD8 APC-Cy7.
- the cells were stained with FOXP3 Alex 488 according to eBioscience‘s FOXP3 staining protocol.
- the % of CD8+ cells were analyzed among viable lymphocytes.
- FOXP3 positive cells was analyzed among CD3+CD4+ T cells. Only data from samples counting >350 cells are included.
- Vaccination Mice were immunized subcutaneously using a 26 1 ⁇ 2 G needle.
- Cultures were stimulated with 2.5nM TNFa daily for 5 days and confirmed to be inflammatory (increased activation of NFKb), insulin resistant (decreased expression of IRS-1) and had a decreased expression of adiponectin.
- Adipose tissue was collected and homogenized using a mechanical sonicator, and the lipid layer was removed. Cell lysates were separated by SDS/PAGE (4) and Western blot performed.
- Antibodies used were rabbit anti-mouse IGF-IR (2 ⁇ g/mL; Genetex), rabbit anti- mouse ATGL (1 ⁇ g/mL; Genetex), rabbit anti-mouse PAI-1 (5 ⁇ g/mL; Genetex) goat anti-mouse FABP4 (1 ⁇ g/ml; Santa Cruz Biotech), rabbit anti-mouse HIF1a (2 ⁇ g/mL; abcam), HRP- conjugated goat anti-rabbit and rabbit anti-goat (diluted 1:10,000; Invitrogen).
- Serum ELISA Measurement of Serum Amyloid A was performed with the SAA Mouse ELISA kit (Invitrogen).
- IL-6 and Leptin were performed with the respective Mouse Duoset ELISAs (Rand D Systems). Cholesterol was measured with the HDL and LDL/VLDL Quantification Kit and triglycerides were measured with the TG Quantification Kit (LS Bio). All ELISAs were performed according to the manufacturer’s directions. [0124] Statistical analysis. Data are expressed as mean + standard error of the mean (SEM). Statistical analysis was performed with Prism 7.0 or 8.0 software (GraphPad). Statistical comparisons between 2 groups were conducted by unpaired, two-tailed t-test. Statistical comparisons between 3 or more groups were conducted by one-way ANOVA followed by a Tukey posthoc analysis to determine statistical significance.
- IL-10-selective epitopes can be identified from aberrantly expressed proteins in adipose tissue in obesity. We sought to identify candidate antigens as targets for inclusion in a multi-epitope, multi-antigen anti-inflammatory vaccine. IFN- ⁇ and IL-10-inducing epitopes were determined from the candidate antigens (Table 3). We have previous determined IL-10 selective epitopes for IGF-IR (p388-402 and p545-558) and HIF1a (p276-295) (4, 5).
- One of six epitopes from FABP4 (p53--67; Fig.1A), 1/5 epitopes in DUSP1 (p29-43; Fig 1B), 2/5 epitopes from ATGL (p93-107, p213-227 and p374-388; Fig 1C) and 6/8 epitopes from PAI-1 (p94-108, p120-134, p158-172, p186-200, p270-284 and p288-302) were identified as inducing primarily IL-10 in PBMC (Fig.1D). No Th2-selective epitopes were identified in IGF-1, 11BHSDB1, SEMA3E or SOCS3. These antigens were not considered further.
- the multi-antigen adipose directed vaccine generated a selective IL-10 response to all antigens in the majority of mice.
- a pool of PAI-1-p94-108 and PAI-1-p158- 172 epitopes (p ⁇ 0.001 for all; Fig.3D), a pool of ATGL-p213-227 and ATGL-p374-388 epitopes (p ⁇ 0.01 for all; Fig.3E) or a pool of IGF-IR-p545 and IGF-IR-p388 epitopes (p ⁇ 0.0001 for all; Fig.3F) generated a significantly increased antigen-specific IL-10 response for each epitope as compared to control. No IFN- ⁇ response was detected with any epitope after vaccination.
- AdVac reduced CD8+ T cell and increased T-regulatory cell levels in the adipose tissue of obese mice.
- IL-10-secreting T-regulatory cells (Treg) which have potent anti-inflammatory effects, were present at very low levels (6).
- Treg IL-10-secreting T-regulatory cells
- AdVac did not alter serum cholesterol, triglyceride or IL-6 levels as compared to the control immunized mice.
- AdVac increased glucose sensitivity. Blood glucose concentrations were significantly lower at all time points evaluated in a GTT for the AdVac- immunized obese mice as compared to the control obese mice (p ⁇ 0.01 for all; Fig.6A). Additionally, the AUC for AdVac-immunized mice was expectedly lower than the control immunized obese mice (p ⁇ 0.0001), but was significantly higher than the AUC in the normal weight mice (p ⁇ 0.0001; Fig.6B). A similar result was observed for an insulin tolerance test (ITT).
- AdVac decreased metabolites in adipose tissue associated with glucose insensitivity and insulin resistance. Adipocytes have been shown to be regulators of glucose homeostasis and can modulate systemic energy balance through alterations in their own metabolism (Rosen, et al. Nature.2006; 14;444(7121):847-53).
- Gluconeogenesis results in the generation of glucose from non-carbohydrate carbon substrates and may aid in providing the high levels of glucose needed for the abnormal metabolism of cancer cells.
- An increase in glycolysis has been shown to be a key metabolic pathway promoting metastasis in breast cancer.
- Fig.8C-8F there were four metabolites in the top pathways that were no different than the levels observed in lean untreated mice.
- Obese TgMMTV-neu develop a greater number of tumors with a faster growth rate than lean TgMMTV-neu mice.
- Fig.9A is a Kaplan-Meier curve demonstrating percent tumor free in obese (lower line) and lean (upper line) mice at 30 weeks. Tumor growth rate (mm 3 /day) for lean or obese mice is shown in Fig.9B.
- AdVac restores insulin sensitivity, reduces systemic leptin levels and results in the development of fewer tumors in obese Tg-MMTV-neu mice. Similar to the diet induced obesity (DIO) model, we observed increased glucose sensitivity in the AdVac-immunized group as compared to the control in obese Tg-MMTV-neu mice. A GTT revealed that the glucose level in the blood before the test was significantly lower in the immunized group as compared to the control (p ⁇ 0.0001).
- the immunized group exhibited less insulin resistance than the control as there was significantly less glucose detected in the blood at every time point measured (p ⁇ 0.0001 for all).
- There was significantly less leptin detected in the serum of mice vaccinated with ADVac as compared to control (Alum; p 0.024; Fig 10B).
- Example 3 Confirming the efficacy and safety of a breast cancer prevention vaccine targeting inflammatory adipocytes.
- Two transgenic mouse models can be used to validate the efficacy of ADVac. First, one can use the TgMMTV-neu mouse, a model of luminal B breast cancer. 7,8 Since mice are not born with lesions, the goal is primary breast cancer prevention. The second model is the TgC3(1)-Tag (C3T) representing “basal like” TNBC. 9 C3T mice are born with DCIS, thus, vaccination would represent breast cancer interception.
- TgMMTV-neu mouse a model of luminal B breast cancer. 7,8 Since mice are not born with lesions, the goal is primary breast cancer prevention.
- the second model is the TgC3(1)-Tag (C3T) representing “basal like” TNBC. 9 C3T mice are born with DCIS, thus, vaccination would represent breast cancer interception.
- mice receiving ADVac will be immunized with 100 ug of each epitope 4 times at 10-14-day intervals starting when the mice become obese.
- Booster vaccines will be given every 6 weeks until study end. Mice will be sacrificed when their tumor reaches 1000mm 3 or at 52 weeks if tumor free.
- Secondary endpoints include the rate of tumor growth in mice developing tumors to ensure ADVac does not accelerate tumor proliferation and multiplicity of mammary tumor development.
- Spleens will be collected at the end of the experiment to assess ADVac antigen specific immunity using IL-10 ELISPOT. Samples of visceral and mammary adipose tissue and tumors will be collected and stored. We will include 30 mice per group. Employing a similar experimental design as described for the TgMMTV-neu model, we will test ADVac immunization in the C3T mouse model using a similar schema for the groups.
- the C3T tumors develop earlier and grow faster than those in the TgMMTV-neu so we will begin vaccination 1 week after starting the animals on the specific diets. The anticipated primary outcome is that we would prevent tumor in 25% of vaccinated mice. 10 Similar analysis will be performed as described for the TgMMTV-neu.
- Immune cells play a critical role in the propagation of adipose inflammation and generation of metabolic dysfunction. 11 We observe significant differences in the incidence of palpable tumors in the TgMMTV-neu mice immunized with ADVac at 31 weeks. For this reason, we will add 10 mice to TgMMTV-neu groups 1-4 described above to collect visceral and mammary adipose and tumor at 31 weeks for the experiments described below.
- mice cohorts We expect samples from the following mice cohorts; (1) tumor bearing lean, (2) tumor bearing obese, (3) tumor bearing obese given the adjuvant control vaccine, (4) ADVac immunized obese protected from tumor growth, and (5) ADVac immunized obese developing tumor despite vaccination.
- ECAR and OCR The relative functional relevance of ECAR and OCR in will be confirmed by forcing the cell to respire using glucose analog 2-deoxy glucose (2-DG) or oligomycin (ATP synthase inhibitor) and rotenone+antimycin A (complex I and II inhibitors) to shut down mitochondrial respiration.
- Trifluoromethoxy carbonylcyanide phenylhydrazone will be used to drive the respiratory chain to function at maximum capacity by collapsing the proton gradient for measuring the cellular spare respiratory capacity (SRC) over basal OCR.
- Etomoxir will be used to inhibit fatty acid oxidation for further confirmation of fatty acids as the primary nutrient source. Metabolic functions of T-cells will be correlated with the metabolites in corresponding tissues.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Microbiology (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Gastroenterology & Hepatology (AREA)
- Molecular Biology (AREA)
- Zoology (AREA)
- Toxicology (AREA)
- Immunology (AREA)
- Biophysics (AREA)
- Mycology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Overexpressed proteins associated with inflammatory adipocytes have been demonstrated in TNF-a inflamed adipocyte lines derived from humans and mice as well as in visceral fat derived from mice fed an inflammatory-generating high fat high sucrose diet. The adipocyte-associated proteins are immunogenic in humans and mice, and can be used as a vaccine that drives Type II T-cells to inflammatory adipose tissue. Compositions and methods for the prevention and treatment of disease associated with metabolic obesity, including cancer, such as breast cancer, and non-alcoholic fatty liver disease (NAFLD) are provided.
Description
TH2 VACCINE-BASED PREVENTION AND TREATMENT OF INFLAMMATION IN OBESITY [0001] This application claims benefit of United States provisional patent application number 63/371,999, filed August 19, 2022, the entire contents of which are incorporated by reference into this application. REFERENCE TO A SEQUENCE LISTING [0002] The content of the XML file of the sequence listing named “UW78_Seq”, which is 41 kb in size, created on August 3, 2023, and electronically submitted herewith the application, is incorporated herein by reference in its entirety. ACKNOWLEDGEMENT OF GOVERNMENT SUPPORT [0003] This invention was made with government support under Grant No. W81XWH-11-1- 0760, awarded by the U.S. Army Medical Research and Materiel Command. The government has certain rights in the invention. BACKGROUND [0004] Obesity is an important risk factor for breast cancer, especially in postmenopausal women. Obesity occurring after menopause increases the risk of developing estrogen receptor positive breast cancer by 20 -40% compared to normal weight women. The dose- response relationship between body mass index (BMI) and postmenopausal cancer consists of a 12% increased risk per 5kg/m2.1 Obesity is associated with the risk of triple negative breast cancer (TNBC) in pre-menopausal women.2 Individuals with a BMI ≥30kg/m2 were shown to have an 82% (95% CI 1.32-2.51) increased risk of triple negative breast cancer (TNBC) compared to women with a BMI ≤25kg/m2. Women with metabolic syndrome, the triad of obesity, Type II diabetes, and hypertension, may be at the highest risk.3,4 BMI is not an accurate measure of adiposity. The term “metabolic obesity” is used to describe individuals who are not obese on the basis of height and weight, but who, like people with overt obesity, are hyperinsulinemic, insulin-resistant, and predisposed to type 2 diabetes, hypertriglyceridemia, premature coronary heart disease+ and an increased risk of breast cancer.6-8 [0005] There remains a need to treat those with metabolic dysregulation that significantly increases their risk of breast cancer, as well as to effectively target obesity-related conditions, including pre-diabetes, cancers, and inflammation.
SUMMARY [0006] The material described herein meets these needs and others by providing a vaccine that drives Type II T-cells to inflammatory adipose tissue. Identified herein are overexpressed proteins associated with inflammatory adipocytes, which are immunogenic in humans and mice. Newly identified antigens, DUSP1, FABP4, PAI-1, and ATGL, and epitopes thereof, can be used in such a vaccine, as well as epitopes of IGF-1R and HIF1a identified herein. Thus, provided are, inter alia, a nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising at least two epitopes selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing. In some embodiments, the polypeptide is selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29- 43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing. In some embodiments, the polypeptide comprises each of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108 and 158-172of PAI-1; (d) amino acids 213-227 and 374- 388 of ATGL; (e) amino acids 276-295 of HIF1a; and (f) amino acids 388-402 and 545-558 of IGF-1R. The amino acid sequences of these epitopes are listed in Table 1. [0007] In some embodiments, the polypeptide is selected from amino acids 53-67 of FABP4, amino acids 29-43 of DUSP1, amino acids 276-295 of HIF1a, and a combination thereof. These epitopes are associated with more IL-10-secreting T cells. [0008] In some embodiments, the polypeptide comprises amino acids 94-108 and/or 158- 172 of PAI-1, amino acids 213-227 and/or 374-388 of ATGL, amino acids 388-402 and/or 545-558 of IGF-1R, or a combination thereof. These epitopes are associated with significantly more antigen-specific IL-10 response. [0009] In some embodiments, the polypeptide comprises one or more epitopes of (a), (b), (c), and (d) as listed above. In some embodiments, the polypeptide comprises at least three epitopes selected from (a), (b), (c), and (d). In some embodiments, the polypeptide comprises at least one of each of (a), (b), (c), and (d).
[0010] In some embodiments, the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f), and optionally further comprises one or more linker sequences disposed between the epitopes. In some embodiments, the linker sequence is GPGPG or GGGS. [0011] In some embodiments, the nucleic acid sequence comprises a promoter sequence. In some embodiments, the nucleic acid sequence comprises a heterologous sequence. In some embodiments, the heterologous sequence encodes a promoter, a transcriptional start site, a translational start site, a detectable marker, a mRNA processing splice site, a polyadenylation sequence, and/or a regulatory element. [0012] Also described herein is a vector comprising the nucleic acid molecule as described herein, wherein the vector is capable of directing expression of the encoded polypeptide(s). In some embodiments, the vector is a DNA plasmid, messenger RNA (mRNA), or viral vector. In some embodiments, the viral vector is a lentiviral vector, an adenoviral vector, or a poxviral vector. In some embodiments, the vector is mRNA. [0013] Additionally provided herein is a polypeptide comprising at least two epitopes selected from the group consisting of the epitopes listed in Table 1. In some embodiments, the epitopes selected include an amino acid sequence having at least 90% identity with any one of the amino acid sequences listed in Table 1. In some embodiments, the polypeptide comprises a combination of at least three, four, five, six, or more of the epitopes listed in Table 1. In some embodiments, the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f). In some embodiments, the polypeptide further comprises one or more linker sequences disposed between the epitopes. [0014] Also provided is a composition comprising the polypeptide described herein. In some embodiments, the composition further comprises an adjuvant. [0015] Additionally, described herein is a method for eliciting a Type 2 immune response directed at inflammation related to obesity, for preventing or treating inflammation in obesity, and/or for preventing or treating disease associated with metabolic obesity in a subject. In some embodiments, the method comprises administering to the subject a composition comprising a nucleic acid molecule, polypeptide, or composition described herein. In some embodiments, the disease associated with metabolic obesity is cancer. In some embodiments, the cancer is breast cancer. In some embodiments, the disease associated with metabolic obesity is non-alcoholic fatty liver disease (NAFLD), metabolic-associated fatty liver disease (MAFLD), or pre-diabetes. [0016] In some embodiments, the subject is human. In some embodiments, the subject is suspected of having, or has been diagnosed with obesity, a disease associated with
metabolic obesity, cancer, such as pre-diabetes/insulin insensitivity, breast cancer, MAFLD, or NAFLD. BRIEF DESCRIPTION OF THE DRAWINGS [0017] FIGS.1A-1D. IL-10-selective epitopes can be identified from aberrantly expressed proteins in adipose tissue in obesity. Th ratio when stimulated with the indicated epitope from (1A) FABP4, (1B) DUSP1 (1C) ATGL or (1D) PAI1. Bars right of “0” denote the Th-2 selective epitopes, and bars left of “0” denote the Th1 selective epitopes. n=10 healthy donors. [0018] FIGS.2A-2D. Overexpressed antigens identified in human obese adipose tissue are also overexpressed in the adipose tissue of obese mice. (2A) Representative Western blots for the indicated proteins in unstimulated differentiated mouse adipocytes or TNFa- stimulated differentiated mouse adipocytes, ***<0.001, ****p<0.0001. (2B) Relative pixel density (± SD) as measured by densitometry for the indicated proteins. (2C) Representative Western blot of cell lysate for the indicated antigen from mice fed a high fat diet (HFD) or normal chow diet (NCD). α/β Tubulin was used as a loading control. (2D) Relative pixel density (± SEM) as measured by densitometry normalized to expression observed in the tissue of mice on a NCD; n=5 mice; **p<0.05, **p<0.01, **p<0.001. [0019] FIGS.3A-3F. IL-10-secreting epitopes identified via in vitro screening generated a Th2-selective response after immunization with each single antigen. Mean (±SEM) IL-10 corrected spots per well (CSPW) in mice vaccinated with a peptide or peptide pool and splenocytes stimulated with the indicated epitope from (3A) FABP4, (3B) DUSP1, (3C) HIF1a, (3D) PAI1, (3E) ATGL and (3F) a IGF-IR presented as box plots showing all points from each individual mice. n=8-12 mice/group; **p<0.01, ***p<0.001, ****p<0.0001. [0020] FIGS.4A-4C. AdVac immunization generated an IL-10 dominant T-cell response to all antigens in the majority of mice. (4A) Corrected IL-10 spots per well (cSPW) in mice immunized with AdVac and splenocytes stimulated with the indicated antigen presented as box and whisker plots, line at median and whiskers minimum to maximum, showing all points. (4B) Percent of mice responding to the indicated antigen. (4C) Percent of mice responding to zero antigens (bottom bar), three antigens (white bar, second from bottom), five antigens (middle bar) or six antigens (uppermost bar) n=10 mice/group; *p<0.05, **p<0.01. [0021] FIG.5. Experimental design for both the male C57BL/6 diet induced obesity model and the female TgMMTV-neu mouse model of mammary cancer. wks=weeks of age.
[0022] FIGS.6A-6D. AdVac increased glucose sensitivity and reduced insulin resistance in obese mice. (6A) Glucose tolerance test (GTT) in lean mice (triangles) or obese mice immunized with adjuvant alone (Control; solid circles) or AdVac (open squares). (6B) Area under the curve (AUC) for the GTT. (6C) Insulin tolerance test (ITT) in lean mice (triangles) or obese mice immunized with control (solid circles) or the AdVac (open squares). (6D) AUC for ITT. n=20 mice/group; **p<0.01,***p<0.001, ****p<0.0001 (compared to control obese). [0023] FIGS.7A-7D. AdVac reduced CD8+ T cell and increased T-regulatory cell levels in the adipose tissue of obese mice. Percent CD8+ T-cells in all viable lymphocytes from (7A) adipose tissue or (7B) spleen from lean mice or obese mice immunized with adjuvant alone (Alum) or the AdVac vaccine. Percent CD4+/FOXP3+ T-regulatory cells from (7C) adipose tissue or (7D) spleen from lean mice or obese mice immunized with Alum or AdVac. All data are presented as box and whisker plots, horizontal line at median, whickers minimum to maximum, showing all points; n=17-20 mice/group; *p,0.05, **p<0.01, ****p<0.0001. [0024] FIGS.8A-8F. ADVac metabolically reprograms the adipose tissue microenvironment. (8A) Heatmap of metabolites in adipose tissue in obese mice immunized with adjuvant alone (Control) or ADVac. (8B) The top four metabolite pathways associated with significantly decreased metabolite levels after ADVac immunization. Normalized metabolite levels for (8C) Glutamine, (8D) Niacinamide, (8E) Glucose-1-phosphate, (8F) Glucose-6-phosphate in obese mice immunized with adjuvant only (control) or ADVac or lean untreated mice presented as box and whisker plots showing all points, horizontal line at median, whiskers minimum to maximum. n=7-10 mice/group; **p<0.01,***p<0.001, ****p<0.0001. [0025] FIGS.9A-9B. Obese TgMMTV-neu develop a greater number of tumors with a faster growth rate than lean TgMMTV-neu mice. (9A) Kaplan-Meier curve demonstrating percent tumor free in control obese (lower line) and obese mice immunized with AdVac (upper line). (9B) Tumor growth rate (mm3/day) for lean or obese mice. n=15 mice/group; **p<0.01, ***p<0.001. [0026] FIGS.10A-10C. AdVac immunization decreases CD8+ T-cells in mammary tissue, lowers serum leptin levels, and results in significant tumor inhibition in obese Tg-MMTV-neu mice. (10A) Percent CD8+ T-cells in viable lymphocytes derived from mammary fat from the indicated treatment group. (10B) Mean (±SEM) serum leptin (ng/ml) from mice immunized with the indicated vaccine. (10C) Kaplan-Meier curve demonstrating percent tumor free in control obese (lower line) and obese mice immunized with AdVac (upper line). n=7-15 mice/group; *p<0.05, **p<0.01, ***p<0.001.
DETAILED DESCRIPTION [0027] The invention described herein is based on the discovery and identification of overexpressed proteins associated with inflammatory adipocytes. The overexpression of these antigens has been demonstrated in TNF-a inflamed adipocyte lines derived from humans and mice as well as in visceral fat derived from mice fed an inflammatory-generating high fat high sucrose (HFHS) diet. The adipocyte-associated proteins are immunogenic in humans and mice. This discovery provides for the development of compositions and methods for the prevention and treatment of disease associated with metabolic obesity, including cancer, such as breast cancer, and non-alcoholic fatty liver disease (NAFLD). Definitions [0028] All scientific and technical terms used in this application have meanings commonly used in the art unless otherwise specified. As used in this application, the following words or phrases have the meanings specified. [0029] As used herein, the term “comprising” is intended to mean that the compositions and methods include the recited elements, but do not exclude others. As used herein, the transitional phrase “consisting essentially of” (and grammatical variants) is to be interpreted as encompassing the recited materials or steps “and those that do not materially affect the basic and novel characteristic(s)” of the recited embodiment. Thus, the term “consisting essentially of” as used herein should not be interpreted as equivalent to “comprising.” “Consisting of” shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions disclosed herein. Aspects defined by each of these transition terms are within the scope of the disclosure herein. [0030] As used herein, a "linker" refers to an amino acid sequence that connects two proteins, polypeptides, peptides, domains, regions, or motifs and may provide a spacer function compatible with interaction of the two sub-binding domains so that the resulting polypeptide retains a specific binding affinity to a target molecule or retains signaling activity. In certain embodiments, a linker is comprised of about two to about 35 amino acids, for instance, or about four to about 20 amino acids or about eight to about 15 amino acids or about 15 to about 25 amino acids. [0031] As used herein, the terms “nucleic acid sequence” or “polynucleotide” refers to nucleotides of any length which are deoxynucleotides (i.e. DNAs), or derivatives thereof; ribonucleotides (i.e. RNAs) or derivatives thereof; or peptide nucleic acids (PNAs) or derivatives thereof. The terms include, without limitation, single-stranded, double-stranded, or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, oligonucleotides (oligos), or other natural, synthetic, modified, mutated or non-natural forms of DNA or RNA.
[0032] The term “vector” refers to, without limitation, a recombinant genetic construct or plasmid or expression construct or expression vector that retains the ability to infect and transduce non-dividing and/or slowly-dividing cells and integrate into the target cell’s genome. The vector may be derived from or based on a wild-type virus. Aspects of this disclosure relate to an adeno-associated virus vector, an adenovirus vector, and a lentivirus vector. [0033] The term “expression control element” as used herein refers to any sequence that regulates the expression of a coding sequence, such as a gene. Exemplary expression control elements include but are not limited to promoters, enhancers, microRNAs, post- transcriptional regulatory elements, polyadenylation signal sequences, and introns. Expression control elements may be, without limitation, constitutive, inducible, repressible, or tissue-specific. A “promoter” is a control sequence that is a region of a polynucleotide sequence at which initiation and rate of transcription are controlled. It may contain genetic elements at which regulatory proteins and molecules may bind such as RNA polymerase and other transcription factors. In some embodiments, expression control by a promoter is tissue-specific. An “enhancer” is a region of DNA that can be bound by activating proteins to increase the likelihood or frequency of transcription. Non-limiting exemplary enhancers and posttranscriptional regulatory elements include the CMV enhancer and WPRE. [0034] The term “multicistronic” or “polycistronic” or “bicistronic” or tricistronic” refers to mRNA with multiple, i.e., double or triple coding areas or exons, and as such will have the capability to express from mRNA two or more, or three or more, or four or more, etc., proteins from a single construct. Multicistronic vectors simultaneously express two or more separate proteins from the same mRNA. The two strategies most widely used for constructing multicistronic configurations are through the use of 1) an IRES or 2) a 2A self- cleaving site. An “IRES” refers to an internal ribosome entry site or portion thereof of viral, prokaryotic, or eukaryotic origin which are used within polycistronic vector constructs. In some embodiments, an IRES is an RNA element that allows for translation initiation in a cap- independent manner. The term “self-cleaving peptides” or “sequences encoding self- cleaving peptides” or “2A self-cleaving site” refer to linking sequences which are used within vector constructs to incorporate sites to promote ribosomal skipping and thus to generate two polypeptides from a single promoter, such self-cleaving peptides include without limitation, T2A, and P2A peptides or sequences encoding the self-cleaving peptides. [0035] “Homology” or “identity” or “similarity” refers to sequence similarity between two peptides or between two nucleic acid molecules. Homology can be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same base or amino acid,
then the molecules are homologous at that position. A degree of homology between sequences is a function of the number of matching or homologous positions shared by the sequences. An “unrelated” or “non-homologous” sequence shares less than 40% identity, or alternatively less than 25% identity, with one of the sequences of disclosed herein. [0036] Percent similarity or percent complementary of any of the disclosed sequences may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG). The GAP program utilizes the alignment method of Needleman and Wunsch (1970). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) which are similar, divided by the total number of symbols in the shorter of the two sequences. The preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non- identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess (1986), (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps. [0037] “Nucleotide sequence” refers to a heteropolymer of deoxyribonucleotides, ribonucleotides, or peptide-nucleic acid sequences that may be assembled from smaller fragments, isolated from larger fragments, or chemically synthesized de novo or partially synthesized by combining shorter oligonucleotide linkers, or from a series of oligonucleotides. [0038] As used herein, the terms “protein”, “peptide”, and “polypeptide” refer to amino acid subunits, amino acid analogs, or peptidomimetics. The subunits may be linked by peptide bonds. In another aspect, the subunit may be linked by other bonds, e.g., ester, ether, etc. As used herein the term “amino acid” refers to either natural and/or unnatural or synthetic amino acids. [0039] As used herein, the term “recombinant expression system” or “recombinant expression vector” refers to a genetic construct for the expression of certain genetic material formed by recombination. [0040] The term "effective amount" or "therapeutically effective amount" or "prophylactically effective amount", refer to an amount of an active agent described herein that is effective to provide the desired/intended result and/or biological activity. Thus, for example, in various embodiments, an effective amount of a composition described herein is an amount that is effective to result in remission or slowing the progression of disease, and/or to improve or to ameliorate symptoms of and/or to treat disease.
[0041] When the disclosure herein relates to a small molecule, polypeptide, protein, polynucleotide, nucleic acid, oligonucleotide, antisense, or miRNA, an equivalent or a biologically equivalent of such is intended within the scope of this disclosure. As used herein, the term “biological equivalent thereof” is intended to be synonymous with “equivalent thereof” when referring to a reference small molecule, polypeptide, protein, polynucleotide, nucleic acid, oligonucleotide, antisense, or miRNA even those reference molecules having minimal homology while still maintaining desired structure or functionality. Unless specifically recited herein, it is contemplated that any nucleic acid, polynucleotide, oligonucleotide, antisense, miRNA, polypeptide, or protein mentioned herein also includes equivalents thereof. For example, an equivalent intends at least about 70% homology or identity, or at least 80 % homology or identity and alternatively, or at least about 85 %, or alternatively at least about 90 %, or alternatively at least about 95 %, or alternatively 98 % percent homology or identity and exhibits substantially equivalent biological activity to the reference protein, polypeptide or nucleic acid. Alternatively, when referring to polynucleotides, an equivalent thereof is a polynucleotide that hybridizes under stringent conditions to the reference polynucleotide or its complement. [0042] In some embodiments disclosed herein, the polypeptide and/or polynucleotide sequences are provided herein for use in gene and protein transfer and expression techniques described herein. Such sequences provided herein can be used to provide the expression product as well as substantially identical sequences that produce a protein that has the same biological properties. These “biologically equivalent” or “biologically active” or “equivalent” polypeptides are encoded by equivalent polynucleotides as described herein. They may possess at least 60%, or alternatively, at least 65%, or alternatively, at least 70%, or alternatively, at least 75%, or alternatively, at least 80%, or alternatively at least 85%, or alternatively at least 90%, or alternatively at least 95% or alternatively at least 98%, identical primary amino acid sequence to the reference polypeptide when compared using sequence identity methods run under default conditions. Specific polynucleotide or polypeptide sequences are provided as examples of particular embodiments. Modifications may be made to the amino acid sequences by using alternate amino acids that have similar charge. Additionally, an equivalent polynucleotide is one that hybridizes under stringent conditions to the reference polynucleotide or its complement or in reference to a polypeptide, a polypeptide encoded by a polynucleotide that hybridizes to the reference encoding polynucleotide under stringent conditions or its complementary strand. Alternatively, an equivalent polypeptide or protein is one that is expressed from an equivalent polynucleotide. [0043] “Hybridization” refers to a reaction in which one or more polynucleotides react to form a complex that is stabilized via hydrogen bonding between the bases of the nucleotide
residues. The hydrogen bonding may occur by Watson-Crick base pairing, Hoogstein binding, or in any other sequence-specific manner. The complex may comprise two strands forming a duplex structure, three or more strands forming a multi-stranded complex, a single self-hybridizing strand, or any combination of these. A hybridization reaction may constitute a step in a more extensive process, such as the initiation of a PC reaction, or the enzymatic cleavage of a polynucleotide by a ribozyme. [0044] Examples of stringent hybridization conditions include: incubation temperatures of about 25°C to about 37°C; hybridization buffer concentrations of about 6x SSC to about 10x SSC; formamide concentrations of about 0% to about 25%; and wash solutions from about 4x SSC to about 8x SSC. Examples of moderate hybridization conditions include: incubation temperatures of about 40°C to about 50°C; buffer concentrations of about 9x SSC to about 2x SSC; formamide concentrations of about 30% to about 50%; and wash solutions of about 5x SSC to about 2x SSC. Examples of high stringency conditions include: incubation temperatures of about 55°C to about 68°C; buffer concentrations of about lx SSC to about 0.1x SSC; formamide concentrations of about 55% to about 75%; and wash solutions of about lx SSC, 0.1x SSC, or deionized water. In general, hybridization incubation times are from 5 minutes to 24 hours, with 1, 2, or more washing steps, and wash incubation times are about 1, 2, or 15 minutes. SSC is 0.15 M NaCl and 15 mM citrate buffer. It is understood that equivalents of SSC using other buffer systems can be employed. [0045] As used herein, “treating” or “treatment” of a disease in a subject refers to (1) preventing the symptoms or disease from occurring in a subject that is predisposed or does not yet display symptoms of the disease; (2) inhibiting the disease or arresting its development; or (3) ameliorating or causing regression of the disease or the symptoms of the disease. As understood in the art, “treatment” is an approach for obtaining beneficial or desired results, including clinical results. For the purposes of the compositions, therapy, nucleic acid molecules, and methods disclosed herein for inducing an immune response, beneficial or desired results can include one or more, but are not limited to, alleviation or amelioration of one or more symptoms of disease associated with metabolic obesity, diminishment of same, stabilized (i.e., not worsening) state of the condition (including disease), delay or slowing of the condition (including disease), progression, amelioration or palliation of a condition (including disease), states of and remission of (whether partial or total) disease associated with metabolic obesity, whether detectable or undetectable. [0046] As used herein, the term "isolated" means that a naturally occurring DNA fragment, DNA molecule, coding sequence, or oligonucleotide is removed from its natural environment, or is a synthetic molecule or cloned product. Preferably, the DNA fragment, DNA molecule, coding sequence, or oligonucleotide is purified, i.e., essentially free from any other DNA
fragment, DNA molecule, coding sequence, or oligonucleotide and associated cellular products or other impurities. [0047] The term “cell” as used herein refers to either a prokaryotic or eukaryotic cell, optionally obtained from a subject or a commercially available source. Cells treated, transfected, transformed, or otherwise in contact with compositions and/or nucleic acid molecules disclosed herein, include without limitation, cells of a human, non-human animal, mammal, or non-human mammal, including without limitation, cells of murine, canine, or non-human primate species. Cells treated, transfected, transformed, or otherwise in contact with compositions and/or nucleic acid molecules disclosed herein are, without limitation, T cells, antigen-presenting cells, and other suitable host cells. [0048] As used herein, the term "subject" includes any human or non-human animal. The term "non-human animal" includes all vertebrates, e.g., mammals and non-mammals, such as non-human primates, horses, sheep, dogs, cows, pigs, chickens, and other veterinary subjects. [0049] As used herein, “a” or “an” means at least one, unless clearly indicated otherwise. [0050] As used herein, to “prevent” or “protect against” a condition or disease means to hinder, reduce or delay the onset or progression of the condition or disease. [0051] The term “encode” as it is applied to nucleic acid sequences refers to a polynucleotide which is said to “encode” a polypeptide, an mRNA, or an effector RNA if, in its native state or when manipulated by methods well known to those skilled in the art, can be transcribed and/or translated to produce the effector RNA, the mRNA, or an mRNA that can for the polypeptide and/or a fragment thereof. The antisense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom. [0052] As used herein, the term “expression” or “gene expression” refers to the process by which polynucleotides are transcribed into mRNA and/or the process by which the transcribed mRNA is subsequently translated into peptides, polypeptides, or proteins. If the polynucleotide is derived from genomic DNA, expression may include splicing of the mRNA in a eukaryotic cell. The expression level of a gene may be determined by measuring the amount of mRNA or protein in a cell or tissue sample; further, the expression level of multiple genes can be determined to establish an expression profile for a particular sample. [0053] As used herein, the term “functional” may be used to modify any molecule, biological, or cellular material to intend that it accomplishes a particular, specified effect. [0054] The term “about,” as used herein when referring to a measurable value such as an amount, level or concentration, for example and without limitation, is meant to encompass
variations of 20%, 10%, 5%, 1 %, 0.5%, or even 0.1 % of the specified amount, or fold differences in levels of a quantifiable comparison with a standard or control or reference material, such as 1-fold, 2-fold, 3-fold, 4-fold…10-fold, 100-fold, etc. of the specified level of comparison. [0055] The terms “acceptable,” “effective,” or “sufficient” when used to describe the selection of any components, ranges, dose forms, etc. disclosed herein intend that said component, range, dose form, etc. is suitable for the disclosed purpose. [0056] The term “adeno-associated virus” or “AAV” as used herein refers to a member of the class of viruses associated with this name and belonging to the genus dependoparvovirus, family Parvoviridae. Multiple serotypes of this virus are known to be suitable for gene delivery; all known serotypes can infect cells from various tissue types. At least 11 or 12, sequentially numbered, are disclosed in the prior art. Non-limiting exemplary serotypes useful in the methods disclosed herein include any of the 11 or 12 serotypes, e.g., AAV2, AAV5, and AAV8, or variant serotypes, e.g. AAV-DJ. The AAV structural particle is composed of 60 protein molecules made up of VP1, VP2, and VP3. Each particle contains approximately 5 VP1 proteins, 5 VP2 proteins and 50 VP3 proteins ordered into an icosahedral structure. [0057] ABBREVIATIONS
Compositions and Nucleic Acid Molecules [0058] Provided are compositions and/or nucleic acid molecules for preventing and treating inflammation associated with obesity, or a disease associated with metabolic obesity, in a subject. Disclosed herein is a nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide that includes one or more of the epitopes described herein. In some embodiments, the polypeptide is selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing. In some embodiments, the polypeptide comprises each of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108 and 158-172of PAI-1; (d) amino acids 213-227 and 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; and (f) amino acids 388-402 and 545- 558 of IGF-1R. The amino acid sequences of these epitopes are listed in Table 1.
Table 1: Epitopes Included in ADVac (indicated) and Candidates for Inclusion in ADVac
*The mature IGF1R protein is cleaved at 30 amino acids at the N-terminus. These amino acid residues are numbered 575-588 when referencing the pro-protein rather than the mature protein sequence. [0059] In some embodiments, the nucleic acid sequence encodes a polypeptide that comprises a combination of one or more epitopes. In some embodiments, the encoded polypeptide comprises one or more of the epitopes listed in Table 1. In some embodiments, the polypeptide comprises the first nine epitopes listed in Table 1. In some embodiments, the polypeptide comprises one or more of the epitopes listed in the latter portion of Table 1 as candidates for inclusion in an ADVac vaccine. In some embodiments, the polypeptide comprises 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more of the epitopes listed in Table 1. [0060] In some embodiments, the nucleic acid sequence optionally further comprises one or more linker sequences disposed between the epitopes. In some embodiments, the linker
sequence is glycine-serine. In some embodiments, the nucleic acid sequence comprises a promoter sequence. In some embodiments, the nucleic acid sequence comprises a heterologous sequence. In some embodiments, the heterologous sequence encodes a promoter, a transcriptional start site, a translational start site, a detectable marker, a mRNA processing splice site, a polyadenylation sequence, and/or a regulatory element. [0061] Such nucleic acid molecules may be delivered by viral or non-viral means. One example of viral delivery is adeno-associated virus (AAV). Other examples include retrovirus, lentivirus, and baculovirus delivery. One example of a non-viral method of delivery is cell penetrating peptide (CPP). Polynucleotide constructs may also be modified, such as through chemical modification, to improve their stability and/or suitability for delivery. In some embodiments, the oligonucleotide is modified by locked nucleic acids and/or phosphorothioate linkages. In some embodiments, a delivery system is selected for improved bioavailability, such as PEGylated liposomes, lipidoids, or biodegradable polymers, as examples. [0062] Viral Vectors [0063] In some embodiments, the vector disclosed herein is a viral vector. In some embodiments, the vector is an adenoviral vector, an adeno-associated viral (AAV) vector, or a lentiviral vector. In some embodiments, the vector is a retroviral vector, an adenoviral/retroviral chimera vector, a herpes simplex viral I or II vector, a parvoviral vector, a reticuloendotheliosis viral vector, a polioviral vector, a papillomaviral vector, a vaccinia viral vector, or any hybrid or chimeric vector incorporating favorable aspects of two or more viral vectors. In some embodiments, the vector further comprises one or more expression control elements operably linked to the polynucleotide. In some embodiments, the vector further comprises one or more selectable markers. [0064] In some embodiments, the vector disclosed herein is an AAV vector with low toxicity. In some embodiments, the AAV vector does not incorporate into the host genome, thereby having a low probability of causing insertional mutagenesis. In some embodiments, the AAV vector can encode a range total polynucleotides from 4.5 kb to 4.75 kb. In some embodiments, exemplary AAV vectors that may be used in any of the herein described compositions, systems, methods, and kits can include an AAV1 vector, a modified AAV1 vector, an AAV2 vector, a modified AAV2 vector, an AAV3 vector, a modified AAV3 vector, an AAV4 vector, a modified AAV4 vector, an AAV5 vector, a modified AAV5 vector, an AAV6 vector, a modified AAV6 vector, an AAV7 vector, a modified AAV7 vector, an AAV8 vector, an AAV9 vector, an AAV.rh10 vector, a modified AAV.rh10 vector, an AAV.rh32/33 vector, a modified AAV.rh32/33 vector, an
AAV.rh43 vector, a modified AAV.rh43 vector, an AAV.rh64R1 vector, and a modified AAV.rh64R1 vector and any combinations or equivalents thereof. [0065] In some embodiments, the vector disclosed herein is a lentiviral vector. In one embodiment, the lentiviral vector is an integrase-competent lentiviral vector (ICLV). In some embodiments, the lentiviral vector can refer to the transgene plasmid vector as well as the transgene plasmid vector in conjunction with related plasmids (e.g., a packaging plasmid, a rev expressing plasmid, an envelope plasmid) as well as a lentiviral-based particle capable of introducing exogenous nucleic acid into a cell through a viral or viral-like entry mechanism. Lentiviral vectors are well-known in the art. In some embodiments, exemplary lentiviral vectors that may be used in relation to any of the herein described compositions, nucleic acid molecules and/or methods, and can include a human immunodeficiency virus (HIV) 1 vector, a modified human immunodeficiency virus (HIV) 1 vector, a human immunodeficiency virus (HIV) 2 vector, a modified human immunodeficiency virus (HIV) 2 vector, a sooty mangabey simian immunodeficiency virus (SIVSM) vector, a modified sooty mangabey simian immunodeficiency virus (SIVSM) vector, a African green monkey simian immunodeficiency virus (SIVAGM) vector, a modified African green monkey simian immunodeficiency virus (SIVAGM) vector, a equine infectious anemia virus (EIAV) vector, a modified equine infectious anemia virus (EIAV) vector, a feline immunodeficiency virus (FIV) vector, a modified feline immunodeficiency virus (FIV) vector, a Visna/maedi virus (VNV/VMV) vector, a modified Visna/maedi virus (VNV/VMV) vector, a caprine arthritis-encephalitis virus (CAEV) vector, a modified caprine arthritis-encephalitis virus (CAEV) vector, a bovine immunodeficiency virus (BIV), or a modified bovine immunodeficiency virus (BIV). [0066] In some embodiments of the compositions and/or nucleic acid molecules and/or methods of the disclosure, a vector of the disclosure is a viral vector. In some embodiments, the viral vector comprises a sequence isolated or derived from a retrovirus. In some embodiments, the viral vector comprises a sequence isolated or derived from a lentivirus. In some embodiments, the viral vector comprises a sequence isolated or derived from an adenovirus. In some embodiments, the viral vector comprises a sequence isolated or derived from an adeno-associated virus (AAV). In some embodiments, the viral vector is replication incompetent. In some embodiments, the viral vector is isolated or recombinant. In some embodiments, the viral vector is self-complementary. [0067] In some embodiments of the compositions and/or nucleic acid molecules and/or methods of the disclosure, the viral vector comprises a sequence isolated or derived from an
adeno-associated virus (AAV). In some embodiments, the viral vector comprises an inverted terminal repeat sequence or a capsid sequence that is isolated or derived from an AAV of serotype AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, or AAV12, or the vector and/or components are derived from a synthetic AAV serotype, such as, without limitation, Anc80 AAV (an ancestor of AAV 1, 2, 6, 8 and 9). In some embodiments, the viral vector is replication incompetent. In some embodiments, the viral vector is isolated or recombinant (rAAV). In some embodiments, the viral vector is self-complementary (scAAV). [0068] In some embodiments of the compositions and methods of the disclosure, a vector of the disclosure is a non-viral vector. In some embodiments, the vector comprises or consists of a nanoparticle, a micelle, a liposome or lipoplex, a polymersome, a polyplex or a dendrimer. [0069] In some embodiments, expression vector or viral vector disclosed herein is used to transfect, transform, or come in contact with a cell which is a eukaryotic cell. In some embodiments, the cell is an animal cell. In some embodiments, the cell is a mammalian cell. In some embodiments, the cell is a bovine, murine, feline, equine, porcine, canine, simian, or human cell. [0070] In some embodiments, a cell is a packaging cell or a producer cell for production of a viral particle. [0071] In some embodiments, provided herein are viral particles comprising, consisting of, or consisting essentially of a vector comprising, consisting of, or consisting essentially of a polynucleotide sequence encoding one or more polypeptides as described herein. [0072] In general, methods of packaging genetic material such as RNA or DNA into one or more vectors is well known in the art. For example, the genetic material may be packaged using a packaging vector and cell lines and introduced via traditional recombinant methods. [0073] In some embodiments, the packaging vector may include, but is not limited to retroviral vector, lentiviral vector, adenoviral vector, and adeno-associated viral vector. The packaging vector contains elements and sequences that facilitate the delivery of genetic materials into cells. For example, the retroviral constructs are packaging plasmids comprising at least one retroviral helper DNA sequence derived from a replication-incompetent retroviral genome encoding in trans all virion proteins required to package a replication incompetent retroviral vector, and for producing virion proteins capable of packaging the replication-incompetent retroviral vector at high titer, without the production of replication-competent helper virus. The retroviral DNA sequence lacks the region encoding the native enhancer and/or promoter of the
viral 5’ LTR of the virus, and lacks both the psi function sequence responsible for packaging helper genome and the 3’ LTR, but encodes a foreign polyadenylation site, for example the SV40 polyadenylation site, and a foreign enhancer and/or promoter which directs efficient transcription in a cell type where virus production is desired. The retrovirus is a leukemia virus such as a Moloney Murine Leukemia Virus (MMLV), the Human Immunodeficiency Virus (HIV), or the Gibbon Ape Leukemia virus (GALV). The foreign enhancer and promoter may be the human cytomegalovirus (HCMV) immediate early (IE) enhancer and promoter, the enhancer and promoter (U3 region) of the Moloney Murine Sarcoma Virus (MMSV), the U3 region of Rous Sarcoma Virus (RSV), the U3 region of Spleen Focus Forming Virus (SFFV), or the HCMV IE enhancer joined to the native Moloney Murine Leukemia Virus (MMLV) promoter. [0074] The retroviral packaging plasmid may consist of two retroviral helper DNA sequences encoded by plasmid-based expression vectors, for example where a first helper sequence contains a cDNA encoding the gag and pol proteins of ecotropic MMLV or GALV and a second helper sequence contains a cDNA encoding the env protein. The Env gene, which determines the host range, may be derived from the genes encoding xenotropic, amphotropic, ecotropic, polytropic (mink focus forming) or 10A1 murine leukemia virus env proteins, or the Gibbon Ape Leukemia Virus (GALV env protein, the Human Immunodeficiency Virus env (gp160) protein, the Vesicular Stomatitus Virus (VSV) G protein, the Human T cell leukemia (HTLV) type I and II env gene products, chimeric envelope gene derived from combinations of one or more of the above env genes or chimeric envelope genes encoding the cytoplasmic and transmembrane of the above env gene products and a monoclonal antibody directed against a specific surface molecule on a desired target cell. Similar vector-based systems may employ other vectors such as sleeping beauty vectors or transposon elements. [0075] The resulting packaged expression systems may then be introduced via an appropriate route of administration, discussed in detail with respect to the method aspects disclosed herein. [0076] Polypeptides [0077] Also provided is a polypeptide that includes one or more epitopes of MCPyV. In some embodiments, the polypeptide is selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing. The amino acid sequences of these epitopes are listed in Table 1. In some embodiments, the epitopes selected
include an amino acid sequence having at least 90% identity with any one of the amino acid sequences listed in Table 1. In some embodiments, the polypeptide comprises a combination of at least three, four, five, six, or more of the epitopes listed in Table 1. In some embodiments, the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f). In some embodiments, the polypeptide further comprises one or more linker sequences disposed between the epitopes. [0078] Pharmaceutical compositions [0079] Pharmaceutical compositions disclosed herein include one or more pharmaceutically or physiologically acceptable carriers, diluents or excipients. Such compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives. Compositions of the disclosure may be formulated for administration as a vaccine, for example, with the inclusion of an adjuvant. [0080] For example, in some embodiments, provided is a composition comprising the polypeptide, wherein the composition further comprises an adjuvant. In some embodiments, the adjuvant elicits T cell responses. In some embodiments, the composition comprises a nucleic acid molecule disclosed herein. [0081] Methods [0082] Described herein are methods for eliciting a Type 2 immune response directed at inflammation related to obesity, for preventing and treating inflammation in obesity, and for preventing and treating disease associated with metabolic obesity in a subject. Also provided are methods for reducing insulin resistance, reducing systemic leptin levels, and reducing tumors associated with obesity. Examples of disease associated with metabolic obesity include, but are not limited to, diabetes, pre-diabetes, non-alcoholic fatty liver disease (NAFLD), metabolic-associated fatty liver disease (MAFLD), colitis, and cancer, including, for example, breast cancer. In a typical embodiment, the method comprises administering to a subject in need thereof a composition as described herein. Such compositions are formulated to deliver a therapeutically effective amount of the epitopes, as described herein. Determination of amounts and appropriate means of administration are determined under the guidance of a treating physician, taking into account an individual patient’s condition.
[0083] Administration and Dosage [0084] The compositions and/or nucleic acid molecules disclosed herein are administered in any suitable manner, often with pharmaceutically acceptable carriers. Suitable methods of administering compositions, compounds, molecules, nucleic acids, and vectors in the context of the present invention to a subject are available, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route. [0085] The dose administered to a patient, in the context of the disclosure herein, should be sufficient to result in a beneficial therapeutic response in the patient over time, or to inhibit disease progression. Thus, the composition is administered to a subject in an amount sufficient to elicit an effective response and/or to alleviate, reduce, cure or at least partially arrest symptoms and/or complications from the disease. An amount adequate to accomplish this is defined as a "therapeutically effective dose." [0086] Routes, order and/or frequency of administration of the therapeutic compositions disclosed herein, as well as dosage, will vary from individual to individual, and may be readily established using standard techniques. In general, an appropriate dosage and treatment regimen provides the active compound(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit. Such a response can be monitored by establishing an improved clinical outcome in treated patients as compared to non-treated patients. EXAMPLES [0087] The following examples are presented to illustrate the present invention and to assist one of ordinary skill in making and using the same. The examples are not intended in any way to otherwise limit the scope of the invention. [0088] Example 1: Type I adipose inflammation reflects an adaptive immune response driving metabolic dysregulation. [0089] The inflammatory infiltrate found in adipose tissue of “metabolically obese” individuals is an adaptive Type I T-cell mediated response that drives metabolic dysfunction.1 Infiltration of CD8 T-cells into fat is an early event.2 Type I cytokines secreted by CD8 T-cells upregulate costimulatory molecules on enlarged adipocytes which, as antigen presenting cells, provide antigenic peptides to CD4 and CD8 T-cells, further stimulating Type I T-cell activation.3-5 Thus,
inflamed adipocytes vaccine against adipocyte associated proteins. Type I CD4 T-cells become essential to maintaining inflammation.6,7 The resulting effector and memory T-cells generated compete for glucose and fatty acids which leads to metabolic dysfunction in both the adipose tissue and the T-cells themselves.8 T-cells are not able to maintain immune surveillance9 and secretion of adipokines promotes epithelial to mesenchymal and malignant transformations.10,11 Losing weight does not solve the problem. Immunologic memory prevents inflammation from resolving even if an individual becomes normal weight. Studies in formerly obese mice show that serum cytokine expression and proinflammatory genes in the mammary fat pad are similar to those in currently obese mice and much higher than in lean controls.12,13 Further, T-cell metabolic defects are not fully corrected with weight loss.14 Similar effects have been observed in humans. While insulin sensitivity is restored after bariatric surgery and weight loss in formerly obese individuals, serum biomarkers of systemic inflammation and pro-inflammatory gene expression in adipose tissue remain15,16 and levels of T-cells are unchanged.17 Obesogenic T- cell memory will confer long-term increases in adipose inflammation in formerly obese individuals.18,19,20 Clinical strategies that either lower Type I T-cells or increase Type II (anti- inflammatory) T-cells in inflammatory adipose would have benefit in preventing breast cancer. [0090] References [0091] 1. Mathis, D. Cell Metab 17, 851-859 (2013). [0092] 2. Nishimura, S., et al. Nat Med 15, 914-920 (2009). [0093] 3. Lumeng, C.N., Bodzin, J.L. & Saltiel, A.R. J Clin Invest 117, 175-184 (2007). [0094] 4. Xiao, L., et al. Int J Obes (Lond) 40, 112-120 (2016). [0095] 5. Missiou, A., et al. Thromb Haemost 103, 788-796 (2010). [0096] 6. Deng, T., et al. Cell Metab 17, 411-422 (2013). [0097] 7. Cho, K.W., et al. Cell Rep 9, 605-617 (2014). [0098] 8. Ringel, A.E., et al. Cell 183, 1848-1866 e1826 (2020). [0099] 9. Zhang, C., et al. Cell Metab 31, 148-161 e145 (2020). [0100] 10. Avtanski, D., et al. Cytokine 120, 155-164 (2019). [0101] 11. Tiwari, P., et al. J Exp Med 216, 1345-1358 (2019). [0102] 12. Rossi, E.L., et al. Cancer Prev Res (Phila) 9, 339-348 (2016). [0103] 13. Zamarron, B.F., et al. Diabetes 66, 392-406 (2017). [0104] 14. Rebeles, J., et al. J Infect Dis 219, 1652-1661 (2019). [0105] 15. Hagman, D.K., et al. Metabolism 70, 12-22 (2017). [0106] 16. Schmitz, J., et al. Mol Metab 5, 328-339 (2016).
[0107] 17. Garcia-Rubio, J., et al. Sci Rep 8, 15203 (2018). [0108] 18. Zou, J., et al. Cell Mol Immunol 15, 630-639 (2018). [0109] 19. Sbierski-Kind, J., et al. J Immunol 205, 45-55 (2020). [0110] 20. Cecil, D.L., et al. Breast Cancer Res Treat 139, 657-665 (2013). [0111] Example 2: Development of a vaccine that drives Type T-cells to inflammatory adipose
tissue. [0112] This Example demonstrates the development of a vaccine that can drive Type II T-cells to inflammatory adipose tissue. We have explored the potential of comprehensive metabolomics profiling and have applied an artificial intelligence approach to a large set of plasma mass spectrometry data collected from 353 breast cancer cases and 141 controls at MD Anderson Cancer Center which revealed a signature involving lipids and other metabolites discriminating cancer cases from controls. Validation of the signature in an independent set of 85 cases and 170 matched controls resulted in an area under the curve (AUC) of 0.812 (CI 0.755-0.870). These and our published data related to breast cancer signatures in circulation justify a discovery strategy focused on breast cancer risk markers associated with obesity. [0113] Antigen identification. Antigens were identified by searching on key words: [(“obese” OR “fat” OR “obesity) AND (“inflammation” OR “inflammatory” OR “immune” OR “immunogenic”) AND (“overexpression”) NOT (“macrophage” OR “neutrophil” OR “innate” OR “monocyte” OR “granulocyte” OR “myeloid” OR “dendritic”) NOT (“nervous system” OR “endocrine system” OR “muscular system” OR “hepatic”) NOT (“expressed in brown adipose tissue”) in PubMed, retrieving 19 candidates. Targets were prioritized if they were associated with hypoxia and/or metabolic syndrome. The final 10 candidates identified are listed in Table 3. [0114] Human subjects. Sample collection was approved by the University of Washington (UW) Human Subjects Division. Peripheral blood mononuclear cells (PBMC) from 10 volunteer controls (4 males and 6 females, median age: 49, range 18-79 years) were collected and cryopreserved as previously described (1). [0115] Analysis of peptide-specific T-cell responses. Peptides, predicted to promiscuously bind human MHCII, were selected using web based algorithms as previously described (Table 3) (2). The peptides were constructed and purified by high-performance liquid chromatography (>90% pure; CPC Scientific). Human PBMC were evaluated by ELISPOT for antigen-specific IFN-gamma (γ) or IL-10 production as previously described (3) using 10 μg/ml peptides. Mouse splenic cells were evaluated by ELISPOT for antigen-specific cytokine secretion as published,
with the following modifications; splenic cells were incubated with antigens for 72hr and spots were developed with the AEC substrate kit (BD Biosciences) (2). Positive responses were defined by a statistically significant difference (p<0.05) between the mean number of spots in the experimental wells and the mean number from no antigen control wells, all performed with 4 replicates. Data are reported as the mean number of spots for each experimental antigen minus the mean number of spots detected in no antigen control wells ± SEM or SD (corrected spots per well: CSPW per 2x105 PBMC or 3.5x105 splenic cells). The ratio of the product of the mean magnitude and percent incidence of response was calculated from the IFN-γ and IL-10 ELISPOT assays (IL-10 incidence X
Ratios greater than 1.4 and with ≤10% donors generating an IFN-γ response defined Th2-selective epitopes and ratios less than 1 defined Th1-selective epitopes. For ease in a graphic representation of the ratio value of Th1 epitopes, the ratio was reversed (IFN-γ/IL-10) and this value was plotted in Figure 1. [0116] Animal model. Animal care and use were in accordance with institutional guidelines. Male, C57BL/6 (Jackson Laboratory) were used in this study. Mice were fed a high fat diet (60% fat calories; Bio-serve) or normal chow from 10 weeks old. Mouse weight was measured once per week for the duration of the study. Ten week old TgMMTV-neu mice were fed a HFHS diet for 4 weeks, then randomized into 2 cohorts, one receiving the adjuvant only (Alum) and one receiving ADVac. Mice were sacrificed at 31 weeks of age or when tumor was >1000mm3. [0117] Metabolic studies. Mice were fasted 4h prior to the metabolic studies. For the glucose tolerance test, mice were injected with 1g glucose per kg body weight and for the insulin tolerance test mice, mice were injected with 0.75 U per kg body weight human insulin (Eli Lilly). Glucose concentration was measured on a glucometer (Freestyle Freedom Lite) from blood collected pre-injection and at 30, 60 and 120 min after injection. [0118] Lipidomics. Ten mg of frozen adipose samples were homogenized in 15% water, 60% methanol, and 25% dichloromethane. The mixture was vortexed for 5 s and 25 μL of the isotope labeled internal standards mixture were added to the tube. Samples were left to incubate at room temperature for 30 min. Next, another 55% water and 45% dichloromethane were added to the tube, followed by gentle vortexing for 5 s, and centrifugation at 2500×g at 15 °C for 10 min. The bottom organic layer was transferred to a new tube and 900 μL of dichloromethane was added to the original tube for a second extraction. The combined extracts were concentrated under nitrogen and reconstituted in 250 μL of the running solution (10 mM ammonium acetate in 50:50 methanol:dichloromethane). Quantitative lipidomics was performed with the Sciex Lipidyzer platform consisting of Shimadzu Nexera X2 LC-30AD pumps, a
Shimadzu Nexera X2 SIL-30AC autosampler, and a Sciex QTRAP® 5500 mass spectrometer equipped with SelexION® for differential mobility spectrometry (DMS).1-propanol was used as the chemical modifier for the DMS. Samples were introduced to the mass spectrometer by flow injection analysis at 8 uL/min. Each sample was injected twice, once with the DMS on (PC /PE/LPC/LPE/SM), and once with the DMS off (CE/CER/DAG/DCER/FFA/HCER/LCER/TAG). The lipid molecular species were measured using multiple reaction monitoring (MRM) and positive/negative polarity switching. Positive ion mode detected lipid classes SM/DAG/CE/CER/DCER/HCER/DCER/TAG and negative ion mode detected lipid classes LPE/LPC/PC/PE/FFA. A total of 1070 lipids and fatty acids were targeted in the analysis. Data were acquired and processed using Analyst 1.6.3 and Lipidomics Workflow Manager 1.0.5.0. [0119] Aqueous metabolomics. Ten mg of frozen adipose tissue was homogenized in 80% Methanol: 9% PBS: 11% H2O. After centrifugation, aqueous extracts were collected and dried at 30°C in a Speed-Vac. All samples were analyzed using an Agilent 7890 GC instrument equipped with a 5977 mass selective detector (MSD), employing an HP-5MS UI GC column. GCMS conditions were used according to the Fiehn library instructions from Agilent. Retention time of individual metabolites were annotated according to known standards. Peak intensities were normalized to total ion count (TIC) and protein concentration and depicted as fold change from control. Data were acquired and processed using MetaboAnalyst 4.0.2. [0120] Isolation of stromal vascular cells and flow cytometry. Epididymal adipose tissue was collected, weighed and put in a C-tube (Miltenyi) containing 5ml DMEM supplemented with 10mg/mL BSA, 0.03mg/mL Liberase Blendzyme 3 (Worthington) and 50 U/mL DNase, then incubated in the gentleMACs dissociator using the manufacturer’s “mTDK1” program. The aqueous slurry was filtered and the red blood cells were lysed. The washed cells (1-2x106) were incubated with a live/dead stain (FVS450; ThermoFisher), washed and incubated with an FC-receptor blocking antibody to reduce non-specific binding. After washing, the cells were stained with fluorochrome-conjugated monoclonal antibodies for phenotyping analyses. The cells were surface-stained with CD3 PE-Cy7, CD4 BV605 and CD8 APC-Cy7. After washing, the cells were stained with FOXP3 Alex 488 according to eBioscience‘s FOXP3 staining protocol. The % of CD8+ cells were analyzed among viable lymphocytes. FOXP3 positive cells was analyzed among CD3+CD4+ T cells. Only data from samples counting >350 cells are included. [0121] Vaccination. Mice were immunized subcutaneously using a 26 ½ G needle. Each mouse was injected with 50 μl of a peptide pool (100 μg each) in Imject Alum Adjuvant (Thermo
Fisher Scientific). To generate an effective immune response to an overexpressed self-antigen, four immunizations were given two weeks apart. [0122] Cell culture and protein expression.3T3-L1 pre-adipocytes were differentiated into adipocytes with 0.5mM IBMX, 1ug/ml insulin, 0.25uM dexamethasone and 2uM rosiglitazone for 10 days. 94% of the culture was positive for oil red-O. Cultures were stimulated with 2.5nM TNFa daily for 5 days and confirmed to be inflammatory (increased activation of NFKb), insulin resistant (decreased expression of IRS-1) and had a decreased expression of adiponectin. Adipose tissue was collected and homogenized using a mechanical sonicator, and the lipid layer was removed. Cell lysates were separated by SDS/PAGE (4) and Western blot performed. Antibodies used were rabbit anti-mouse IGF-IR (2 μg/mL; Genetex), rabbit anti- mouse ATGL (1 μg/mL; Genetex), rabbit anti-mouse PAI-1 (5 μg/mL; Genetex) goat anti-mouse FABP4 (1 μg/ml; Santa Cruz Biotech), rabbit anti-mouse HIF1a (2 μg/mL; abcam), HRP- conjugated goat anti-rabbit and rabbit anti-goat (diluted 1:10,000; Invitrogen). [0123] Serum ELISA. Measurement of Serum Amyloid A was performed with the SAA Mouse ELISA kit (Invitrogen). Measurement of IL-6 and Leptin was performed with the respective Mouse Duoset ELISAs (Rand D Systems). Cholesterol was measured with the HDL and LDL/VLDL Quantification Kit and triglycerides were measured with the TG Quantification Kit (LS Bio). All ELISAs were performed according to the manufacturer’s directions. [0124] Statistical analysis. Data are expressed as mean + standard error of the mean (SEM). Statistical analysis was performed with Prism 7.0 or 8.0 software (GraphPad). Statistical comparisons between 2 groups were conducted by unpaired, two-tailed t-test. Statistical comparisons between 3 or more groups were conducted by one-way ANOVA followed by a Tukey posthoc analysis to determine statistical significance. The value of p < 0.05 was considered statistically significant. [0125] Table 2: Genes Associated With Obesity, Metabolic Syndrome, and/or Type 2 Diabetes
[0126] RESULTS [0127] IL-10-selective epitopes can be identified from aberrantly expressed proteins in adipose tissue in obesity. We sought to identify candidate antigens as targets for inclusion in a multi-epitope, multi-antigen anti-inflammatory vaccine. IFN-γ and IL-10-inducing epitopes were determined from the candidate antigens (Table 3). We have previous determined IL-10 selective epitopes for IGF-IR (p388-402 and p545-558) and HIF1a (p276-295) (4, 5). One of six epitopes from FABP4 (p53--67; Fig.1A), 1/5 epitopes in DUSP1 (p29-43; Fig 1B), 2/5 epitopes from ATGL (p93-107, p213-227 and p374-388; Fig 1C) and 6/8 epitopes from PAI-1 (p94-108, p120-134, p158-172, p186-200, p270-284 and p288-302) were identified as inducing primarily IL-10 in PBMC (Fig.1D). No Th2-selective epitopes were identified in IGF-1, 11BHSDB1, SEMA3E or SOCS3. These antigens were not considered further. [0128] Table 3: Candidate Antigens, Identification Codes and Epitope Locations
[0129] We next questioned whether the candidate antigens were truly upregulated in inflammatory adipocytes. Protein expression of HIF1a, IGF-IR, PAI-1 and FABP4 were upregulated after rmTNF-a stimulation (p<0.0001 for all) compared to resting control cells (Fig. 2A and 2B). Expression of ATGL was significantly decreased after stimulation compared to control (p<0.0001). We were unable to perform protein expression for DUSP1, but we observed a significant increase in gene expression after stimulation with TNFa as compared to the non- inflammatory control (p<0.0001). In similar experiments, we questioned whether these proteins
would also be upregulated in inflammatory fat derived from obese mice, providing us with a model to test adipocyte targeting Th2 selective vaccines. Western blot analysis demonstrated that the majority of candidate antigens evaluated were overexpressed in inflammatory fat as compared to controls (Fig 2C and 2D). Western blot analysis revealed increased protein expression of HIF1a (p=0.018), PAI-1 (p<0.0001) and FABP4 (p<0.0001) compared to expression in the adipose tissue from normal weight mice. Protein expression for IGF-IR and ATGL was not increased in obese adipose tissue (Fig.2C and 2D). [0130] The multi-antigen adipose directed vaccine generated a selective IL-10 response to all antigens in the majority of mice. We initially verified that individual epitopes from each antigen, highly homologous between mouse and human (Table 4), could generate an IL-10 with little to no IFN-γ response after immunization. There were significantly more IL-10-secreting T cells observed when splenocytes from FABP4-p53-67 vaccinated mice (p<0.0001; Fig.3A), DUSP1-p29-vaccinated mice (p<0.0001; Fig.3B) or HIF1a-p276-295 vaccinated mice (p=0.0004; Fig.3C) were stimulated with the vaccinating epitope as compared to the negative control epitope HIVp52-66. Similarly, vaccination with a pool of PAI-1-p94-108 and PAI-1-p158- 172 epitopes (p<0.001 for all; Fig.3D), a pool of ATGL-p213-227 and ATGL-p374-388 epitopes (p<0.01 for all; Fig.3E) or a pool of IGF-IR-p545 and IGF-IR-p388 epitopes (p<0.0001 for all; Fig.3F) generated a significantly increased antigen-specific IL-10 response for each epitope as compared to control. No IFN-γ response was detected with any epitope after vaccination. [0131] Table 4: Homology for Candidate Antigens Identified as Vaccine Targets
[0132] Immunization with multi-antigen adipose direct vaccine, which we refer to as AdVac, generated a significant IL-10 response for each antigen as compared to the negative control epitope (p<0.01 for all; Fig.4A). There was no antigenic competition with AdVac. The median incidence of response to any antigen was 80% (range 70-90%; Fig.4B). The majority of mice
responded to all antigens. Sixty percent of mice induced a significant IL-10 response to all six antigens, while an additional 20% responded to a total of five antigens (Fig.4C). [0133] AdVac reduced CD8+ T cell and increased T-regulatory cell levels in the adipose tissue of obese mice. An evaluation of adipose infiltrating T-cell phenotypes in obese and lean individuals revealed Th1 and CD8 T-cells predominate in obese fat and are associated with insulin resistance. Furthermore, IL-10-secreting T-regulatory cells (Treg), which have potent anti-inflammatory effects, were present at very low levels (6). Thus, we evaluated CD8+ and Treg populations infiltrating the adipose tissue in immunized obese mice and compared those levels with normal weight mice. Significantly fewer CD8+ T-cells were observed in the adipose tissue of obese mice immunized with the AdVac (mean; 7±0.8%) as compared to the control (11±1.1%; p=0.0011; Fig 7A). While there were still significantly more CD8+ T cells in the adipose of AdVac-immunized obese mice as compared to the normal weight control (mean, 4.1±0.3%p=0.043), 40% of the AdVac-immunized mice had CD8+ T-cell levels within the mean and 2SD of that observed in the normal weight mice. In contrast, none of the control immunized obese mice had T-cells within this range. The decrease in CD8+ T-cells after vaccination was specific for adipose tissue as no change in this T-cell population was observed in matched spleen (Fig.7B). Though immunization with the AdVac did not increase Treg in the adipose tissue of obese mice to the levels detected in normal weight mice (56±5.6%; p=0.0062), there was a significant increase in this T-cell population in the adipose tissue of mice immunized with AdVac (27±3.7%) as compared to control mice (16±1.5%; p=0.031; Fig.7C). No changes in Treg levels were observed after vaccination in matched spleen (Fig.7D). [0134] AdVac increased glucose sensitivity and reduced insulin resistance in obese mice. AdVac did not alter serum cholesterol, triglyceride or IL-6 levels as compared to the control immunized mice. However, AdVac increased glucose sensitivity. Blood glucose concentrations were significantly lower at all time points evaluated in a GTT for the AdVac- immunized obese mice as compared to the control obese mice (p<0.01 for all; Fig.6A). Additionally, the AUC for AdVac-immunized mice was expectedly lower than the control immunized obese mice (p<0.0001), but was significantly higher than the AUC in the normal weight mice (p<0.0001; Fig.6B). A similar result was observed for an insulin tolerance test (ITT). Blood glucose was lower at all time points during the study in the mice treated with AdVac (p<0.001 for all; Fig.6C). The AUC for the mice immunized with AdVac was significantly decreased as compared to the control obese mice (p<0.0001), but was nevertheless higher than measured for the normal weight mouse (p<0.0001; Fig.6D).
[0135] AdVac decreased metabolites in adipose tissue associated with glucose insensitivity and insulin resistance. Adipocytes have been shown to be regulators of glucose homeostasis and can modulate systemic energy balance through alterations in their own metabolism (Rosen, et al. Nature.2006; 14;444(7121):847-53). We next questioned if immunization induced changes in the metabolome or lipidome of the adipose tissue. While only two lipids were significantly lower in the adipose of the immunized mice, the levels of 50 aqueous metabolites were significantly lower in tissue from mice immunized with ADVac as compared to control (Fig.8A). The top four pathways modulated by vaccination as compared to control (p>-log1.5; Fig 8B) demonstrated a decrease in metabolites associated with glycolysis, an important pathway in cancer cell metabolism and growth. In the Warburg effect, it has been observed that cancer cells do not produce energy normally through oxidative phosphorylation but rather through aerobic glycolysis, taking up high levels of glucose, followed by lactic acid fermentation. Pyruvate is the terminal product of glycolysis and nicotinamide adenine dinucleotide is an important coenzyme that regulates glycolysis. Gluconeogenesis results in the generation of glucose from non-carbohydrate carbon substrates and may aid in providing the high levels of glucose needed for the abnormal metabolism of cancer cells. An increase in glycolysis has been shown to be a key metabolic pathway promoting metastasis in breast cancer. Of note, after vaccination, there were four metabolites in the top pathways that were no different than the levels observed in lean untreated mice (Fig.8C-8F). [0136] Obese TgMMTV-neu develop a greater number of tumors with a faster growth rate than lean TgMMTV-neu mice. Fig.9A is a Kaplan-Meier curve demonstrating percent tumor free in obese (lower line) and lean (upper line) mice at 30 weeks. Tumor growth rate (mm3/day) for lean or obese mice is shown in Fig.9B. [0137] AdVac restores insulin sensitivity, reduces systemic leptin levels and results in the development of fewer tumors in obese Tg-MMTV-neu mice. Similar to the diet induced obesity (DIO) model, we observed increased glucose sensitivity in the AdVac-immunized group as compared to the control in obese Tg-MMTV-neu mice. A GTT revealed that the glucose level in the blood before the test was significantly lower in the immunized group as compared to the control (p<0.0001). The immunized group exhibited less insulin resistance than the control as there was significantly less glucose detected in the blood at every time point measured (p<0.0001 for all). After the obese mice were immunized, significantly fewer CD8+ cells were observed in the mammary adipose tissue as compared to the control obese (p=0.001; Fig.10A). There was significantly less leptin detected in the serum of mice vaccinated with ADVac as
compared to control (Alum; p=0.024; Fig 10B). The median age of tumor development was 25 weeks in the control cohort and 29 weeks in the ADVac-immunized group (p=0.009; Fig 10C). Sixty percent (9/15) of the vaccinated mice were tumor free at the planned study termination, whereas 100% of the control mice had developed tumor before the study endpoint. [0138] References [0139] 1. Disis ML, et al. J Immunol Methods.2006;308:13-8. [0140] 2. Park KH, et al. Cancer Res.2008;68:8400-9. [0141] 3. Cecil DL, et al. Cancer Res.2014;74:2710-8. [0142] 4. Cecil DL, et al. Clin Cancer Res.2017;23:3396-404. [0143] 5. Cecil DL, et.al. J Immunother Cancer 2021 Mar;9(3):e002355 [0144] 6. McLaughlin T, et al. Arterioscler Thromb Vasc Biol.2014;34:2637-43. [0145] Example 3: Confirming the efficacy and safety of a breast cancer prevention vaccine targeting inflammatory adipocytes. [0146] Two transgenic mouse models can be used to validate the efficacy of ADVac. First, one can use the TgMMTV-neu mouse, a model of luminal B breast cancer.7,8 Since mice are not born with lesions, the goal is primary breast cancer prevention. The second model is the TgC3(1)-Tag (C3T) representing “basal like” TNBC.9 C3T mice are born with DCIS, thus, vaccination would represent breast cancer interception. We have shown feeding TgMMTV-neu mice a HFHS diet leads to insulin resistance and acceleration of mammary cancer onset, with a median age of tumor development of 25 weeks in the HFHS cohort and 28.4 weeks in the normal chow cohort (p=0.0011). HSHS diet fed mice had a faster tumor growth (mean, 12.1 mm3/day) compared to tumors from lean mice (mean, 4.2 mm3/day; p=0.004 and increased tumor multiplicity. [0147] Table 5: Experimental Groups
[0148] In the TgMMTV-neu model, we will evaluate 5 experimental groups (Table 5). We include Group 5 to document that antigen specific IL-10 secreting T-cells are maintained during active immunization and to assess the immunologic efficacy of vaccine boosters. We will use antigen specific T-cell lines from Group 5 to determine the specific T-cell receptors used to recognize the antigens in ADVac. Defining specific TCRVbeta (b) will allow us to track the distribution of vaccine induced T-cells. Mice receiving ADVac will be immunized with 100 ug of each epitope 4 times at 10-14-day intervals starting when the mice become obese. Booster vaccines will be given every 6 weeks until study end. Mice will be sacrificed when their tumor reaches 1000mm3 or at 52 weeks if tumor free. We hypothesize that we will be able to duplicate a 60% protection rate. Secondary endpoints include the rate of tumor growth in mice developing tumors to ensure ADVac does not accelerate tumor proliferation and multiplicity of mammary tumor development. Spleens will be collected at the end of the experiment to assess ADVac antigen specific immunity using IL-10 ELISPOT. Samples of visceral and mammary adipose tissue and tumors will be collected and stored. We will include 30 mice per group. Employing a similar experimental design as described for the TgMMTV-neu model, we will test ADVac immunization in the C3T mouse model using a similar schema for the groups. The C3T tumors develop earlier and grow faster than those in the TgMMTV-neu so we will begin vaccination 1 week after starting the animals on the specific diets. The anticipated primary outcome is that we would prevent tumor in 25% of vaccinated mice.10 Similar analysis will be performed as described for the TgMMTV-neu. [0149] Immune cells play a critical role in the propagation of adipose inflammation and generation of metabolic dysfunction.11 We observe significant differences in the incidence of palpable tumors in the TgMMTV-neu mice immunized with ADVac at 31 weeks. For this reason, we will add 10 mice to TgMMTV-neu groups 1-4 described above to collect visceral and mammary adipose and tumor at 31 weeks for the experiments described below. We expect samples from the following mice cohorts; (1) tumor bearing lean, (2) tumor bearing obese, (3) tumor bearing obese given the adjuvant control vaccine, (4) ADVac immunized obese protected from tumor growth, and (5) ADVac immunized obese developing tumor despite vaccination. [0150] We will use mass cytometry (CyTOF) to evaluate vaccine induced changes in immune cells subsets and activation status. Using broad and unbiased cellular profiling, our goal will be to discern differences in cellular compositions or phenotypes between vaccine protected and
unprotected mice. We will also have material from tumor bearing lean and tumor bearing obese mice as comparisons. The Newell lab has extensive experience in the use of mass cytometry to study various tissues and cell types including multiple tumor types in both mouse and human.12,13 To greatly reduce batch effects, normalization beads14 and frozen aliquots of antibody cocktail15 will be used. We will employ the 10x Genomics single-cell sequencing-based multiomics approach to more deeply compare the protein and gene expression profiles between protected and unprotected mice. For these experiments, cells will be stained with a cocktail of CITE-seq antibodies (Biolegend, inclusive of all markers in the mass cytometry panel) prior to live cell FACS sorting and analysis using the 5’ 10x Genomics immune profiling platform. [0151] We will perform metabolic analysis on visceral and adipose tissue as described to identify significant differences between ADVac protected and unprotected mice. Systems level analysis of cellular metabolic function will be conducted by measuring glucose and fatty acid energy metabolism in real-time using the Agilent Seahorse XF 96 extracellular flux analyzer. Direct ex vivo samples of purified CD4 and CD8 T cells will be isolated from mammary fat, visceral fat, tumors and spleens of obese mice immunized with alum vaccine or ADVac and compared with purified T-cells from matching tissues isolated from unimmunized obese and lean control mice. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) will be measured as read-outs of mitochondrial respiration and glycolysis, respectively by the Kalia laboratory. The relative functional relevance of ECAR and OCR in will be confirmed by forcing the cell to respire using glucose analog 2-deoxy glucose (2-DG) or oligomycin (ATP synthase inhibitor) and rotenone+antimycin A (complex I and II inhibitors) to shut down mitochondrial respiration. Trifluoromethoxy carbonylcyanide phenylhydrazone will be used to drive the respiratory chain to function at maximum capacity by collapsing the proton gradient for measuring the cellular spare respiratory capacity (SRC) over basal OCR. Etomoxir will be used to inhibit fatty acid oxidation for further confirmation of fatty acids as the primary nutrient source. Metabolic functions of T-cells will be correlated with the metabolites in corresponding tissues. [0152] References [0153] 7. Guy, C.T., et al. Proc Natl Acad Sci U S A 89, 10578-10582 (1992). [0154] 8. Herschkowitz, J.I., et al. Genome Biol 8, R76 (2007). [0155] 9. Maroulakou, I.G., et al. Proc Natl Acad Sci U S A 91, 11236-11240 (1994). [0156] 10. Gad, E., et al. Breast Cancer Res Treat 148, 501-510 (2014). [0157] 11. Turbitt, W.J., et al. Immunol Rev 295, 203-219 (2020). [0158] 12. Li, S., et al. Proc Natl Acad Sci U S A 118(2021).
[0159] 13. Cheng, Y., et al. Immunity 54, 1825-1840 e1827 (2021). [0160] 14. Finck, R., et al. Cytometry A 83, 483-494 (2013). [0161] 15. Schulz, A.R., et al. Cytometry A 95, 910-916 (2019). [0162] Throughout this application various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to describe more fully the state of the art to which this invention pertains. [0163] From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.
Claims
What is claimed is: 1. A nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising at least two epitopes selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing.
2. The nucleic acid molecule of claim 1, wherein the polypeptide is selected from amino acids 53-67 of FABP4, amino acids 29-43 of DUSP1, amino acids 276-295 of HIF1a, and a combination thereof.
3. The nucleic acid molecule of claim 1, wherein the polypeptide comprises amino acids 94-108 and/or 158-172 of PAI-1, amino acids 213-227 and/or 374-388 of ATGL, amino acids 388-402 and/or 545-558 of IGF-1R, or a combination thereof.
4. The nucleic acid molecule of claim 1, wherein the polypeptide comprises at least one of each of (a), (b), (c), and (d).
5. The nucleic acid molecule of claim 1, wherein the polypeptide comprises each of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108 and 158-172 of PAI-1; (d) amino acids 213-227 and 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; and (f) amino acids 388-402 and 545-558 of IGF-1R.
6. The nucleic acid molecule of claim 1, wherein the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f), and optionally further comprises a linker sequence disposed between the epitopes.
7. The nucleic acid molecule of claim 6, wherein the linker sequence is selected from: GPGPG and GGGS.
8. The nucleic acid molecule of claim 1, wherein the nucleic acid sequence comprises a promoter sequence.
9. The nucleic acid molecule of claim 1, wherein the nucleic acid sequence comprises a heterologous sequence.
10. The nucleic acid molecule of claim 9, wherein the heterologous sequence encodes a promoter, a transcriptional start site, a translational start site, a detectable marker, a mRNA processing splice site, a polyadenylation sequence, and/or a regulatory element.
11. A vector comprising the nucleic acid molecule of any one of claims 1 to 10, wherein the vector is capable of directing expression of the encoded polypeptide(s).
12. The vector of claim 11, wherein the vector is a DNA plasmid, messenger RNA (mRNA), or viral vector.
13. The vector of claim 12, wherein the viral vector is a lentiviral vector, an adenoviral vector, or a poxviral vector.
14 The vector of claim 12, where in the vector is mRNA.
15. A polypeptide comprising at least two epitopes selected from the group consisting of: (a) amino acids 53-67 of FABP4; (b) amino acids 29-43 of DUSP1; (c) amino acids 94-108, 120-134, 158-172, 186-200, 270-284 and/or 288-302 of PAI-1; (d) amino acids 93-107, 213-227 and/or 374-388 of ATGL; (e) amino acids 276-295 of HIF1a; (f) amino acids 388-402 and/or 545-558 of IGF-1R; and (g) an amino acid sequence having at least 90% identity with any one of the foregoing.
16. A composition comprising the polypeptide of claim 15, wherein the polypeptide comprises at least one of each of (a), (b), (c), (d), (e), and (f), and optionally further comprises one or more linker sequences disposed between the epitopes.
17. A composition comprising the polypeptide of claim 15, wherein the composition further comprises an adjuvant.
18. A method for eliciting a Type 2 immune response directed at inflammation related to obesity, for preventing or treating inflammation in obesity, and/or for preventing or treating disease associated with metabolic obesity in a subject, the method comprising administering to the subject a composition comprising the nucleic acid molecule of claim 1, or the composition of claim 15.
19. The method of claim 18, wherein the disease associated with metabolic obesity is cancer.
20. The method of claim 19, wherein the cancer is breast cancer.
21. The method of claim 18, wherein the disease associated with metabolic obesity is non-alcoholic fatty liver disease (NAFLD).
22. The method of claim 18, wherein the disease associated with metabolic obesity is pre-diabetes.
23. The method of any one of claims 18-22, wherein the subject is human.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202263371999P | 2022-08-19 | 2022-08-19 | |
US63/371,999 | 2022-08-19 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2024040025A2 true WO2024040025A2 (en) | 2024-02-22 |
WO2024040025A3 WO2024040025A3 (en) | 2024-04-25 |
Family
ID=89942345
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2023/072164 WO2024040025A2 (en) | 2022-08-19 | 2023-08-14 | Th2 vaccine-based prevention and treatment of inflammation in obesity |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024040025A2 (en) |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2010504094A (en) * | 2006-09-25 | 2010-02-12 | エスジェイ バイオメッド インコーポレイテッド | Anti-obesity immunogenic hybrid polypeptide and anti-obesity vaccine composition comprising the same (Anti-ObseImmunogenetic HybridPolypeptidesAndAnti-ObaseVaccineComposingComprisingTheSame) |
WO2009149931A2 (en) * | 2008-06-11 | 2009-12-17 | Technische Universität Dresden | Fatty acid binding protein in the pathogenesis of cardiac dysfunction |
CN110366559B (en) * | 2017-02-28 | 2023-08-01 | 皮尔斯生物技术公司 | Detection and quantification of RAS-RAF-MAPK pathway proteins |
-
2023
- 2023-08-14 WO PCT/US2023/072164 patent/WO2024040025A2/en unknown
Also Published As
Publication number | Publication date |
---|---|
WO2024040025A3 (en) | 2024-04-25 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2023202423A1 (en) | Alphavirus neoantigen vectors | |
KR20210013105A (en) | Shared antigen | |
EP3402483B1 (en) | Methods and compositons for the activation of gamma-delta t-cells | |
US10688164B2 (en) | CMV vectors comprising microRNA recognition elements | |
PT2155243E (en) | Compositions and methods comprising klk3, psca, or folh1 antigen | |
Voutetakis et al. | Salivary glands as a potential gene transfer target for gene therapeutics of some monogenetic endocrine disorders | |
JP2009502150A (en) | Polynucleotides encoding MHC class I restricted hTERT epitopes, analogs or polyepitope thereof | |
EP3062824B1 (en) | A telomerase encoding dna vaccine | |
CN108135977B (en) | Antitumor immunity enhancing composition containing adenovirus simultaneously expressing IL-12 and shVEGF | |
KR20220016137A (en) | modified adenovirus | |
KR20210090650A (en) | Alphavirus neoantigen vectors and interferon inhibitors | |
KR20220041844A (en) | HIV antigen and MHC complex | |
US20220313643A1 (en) | Methods and Compositions for Treating Cancer | |
JP2019077676A (en) | Cancer vaccine for cats | |
KR20230029673A (en) | Nucleic acid artificial mini-proteome library | |
Krotova et al. | Tumor antigen-loaded AAV vaccine drives protective immunity in a melanoma animal model | |
JP5938143B2 (en) | Lentiviral vector containing MHC class I promoter | |
KR20080098585A (en) | Treatment of epstein-barr virus-associated diseases | |
WO2024040025A2 (en) | Th2 vaccine-based prevention and treatment of inflammation in obesity | |
EP2869828B1 (en) | Vault immunotherapy | |
WO2019022160A1 (en) | Nucleic acid structure for gene expression, and use thereof | |
RU2709659C1 (en) | Immunobiological agent and a method for use thereof for inducing specific immunity to the middle eastern respiratory syndrome virus (versions) | |
WO2022167831A1 (en) | Sars-cov-2 immunogenic compositions, vaccines, and methods | |
CN109923212B (en) | Lentiviral vector for expression of Hepatitis B Virus (HBV) antigen | |
WO2024196664A1 (en) | Dna vaccine targeting overexpressed antigens in colorectal cancer |
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: 23855592 Country of ref document: EP Kind code of ref document: A2 |