WO2024117232A1 - Complexe immunogène et composition pharmaceutique - Google Patents
Complexe immunogène et composition pharmaceutique Download PDFInfo
- Publication number
- WO2024117232A1 WO2024117232A1 PCT/JP2023/042983 JP2023042983W WO2024117232A1 WO 2024117232 A1 WO2024117232 A1 WO 2024117232A1 JP 2023042983 W JP2023042983 W JP 2023042983W WO 2024117232 A1 WO2024117232 A1 WO 2024117232A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- ova
- mdo
- antigen
- administration
- mannoprotein
- Prior art date
Links
- 230000002163 immunogen Effects 0.000 title claims abstract description 71
- 239000008194 pharmaceutical composition Substances 0.000 title claims abstract description 36
- 102100024023 Histone PARylation factor 1 Human genes 0.000 claims abstract description 138
- 101001047783 Homo sapiens Histone PARylation factor 1 Proteins 0.000 claims abstract description 131
- 102000036639 antigens Human genes 0.000 claims abstract description 96
- 108091007433 antigens Proteins 0.000 claims abstract description 96
- 239000000427 antigen Substances 0.000 claims abstract description 95
- 239000002105 nanoparticle Substances 0.000 claims description 112
- 206010020751 Hypersensitivity Diseases 0.000 claims description 46
- 108060003393 Granulin Proteins 0.000 claims description 31
- 239000013566 allergen Substances 0.000 claims description 21
- 230000007815 allergy Effects 0.000 claims description 20
- 239000002955 immunomodulating agent Substances 0.000 claims description 20
- 229940121354 immunomodulator Drugs 0.000 claims description 20
- 229920000057 Mannan Polymers 0.000 claims description 18
- 208000026935 allergic disease Diseases 0.000 claims description 18
- 230000002584 immunomodulator Effects 0.000 claims description 16
- 229940044601 receptor agonist Drugs 0.000 claims description 12
- 239000000018 receptor agonist Substances 0.000 claims description 12
- 208000023275 Autoimmune disease Diseases 0.000 claims description 9
- 150000004945 aromatic hydrocarbons Chemical class 0.000 claims description 4
- 229940121372 histone deacetylase inhibitor Drugs 0.000 claims description 4
- 239000003276 histone deacetylase inhibitor Substances 0.000 claims description 4
- 229940124302 mTOR inhibitor Drugs 0.000 claims description 4
- 239000003628 mammalian target of rapamycin inhibitor Substances 0.000 claims description 4
- 102000009310 vitamin D receptors Human genes 0.000 claims description 4
- 108050000156 vitamin D receptors Proteins 0.000 claims description 4
- 102000034527 Retinoid X Receptors Human genes 0.000 claims description 2
- 108010038912 Retinoid X Receptors Proteins 0.000 claims description 2
- 102000003702 retinoic acid receptors Human genes 0.000 claims description 2
- 108090000064 retinoic acid receptors Proteins 0.000 claims description 2
- 101000609767 Dromaius novaehollandiae Ovalbumin Proteins 0.000 description 138
- 238000011282 treatment Methods 0.000 description 47
- 239000000243 solution Substances 0.000 description 37
- 210000004443 dendritic cell Anatomy 0.000 description 36
- 108091006905 Human Serum Albumin Proteins 0.000 description 34
- 102000008100 Human Serum Albumin Human genes 0.000 description 34
- 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 33
- 239000002953 phosphate buffered saline Substances 0.000 description 31
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 30
- 230000001965 increasing effect Effects 0.000 description 29
- 230000002829 reductive effect Effects 0.000 description 29
- 210000005259 peripheral blood Anatomy 0.000 description 28
- 239000011886 peripheral blood Substances 0.000 description 28
- 238000010438 heat treatment Methods 0.000 description 27
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 25
- 241000699670 Mus sp. Species 0.000 description 23
- 230000000694 effects Effects 0.000 description 23
- 125000003396 thiol group Chemical group [H]S* 0.000 description 23
- 238000000034 method Methods 0.000 description 22
- 210000003651 basophil Anatomy 0.000 description 21
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 20
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 19
- 229960002930 sirolimus Drugs 0.000 description 19
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 19
- 239000002245 particle Substances 0.000 description 18
- 238000002360 preparation method Methods 0.000 description 17
- 108090000765 processed proteins & peptides Proteins 0.000 description 17
- 235000018102 proteins Nutrition 0.000 description 17
- 102000004169 proteins and genes Human genes 0.000 description 17
- 108090000623 proteins and genes Proteins 0.000 description 17
- 238000007920 subcutaneous administration Methods 0.000 description 17
- 230000004913 activation Effects 0.000 description 16
- 230000009257 reactivity Effects 0.000 description 16
- 239000011780 sodium chloride Substances 0.000 description 16
- 208000030961 allergic reaction Diseases 0.000 description 15
- 108010062580 Concanavalin A Proteins 0.000 description 14
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 14
- 239000012530 fluid Substances 0.000 description 13
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 12
- 210000001744 T-lymphocyte Anatomy 0.000 description 12
- 230000000638 stimulation Effects 0.000 description 12
- 229940037003 alum Drugs 0.000 description 11
- 210000004027 cell Anatomy 0.000 description 11
- 210000004072 lung Anatomy 0.000 description 11
- 230000003449 preventive effect Effects 0.000 description 11
- 208000035285 Allergic Seasonal Rhinitis Diseases 0.000 description 10
- 241000218645 Cedrus Species 0.000 description 10
- 229960002685 biotin Drugs 0.000 description 10
- 235000020958 biotin Nutrition 0.000 description 10
- 239000011616 biotin Substances 0.000 description 10
- 239000007853 buffer solution Substances 0.000 description 10
- 239000008363 phosphate buffer Substances 0.000 description 10
- 102000004196 processed proteins & peptides Human genes 0.000 description 10
- 239000000126 substance Substances 0.000 description 10
- 230000001225 therapeutic effect Effects 0.000 description 10
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 10
- 230000015572 biosynthetic process Effects 0.000 description 9
- 239000004615 ingredient Substances 0.000 description 9
- 239000000203 mixture Substances 0.000 description 9
- 206010048908 Seasonal allergy Diseases 0.000 description 8
- 239000000872 buffer Substances 0.000 description 8
- VHJLVAABSRFDPM-QWWZWVQMSA-N dithiothreitol Chemical compound SC[C@@H](O)[C@H](O)CS VHJLVAABSRFDPM-QWWZWVQMSA-N 0.000 description 8
- 239000002158 endotoxin Substances 0.000 description 8
- 238000002474 experimental method Methods 0.000 description 8
- 229920006008 lipopolysaccharide Polymers 0.000 description 8
- 239000007800 oxidant agent Substances 0.000 description 8
- 201000004338 pollen allergy Diseases 0.000 description 8
- 229920001184 polypeptide Polymers 0.000 description 8
- 238000012360 testing method Methods 0.000 description 8
- 238000000108 ultra-filtration Methods 0.000 description 8
- 238000011725 BALB/c mouse Methods 0.000 description 7
- 101000897042 Homo sapiens Nucleotide pyrophosphatase Proteins 0.000 description 7
- 108010002350 Interleukin-2 Proteins 0.000 description 7
- 108010031099 Mannose Receptor Proteins 0.000 description 7
- 102100021969 Nucleotide pyrophosphatase Human genes 0.000 description 7
- 230000008859 change Effects 0.000 description 7
- 238000004925 denaturation Methods 0.000 description 7
- 230000036425 denaturation Effects 0.000 description 7
- 230000001939 inductive effect Effects 0.000 description 7
- 108010005335 mannoproteins Proteins 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 238000002156 mixing Methods 0.000 description 7
- 239000013642 negative control Substances 0.000 description 7
- 239000000523 sample Substances 0.000 description 7
- 210000002966 serum Anatomy 0.000 description 7
- KIUMMUBSPKGMOY-UHFFFAOYSA-N 3,3'-Dithiobis(6-nitrobenzoic acid) Chemical compound C1=C([N+]([O-])=O)C(C(=O)O)=CC(SSC=2C=C(C(=CC=2)[N+]([O-])=O)C(O)=O)=C1 KIUMMUBSPKGMOY-UHFFFAOYSA-N 0.000 description 6
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 6
- 102000004388 Interleukin-4 Human genes 0.000 description 6
- 108090000978 Interleukin-4 Proteins 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 208000003455 anaphylaxis Diseases 0.000 description 6
- 230000027455 binding Effects 0.000 description 6
- 238000009826 distribution Methods 0.000 description 6
- 239000002552 dosage form Substances 0.000 description 6
- 229940079593 drug Drugs 0.000 description 6
- 239000003814 drug Substances 0.000 description 6
- 238000011534 incubation Methods 0.000 description 6
- 230000006698 induction Effects 0.000 description 6
- 230000008595 infiltration Effects 0.000 description 6
- 238000001764 infiltration Methods 0.000 description 6
- 229940028885 interleukin-4 Drugs 0.000 description 6
- 210000003289 regulatory T cell Anatomy 0.000 description 6
- 241000218692 Cryptomeria Species 0.000 description 5
- 108010002616 Interleukin-5 Proteins 0.000 description 5
- 108090001005 Interleukin-6 Proteins 0.000 description 5
- 102000043131 MHC class II family Human genes 0.000 description 5
- 108091054438 MHC class II family Proteins 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- 108010058846 Ovalbumin Proteins 0.000 description 5
- 239000004480 active ingredient Substances 0.000 description 5
- 239000003085 diluting agent Substances 0.000 description 5
- 239000003937 drug carrier Substances 0.000 description 5
- 239000007928 intraperitoneal injection Substances 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 210000005087 mononuclear cell Anatomy 0.000 description 5
- 229940092253 ovalbumin Drugs 0.000 description 5
- 230000004044 response Effects 0.000 description 5
- PYWVYCXTNDRMGF-UHFFFAOYSA-N rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- 239000012224 working solution Substances 0.000 description 5
- 206010002198 Anaphylactic reaction Diseases 0.000 description 4
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 4
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 4
- 108090000176 Interleukin-13 Proteins 0.000 description 4
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 4
- 102000009389 Prostaglandin D receptors Human genes 0.000 description 4
- 108050000258 Prostaglandin D receptors Proteins 0.000 description 4
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 4
- 210000001185 bone marrow Anatomy 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 4
- 210000003979 eosinophil Anatomy 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- 230000028993 immune response Effects 0.000 description 4
- 230000002401 inhibitory effect Effects 0.000 description 4
- 230000005764 inhibitory process Effects 0.000 description 4
- 238000011068 loading method Methods 0.000 description 4
- 210000000440 neutrophil Anatomy 0.000 description 4
- 239000013641 positive control Substances 0.000 description 4
- 230000001629 suppression Effects 0.000 description 4
- 206010002199 Anaphylactic shock Diseases 0.000 description 3
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 3
- 102000004127 Cytokines Human genes 0.000 description 3
- 108090000695 Cytokines Proteins 0.000 description 3
- 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 3
- 238000002965 ELISA Methods 0.000 description 3
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 3
- 108010090804 Streptavidin Proteins 0.000 description 3
- 238000002835 absorbance Methods 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 238000007818 agglutination assay Methods 0.000 description 3
- 230000002776 aggregation Effects 0.000 description 3
- 238000004220 aggregation Methods 0.000 description 3
- SHGAZHPCJJPHSC-YCNIQYBTSA-N all-trans-retinoic acid Chemical compound OC(=O)\C=C(/C)\C=C\C=C(/C)\C=C\C1=C(C)CCCC1(C)C SHGAZHPCJJPHSC-YCNIQYBTSA-N 0.000 description 3
- 201000009961 allergic asthma Diseases 0.000 description 3
- 230000030741 antigen processing and presentation Effects 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 208000006673 asthma Diseases 0.000 description 3
- 238000006664 bond formation reaction Methods 0.000 description 3
- 210000002798 bone marrow cell Anatomy 0.000 description 3
- 239000003638 chemical reducing agent Substances 0.000 description 3
- 238000001816 cooling Methods 0.000 description 3
- 239000006185 dispersion Substances 0.000 description 3
- 238000002296 dynamic light scattering Methods 0.000 description 3
- 238000002073 fluorescence micrograph Methods 0.000 description 3
- 235000013305 food Nutrition 0.000 description 3
- 210000002865 immune cell Anatomy 0.000 description 3
- 238000010348 incorporation Methods 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 239000011259 mixed solution Substances 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000007935 neutral effect Effects 0.000 description 3
- 238000007254 oxidation reaction Methods 0.000 description 3
- -1 permanganate Chemical compound 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- 239000013573 pollen allergen Substances 0.000 description 3
- 238000002203 pretreatment Methods 0.000 description 3
- 238000011002 quantification Methods 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 150000003839 salts Chemical class 0.000 description 3
- 239000012723 sample buffer Substances 0.000 description 3
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 3
- 239000002904 solvent Substances 0.000 description 3
- 238000003860 storage Methods 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 3
- CXYYHBMOVJJZTD-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) 3',6'-bis(dimethylamino)-3-oxospiro[2-benzofuran-1,9'-xanthene]-5-carboxylate Chemical compound C=1C(N(C)C)=CC=C2C=1OC1=CC(N(C)C)=CC=C1C2(C1=CC=2)OC(=O)C1=CC=2C(=O)ON1C(=O)CCC1=O CXYYHBMOVJJZTD-UHFFFAOYSA-N 0.000 description 2
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 2
- PWKSKIMOESPYIA-UHFFFAOYSA-N 2-acetamido-3-sulfanylpropanoic acid Chemical compound CC(=O)NC(CS)C(O)=O PWKSKIMOESPYIA-UHFFFAOYSA-N 0.000 description 2
- UAIUNKRWKOVEES-UHFFFAOYSA-N 3,3',5,5'-tetramethylbenzidine Chemical compound CC1=C(N)C(C)=CC(C=2C=C(C)C(N)=C(C)C=2)=C1 UAIUNKRWKOVEES-UHFFFAOYSA-N 0.000 description 2
- 102000008096 B7-H1 Antigen Human genes 0.000 description 2
- 108010074708 B7-H1 Antigen Proteins 0.000 description 2
- 238000011746 C57BL/6J (JAX™ mouse strain) Methods 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 238000008157 ELISA kit Methods 0.000 description 2
- SXRSQZLOMIGNAQ-UHFFFAOYSA-N Glutaraldehyde Chemical compound O=CCCCC=O SXRSQZLOMIGNAQ-UHFFFAOYSA-N 0.000 description 2
- 108010053070 Glutathione Disulfide Proteins 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 239000007995 HEPES buffer Substances 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 206010061218 Inflammation Diseases 0.000 description 2
- 102100037850 Interferon gamma Human genes 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 108090000174 Interleukin-10 Proteins 0.000 description 2
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 2
- 108090001090 Lectins Proteins 0.000 description 2
- 102000004856 Lectins Human genes 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 239000012901 Milli-Q water Substances 0.000 description 2
- 108090000854 Oxidoreductases Proteins 0.000 description 2
- 102000004316 Oxidoreductases Human genes 0.000 description 2
- 229930040373 Paraformaldehyde Natural products 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 229940096885 Retinoic acid receptor agonist Drugs 0.000 description 2
- 229940121908 Retinoid X receptor agonist Drugs 0.000 description 2
- 241000198071 Saccharomyces cariocanus Species 0.000 description 2
- 241001123227 Saccharomyces pastorianus Species 0.000 description 2
- 239000007983 Tris buffer Substances 0.000 description 2
- WQZGKKKJIJFFOK-PQMKYFCFSA-N alpha-D-mannose Chemical compound OC[C@H]1O[C@H](O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-PQMKYFCFSA-N 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 2
- 210000000805 cytoplasm Anatomy 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 239000000706 filtrate Substances 0.000 description 2
- 239000008103 glucose Substances 0.000 description 2
- YPZRWBKMTBYPTK-BJDJZHNGSA-N glutathione disulfide Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@H](C(=O)NCC(O)=O)CSSC[C@@H](C(=O)NCC(O)=O)NC(=O)CC[C@H](N)C(O)=O YPZRWBKMTBYPTK-BJDJZHNGSA-N 0.000 description 2
- 239000008187 granular material Substances 0.000 description 2
- 238000004128 high performance liquid chromatography Methods 0.000 description 2
- WQYVRQLZKVEZGA-UHFFFAOYSA-N hypochlorite Chemical compound Cl[O-] WQYVRQLZKVEZGA-UHFFFAOYSA-N 0.000 description 2
- 230000036737 immune function Effects 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 229960003444 immunosuppressant agent Drugs 0.000 description 2
- 230000001861 immunosuppressant effect Effects 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 238000009169 immunotherapy Methods 0.000 description 2
- 230000001976 improved effect Effects 0.000 description 2
- 230000004054 inflammatory process Effects 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 230000004073 interleukin-2 production Effects 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 239000002523 lectin Substances 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 229910001629 magnesium chloride Inorganic materials 0.000 description 2
- 229940050906 magnesium chloride hexahydrate Drugs 0.000 description 2
- DHRRIBDTHFBPNG-UHFFFAOYSA-L magnesium dichloride hexahydrate Chemical compound O.O.O.O.O.O.[Mg+2].[Cl-].[Cl-] DHRRIBDTHFBPNG-UHFFFAOYSA-L 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 229910000403 monosodium phosphate Inorganic materials 0.000 description 2
- 235000019799 monosodium phosphate Nutrition 0.000 description 2
- 239000002102 nanobead Substances 0.000 description 2
- 238000005457 optimization Methods 0.000 description 2
- 229920002866 paraformaldehyde Polymers 0.000 description 2
- OQUKIQWCVTZJAF-UHFFFAOYSA-N phenol;sulfuric acid Chemical compound OS(O)(=O)=O.OC1=CC=CC=C1 OQUKIQWCVTZJAF-UHFFFAOYSA-N 0.000 description 2
- 230000034190 positive regulation of NF-kappaB transcription factor activity Effects 0.000 description 2
- FGIUAXJPYTZDNR-UHFFFAOYSA-N potassium nitrate Chemical compound [K+].[O-][N+]([O-])=O FGIUAXJPYTZDNR-UHFFFAOYSA-N 0.000 description 2
- XJMOSONTPMZWPB-UHFFFAOYSA-M propidium iodide Chemical compound [I-].[I-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CCC[N+](C)(CC)CC)=C1C1=CC=CC=C1 XJMOSONTPMZWPB-UHFFFAOYSA-M 0.000 description 2
- 238000002731 protein assay Methods 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- AJPJDKMHJJGVTQ-UHFFFAOYSA-M sodium dihydrogen phosphate Chemical compound [Na+].OP(O)([O-])=O AJPJDKMHJJGVTQ-UHFFFAOYSA-M 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 239000012192 staining solution Substances 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 235000011149 sulphuric acid Nutrition 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 150000003573 thiols Chemical class 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- WAEXFXRVDQXREF-UHFFFAOYSA-N vorinostat Chemical group ONC(=O)CCCCCCC(=O)NC1=CC=CC=C1 WAEXFXRVDQXREF-UHFFFAOYSA-N 0.000 description 2
- 229960000237 vorinostat Drugs 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 1
- KDDXOGDIPZSCTM-UHFFFAOYSA-N 2-[1H-indol-3-yl(oxo)methyl]-4-thiazolecarboxylic acid methyl ester Chemical group COC(=O)C1=CSC(C(=O)C=2C3=CC=CC=C3NC=2)=N1 KDDXOGDIPZSCTM-UHFFFAOYSA-N 0.000 description 1
- GHCZTIFQWKKGSB-UHFFFAOYSA-N 2-hydroxypropane-1,2,3-tricarboxylic acid;phosphoric acid Chemical compound OP(O)(O)=O.OC(=O)CC(O)(C(O)=O)CC(O)=O GHCZTIFQWKKGSB-UHFFFAOYSA-N 0.000 description 1
- HJCMDXDYPOUFDY-WHFBIAKZSA-N Ala-Gln Chemical compound C[C@H](N)C(=O)N[C@H](C(O)=O)CCC(N)=O HJCMDXDYPOUFDY-WHFBIAKZSA-N 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 108010032595 Antibody Binding Sites Proteins 0.000 description 1
- 235000003261 Artemisia vulgaris Nutrition 0.000 description 1
- 240000006891 Artemisia vulgaris Species 0.000 description 1
- 241000238421 Arthropoda Species 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 208000023328 Basedow disease Diseases 0.000 description 1
- 244000274847 Betula papyrifera Species 0.000 description 1
- 235000009113 Betula papyrifera Nutrition 0.000 description 1
- 235000009109 Betula pendula Nutrition 0.000 description 1
- 235000010928 Betula populifolia Nutrition 0.000 description 1
- 235000002992 Betula pubescens Nutrition 0.000 description 1
- BTBUEUYNUDRHOZ-UHFFFAOYSA-N Borate Chemical compound [O-]B([O-])[O-] BTBUEUYNUDRHOZ-UHFFFAOYSA-N 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 238000009010 Bradford assay Methods 0.000 description 1
- 241000222120 Candida <Saccharomycetales> Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- LZZYPRNAOMGNLH-UHFFFAOYSA-M Cetrimonium bromide Chemical compound [Br-].CCCCCCCCCCCCCCCC[N+](C)(C)C LZZYPRNAOMGNLH-UHFFFAOYSA-M 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 241000218691 Cupressaceae Species 0.000 description 1
- 108010037897 DC-specific ICAM-3 grabbing nonintegrin Proteins 0.000 description 1
- 206010012438 Dermatitis atopic Diseases 0.000 description 1
- ZNZYKNKBJPZETN-WELNAUFTSA-N Dialdehyde 11678 Chemical compound N1C2=CC=CC=C2C2=C1[C@H](C[C@H](/C(=C/O)C(=O)OC)[C@@H](C=C)C=O)NCC2 ZNZYKNKBJPZETN-WELNAUFTSA-N 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 108010087819 Fc receptors Proteins 0.000 description 1
- 102000009109 Fc receptors Human genes 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 208000004262 Food Hypersensitivity Diseases 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 241000282575 Gorilla Species 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 208000015023 Graves' disease Diseases 0.000 description 1
- 208000030836 Hashimoto thyroiditis Diseases 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 1
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 1
- 102000003839 Human Proteins Human genes 0.000 description 1
- 108090000144 Human Proteins Proteins 0.000 description 1
- 102100022297 Integrin alpha-X Human genes 0.000 description 1
- 241001159781 Kazachstania spencerorum Species 0.000 description 1
- 241000039979 Kazachstania turicensis Species 0.000 description 1
- 241001123232 Kazachstania unispora Species 0.000 description 1
- 241000965982 Kazachstania zonata Species 0.000 description 1
- 239000004201 L-cysteine Substances 0.000 description 1
- 235000013878 L-cysteine Nutrition 0.000 description 1
- 241000235087 Lachancea kluyveri Species 0.000 description 1
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 1
- 238000011779 Ly5.1 mouse Methods 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 101000746372 Mus musculus Granulocyte-macrophage colony-stimulating factor Proteins 0.000 description 1
- 101001002703 Mus musculus Interleukin-4 Proteins 0.000 description 1
- 101000932477 Mus musculus Receptor-type tyrosine-protein kinase FLT3 Proteins 0.000 description 1
- 241000282339 Mustela Species 0.000 description 1
- 241001123224 Naumovozyma dairenensis Species 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 240000007594 Oryza sativa Species 0.000 description 1
- 235000007164 Oryza sativa Nutrition 0.000 description 1
- CBENFWSGALASAD-UHFFFAOYSA-N Ozone Chemical compound [O-][O+]=O CBENFWSGALASAD-UHFFFAOYSA-N 0.000 description 1
- 101150051118 PTM1 gene Proteins 0.000 description 1
- 241000282579 Pan Species 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 241000235648 Pichia Species 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 240000001341 Reynoutria japonica Species 0.000 description 1
- 235000018167 Reynoutria japonica Nutrition 0.000 description 1
- 206010039085 Rhinitis allergic Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 241000235070 Saccharomyces Species 0.000 description 1
- 241000235072 Saccharomyces bayanus Species 0.000 description 1
- 241000877399 Saccharomyces chevalieri Species 0.000 description 1
- 241001063879 Saccharomyces eubayanus Species 0.000 description 1
- 241001407717 Saccharomyces norbensis Species 0.000 description 1
- 241001123228 Saccharomyces paradoxus Species 0.000 description 1
- 241000582914 Saccharomyces uvarum Species 0.000 description 1
- 241000235343 Saccharomycetales Species 0.000 description 1
- 206010070834 Sensitisation Diseases 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 208000004732 Systemic Vasculitis Diseases 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 102000013530 TOR Serine-Threonine Kinases Human genes 0.000 description 1
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 1
- NAVMQTYZDKMPEU-UHFFFAOYSA-N Targretin Chemical group CC1=CC(C(CCC2(C)C)(C)C)=C2C=C1C(=C)C1=CC=C(C(O)=O)C=C1 NAVMQTYZDKMPEU-UHFFFAOYSA-N 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 210000004241 Th2 cell Anatomy 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 208000024780 Urticaria Diseases 0.000 description 1
- 206010047115 Vasculitis Diseases 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- QYSXJUFSXHHAJI-XFEUOLMDSA-N Vitamin D3 Natural products C1(/[C@@H]2CC[C@@H]([C@]2(CCC1)C)[C@H](C)CCCC(C)C)=C/C=C1\C[C@@H](O)CCC1=C QYSXJUFSXHHAJI-XFEUOLMDSA-N 0.000 description 1
- 238000010521 absorption reaction Methods 0.000 description 1
- 239000008351 acetate buffer Substances 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 208000037883 airway inflammation Diseases 0.000 description 1
- 239000003513 alkali Substances 0.000 description 1
- 230000009285 allergic inflammation Effects 0.000 description 1
- 201000010105 allergic rhinitis Diseases 0.000 description 1
- 201000010435 allergic urticaria Diseases 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
- 229940035676 analgesics Drugs 0.000 description 1
- 239000000730 antalgic agent Substances 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 229940121363 anti-inflammatory agent Drugs 0.000 description 1
- 239000002260 anti-inflammatory agent Substances 0.000 description 1
- 230000001754 anti-pyretic effect Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 239000002221 antipyretic Substances 0.000 description 1
- 229940125716 antipyretic agent Drugs 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 125000000613 asparagine group Chemical group N[C@@H](CC(N)=O)C(=O)* 0.000 description 1
- 201000008937 atopic dermatitis Diseases 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 235000013361 beverage Nutrition 0.000 description 1
- 229960002938 bexarotene Drugs 0.000 description 1
- 230000002902 bimodal effect Effects 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 230000036760 body temperature Effects 0.000 description 1
- 210000000621 bronchi Anatomy 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 235000011148 calcium chloride Nutrition 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 210000002421 cell wall Anatomy 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 239000007979 citrate buffer Substances 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 230000000139 costimulatory effect Effects 0.000 description 1
- 230000008878 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- 238000000586 desensitisation Methods 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 201000010099 disease Diseases 0.000 description 1
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 239000006196 drop Substances 0.000 description 1
- 239000000428 dust Substances 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 108700014844 flt3 ligand Proteins 0.000 description 1
- 235000020932 food allergy Nutrition 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 235000003599 food sweetener Nutrition 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 239000003979 granulating agent Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 238000010562 histological examination Methods 0.000 description 1
- 230000028996 humoral immune response Effects 0.000 description 1
- 239000000416 hydrocolloid Substances 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000006058 immune tolerance Effects 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 239000012678 infectious agent Substances 0.000 description 1
- 208000027866 inflammatory disease Diseases 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 230000031261 interleukin-10 production Effects 0.000 description 1
- 230000031146 intracellular signal transduction Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 238000012417 linear regression Methods 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 125000000311 mannosyl group Chemical group C1([C@@H](O)[C@@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- 241001515942 marmosets Species 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 229940035032 monophosphoryl lipid a Drugs 0.000 description 1
- 239000007923 nasal drop Substances 0.000 description 1
- 229940100662 nasal drops Drugs 0.000 description 1
- 210000005170 neoplastic cell Anatomy 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- 230000009871 nonspecific binding Effects 0.000 description 1
- 239000002417 nutraceutical Substances 0.000 description 1
- 235000021436 nutraceutical agent Nutrition 0.000 description 1
- 150000002482 oligosaccharides Polymers 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 230000001590 oxidative effect Effects 0.000 description 1
- YPZRWBKMTBYPTK-UHFFFAOYSA-N oxidized gamma-L-glutamyl-L-cysteinylglycine Natural products OC(=O)C(N)CCC(=O)NC(C(=O)NCC(O)=O)CSSCC(C(=O)NCC(O)=O)NC(=O)CCC(N)C(O)=O YPZRWBKMTBYPTK-UHFFFAOYSA-N 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- VLTRZXGMWDSKGL-UHFFFAOYSA-M perchlorate Inorganic materials [O-]Cl(=O)(=O)=O VLTRZXGMWDSKGL-UHFFFAOYSA-M 0.000 description 1
- VLTRZXGMWDSKGL-UHFFFAOYSA-N perchloric acid Chemical compound OCl(=O)(=O)=O VLTRZXGMWDSKGL-UHFFFAOYSA-N 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 239000008055 phosphate buffer solution Substances 0.000 description 1
- 230000001766 physiological effect Effects 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 235000010333 potassium nitrate Nutrition 0.000 description 1
- 239000004323 potassium nitrate Substances 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 238000004445 quantitative analysis Methods 0.000 description 1
- 239000009342 ragweed pollen Substances 0.000 description 1
- 230000035484 reaction time Effects 0.000 description 1
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 1
- 229930002330 retinoic acid Natural products 0.000 description 1
- 238000010242 retro-orbital bleeding Methods 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 235000009566 rice Nutrition 0.000 description 1
- 210000005241 right ventricle Anatomy 0.000 description 1
- 238000012502 risk assessment Methods 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 239000012488 sample solution Substances 0.000 description 1
- 230000008313 sensitization Effects 0.000 description 1
- 230000001235 sensitizing effect Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 238000005549 size reduction Methods 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 239000012086 standard solution Substances 0.000 description 1
- 230000004936 stimulating effect Effects 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 238000011421 subcutaneous treatment Methods 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000013589 supplement Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000375 suspending agent Substances 0.000 description 1
- 239000003765 sweetening agent Substances 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 125000000341 threoninyl group Chemical group [H]OC([H])(C([H])([H])[H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 210000002303 tibia Anatomy 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 210000003437 trachea Anatomy 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 235000005282 vitamin D3 Nutrition 0.000 description 1
- 239000011647 vitamin D3 Substances 0.000 description 1
- QYSXJUFSXHHAJI-YRZJJWOYSA-N vitamin D3 Chemical group C1(/[C@@H]2CC[C@@H]([C@]2(CCC1)C)[C@H](C)CCCC(C)C)=C\C=C1\C[C@@H](O)CCC1=C QYSXJUFSXHHAJI-YRZJJWOYSA-N 0.000 description 1
- 229940021056 vitamin d3 Drugs 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Images
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
-
- 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
- A61K39/35—Allergens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/62—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/69—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
Definitions
- the present invention relates to immunogenic conjugates and pharmaceutical compositions.
- This application claims priority based on Japanese Patent Application No. 2022-191916, filed on November 30, 2022, the contents of which are incorporated herein by reference.
- Allergies are inflammatory diseases in which an excessive immune response occurs against specific allergen proteins.
- the current radical treatment for allergies is desensitization therapy, which involves continuously administering small amounts of allergens and using dendritic cells (DCs) to induce regulatory T cells (Tregs) and establish immune tolerance.
- DCs dendritic cells
- Tregs regulatory T cells
- current drugs use the allergens as is, and there is a risk that the allergens will bind to IgE antibodies, causing a severe allergic reaction. Therefore, current drugs can only be administered in small amounts, which causes treatment to take a long time. If it were possible to deliver large amounts of allergens to DCs while avoiding binding to IgE antibodies, these problems could be solved.
- Non-Patent Document 1 As a ligand for MR, mannoprotein (MAN) derived from budding yeast, which contains highly branched mannose polymers and shows high affinity for MR, is considered to be effective.
- MAN mannoprotein derived from budding yeast, which contains highly branched mannose polymers and shows high affinity for MR, is considered to be effective.
- Patent Document 1 describes an immunogenic complex in which MAN is bound to an allergen via a reactive dialdehyde.
- the present invention aims to provide an immunogenic complex that has low reactivity to IgE antibodies and is highly capable of inducing tolerant DCs, and a pharmaceutical composition containing the immunogenic complex.
- the present invention includes the following aspects.
- An immunogenic complex comprising an antigen and a mannoprotein, wherein the antigen and the mannoprotein are bound by a disulfide bond.
- the immunogenic complex according to [1] wherein the immunogenic complex forms a nanoparticle comprising a shell and a core, the shell comprising the mannan chain of the mannoprotein, and the core comprising the antigen.
- the antigen is an allergen or an autoantigen.
- the immunomodulator is at least one selected from the group consisting of a retinoic acid receptor agonist, a retinoid X receptor agonist, a vitamin D receptor agonist, an aromatic hydrocarbon receptor agonist, a histone deacetylase inhibitor, and an mTOR inhibitor.
- a pharmaceutical composition comprising the immunogenic complex according to any one of [1] to [6].
- the present invention provides an immunogenic complex that has low reactivity to IgE antibodies and high ability to induce tolerant DCs, and a pharmaceutical composition containing the immunogenic complex.
- An example of a method for preparing an immunogenic complex of one embodiment is shown.
- An example of a method for preparing an immunogenic complex of one embodiment is shown.
- the results of evaluating the effect of heating temperature and heating time on the particle size of disulfide-crosslinked MAN-OVA nanoparticles (MDO) are shown. "65°C”, "75°C”, and "85°C” indicate heating temperatures.
- OVA ovalbumin; 5 min: MDO (heated for 5 minutes); 15 min: MDO (heated for 15 minutes); 25 min: MDO (heated for 25 minutes); 60 min: MDO (heated for 60 minutes).
- OVA dNP disulfide cross-linked OVA nanoparticles
- MDO(5) MDO with 5 mg/mL MAN
- MDO(10) MDO with 10 mg/mL MAN
- MDO(15) MDO with 15 mg/mL MAN
- MDO(20) MDO with 20 mg/mL MAN.
- MDO-15 w/o H 2 O 2 MDO with 15 mg/mL MAN (without hydrogen peroxide treatment); MDO-15 w H 2 O 2 : MDO with 15 mg/mL MAN (with hydrogen peroxide treatment).
- SDS treatment SDS + DDT treatment.
- OVA ovalbumin
- Man mannoprotein
- OVA dNP disulfide-crosslinked OVA nanoparticles
- MO(5) MAN-OVA nanoparticles (MO) with 5 mg/mL MAN
- MDO(5) MDO with 10 mg/mL MAN
- MO(15) MO with 15 mg/mL MAN
- OVA ovalbumin
- OVA dNP disulfide-crosslinked OVA nanoparticles
- MDO disulfide-crosslinked MAN-OVA nanoparticles
- MGO glutaraldehyde-crosslinked MAN-OVA nanoparticles.
- Fluorescence microscopy images evaluating the uptake of each nanoparticle into DCs are shown.
- the results of evaluating antigen presentation by bone marrow dendritic cells (BMDCs) stimulated with each nanoparticle, based on IL-2 production, are shown.
- the results of evaluating IL-10 production from bone marrow dendritic cells (BMDCs) stimulated with each type of nanoparticle are shown.
- ATRA reductase RALDH
- MDO (1:1): mixed with MP:OVA 1:1
- MDO (3:1): mixed with MP:OVA 3:1
- MDO (4:1): mixed with MP:OVA 4:1.
- the results of evaluating the reactivity of each nanoparticle with anti-OVA antibody are shown.
- the administration schedule of the experiment evaluating the risk of anaphylactic shock due to administration of each nanoparticle is shown below.
- Alum Alum.
- the results of evaluating the change in rectal temperature after administration of each nanoparticle as an index of anaphylactic shock are shown.
- 1 shows the administration schedule for an experiment evaluating the preventive effect of each nanoparticle on airway allergic reaction by pre-oral administration.
- the results of evaluating the preventive effect of airway allergic reactions by pre-oral administration of each nanoparticle are shown.
- the results of evaluating the preventive effect of airway allergic reactions by pre-oral administration of each nanoparticle are shown.
- the results of evaluating the preventive effect of airway allergic reactions by pre-oral administration of each nanoparticle are shown.
- 1 shows the administration schedule for an experiment evaluating the preventive effect of each nanoparticle on airway allergic reaction by pre-subcutaneous administration.
- the results of evaluating the preventive effect of airway allergic reactions by pre-subcutaneous administration of each nanoparticle are shown.
- the results of evaluating the preventive effect of airway allergic reactions by pre-subcutaneous administration of each nanoparticle are shown.
- the results of evaluating the preventive effect of airway allergic reactions by pre-subcutaneous administration of each nanoparticle are shown.
- the results of evaluating the preventive effect of airway allergic reactions by pre-subcutaneous administration of each nanoparticle are shown. 1 shows the administration schedule for an experiment evaluating the therapeutic effect of each nanoparticle on airway allergic reaction by post-oral administration.
- the results of evaluating the therapeutic effect of each nanoparticle on airway allergic reaction after post-oral administration are shown.
- the results of evaluating the therapeutic effect of each nanoparticle on airway allergic reaction after post-oral administration are shown.
- the results of evaluating the therapeutic effect of each nanoparticle on airway allergic reaction after post-oral administration are shown.
- 1 shows the administration schedule for an experiment evaluating the therapeutic effect of each nanoparticle on airway allergic reaction by post-subcutaneous administration.
- the results of evaluating the therapeutic effect of each nanoparticle on airway allergic reaction after post-subcutaneous administration are shown.
- the results of evaluating the therapeutic effect of each nanoparticle on airway allergic reaction after post-subcutaneous administration are shown.
- the results of evaluating the therapeutic effect of each nanoparticle on airway allergic reaction after post-subcutaneous administration are shown.
- the results of evaluating the therapeutic effect of each nanoparticle on airway allergic reaction after post-subcutaneous administration are shown.
- the results of evaluating the inhibition of mononuclear cell infiltration by administration of each nanoparticle are shown.
- 1 shows the administration schedule for an experiment evaluating the inhibitory effect of pre-oral administration of each nanoparticle on type IV allergic response.
- the results of evaluating the inhibitory effect of pre-oral administration of each nanoparticle on type IV allergic response are shown.
- the figures show the results of evaluating the effect of the OVA content on the particle size of disulfide-crosslinked HSA (human serum albumin)-OVA nanoparticles (HO): HO-0: HO with 0% OVA content; HO-5: HO with 5% OVA content; HO-10: HO with 10% OVA content; HO-20: HO with 20% OVA content; HO-50: HO with 50% OVA content.
- Non-reduced SDS treatment; reduced: SDS + DDT treatment.
- HO-5 HO with 5% OVA content
- HO-10 HO with 10% OVA content
- HO-20 HO with 20% OVA content
- HO-50 HO with 50% OVA content
- MHO-10 10 mg/mL disulfide-crosslinked MAN-HSA-OVA nanoparticles (MHO) of MAN.
- MHO-0 0 mg/mL MHO of MAN
- MHO-5 5 mg/mL MHO of MAN
- MHO-7.5 7.5 mg/mL MHO of MAN
- MHO-10 10 mg/mL MHO of MAN.
- Fluorescence microscopy images evaluating the uptake of each nanoparticle into DCs are shown.
- the results of evaluating the change in DC phenotype due to stimulation with each nanoparticle are shown below: (a) MHC class II (IA b ) expression; (b) CD80 expression; (c) CD86 expression; (d) PD-L1 expression.
- MHO-10-R MHO-10 + rapamycin.
- the antigen presentation of BMDCs stimulated with each nanoparticle was evaluated based on IL-2 production.
- the results of evaluating the reactivity of each nanoparticle with anti-OVA antibody are shown. 1 shows the results of a basophil activation test in peripheral blood from healthy subjects.
- the peripheral blood from healthy subjects after each stimulation was stained with CRTH2 (CD294)-FITC, CD203c-PE, and CD3-PC7, and analyzed by a flow cytometer.
- the figures show the results of a basophil activation test in peripheral blood from patients with Japanese cedar pollen allergy.
- the figures show the results of staining peripheral blood from healthy subjects with CRTH2 (CD294)-FITC, CD203c-PE, and CD3-PC7 after each stimulation and analyzing the blood using a flow cytometer.
- 1 shows the results of a basophil activation test in peripheral blood from healthy subjects and from patients with cedar pollen allergy.
- the term “comprises” means that it may contain components other than the target component.
- the term “consists of” means that it does not contain components other than the target component.
- the term “consists essentially of” means that it does not contain components other than the target component in a form that exerts a special function (such as a form that completely loses the effect of the invention). In this specification, when “comprises,” it includes aspects that "consist of” and aspects that “consist essentially of.”
- a numerical range expressed using " ⁇ ” means that the range includes the numbers written before and after " ⁇ " as the lower and upper limits.
- a first aspect of the present disclosure is an immunogenic complex.
- the immunogenic complex of this aspect includes an antigen and a mannoprotein.
- the antigen and the mannoprotein are bound by a disulfide bond.
- Antigen refers to any substance capable of inducing an immune response, such as humoral and cellular immune responses, in a target organism.
- An antigen may be any substance capable of inducing proliferation, activation, maturation, etc. of immune cells when in contact with immune cells, or any substance capable of inducing cytokine production, antibody production, etc. from immune cells.
- the antigen is a protein or peptide.
- An antigen may be a protein that may be an allergen, a protein of an infectious agent (such as a virus, bacteria, fungus, etc.), a protein derived from a neoplastic cell (such as a tumor cell), a peptide or fragment of said protein, or a recombinant protein of said protein.
- An antigen may be an autoantigen.
- the antigen is an allergen or an autoantigen.
- Autoantigen refers to a substance possessed by an individual itself that may be an antigen.
- allergen refers to a substance that can cause an allergy in an individual.
- An allergen is a substance that is recognized as a foreign substance by an individual's immune system and causes an immune response, primarily the production of immunoglobulin E (IgE).
- allergens include pollen allergens, arthropod-derived allergens, food-derived allergens, and allergens present in insect saliva, nails, or needles.
- the allergen is a pollen allergen.
- pollen allergens include allergens derived from cedar pollen, cypress pollen, white birch pollen, rice pollen, ragweed pollen, mugwort pollen, and Japanese knotweed pollen.
- Antigens may be used alone or in combination of two or more types.
- Mannoproteins are glycoproteins composed of mannan and protein. Mannan is a polysaccharide whose main component is mannose. Mannoproteins are known in nature as one of the components of the cell walls of microorganisms, particularly yeast. For example, in yeast, mannoproteins exist in a structure in which a polypeptide chain is bound to the end of ⁇ -mannan.
- mannoproteins are composed of an internal chain portion N-glycosidically linked to an asparagine residue of a polypeptide, a polymeric mannan chain portion on the outside of the internal chain portion, and an oligosaccharide chain portion O-glycosidically linked to a serine or threonine residue of a peptide.
- Mannoproteins may be obtained from natural sources or by known chemical synthesis methods. Mannoproteins may be obtained, for example, from yeast.
- yeast include Saccharomyces, Pichia, and Candida, with Saccharomyces yeasts being preferred.
- Saccharomyces yeasts include S. bayanus, S. boulardii, S. bulderi, S. cariocanus, S. cariocus, S. cerevisiae, S. chevalieri, S. dairenensis, S. S. ellipsoides, S. eubayanus, S. exigus, S. florentinus, S. kluyveri, S. martiniae, S. monacensis, S.
- yeast is S. cerevisiae.
- the method for obtaining mannoprotein is not particularly limited, and known methods can be used.
- Methods for obtaining mannoprotein from yeast include, for example, a method that utilizes the solubility of mannoprotein in water. Examples include a method of extracting mannoprotein from yeast with hot water, and a method of extracting mannoprotein from yeast with alkali.
- mannoprotein can be obtained by extracting mannoprotein from yeast with hot water, followed by precipitation with cetablon (hexadecyltrimethylammonium bromide). Commercially available mannoprotein may be used.
- the antigen and the mannoprotein are bound by a disulfide bond.
- a thiol group (-SH) of the antigen and a thiol group of the polypeptide chain of the mannoprotein are bound by a disulfide bond to form a nanoparticle.
- the antigen is a protein or peptide
- the thiol group may be a thiol group of a cysteine residue of the protein or peptide.
- the thiol group of the mannoprotein may be a thiol group of a cysteine residue of the polypeptide chain of the mannoprotein.
- Disulfide bonds can be formed between thiol groups of the antigen; between a thiol group of the antigen and a thiol group of the mannoprotein; and between thiol groups of the mannoprotein.
- the immunogenic complex of this embodiment may form nanoparticles by binding the antigen and the mannoprotein via a disulfide bond.
- the size of the nanoparticles is not particularly limited, but may have an average particle diameter of about 10 to 2000 nm, for example. The average particle diameter tends to increase when the amount of mannoprotein used is increased.
- the average particle diameter of the nanoparticles may be about 30 to 1000 nm, about 50 to 500 nm, or about 50 to 300 nm.
- the average particle diameter of the nanoparticles can be measured using a dynamic light scattering spectrophotometer.
- the particle diameter polydispersity (PDI) of the nanoparticles is not particularly limited, but may be, for example, about 0.1 to 0.8, or about 0.2 to 0.6.
- the particle diameter polydispersity of the nanoparticles can be measured using a dynamic light scattering spectrophotometer.
- the nanoparticle comprises a shell and a core.
- the shell of the nanoparticle comprises mannan chains of a mannoprotein.
- the core of the nanoparticle comprises an antigen.
- the core may further comprise a polypeptide chain of a mannoprotein.
- the core of the nanoparticle comprises an antigen and a polypeptide chain of a mannoprotein, which are linked by disulfide bonds to form the core (see FIG. 1).
- the immunogenic complex of this embodiment may contain optional components in addition to the antigen and mannoprotein, such as matrix proteins, immunomodulators, and the like.
- Microx protein refers to a protein other than the antigen of interest that forms an immunogenic complex together with the antigen and mannoprotein.
- the matrix protein can be used, for example, when the cost of the antigen is high, to allow a small amount of antigen to form an immunogenic complex.
- the matrix protein is not particularly limited. In order to prevent the induction of an unexpected immune response, it is preferable to use a protein derived from the same organism as the subject of administration of the immunogenic complex.
- a human protein can be used as the matrix protein.
- a specific example of the matrix protein is albumin.
- human serum albumin can be preferably used as the matrix protein.
- the matrix protein may be used alone or in combination of two or more kinds.
- the matrix protein is present in the core of the nanoparticle and is bound to either or both of the antigen and the polypeptide chain of the mannoprotein by disulfide bonds to form the core together with them.
- Immunomodulator means a drug that has the effect of modulating immune function.
- the immunomodulator is an immunosuppressant that suppresses immune function.
- immunomodulators include retinoic acid receptor agonists, retinoid X receptor agonists, vitamin D receptor agonists, aromatic hydrocarbon receptor agonists, histone deacetylase inhibitors, and mTOR inhibitors.
- An "agonist” is a compound that binds to a receptor and activates an intracellular signal transduction system.
- An example of a retinoic acid receptor agonist is all-trans-retinoic acid.
- An example of a retinoid X receptor agonist is bexarotene.
- An example of a vitamin D receptor agonist is vitamin D3.
- An example of an aromatic hydrocarbon receptor agonist is 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester.
- An example of a histone deacetylase inhibitor is suberoylanilide hydroxamic acid (SAHA).
- An example of an mTOR inhibitor is rapamycin.
- the immunomodulatory agent may be used alone or in combination of two or more kinds.
- the immunomodulator is thought to be present in the core of the nanoparticle due to intermolecular forces, etc.
- FIG. 1 An example of a method for producing an immunogenic complex according to an embodiment is shown in Fig. 1.
- Fig. 1 an antigen and mannoprotein (MAN) are used as materials for the immunogenic complex.
- Fig. 1 shows only one thiol group each of the antigen and mannoprotein, but each of the antigen and mannoprotein may have two or more thiol groups.
- Figure 2 shows an example of a method for producing an immunogenic complex according to one embodiment.
- an antigen, mannoprotein (MAN), and matrix protein are used as materials for the immunogenic complex.
- the antigen, mannoprotein, and matrix protein are shown to have only one thiol group each, but each may have two or more thiol groups.
- the method for producing the immunogenic complex may include, for example, a step of heating a solution in which an antigen and a mannoprotein are dissolved to form nanoparticles (hereinafter also referred to as “step (i)”), and a step of oxidizing the nanoparticles obtained in step (i) (hereinafter also referred to as “step (ii)").
- the antigen and mannoprotein are dissolved in an appropriate buffer solution to prepare a solution containing the antigen and mannoprotein.
- the buffer solution is not particularly limited, and can be one commonly used in the field of biochemistry.
- the buffer solution include phosphate buffer (PB), phosphate buffered saline (PBS), acetate buffer, citrate buffer, citrate phosphate buffer, borate buffer, tartaric acid ornamental solution, Tris buffer, HEPES buffer, etc.
- the buffer solution preferably contains a salt (e.g., sodium chloride).
- the salt concentration include 10 to 100 mM, preferably 20 to 80 mM, more preferably 30 to 70 mM, and even more preferably 40 to 60 mM.
- the pH of the buffer solution is not particularly limited, but is preferably near neutral, and preferably 6.0 to 8.0.
- a specific example of the buffer solution is 10 mM phosphate buffer solution (pH 7.4) to which 50 mM NaCl has been added.
- the mixing ratio of the antigen and the mannoprotein is not particularly limited. When the ratio of the mannoprotein is increased, the size of the nanoparticles tends to increase.
- the mannoprotein concentration in the antigen/mannoprotein solution is not particularly limited, but may be, for example, 1 to 50 mg/mL, preferably 5 to 30 mg/mL, more preferably 5 to 20 mg/mL, and even more preferably 5 to 15 mg/mL.
- the mannoprotein concentration is within the above-mentioned preferred range, the PDI tends to be small.
- the antigen concentration in the antigen/mannoprotein solution is not particularly limited, but may be, for example, 1 to 50 mg/mL, preferably 1 to 10 mg/mL, and more preferably 1 to 5 mg/mL.
- the matrix protein When a matrix protein is used, the matrix protein is dissolved in a buffer together with the antigen and mannoprotein to prepare a solution containing the antigen, mannoprotein and matrix protein (see FIG. 2).
- the total mass of the antigen and matrix protein can be considered as the mass of the antigen mentioned above.
- the ratio of the antigen to the total mass (100% by mass) of the antigen and the matrix protein is, for example, 1 to 90% by mass, and may be 3 to 80% by mass, 5 to 70% by mass, 10 to 60% by mass, 15 to 50% by mass, 15 to 40% by mass, or 20 to 40% by mass.
- the immunomodulator When an immunomodulator is used, the immunomodulator is dissolved in a buffer solution together with the antigen and mannoprotein to prepare a solution containing the antigen, mannoprotein and immunomodulator (and optionally the matrix protein).
- the amount of immunomodulator used is not particularly limited and can be appropriately selected depending on the type of immunomodulator.
- the concentration of rapamycin in the solution can be, for example, 1 to 100 ⁇ g/mL, or can be 5 to 50 ⁇ g/mL or 10 to 30 ⁇ g/mL.
- the heat treatment temperature can be set appropriately depending on the denaturation temperature of the antigen (and matrix protein).
- the heat treatment temperature can be set to a temperature at which the antigen (and matrix protein) denatures. By setting the heat treatment temperature to a temperature at which the antigen (and matrix protein) denatures, nanoparticles are stably formed. In addition, reactivity of the antigen with the antibody is suppressed.
- Specific examples of heat treatment temperatures include 60 to 90°C, with 65 to 90°C being preferred, 70 to 90°C being more preferred, and 75 to 85°C being even more preferred.
- the heat treatment time can be set to a time sufficient for denaturing the antigen (and matrix protein) at the heat treatment temperature.
- the heat treatment time can be, for example, 1 minute or more, preferably 5 minutes or more, more preferably 10 minutes or more, even more preferably 15 minutes or more, and particularly preferably 20 minutes or more.
- the treatment time may be 30 minutes or more, or may be 60 minutes or more.
- the upper limit of the treatment time is not particularly limited, but can be, for example, 100 minutes or less, or 80 minutes or less.
- step (i) the antigen and mannoprotein, and optionally the matrix protein and the immunomodulator, are aggregated in the solution.
- Pre-NP nanoparticle
- the mixture may be cooled using an ice bath or the like. Cooling can stop the formation of nanoparticles.
- the oxidation treatment of the nanoparticles (Pre-NP) can be carried out using an oxidizing agent.
- the oxidizing agent include hydrogen peroxide, oxidized glutathione (GSSG), potassium nitrate, perchlorate, hypochlorite, nitric acid, permanganate, ozone, etc.
- the oxidizing agent is hydrogen peroxide.
- the oxidizing agent may be used alone or in combination of two or more kinds.
- the amount of the oxidizing agent used can be appropriately set depending on the type of the oxidizing agent.
- the oxidizing agent is hydrogen peroxide, it can be added to the solution obtained in step (i) so that the final concentration becomes, for example, 0.1 to 1 mass %.
- the reaction conditions for the oxidation reaction may be any conditions that allow the thiol groups in the nanoparticles to form disulfide bonds.
- the reaction temperature is, for example, 0 to 30°C, preferably 1 to 20°C, more preferably 2 to 10°C, and even more preferably 3 to 5°C.
- the reaction time is, for example, 1 hour or more, preferably 5 hours or more, more preferably 8 hours or more, even more preferably 10 hours or more, and particularly preferably 12 hours or more.
- the upper limit of the treatment time is not particularly limited, but can be, for example, 30 hours or less, or 20 hours or less.
- step (ii) disulfide bonds are formed between the thiol groups in the nanoparticles. This makes it possible to obtain nanoparticles of an immunogenic complex in which the antigen and mannoprotein are bound by disulfide bonds.
- nanoparticles of the immunogenic complex may be collected by ultrafiltration or the like.
- ultrafiltration By performing ultrafiltration, free antigens, mannoproteins, oxidizing agents, matrix proteins, immunomodulators, etc. can be removed.
- an ultrafiltration membrane with a molecular weight cutoff of 100 kDa can be used.
- the recovered nanoparticles of the immunogenic complex may be freeze-dried and stored.
- the nanoparticles may be suspended in a 1% by weight glucose solution and freeze-dried.
- the immunogenic complex has a shape in which the antigen is covered with mannan chains of mannoprotein. Therefore, it is easily taken up into dendritic cells by the mannose receptor (MR) and CD209 of DC.
- the immunogenic complex taken up into DC is cleaved within the DC as the disulfide bonds are reduced to thiol groups. This releases the antigen from the immunogenic complex.
- the antigen released into the DC cell is degraded within the DC cell to a peptide of about 15 to 30 amino acid residues. This peptide is presented by MHC molecules as a T cell epitope. This induces tolerant DC.
- the immunogenic complex of this embodiment has low reactivity with antigen-specific antibodies because the antigen is not exposed. Furthermore, by thermally denaturing the antigen during the preparation of the immunogenic complex, the reactivity with antigen-specific antibodies is further reduced. This suppresses binding between the antigen and IgE antibodies. Therefore, with the immunogenic complex of this embodiment, it is possible to administer many antigens to the subject while avoiding binding with IgE antibodies.
- a second aspect of the present disclosure is a pharmaceutical composition.
- the pharmaceutical composition of this aspect contains the immunogenic complex of the first aspect.
- the pharmaceutical composition of this aspect contains the immunogenic complex of the first aspect as an active ingredient.
- the pharmaceutical composition of this embodiment may contain an optional component in addition to the immunogenic complex of the first embodiment.
- the optional component may include, for example, a pharma- ceutically acceptable carrier.
- pharmaceutical-ceutically acceptable carrier refers to a carrier that does not inhibit the physiological activity of the active ingredient and does not show substantial toxicity to the subject to which it is administered.
- not substantially toxic refers to a carrier that does not show toxicity to the subject to which it is administered at a dose that is normally used.
- the pharma-ceutically acceptable carrier is a carrier that does not inhibit the immunogenicity of the immunogenic complex of the first embodiment and does not show substantial toxicity to the subject to which it is administered.
- the pharma-ceutically acceptable carrier includes any known pharma-ceutically acceptable component that is typically considered to be an inactive component.
- the pharma-ceutically acceptable carrier is not particularly limited, but examples thereof include solvents, diluents, vehicles, excipients, flow enhancers, binders, granulating agents, dispersing agents, suspending agents, wetting agents, lubricants, disintegrants, solubilizers, stabilizers, emulsifiers, and fillers.
- the pharma- ceutical acceptable carrier may be used alone or in combination of two or more kinds.
- the pharmaceutical composition of this embodiment may contain, for example, a buffer solution as a solvent. Examples of the buffer solution include those mentioned above.
- the pharmaceutical composition may contain other ingredients in addition to the above ingredients.
- the other ingredients are not particularly limited, and any ingredient commonly used in the pharmaceutical field may be used without any particular restriction.
- examples of the other ingredients include pharmaceutical additives other than those mentioned above.
- pharmaceutical additives include, but are not limited to, preservatives (e.g., antioxidants), chelating agents, flavoring agents, sweeteners, thickeners, buffers, colorants, etc. These ingredients may be used alone or in combination of two or more kinds.
- the pharmaceutical composition may contain an active ingredient other than the immunogenic complex of the first aspect.
- the active ingredient include, but are not limited to, antiviral agents, antibiotics, anti-inflammatory agents, antipyretics, analgesics, etc.
- One type of active ingredient may be used alone, or two or more types may be used in combination.
- the pharmaceutical composition may contain an adjuvant.
- adjuvants include, but are not limited to, aluminum hydroxide, calcium phosphate, monophosphoryl lipid A, chitosan, etc.
- One type of adjuvant may be used alone, or two or more types may be used in combination.
- the dosage form of the pharmaceutical composition is not particularly limited, and may be any dosage form commonly used as a pharmaceutical preparation.
- the pharmaceutical composition of this embodiment may be an oral preparation or a parenteral preparation.
- Oral preparations include, for example, tablets, coated tablets, pills, powders, granules, capsules, syrups, fine granules, liquids, drops, emulsions, etc.
- Parenteral preparations include, for example, injections, suppositories, nasal drops, enteral preparations, inhalants, etc.
- Pharmaceutical compositions of these dosage forms may be formulated according to standard methods (for example, methods described in the Japanese Pharmacopoeia).
- the route of administration of the pharmaceutical composition of this embodiment is not particularly limited, and may be oral or parenteral.
- parenteral administration include sublingual administration, intravenous administration, intranasal administration, subcutaneous administration, intradermal administration, intramuscular administration, intraperitoneal administration, and enteral administration.
- the pharmaceutical composition can be administered in a therapeutically effective amount of the immunogenic complex of the first aspect.
- therapeutically effective amount means an amount of a drug effective for treating or preventing a target disease.
- a therapeutically effective amount of an immunogenic complex can be an amount effective for inducing tolerant DCs.
- the therapeutically effective amount may be appropriately determined depending on the symptoms, body weight, age, and sex of the patient, as well as the dosage form and administration method of the pharmaceutical composition.
- the pharmaceutical composition can be administered in a single dose of 0.001 to 1000 mg of antigen per kg of the body weight of the subject. The dose may be 0.005 to 500 mg/kg, 0.01 to 300 mg/kg, 0.02 to 200 mg/kg, or 0.03 to 100 mg/kg.
- the pharmaceutical composition may contain a therapeutically effective amount of the immunogenic complex per unit dosage form.
- the content of the immunogenic complex in the pharmaceutical composition may be 0.01 to 90% by mass, 0.05 to 80% by mass, or 0.1 to 60% by mass.
- the pharmaceutical composition may be administered in a single dose or repeatedly.
- the administration interval may be appropriately determined depending on the symptoms, weight, age, and sex of the patient, as well as the dosage form of the pharmaceutical composition and the administration method.
- the administration interval may be, for example, every few hours, 2-3 times a day, once a day, once every 2-3 days, once a week, once a month, once every few months, etc.
- the subject to which the pharmaceutical composition is administered is not particularly limited.
- the subject to which the pharmaceutical composition is administered is preferably a mammal, and may be a human or a non-human mammal.
- non-human mammals include primates (monkeys, gorillas, chimpanzees, marmosets, etc.), rodents (mice, rats, guinea pigs, hamsters, etc.), pets (dogs, cats, rabbits, ferrets, etc.), and livestock (cows, pigs, horses, goats, sheep, etc.).
- the pharmaceutical composition can be used for treating or preventing allergies or autoimmune diseases.
- the allergies or autoimmune diseases to which the pharmaceutical composition can be applied are not particularly limited. Examples of allergies include, but are not limited to, hay fever, food allergies, dust mite allergies, atopic dermatitis, allergic asthma, allergic rhinitis, allergic urticaria, and the like. Examples of autoimmune diseases include, but are not limited to, Graves' disease, rheumatoid arthritis, Hashimoto's thyroiditis, type 1 diabetes, systemic lupus erythematosus, vasculitis, and the like.
- the disclosure provides a method of treating or preventing allergy or an autoimmune disease comprising administering to a subject an immunogenic conjugate of the first aspect.
- the immunogenic conjugate may be administered in a therapeutically effective amount.
- the disclosure provides an immunogenic conjugate of the first aspect for treating or preventing an allergy or autoimmune disease.
- the disclosure provides the use of the immunogenic complex of the first aspect in the manufacture of a pharmaceutical composition for treating or preventing an allergy or an autoimmune disease.
- the disclosure provides the use of the immunogenic conjugate of the first aspect for treating or preventing allergy or an autoimmune disease.
- Ovalbumin Ovalbumin
- MAN mannoprotein
- ConA concanavalin A
- Tris tris(hydroxymethyl)aminomethane
- LPS lipopolysaccharide
- Hydrogen peroxide H2O2 , 30 wt% in water
- dithiothreitol DTT
- sulfuric acid 98%, H2SO4
- calcium chloride CaCl2
- magnesium chloride hexahydrate MgCl2 ⁇ 6H2O
- sodium dodecyl sulfate SDS
- glycine sodium chloride
- NaCl NaCl
- Mildform 10N 10% formalin neutral buffer
- CBB solution for protein assay 4% paraformaldehyde solution (PFA), disodium hydrogen phosphate, and sodium dihydrogen phosphate were purchased from Nacalai Tesque, Inc.
- D-glucose was purchased from Tokyo Chemical Industry Co., Ltd.
- Biotin anti-mouse IgE antibody, biotin anti-mouse IgG1 antibody, and biotin anti-mouse IgG2a antibody were purchased from BioLegend.
- NPs Mannan-Coated OVA Nanoparticles
- PB phosphate buffer
- OVA and MAN were mixed at different concentrations to obtain working solutions. In the following tests, the concentrations of OVA and MAN are expressed using the final concentrations in the working solutions.
- Protein nanoparticles (NPs) were prepared by heating 1 mL of the working solution at 65°C, 75°C, or 85°C. The heating times were 0, 5, 15, 25, or 60 minutes, respectively. The NP formation was stopped by cooling in an ice bath.
- MAN-OVA NPs (MO) without disulfide bonds.
- H 2 O 2 was added to the MO solution up to 0.3 wt %, and the mixture was reacted overnight at 4°C to form intermolecular disulfide bonds.
- Free OVA, MAN, and H2O2 were removed by ultrafiltration (100 kDa Amicon, Merck Millipore) to recover disulfide-crosslinked MAN-OVA NPs (MDO).
- MDO was freeze-dried in a 1 wt% glucose solution to ensure long-term storage.
- 20 mg/mL OVA and 20 mg/mL MAN were mixed in equal amounts and conjugated with glutaraldehyde (final concentration 25 mM) to prepare a MAN-OVA conjugate (MGO).
- Nanoparticle size and zeta potential The particle size, particle size distribution and zeta potential of the nanoparticles were measured at 25° C. using a dynamic light scattering spectrophotometer (DLS, Zetasizer Pro ZSU3200).
- OVA and MAN OVA concentration were measured using a protein assay CBB solution based on the Bradford method.
- OVA standard solution was serially diluted 2-fold, and a standard curve was created by linear regression. Samples were mixed with CBB solution and incubated at room temperature for 10 minutes. Absorbance was measured at 595 nm using a plate reader (Infinite200PRO M Plex, Tecan).
- a phenol-sulfuric acid assay was used to measure MAN concentration.
- a standard curve was prepared by gradient dilution of MAN. 5% (w/v) phenol was mixed with the sample, and H2SO4 was added and mixed well. After incubation at room temperature for 40 minutes, the absorbance was measured at 490 nm.
- ConA agglutination assay was performed as previously reported (Cui, Z.; J Control Release 2002, 81 (1-2), 173-184.). Briefly, ConA was dissolved in phosphate-buffered saline (PBS, 10 mM, pH 7.4), and 5 mM CaCl2 and 5 mM MgCl2 were added to obtain a 1 mg/mL ConA solution. 40 ⁇ L of sample was mixed with 200 ⁇ L of ConA solution. Turbidity was measured at 36 nm for 300 seconds.
- PBS phosphate-buffered saline
- ⁇ Mouse> C57BL/6J mice (7-8 weeks old, male) and BALB/c mice (6-8 weeks old, female) were purchased from Kyudou Co., Ltd.
- OT-II Tg/wt,Ly5.1/Ly5.1 mice were fed CE-2 (Kyudou Co., Ltd.) and had access to bedding and water.
- Cage changing was performed in a laminar flow hood. Mice were housed under a 12-hour light/12-hour dark cycle. Animal experiments were performed in accordance with the guidelines of the Kyushu University Animal Care and Management Committee.
- BMDCs Myeloid Dendritic Cells
- BMDCs were first negatively selected from bone marrow cells with biotin anti-mouse F4/80 and streptavidin nanobeads, and then positively selected with anti-mouse CD11c nanobeads.
- OVA-rhodamine was used to prepare fluorescently labeled NPs.
- BMDCs (2 ⁇ 10 5 cells/well) were seeded in 12-well plates and incubated with rhodamine-labeled OVA, OVA dNPs (disulfide-crosslinked OVA nanoparticles), MDO, and MGO for different times.
- MAN was added to the medium at a final concentration of 1 mg/mL 30 min before sample addition. Dead cells were stained with SYTOX TM Green (Thermo Fisher Scientific).
- MFI mean fluorescence intensity
- BMDCs were seeded in a 96-well plate (5 ⁇ 10 4 cells/well) and pulsed with PBS, OVA, MDO, or MGO for 3 hours. Then, BMDCs and OT-II CD4 + T cells (2.5 ⁇ 10 5 cells/well) were cocultured for 24 hours.
- OT-II CD4 + T cells were isolated from the spleen of OT-II mice using a Mouse CD4 T Cell Isolation Kit (Biolegend). The IL-2 concentration in the culture medium was measured using an IL-2 Mouse ELISA Kit (Thermo Fisher Scientific).
- mice were sensitized with 200 ⁇ L PBS containing 10 ⁇ g OVA and 1 mg alum by intraperitoneal injection three times at 7-day intervals. Seven days after the last sensitization, the mice were sacrificed and peripheral blood was collected by retro-orbital bleeding. Serum was obtained from the peripheral blood.
- An indirect immunoassay (indirect-ELISA) protocol was used. 100 ⁇ L of OVA, OVA-dNP, MDO, and MGO (as 100 ⁇ g/mL of OVA) were coated onto a 96-well plate at 4°C for 24 hours. After blocking with ELISA-ELISPOT diluent, serially diluted sera were added to the plate and incubated overnight at 4°C. Biotin anti-mouse IgE, biotin anti-mouse IgG1, and biotin anti-mouse IgG2a antibodies were added to the plate, respectively. After incubation, streptavidin-HRP was added, and TMB solution was used as a substrate. Finally, the reaction was stopped, and the optical density (OD) of each well was measured at 450 nm.
- mice were sensitized by intraperitoneal injection of 200 ⁇ L PBS containing 10 ⁇ g OVA and 1 mg alum on days 0 and 14. On day 28, mice were intraperitoneally injected with 200 ⁇ g OVA, OVA dNP, MDO, and MGO (containing 200 ⁇ g OVA). After intraperitoneal injection, rectal temperature was measured every 10 min using a thermometer (Physitemp PTM1, Muromachi Kikai Co., Ltd.).
- mice received (a) oral administration of PBS, OVA, MDO and MGO (containing 0.5 mg OVA) on days 0-4 and 7-11, or (b) subcutaneous administration of PBS, OVA, MDO and MGO (containing 20 ⁇ g OVA) on days 0, 2, 4, 7, 9 and 11.
- mice were sensitized by intraperitoneal injection of 10 ⁇ g OVA and 1 mg alum.
- mice were intranasally administered 25 ⁇ g OVA.
- mice were sacrificed and bronchoalveolar lavage fluid (BAL), peripheral blood and lung tissue were collected.
- BAL bronchoalveolar lavage fluid
- mice were first sensitizing mice with intraperitoneal injections of 10 ⁇ g OVA and 1 mg alum on days 0 and 7. Next, mice received (a) oral administration of PBS, OVA, MDO, and MGO (containing 0.5 mg OVA) on days 14-18 and 21-25, or (b) subcutaneous administration of PBS, OVA, MDO, and MGO (containing 20 ⁇ g OVA) on days 14, 16, 18, 21, 23, and 25. Finally, mice were intranasally challenged with 25 ⁇ g OVA on days 32-35. Mice were sacrificed on day 36, and bronchoalveolar lavage fluid (BAL), peripheral blood, and lung tissue were collected.
- BAL bronchoalveolar lavage fluid
- cytokines in BAL fluid IL-4, IL-5, IL-6, IL-10, IL-13, and IFN- ⁇ in the BAL fluid were detected using a BD Cytometric Bead Array (BD Life Sciences-Biosciences), followed by measurement using a flow cytometer (CyroFLEX-S, Beckman Coulter Inc.).
- thiol groups were not detected in native OVA (0 min), but after heating, thiol groups appeared on the OVA surface and became detectable.
- the free thiol group concentration of NPs not treated with H2O2 was significantly higher than that of dNPs treated with H2O2 (Fig . 6), indicating the formation of disulfide bonds in dNPs.
- the addition of MAN reduced the detectable thiol groups, which may be due to the inhibition of the reaction of DTNB with SH by the coating of MAN.
- ConA is a tetrameric lectin with four binding sites for ⁇ -mannose.
- the turbidity of OVA dNP did not change significantly.
- the turbidity of MDO increased significantly, and the turbidity increased with increasing MAN content. This suggested that a large amount of mannan was present on the surface of MDO-20. Due to the unimodal distribution and high MAN content of MDO-15, MDO-15 was used in subsequent experiments.
- BMDCs ⁇ Induction of regulatory T cells by MDO>
- T cell stimulating ability of BMDCs was measured using OT-II CD4 + T cells that have a specific TCR that recognizes the OVA peptide/I-AB complex and secrete IL-2 in response to it.
- BMDCs were pulsed with OVA, MDO, and MGO for 3 hours, respectively, and then co-cultured with OT-II CD4 + T cells for 24 hours, and the secreted IL-2 concentration in the medium was measured using an ELISA kit.
- MDO treatment significantly increased the IL-2 concentration compared to OVA treatment and MGO treatment ( Figure 11).
- MDO treatment increased the IL-10 concentration compared to OVA treatment and MGO treatment ( Figure 12).
- MDO treatment increased the activity of ATRA reductase (RALDH) compared to OVA treatment ( Figure 13).
- MGO and MDO treatments increased the proportion of regulatory T cells compared to OVA treatment ( Figure 14).
- mice were administered two routes of administration (oral and subcutaneous).
- oral administration mice were administered PBS, 500 ⁇ g OVA, or 500 ⁇ g OVA as MDO or MGO on days 0-4 and 7-11.
- Mice were sensitized by subcutaneous injection of OVA and alum on days 14 and 21, and finally administered OVA intranasally on days 28-31 ( Figure 18).
- serum, bronchoalveolar lavage fluid (BAL), and lungs were collected.
- Anti-OVA IgE levels were reduced by OVA pre-treatment and more strongly by MDO and MGO pre-treatment ( Figure 19).
- Anti-OVA IgG2a levels were significantly increased by MDO treatment, whereas IgG1 levels were not different (Figure 19).
- Pre-administration of OVA, MDO, and MGO suppressed the increase in eosinophils and neutrophils and reduced the total cell count in the BAL fluid, but no significant difference was observed in the number of macrophage cells ( Figure 20).
- the levels of IL-4, IL-5, IL-6, and IL-13 mediated by Th2 cells in the BAL fluid were significantly reduced by MDO pre-administration and MGO pre-administration compared to PBS treatment ( Figure 21).
- Pre-administration of OVA also reduced Th2 cytokines, but not as significantly as MDO pre-administration and MGO pre-administration.
- mice were given 20 ⁇ g of PBS or OVA, or 20 ⁇ g of OVA as MDO or MGO on days 0, 2, 4, 7, 9, and 11.
- the subsequent course was the same as for oral administration ( Figure 22).
- Pre-administration of OVA, pre-administration of MDO, and pre-administration of MGO showed similar IgE production inhibitory effects (Figure 23).
- IgG2a levels increased with MDO pre-administration, but IgG1 levels did not differ among all groups.
- IgG2a/IgE and IgG2a/IgG1 values indicated that MDO pre-administration and MGO pre-administration showed superior IgE inhibitory effects ( Figure 23).
- mice were sensitized with OVA and alum on days 0 and 7 and orally administered PBS, 20 ⁇ g OVA, or 500 ⁇ g OVA as MDO or MGO on days 14-18 and 21-25 (Figure 26).
- Nasal administration of OVA was performed on days 32-35.
- Anti-OVA IgE levels were decreased after OVA post-administration and MDO post-administration ( Figure 27). There was a tendency for a decrease after MGO post-administration, but there was no statistical difference (Figure 27).
- mice were administered PBS, 20 ⁇ g OVA, or 20 ⁇ g OVA as MDO or MGO on days 14, 16, 18, 21, 23, and 25 (Figure 30).
- MDO post-administration reduced anti-OVA IgE production ( Figure 31).
- BAL fluid neutrophil, eosinophil, and total cell counts were reduced by MDO post-administration ( Figure 32).
- IL-4, IL-5, and IL-6 were reduced by MDO post-administration (Figure 33).
- Mononuclear cell infiltration was reduced by OVA pre-administration and MGO post-administration, but more dramatically by MDO post-administration (Figure 34: subcutaneous treatment).
- HSA Human serum albumin
- OVA ovalbumin
- MAN mannoprotein
- ConA concanavalin A
- LPS lipopolysaccharide
- Hydrogen peroxide H 2 O 2 , 30 wt % in water
- sulfuric acid (98%, H 2 SO 4 )
- calcium chloride CaCl 2
- magnesium chloride hexahydrate MgCl 2 ⁇ 6H 2 O
- sodium chloride NaCl
- DTNB 2,2'-dinitro-5,5'-dithiodibenzoic acid
- 5(6)-Carboxytetramethylrhodamine N-succinimidyl ester HNS-Rho was purchased from Thermo Fisher Scientific.
- NuPAGE sample reducing agent (10x), NuPAGE LDS sample buffer (4x), ELISA-ELISPOT diluent (5x), TMB (3,3',5,5'-tetramethylbenzidine) solution (1x), and HRP-conjugated Streptavidin (horseradish peroxidase) were purchased from Invitrogen.
- QuickBlue staining solution was purchased from Bio Dynamics Institute Co., Ltd.
- 4% paraformaldehyde solution (PFA), disodium hydrogen phosphate, and sodium dihydrogen phosphate were purchased from Nacalai Tesque, Inc.
- Biotin anti-mouse IgE antibody, biotin anti-mouse IgG1 antibody, and biotin anti-mouse IgG2a antibody were purchased from BioLegend.
- HSA NPs were prepared under various conditions [buffer (10 mM phosphate buffer containing 0 mM, 50 mM or 135 mM NaCl (pH 7.4)]; heating temperature (65°C, 75°C or 85°C); heating time (5 min, 10 min, 30 min or 60 min)]. HSA was dissolved in the buffer and left at 4°C overnight. The HSA dispersion was filtered through a 0.22 ⁇ m membrane to remove aggregates. 1 mL of the HSA solution was heated in a water bath and immediately cooled in an ice bath.
- H2O2 was added to the NP solution to a final concentration of 0.3 wt%, and the solution was kept at 4°C overnight to form disulfide bonds inside the NPs.
- Free HSA and H2O2 were removed by ultrafiltration (100 kDa Amicon, Merck Millipore).
- HSA-OVA NP(HO) HSA-OVA NP(HO)
- rapamycin 10 ⁇ L of rapamycin (1 mg/mL) was added to 1 mL of the mixed solution of MAN, HSA, and OVA.
- the buffer used for dissolving HSA, OVA, and MAN was 10 mM PB + 50 mM NaCl (pH 7.4).
- the mixed solution was heated at 85°C for 60 minutes. After cooling the mixed solution, H 2 O 2 was added, and the solution was left to stand overnight at 4°C, and the NP solution was collected. Ultrafiltration was performed using a 100 kDa filter to collect the NP.
- Rapamycin was quantified by a validated high performance liquid chromatography (HPLC) method using an Inertsil C8 column (5 ⁇ m, 4.6 mm ⁇ 150 mm) with an Elite Lachrom L-2455 diode array detector (Hitachi). The mobile phase was changed from 70% methanol and 30% Milli-Q water to 90% methanol and 10% Milli-Q water within 20 min. The flow rate was set at 1.0 mL/min, injection volume was 20 ⁇ L, and column oven temperature was set at 25° C. Rapamycin was detected using a photodiode array detector at a wavelength of 277 nm.
- HPLC high performance liquid chromatography
- BMDC phenotype analysis BMDCs were seeded in 12-well plates (3 ⁇ 10 6 cells/well), stimulated with 100 ng/mL LPS for 24 hours, and treated with PBS, OVA, HO-20, MHO-10, or MHO-10-R (MHO-10 encapsulated with rapamycin) for 24 hours. BMDCs were collected, and nonspecific binding of immunoglobulins to Fc receptors was inhibited with anti-mouse CD16/32 antibody. BMDCs were stained with anti-mouse I-Ab-FITC, CD86-APC, CD80-BV421, and propidium iodide (PI) (Biolegend). Subsequent analysis was performed by flow cytometry.
- PI propidium iodide
- NP preparation conditions The optimal conditions for preparing NPs from HSA, a matrix protein, were screened. HSA is neutral and negatively charged, so HSA aggregates are stably dispersed. The HSA solution was heated at a specific temperature (65-85°C) for up to 60 minutes. Then, H2O2 was added to crosslink the disulfide bonds. The size of HSA NPs increased with heating temperature (data not shown). This is thought to be due to the progression of denaturation with temperature. HSA is known to denature at 65-80°C. Since HSA is expected to be completely denatured and aggregation promoted at 85°C, 85°C was selected as the preparation temperature. When sodium chloride is added, the aggregate size increases, possibly due to the suppression of electrostatic repulsion. 50 mM NaCl was selected because excess sodium chloride destabilizes NPs.
- the effect of the amount of MAN added on the preparation conditions of HSA/OVA NPs was evaluated with a fixed HSA/OVA ratio of 4:1.
- the particle size and PDI of the obtained NPs (MHO series) increased with increasing MAN concentration ( Figure 39, Tables 3 and 4).
- the MAN content in MHO quantified by the phenol-sulfuric acid method increased with increasing MAN concentration, indicating that the incorporation of MAN into NPs is concentration-dependent (Tables 3 and 4). Since the mannan portion of MAN is hydrophilic, mannan coats the surface of the NPs.
- MHO-10 was dispersed in a buffer solution (10 mM PB + 50 mM NaCl, pH 7.4) and storage stability at 4°C was confirmed. No size change was observed after storage for at least 28 days (data not shown).
- rapamycin an immunosuppressant
- HPLC analysis of the rapamycin remaining in the filtrate of the NP dispersion we confirmed that rapamycin had been incorporated almost quantitatively (data not shown).
- NPs (MHCryj) were prepared by the above method using 4.5 mg/mL HSA, 0.5 mg/mL Cry j, and 10 mg/mL MAN. MHCryj was slightly larger than MHO-10, but showed a uniform size distribution ( Figure 41, Table 5). In the PAGE analysis of MHCryj, bands of Cry j and HSA were observed under reducing conditions ( Figure 42), and no bands were observed under non-reducing conditions.
- NPs have the effect of inducing tolerant DCs.
- BMCs stimulated with LPS expressed high levels of MHC class II (I-A b ), costimulatory factors CD80 and CD86, indicating that BMDCs were mature.
- I-A b MHC class II
- CD80 and CD86 costimulatory factors
- MHO-10 and MHO-10-R have the ability to suppress the activation of BMDCs, and that MHO-10-R may induce tolerant DCs due to the effect of rapamycin.
- MAN induces signal transduction via mannose receptor and DC-SIGN, causing activation of NF- ⁇ B. Rapamycin enhances activation of NF- ⁇ B through inhibition of mammalian target of rapamycin, which is a signal transduction pathway different from that of MAN. Therefore, MAN and rapamycin may synergistically induce tolerant DC.
- ⁇ Reactivity to anti-OVA antibodies> The reactivity of HO and MHO with anti-OVA antibodies was examined by ELISA. Anti-OVA antibodies were obtained from the serum of mice in which OVA-specific allergy was induced. As shown in FIG. 47, HO-20 and MHO-10 had much lower reactivity with anti-OVA antibodies than OVA, and there was no significant difference between these two NPs. This result indicates that MHO induces a weak IgE-mediated allergic reaction. In allergy treatment, this indicates that MHO is safer than native antigens used in conventional antigen-specific immunotherapy.
- Example 3 To evaluate whether Cry j NP induces an allergic reaction, a basophil activation test was performed using human peripheral blood.
- MHCryj Materials and Methods ⁇ Preparation of MHCryj> NP (MHCryj) was prepared in the same manner as above using 4.75 mg/mL HSA, 0.25 mg/mL Cry j 1/2 (a mixture of Cry j 1 and Cry j 2), and 7.5 mg/mL MAN. HSA and Cry j 1/2 were dissolved in PB and left to stand overnight at 4°C. The solution was filtered through a 0.22 ⁇ m membrane to remove aggregates. The solution was then heated in a water bath (85°C, 10 minutes) and immediately cooled in an ice bath.
- H 2 O 2 was added to the solution to a final concentration of 0.1 wt%, and the solution was kept at 4°C overnight to form disulfide bonds inside the NP.
- Free HSA and H 2 O 2 were removed by ultrafiltration (100 kDa Amicon, Merck Millipore).
- the size and PDI of the obtained MHCryj were as follows: Size: 123.6 ⁇ 1.6 (d.nm) PDI: 0.34 ⁇ 0.04
- the basophil activation test was performed using an Allergenecity kit (BECKMAN COULTER, Tokyo, Japan) according to the protocol attached to the Allergenecity kit.
- peripheral blood from healthy subjects and peripheral blood from patients with cedar pollen allergy were used, collected using blood collection tubes containing ethylenediaminetetraacetic acid (EDTA).
- EDTA ethylenediaminetetraacetic acid
- Peripheral blood was stimulated using Cry j 1/2 or MHCryj as an allergen.
- Cry j 1/2 was diluted with PBS so that the amount of Cry j 1/2 added per 100 ⁇ L of peripheral blood was 0.01 ⁇ g, 0.1 ⁇ g, or 1 ⁇ g, and 20 ⁇ L was added to 100 ⁇ L of peripheral blood.
- MHCryj was diluted with PBS so that the amount of Cry j 1/2 added per 100 ⁇ L of peripheral blood was 0.01 ⁇ g, 0.1 ⁇ g, or 1 ⁇ g, and 20 ⁇ L was added to 100 ⁇ L of peripheral blood.
- Anti-IgE antibody was used as a positive control.
- PBS was used as a negative control.
- the samples used to stimulate each peripheral blood are summarized in Table 6. The values in Table 6 indicate the amount of Cry j 1/2 added ( ⁇ g) per 100 ⁇ L of peripheral blood.
- each peripheral blood sample was stained with CRTH2 (CD294)-FITC, CD203c-PE, and CD3-PC7, and analyzed by a flow cytometer. Basophil activation was evaluated based on the analysis results. The rate of basophil activation was determined by calculating the percentage of basophils with increased CD203c fluorescence intensity compared to the negative control (PBS) in the CRTH2 (CD294) and CD203c double positive basophil population in the CD3 negative mononuclear cell region.
- PBS negative control
- CD203c double positive basophil population in the CD3 negative mononuclear cell region.
- the positive control increased the fluorescence intensity of CD203c compared to the negative control (PBS), and an increase in the proportion of activated basophils was confirmed.
- stimulation with Cry j (0.01 ⁇ g, 0.1 ⁇ g, 1 ⁇ g) also increased the fluorescence intensity of CD203c compared to the negative control (PBS), and an increase in the proportion of activated basophils was confirmed ( Figures 48 and 50, right).
- the present invention provides an immunogenic complex that has low reactivity to IgE antibodies and high ability to induce tolerant DCs, and a pharmaceutical composition containing the immunogenic complex.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical & Material Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Epidemiology (AREA)
- Immunology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Peptides Or Proteins (AREA)
Abstract
L'invention concerne un complexe immunogène comprenant un antigène et une mannoprotéine, ledit antigène et ladite mannoprotéine étant liés par un pont disulfure. L'invention concerne également une composition pharmaceutique comprenant ledit complexe immunogène.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2022191916 | 2022-11-30 | ||
JP2022-191916 | 2022-11-30 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024117232A1 true WO2024117232A1 (fr) | 2024-06-06 |
Family
ID=91323900
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2023/042983 WO2024117232A1 (fr) | 2022-11-30 | 2023-11-30 | Complexe immunogène et composition pharmaceutique |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024117232A1 (fr) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JPH0393730A (ja) * | 1989-09-02 | 1991-04-18 | Hayashibara Biochem Lab Inc | 減感作剤 |
JP2013527854A (ja) * | 2010-05-10 | 2013-07-04 | アセンド・バイオファーマシューティカルズ・ピーティーワイ・リミテッド | 免疫刺激組成物及びワクチン組成物 |
JP2016515603A (ja) * | 2013-04-03 | 2016-05-30 | インムノテック・ソシエダッド・リミターダInmunotek, S.L. | ワクチン接種における使用のための免疫原性複合体およびこれを得る方法 |
-
2023
- 2023-11-30 WO PCT/JP2023/042983 patent/WO2024117232A1/fr unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JPH0393730A (ja) * | 1989-09-02 | 1991-04-18 | Hayashibara Biochem Lab Inc | 減感作剤 |
JP2013527854A (ja) * | 2010-05-10 | 2013-07-04 | アセンド・バイオファーマシューティカルズ・ピーティーワイ・リミテッド | 免疫刺激組成物及びワクチン組成物 |
JP2016515603A (ja) * | 2013-04-03 | 2016-05-30 | インムノテック・ソシエダッド・リミターダInmunotek, S.L. | ワクチン接種における使用のための免疫原性複合体およびこれを得る方法 |
Non-Patent Citations (4)
Title |
---|
BENITO-VILLALVILLA CRISTINA, SORIA IRENE, SUBIZA JOSé LUIS, PALOMARES OSCAR: "Novel vaccines targeting dendritic cells by coupling allergoids to mannan", ALLERGO JOURNAL INTERNATIONAL, vol. 27, no. 8, 1 December 2018 (2018-12-01), pages 256 - 262, XP055780369, DOI: 10.1007/s40629-018-0069-8 * |
LI SHUNYI, MURAKAMI DAISUKE, NAGATOISHI SATORU, LIU YIWEI, TSUMOTO KOUHEI, KATAYAMA YOSHIKI, MORI TAKESHI: "One-pot preparation of mannan-coated antigen nanoparticles using human serum albumin as a matrix for tolerance induction", JOURNAL OF COLLOID AND INTERFACE SCIENCE, ACADEMIC PRESS,INC., US, vol. 649, 1 November 2023 (2023-11-01), US , pages 955 - 965, XP093177016, ISSN: 0021-9797, DOI: 10.1016/j.jcis.2023.06.170 * |
LI SHUNYI, TORIUMI HIROKI, TAKAHASHI DAISUKE, KAMASAKI TOMOKO, FUJIOKA YOICHIRO, NAGATOISHI SATORU, LI JINTING, LIU YIWEI, HOSOKAW: "Safe and efficient oral allergy immunotherapy using one-pot-prepared mannan-coated allergen nanoparticles", BIOMATERIALS, ELSEVIER, AMSTERDAM, NL, vol. 303, 1 December 2023 (2023-12-01), AMSTERDAM, NL , pages 122381, XP093177019, ISSN: 0142-9612, DOI: 10.1016/j.biomaterials.2023.122381 * |
SIRVENT SOFIA; SORIA IRENE; CIRAUQUI CRISTINA; CASES BARBARA; MANZANO ANA I.; DIEZ-RIVERO CARMEN M.; RECHE PEDRO A.; LóPEZ-RE: "Novel vaccines targeting dendritic cells by coupling allergoids to nonoxidized mannan enhance allergen uptake and induce functional regulatory T cells through programmed death ligand 1", JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, ELSEVIER, AMSTERDAM, NL, vol. 138, no. 2, 13 April 2016 (2016-04-13), AMSTERDAM, NL , pages 558, XP029672342, ISSN: 0091-6749, DOI: 10.1016/j.jaci.2016.02.029 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20240000830A1 (en) | Immune-modifying nanoparticles for the treatment of inflammatory diseases | |
US20210268082A1 (en) | METHOD OF INDUCING IMMUNE TOLERANCE BY ADMINISTERING ACTIVE PRINCIPLE-LOADING RBC's | |
JP5191234B2 (ja) | 免疫疾患の予防ないし治療剤および方法 | |
US20120189700A1 (en) | Nanoparticle Based Immunological Stimulation | |
Afify et al. | Bovine holo-beta-lactoglobulin cross-protects against pollen allergies in an innate manner in BALB/c mice: potential model for the farm effect | |
JP2021165273A (ja) | ペプチドコンジュゲート粒子 | |
JP2010505883A (ja) | 免疫応答を調節するための組成物および方法 | |
Wang et al. | Total coumarins from Urtica dentata Hand prevent murine autoimmune diabetes via suppression of the TLR4-signaling pathways | |
US20220031632A1 (en) | Nanoparticle compositions and uses thereof | |
JP2003527438A (ja) | アレルギーの予防及び/又は治療用組成物及びアレルギーの予防及び/又は治療方法 | |
WO2024117232A1 (fr) | Complexe immunogène et composition pharmaceutique | |
US20230381183A1 (en) | USE OF FOLIC ACID AND FOLATE MODIFICATION IN INDUCING B-CELL IMMUNE TOLERANCE AND TARGETING mIgM-POSITIVELY-EXPRESSED B-CELL LYMPHOMA | |
Pali-Schöll et al. | Formulations for allergen immunotherapy in human and veterinary patients: new candidates on the horizon | |
JP2010527354A (ja) | 免疫調節化合物によるアレルギー性疾患の治療 | |
Garn et al. | Bovine Holo-Beta-Lactoglobulin Cross-Protects Against Pollen Allergies in an Innate Manner in BALB/c Mice: Potential Model for the Farm Effect | |
RU2782223C2 (ru) | Композиция для индуцирования специфической иммунологической толерантности | |
WO2019246408A2 (fr) | Immunothérapie à base de peptide lié à un anticorps pour le diabète de type 1 |
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: 23897878 Country of ref document: EP Kind code of ref document: A1 |