EP2453911A1 - Composition de nanomédicament immunogène, préparation et utilisations de cette composition - Google Patents
Composition de nanomédicament immunogène, préparation et utilisations de cette compositionInfo
- Publication number
- EP2453911A1 EP2453911A1 EP10719414A EP10719414A EP2453911A1 EP 2453911 A1 EP2453911 A1 EP 2453911A1 EP 10719414 A EP10719414 A EP 10719414A EP 10719414 A EP10719414 A EP 10719414A EP 2453911 A1 EP2453911 A1 EP 2453911A1
- Authority
- EP
- European Patent Office
- Prior art keywords
- pei
- pharmaceutical composition
- macromolecule
- nanocomp
- nucleic acid
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Withdrawn
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 139
- 238000002360 preparation method Methods 0.000 title claims abstract description 24
- 230000002163 immunogen Effects 0.000 title description 9
- 229920002873 Polyethylenimine Polymers 0.000 claims abstract description 114
- 239000002105 nanoparticle Substances 0.000 claims abstract description 70
- 238000000034 method Methods 0.000 claims abstract description 64
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 50
- 229920002521 macromolecule Polymers 0.000 claims abstract description 47
- 239000012669 liquid formulation Substances 0.000 claims abstract description 27
- 238000009472 formulation Methods 0.000 claims description 91
- 239000000243 solution Substances 0.000 claims description 64
- 102000039446 nucleic acids Human genes 0.000 claims description 63
- 108020004707 nucleic acids Proteins 0.000 claims description 63
- 150000007523 nucleic acids Chemical class 0.000 claims description 63
- 108091007433 antigens Proteins 0.000 claims description 57
- 102000036639 antigens Human genes 0.000 claims description 57
- 239000000427 antigen Substances 0.000 claims description 56
- 239000013612 plasmid Substances 0.000 claims description 51
- 230000004071 biological effect Effects 0.000 claims description 44
- 208000024891 symptom Diseases 0.000 claims description 36
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 32
- 238000011282 treatment Methods 0.000 claims description 32
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 claims description 29
- 208000026935 allergic disease Diseases 0.000 claims description 29
- 239000000594 mannitol Substances 0.000 claims description 29
- 229920000642 polymer Polymers 0.000 claims description 29
- 208000015181 infectious disease Diseases 0.000 claims description 27
- 235000010355 mannitol Nutrition 0.000 claims description 26
- 206010020751 Hypersensitivity Diseases 0.000 claims description 25
- 229930195725 Mannitol Natural products 0.000 claims description 25
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Triethanolamine Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 claims description 25
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 25
- 150000005846 sugar alcohols Chemical class 0.000 claims description 24
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 23
- 239000002245 particle Substances 0.000 claims description 23
- 239000002904 solvent Substances 0.000 claims description 23
- 201000010099 disease Diseases 0.000 claims description 22
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 20
- 230000002265 prevention Effects 0.000 claims description 19
- JUJWROOIHBZHMG-UHFFFAOYSA-N pyridine Substances C1=CC=NC=C1 JUJWROOIHBZHMG-UHFFFAOYSA-N 0.000 claims description 19
- 201000010105 allergic rhinitis Diseases 0.000 claims description 18
- 230000007815 allergy Effects 0.000 claims description 18
- 229920001184 polypeptide Polymers 0.000 claims description 18
- 206010039085 Rhinitis allergic Diseases 0.000 claims description 17
- 239000007864 aqueous solution Substances 0.000 claims description 15
- 230000015556 catabolic process Effects 0.000 claims description 15
- 150000001875 compounds Chemical class 0.000 claims description 15
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 claims description 15
- 238000006731 degradation reaction Methods 0.000 claims description 14
- 230000028993 immune response Effects 0.000 claims description 14
- 241000606161 Chlamydia Species 0.000 claims description 13
- 150000001720 carbohydrates Chemical class 0.000 claims description 13
- 238000002347 injection Methods 0.000 claims description 13
- 239000007924 injection Substances 0.000 claims description 13
- 239000000126 substance Substances 0.000 claims description 12
- 239000000428 dust Substances 0.000 claims description 11
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims description 11
- 235000014633 carbohydrates Nutrition 0.000 claims description 10
- 239000013572 airborne allergen Substances 0.000 claims description 9
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 9
- 125000000217 alkyl group Chemical group 0.000 claims description 8
- GUBGYTABKSRVRQ-XABDMIIQSA-N alpha-D-Manp-(1->4)-alpha-D-Manp Chemical group OC[C@H]1O[C@H](O[C@H]2[C@H](O)[C@H](O)[C@@H](O)O[C@@H]2CO)[C@@H](O)[C@@H](O)[C@@H]1O GUBGYTABKSRVRQ-XABDMIIQSA-N 0.000 claims description 8
- 230000003834 intracellular effect Effects 0.000 claims description 8
- 150000003839 salts Chemical class 0.000 claims description 8
- 206010027654 Allergic conditions Diseases 0.000 claims description 7
- 239000003054 catalyst Substances 0.000 claims description 7
- UMJSCPRVCHMLSP-UHFFFAOYSA-N pyridine Natural products COC1=CC=CN=C1 UMJSCPRVCHMLSP-UHFFFAOYSA-N 0.000 claims description 7
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 claims description 6
- 208000030773 pneumonia caused by chlamydia Diseases 0.000 claims description 6
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 claims description 5
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 claims description 5
- 108010000912 Egg Proteins Proteins 0.000 claims description 5
- 102000002322 Egg Proteins Human genes 0.000 claims description 5
- NNTOJPXOCKCMKR-UHFFFAOYSA-N boron;pyridine Chemical compound [B].C1=CC=NC=C1 NNTOJPXOCKCMKR-UHFFFAOYSA-N 0.000 claims description 5
- 238000006268 reductive amination reaction Methods 0.000 claims description 5
- 239000000600 sorbitol Substances 0.000 claims description 5
- 235000010356 sorbitol Nutrition 0.000 claims description 5
- 238000010257 thawing Methods 0.000 claims description 5
- QCVGEOXPDFCNHA-UHFFFAOYSA-N 5,5-dimethyl-2,4-dioxo-1,3-oxazolidine-3-carboxamide Chemical group CC1(C)OC(=O)N(C(N)=O)C1=O QCVGEOXPDFCNHA-UHFFFAOYSA-N 0.000 claims description 4
- 208000004262 Food Hypersensitivity Diseases 0.000 claims description 4
- 206010016946 Food allergy Diseases 0.000 claims description 4
- 239000002253 acid Substances 0.000 claims description 4
- 235000014103 egg white Nutrition 0.000 claims description 4
- 210000000969 egg white Anatomy 0.000 claims description 4
- 235000020932 food allergy Nutrition 0.000 claims description 4
- 238000002156 mixing Methods 0.000 claims description 4
- HBAQYPYDRFILMT-UHFFFAOYSA-N 8-[3-(1-cyclopropylpyrazol-4-yl)-1H-pyrazolo[4,3-d]pyrimidin-5-yl]-3-methyl-3,8-diazabicyclo[3.2.1]octan-2-one Chemical class C1(CC1)N1N=CC(=C1)C1=NNC2=C1N=C(N=C2)N1C2C(N(CC1CC2)C)=O HBAQYPYDRFILMT-UHFFFAOYSA-N 0.000 claims description 3
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 claims description 3
- TVXBFESIOXBWNM-UHFFFAOYSA-N Xylitol Natural products OCCC(O)C(O)C(O)CCO TVXBFESIOXBWNM-UHFFFAOYSA-N 0.000 claims description 3
- 239000000654 additive Substances 0.000 claims description 3
- 230000001939 inductive effect Effects 0.000 claims description 3
- 239000012931 lyophilized formulation Substances 0.000 claims description 3
- HEBKCHPVOIAQTA-UHFFFAOYSA-N meso ribitol Natural products OCC(O)C(O)C(O)CO HEBKCHPVOIAQTA-UHFFFAOYSA-N 0.000 claims description 3
- 239000006174 pH buffer Substances 0.000 claims description 3
- 239000000811 xylitol Substances 0.000 claims description 3
- HEBKCHPVOIAQTA-SCDXWVJYSA-N xylitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)CO HEBKCHPVOIAQTA-SCDXWVJYSA-N 0.000 claims description 3
- 235000010447 xylitol Nutrition 0.000 claims description 3
- 229960002675 xylitol Drugs 0.000 claims description 3
- 208000035143 Bacterial infection Diseases 0.000 claims description 2
- 125000002853 C1-C4 hydroxyalkyl group Chemical group 0.000 claims description 2
- 201000005019 Chlamydia pneumonia Diseases 0.000 claims description 2
- 208000017667 Chronic Disease Diseases 0.000 claims description 2
- 208000022362 bacterial infectious disease Diseases 0.000 claims description 2
- 125000002768 hydroxyalkyl group Chemical group 0.000 claims description 2
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 claims description 2
- 229930182842 L-mannitol Natural products 0.000 claims 1
- FBPFZTCFMRRESA-BXKVDMCESA-N L-mannitol Chemical compound OC[C@H](O)[C@H](O)[C@@H](O)[C@@H](O)CO FBPFZTCFMRRESA-BXKVDMCESA-N 0.000 claims 1
- 238000009169 immunotherapy Methods 0.000 abstract description 31
- 229960005486 vaccine Drugs 0.000 abstract description 31
- 108090000623 proteins and genes Proteins 0.000 description 68
- 108020004414 DNA Proteins 0.000 description 57
- 102000004169 proteins and genes Human genes 0.000 description 56
- 235000018102 proteins Nutrition 0.000 description 54
- 230000000694 effects Effects 0.000 description 45
- 210000004027 cell Anatomy 0.000 description 43
- 241000699670 Mus sp. Species 0.000 description 32
- 230000014509 gene expression Effects 0.000 description 32
- 108010058846 Ovalbumin Proteins 0.000 description 31
- 229940092253 ovalbumin Drugs 0.000 description 31
- 239000013566 allergen Substances 0.000 description 30
- 238000002255 vaccination Methods 0.000 description 30
- 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 26
- 101710164702 Major outer membrane protein Proteins 0.000 description 25
- 108010061629 Dermatophagoides pteronyssinus antigen p 1 Proteins 0.000 description 24
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 24
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 24
- 229960001031 glucose Drugs 0.000 description 24
- 125000002091 cationic group Chemical group 0.000 description 19
- 230000003053 immunization Effects 0.000 description 19
- 238000002649 immunization Methods 0.000 description 19
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 18
- 241001465754 Metazoa Species 0.000 description 18
- 239000000872 buffer Substances 0.000 description 18
- 230000003449 preventive effect Effects 0.000 description 18
- 238000003860 storage Methods 0.000 description 18
- 230000015572 biosynthetic process Effects 0.000 description 17
- 241001647372 Chlamydia pneumoniae Species 0.000 description 16
- 241000699666 Mus <mouse, genus> Species 0.000 description 16
- 201000001320 Atherosclerosis Diseases 0.000 description 15
- 241000894006 Bacteria Species 0.000 description 15
- 238000002474 experimental method Methods 0.000 description 15
- 108020004705 Codon Proteins 0.000 description 14
- 108020004635 Complementary DNA Proteins 0.000 description 14
- 238000010804 cDNA synthesis Methods 0.000 description 14
- 239000002299 complementary DNA Substances 0.000 description 14
- 239000008121 dextrose Substances 0.000 description 14
- 230000001965 increasing effect Effects 0.000 description 14
- 238000012360 testing method Methods 0.000 description 14
- 230000001225 therapeutic effect Effects 0.000 description 14
- 241000498849 Chlamydiales Species 0.000 description 13
- 229960004784 allergens Drugs 0.000 description 13
- 235000001014 amino acid Nutrition 0.000 description 13
- 208000010668 atopic eczema Diseases 0.000 description 13
- 239000000047 product Substances 0.000 description 13
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 12
- 230000002009 allergenic effect Effects 0.000 description 12
- 230000000172 allergic effect Effects 0.000 description 12
- 239000012528 membrane Substances 0.000 description 12
- 244000052769 pathogen Species 0.000 description 12
- -1 2-substituted 2-oxazoline Chemical class 0.000 description 11
- 150000001413 amino acids Chemical class 0.000 description 11
- 238000006243 chemical reaction Methods 0.000 description 11
- 230000003247 decreasing effect Effects 0.000 description 11
- 108020001507 fusion proteins Proteins 0.000 description 11
- 102000037865 fusion proteins Human genes 0.000 description 11
- 210000004379 membrane Anatomy 0.000 description 11
- 206010061041 Chlamydial infection Diseases 0.000 description 10
- 102100038132 Endogenous retrovirus group K member 6 Pro protein Human genes 0.000 description 10
- 241000282414 Homo sapiens Species 0.000 description 10
- 230000002776 aggregation Effects 0.000 description 10
- 238000004220 aggregation Methods 0.000 description 10
- 230000000890 antigenic effect Effects 0.000 description 10
- 108091005804 Peptidases Proteins 0.000 description 9
- 239000004365 Protease Substances 0.000 description 9
- 206010070834 Sensitisation Diseases 0.000 description 9
- 238000013459 approach Methods 0.000 description 9
- 230000008569 process Effects 0.000 description 9
- 230000008313 sensitization Effects 0.000 description 9
- 238000001890 transfection Methods 0.000 description 9
- 208000007190 Chlamydia Infections Diseases 0.000 description 8
- WYURNTSHIVDZCO-UHFFFAOYSA-N Tetrahydrofuran Chemical compound C1CCOC1 WYURNTSHIVDZCO-UHFFFAOYSA-N 0.000 description 8
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 8
- 238000005516 engineering process Methods 0.000 description 8
- 238000007912 intraperitoneal administration Methods 0.000 description 8
- 238000004519 manufacturing process Methods 0.000 description 8
- 230000001717 pathogenic effect Effects 0.000 description 8
- 239000007858 starting material Substances 0.000 description 8
- NOWKCMXCCJGMRR-UHFFFAOYSA-N Aziridine Chemical compound C1CN1 NOWKCMXCCJGMRR-UHFFFAOYSA-N 0.000 description 7
- 238000011725 BALB/c mouse Methods 0.000 description 7
- 238000002965 ELISA Methods 0.000 description 7
- 101710098399 Outer membrane protein YopN Proteins 0.000 description 7
- 238000001476 gene delivery Methods 0.000 description 7
- 239000008103 glucose Substances 0.000 description 7
- 210000004988 splenocyte Anatomy 0.000 description 7
- 108010077805 Bacterial Proteins Proteins 0.000 description 6
- 108090000695 Cytokines Proteins 0.000 description 6
- 102000004127 Cytokines Human genes 0.000 description 6
- 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 6
- 206010061218 Inflammation Diseases 0.000 description 6
- 102100037850 Interferon gamma Human genes 0.000 description 6
- 108010074328 Interferon-gamma Proteins 0.000 description 6
- 241000700605 Viruses Species 0.000 description 6
- 238000010171 animal model Methods 0.000 description 6
- 238000010668 complexation reaction Methods 0.000 description 6
- 238000011161 development Methods 0.000 description 6
- 239000003814 drug Substances 0.000 description 6
- 238000002296 dynamic light scattering Methods 0.000 description 6
- 210000000987 immune system Anatomy 0.000 description 6
- 230000036039 immunity Effects 0.000 description 6
- 230000004054 inflammatory process Effects 0.000 description 6
- 230000003993 interaction Effects 0.000 description 6
- 210000004072 lung Anatomy 0.000 description 6
- 239000002773 nucleotide Substances 0.000 description 6
- 125000003729 nucleotide group Chemical group 0.000 description 6
- 230000002829 reductive effect Effects 0.000 description 6
- 230000028327 secretion Effects 0.000 description 6
- 239000008223 sterile water Substances 0.000 description 6
- 235000000346 sugar Nutrition 0.000 description 6
- 239000013598 vector Substances 0.000 description 6
- 208000035086 Chlamydophila Infections Diseases 0.000 description 5
- 241000238740 Dermatophagoides pteronyssinus Species 0.000 description 5
- 239000002671 adjuvant Substances 0.000 description 5
- 239000003242 anti bacterial agent Substances 0.000 description 5
- 229940088710 antibiotic agent Drugs 0.000 description 5
- 208000006673 asthma Diseases 0.000 description 5
- 239000000460 chlorine Substances 0.000 description 5
- 229940079593 drug Drugs 0.000 description 5
- 239000012636 effector Substances 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 230000006698 induction Effects 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 238000005259 measurement Methods 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- 239000000178 monomer Substances 0.000 description 5
- 230000002085 persistent effect Effects 0.000 description 5
- 210000002966 serum Anatomy 0.000 description 5
- 206010041232 sneezing Diseases 0.000 description 5
- 210000001519 tissue Anatomy 0.000 description 5
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 4
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 4
- 108060003951 Immunoglobulin Proteins 0.000 description 4
- 229910020889 NaBH3 Inorganic materials 0.000 description 4
- 108091028043 Nucleic acid sequence Proteins 0.000 description 4
- 229910019142 PO4 Inorganic materials 0.000 description 4
- 238000000246 agarose gel electrophoresis Methods 0.000 description 4
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 4
- 229910021502 aluminium hydroxide Inorganic materials 0.000 description 4
- 150000001412 amines Chemical class 0.000 description 4
- 230000006399 behavior Effects 0.000 description 4
- 230000008901 benefit Effects 0.000 description 4
- 229940077731 carbohydrate nutrients Drugs 0.000 description 4
- 229920006317 cationic polymer Polymers 0.000 description 4
- 230000004700 cellular uptake Effects 0.000 description 4
- 210000000805 cytoplasm Anatomy 0.000 description 4
- 102000018358 immunoglobulin Human genes 0.000 description 4
- 229940072221 immunoglobulins Drugs 0.000 description 4
- 230000001024 immunotherapeutic effect Effects 0.000 description 4
- 230000002458 infectious effect Effects 0.000 description 4
- 210000004962 mammalian cell Anatomy 0.000 description 4
- 235000021317 phosphate Nutrition 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 239000000523 sample Substances 0.000 description 4
- 241000894007 species Species 0.000 description 4
- 238000003756 stirring Methods 0.000 description 4
- 150000008163 sugars Chemical class 0.000 description 4
- YLQBMQCUIZJEEH-UHFFFAOYSA-N tetrahydrofuran Natural products C=1C=COC=1 YLQBMQCUIZJEEH-UHFFFAOYSA-N 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 4
- 239000013603 viral vector Substances 0.000 description 4
- GUBGYTABKSRVRQ-PZPXDAEZSA-N 4β-mannobiose Chemical compound O[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-PZPXDAEZSA-N 0.000 description 3
- 102000013918 Apolipoproteins E Human genes 0.000 description 3
- 108010025628 Apolipoproteins E Proteins 0.000 description 3
- 108010074051 C-Reactive Protein Proteins 0.000 description 3
- 102100032752 C-reactive protein Human genes 0.000 description 3
- 108091026890 Coding region Proteins 0.000 description 3
- 229940021995 DNA vaccine Drugs 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 3
- DKXNBNKWCZZMJT-UHFFFAOYSA-N O4-alpha-D-Mannopyranosyl-D-mannose Natural products O=CC(O)C(O)C(C(O)CO)OC1OC(CO)C(O)C(O)C1O DKXNBNKWCZZMJT-UHFFFAOYSA-N 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 239000007983 Tris buffer Substances 0.000 description 3
- 238000002835 absorbance Methods 0.000 description 3
- 230000004913 activation Effects 0.000 description 3
- 239000008186 active pharmaceutical agent Substances 0.000 description 3
- 229940074608 allergen extract Drugs 0.000 description 3
- 208000030961 allergic reaction Diseases 0.000 description 3
- 125000003277 amino group Chemical group 0.000 description 3
- 125000000129 anionic group Chemical group 0.000 description 3
- 230000001580 bacterial effect Effects 0.000 description 3
- 230000003197 catalytic effect Effects 0.000 description 3
- 239000006285 cell suspension Substances 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 230000001684 chronic effect Effects 0.000 description 3
- 238000010276 construction Methods 0.000 description 3
- 235000018417 cysteine Nutrition 0.000 description 3
- 210000000172 cytosol Anatomy 0.000 description 3
- 231100000433 cytotoxic Toxicity 0.000 description 3
- 230000001472 cytotoxic effect Effects 0.000 description 3
- 239000000850 decongestant Substances 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 3
- 239000013604 expression vector Substances 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 230000036732 histological change Effects 0.000 description 3
- 150000003840 hydrochlorides Chemical class 0.000 description 3
- 230000009610 hypersensitivity Effects 0.000 description 3
- 210000002865 immune cell Anatomy 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 230000007774 longterm Effects 0.000 description 3
- 210000004698 lymphocyte Anatomy 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 239000002609 medium Substances 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 238000010172 mouse model Methods 0.000 description 3
- 229910052757 nitrogen Inorganic materials 0.000 description 3
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 3
- 239000010452 phosphate Substances 0.000 description 3
- 239000002244 precipitate Substances 0.000 description 3
- 150000003141 primary amines Chemical class 0.000 description 3
- 230000000069 prophylactic effect Effects 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 238000003118 sandwich ELISA Methods 0.000 description 3
- 238000007086 side reaction Methods 0.000 description 3
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Natural products 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 3
- 238000007920 subcutaneous administration Methods 0.000 description 3
- 239000006228 supernatant Substances 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- GUBGYTABKSRVRQ-TYAPZPMWSA-N 4α-mannobiose Chemical compound O[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-TYAPZPMWSA-N 0.000 description 2
- 108010088751 Albumins Proteins 0.000 description 2
- 102000009027 Albumins Human genes 0.000 description 2
- 208000035285 Allergic Seasonal Rhinitis Diseases 0.000 description 2
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 2
- 101710205625 Capsid protein p24 Proteins 0.000 description 2
- 241001185363 Chlamydiae Species 0.000 description 2
- 208000035473 Communicable disease Diseases 0.000 description 2
- 108091035707 Consensus sequence Proteins 0.000 description 2
- 108010041986 DNA Vaccines Proteins 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- NTYJJOPFIAHURM-UHFFFAOYSA-N Histamine Chemical group NCCC1=CN=CN1 NTYJJOPFIAHURM-UHFFFAOYSA-N 0.000 description 2
- 206010020772 Hypertension Diseases 0.000 description 2
- 108090000978 Interleukin-4 Proteins 0.000 description 2
- 108010002616 Interleukin-5 Proteins 0.000 description 2
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 2
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 2
- 229930182816 L-glutamine Natural products 0.000 description 2
- 102000007330 LDL Lipoproteins Human genes 0.000 description 2
- 108010007622 LDL Lipoproteins Proteins 0.000 description 2
- 108010052285 Membrane Proteins Proteins 0.000 description 2
- 101710163270 Nuclease Proteins 0.000 description 2
- 101710177166 Phosphoprotein Proteins 0.000 description 2
- 206010035664 Pneumonia Diseases 0.000 description 2
- 241000238711 Pyroglyphidae Species 0.000 description 2
- 238000002123 RNA extraction Methods 0.000 description 2
- 206010039094 Rhinitis perennial Diseases 0.000 description 2
- 208000036284 Rhinitis seasonal Diseases 0.000 description 2
- 108700028909 Serum Amyloid A Proteins 0.000 description 2
- 102000054727 Serum Amyloid A Human genes 0.000 description 2
- 101710149279 Small delta antigen Proteins 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 230000024932 T cell mediated immunity Effects 0.000 description 2
- 102100022563 Tubulin polymerization-promoting protein Human genes 0.000 description 2
- 108010069584 Type III Secretion Systems Proteins 0.000 description 2
- 108700005077 Viral Genes Proteins 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 230000001154 acute effect Effects 0.000 description 2
- 235000004279 alanine Nutrition 0.000 description 2
- 150000001298 alcohols Chemical class 0.000 description 2
- GZCGUPFRVQAUEE-SLPGGIOYSA-N aldehydo-D-glucose Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C=O GZCGUPFRVQAUEE-SLPGGIOYSA-N 0.000 description 2
- 201000009961 allergic asthma Diseases 0.000 description 2
- 230000030741 antigen processing and presentation Effects 0.000 description 2
- 229940125715 antihistaminic agent Drugs 0.000 description 2
- 239000000739 antihistaminic agent Substances 0.000 description 2
- 239000004599 antimicrobial Substances 0.000 description 2
- 230000036523 atherogenesis Effects 0.000 description 2
- 201000008937 atopic dermatitis Diseases 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 210000004369 blood Anatomy 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 239000008366 buffered solution Substances 0.000 description 2
- 230000003139 buffering effect Effects 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 201000000902 chlamydia Diseases 0.000 description 2
- 208000012538 chlamydia trachomatis infectious disease Diseases 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 238000005056 compaction Methods 0.000 description 2
- 229920001577 copolymer Polymers 0.000 description 2
- 239000003246 corticosteroid Substances 0.000 description 2
- 229960001334 corticosteroids Drugs 0.000 description 2
- 239000012043 crude product Substances 0.000 description 2
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 229940124581 decongestants Drugs 0.000 description 2
- 230000007423 decrease Effects 0.000 description 2
- 238000004925 denaturation Methods 0.000 description 2
- 230000036425 denaturation Effects 0.000 description 2
- 210000004443 dendritic cell Anatomy 0.000 description 2
- 230000001419 dependent effect Effects 0.000 description 2
- 238000000586 desensitisation Methods 0.000 description 2
- GLYLMXARZJNUEY-UHFFFAOYSA-N dichloromethane;methanol;hydrate Chemical compound O.OC.ClCCl GLYLMXARZJNUEY-UHFFFAOYSA-N 0.000 description 2
- 238000000914 diffusion-ordered spectroscopy Methods 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 150000002016 disaccharides Chemical class 0.000 description 2
- 238000004090 dissolution Methods 0.000 description 2
- 210000001163 endosome Anatomy 0.000 description 2
- 210000003038 endothelium Anatomy 0.000 description 2
- 239000002158 endotoxin Substances 0.000 description 2
- 230000008029 eradication Effects 0.000 description 2
- 239000000284 extract Substances 0.000 description 2
- 230000002349 favourable effect Effects 0.000 description 2
- 239000012091 fetal bovine serum Substances 0.000 description 2
- 210000000497 foam cell Anatomy 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 239000001963 growth medium Substances 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 229920001519 homopolymer Polymers 0.000 description 2
- 229940046533 house dust mites Drugs 0.000 description 2
- 230000028996 humoral immune response Effects 0.000 description 2
- 230000002519 immonomodulatory effect Effects 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 208000027866 inflammatory disease Diseases 0.000 description 2
- 239000000543 intermediate Substances 0.000 description 2
- 238000011813 knockout mouse model Methods 0.000 description 2
- 210000001821 langerhans cell Anatomy 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 239000008176 lyophilized powder Substances 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 238000007726 management method Methods 0.000 description 2
- 238000002483 medication Methods 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- 230000003278 mimic effect Effects 0.000 description 2
- 150000002772 monosaccharides Chemical class 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 230000003472 neutralizing effect Effects 0.000 description 2
- 210000001331 nose Anatomy 0.000 description 2
- 239000012038 nucleophile Substances 0.000 description 2
- 238000005457 optimization Methods 0.000 description 2
- 230000008520 organization Effects 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 230000003950 pathogenic mechanism Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 210000003800 pharynx Anatomy 0.000 description 2
- 229920000768 polyamine Polymers 0.000 description 2
- 229920001601 polyetherimide Polymers 0.000 description 2
- 238000006116 polymerization reaction Methods 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 239000011148 porous material Substances 0.000 description 2
- 238000001556 precipitation Methods 0.000 description 2
- 239000002243 precursor Substances 0.000 description 2
- 238000004321 preservation Methods 0.000 description 2
- 230000002797 proteolythic effect Effects 0.000 description 2
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 238000006722 reduction reaction Methods 0.000 description 2
- 210000003289 regulatory T cell Anatomy 0.000 description 2
- 108091008146 restriction endonucleases Proteins 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 206010039083 rhinitis Diseases 0.000 description 2
- 150000003335 secondary amines Chemical class 0.000 description 2
- 125000000467 secondary amino group Chemical group [H]N([*:1])[*:2] 0.000 description 2
- 230000003248 secreting effect Effects 0.000 description 2
- 210000003491 skin Anatomy 0.000 description 2
- 210000004989 spleen cell Anatomy 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 230000009897 systematic effect Effects 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- 150000003573 thiols Chemical class 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- 238000001665 trituration Methods 0.000 description 2
- 230000003612 virological effect Effects 0.000 description 2
- 230000003442 weekly effect Effects 0.000 description 2
- 238000001262 western blot Methods 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- OJHZNMVJJKMFGX-BWCYBWMMSA-N (4r,4ar,7ar,12bs)-9-methoxy-3-methyl-1,2,4,4a,5,6,7a,13-octahydro-4,12-methanobenzofuro[3,2-e]isoquinoline-7-one;(2r,3r)-2,3-dihydroxybutanedioic acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O.C([C@H]1[C@H](N(CC[C@@]112)C)C3)CC(=O)[C@@H]1OC1=C2C3=CC=C1OC OJHZNMVJJKMFGX-BWCYBWMMSA-N 0.000 description 1
- 238000005160 1H NMR spectroscopy Methods 0.000 description 1
- IMSODMZESSGVBE-UHFFFAOYSA-N 2-Oxazoline Chemical compound C1CN=CO1 IMSODMZESSGVBE-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 101710132383 66 kDa protein Proteins 0.000 description 1
- 241000238876 Acari Species 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 208000024827 Alzheimer disease Diseases 0.000 description 1
- 244000036975 Ambrosia artemisiifolia Species 0.000 description 1
- 235000003129 Ambrosia artemisiifolia var elatior Nutrition 0.000 description 1
- 238000010953 Ames test Methods 0.000 description 1
- 231100000039 Ames test Toxicity 0.000 description 1
- 240000002470 Amphicarpaea bracteata Species 0.000 description 1
- 208000012657 Atopic disease Diseases 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 230000028728 B cell mediated immunity Effects 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 208000019300 CLIPPERS Diseases 0.000 description 1
- 101100297345 Caenorhabditis elegans pgl-2 gene Proteins 0.000 description 1
- BHPQYMZQTOCNFJ-UHFFFAOYSA-N Calcium cation Chemical compound [Ca+2] BHPQYMZQTOCNFJ-UHFFFAOYSA-N 0.000 description 1
- 206010007269 Carcinogenicity Diseases 0.000 description 1
- 208000024172 Cardiovascular disease Diseases 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108091006146 Channels Proteins 0.000 description 1
- 101100024012 Chlamydia pneumoniae ompA gene Proteins 0.000 description 1
- 241001647378 Chlamydia psittaci Species 0.000 description 1
- 241000606153 Chlamydia trachomatis Species 0.000 description 1
- 241000606069 Chlamydiaceae Species 0.000 description 1
- 241001065357 Chlamydophila pneumoniae TW-183 Species 0.000 description 1
- ZAMOUSCENKQFHK-UHFFFAOYSA-N Chlorine atom Chemical compound [Cl] ZAMOUSCENKQFHK-UHFFFAOYSA-N 0.000 description 1
- 208000006545 Chronic Obstructive Pulmonary Disease Diseases 0.000 description 1
- 208000014085 Chronic respiratory disease Diseases 0.000 description 1
- 206010009137 Chronic sinusitis Diseases 0.000 description 1
- 206010010744 Conjunctivitis allergic Diseases 0.000 description 1
- 206010011224 Cough Diseases 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 238000011238 DNA vaccination Methods 0.000 description 1
- 206010012434 Dermatitis allergic Diseases 0.000 description 1
- 206010012438 Dermatitis atopic Diseases 0.000 description 1
- 241000238713 Dermatophagoides farinae Species 0.000 description 1
- 108010061608 Dermatophagoides pteronyssinus antigen p 2 Proteins 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 208000004739 Egg Hypersensitivity Diseases 0.000 description 1
- 101710091045 Envelope protein Proteins 0.000 description 1
- 102000010911 Enzyme Precursors Human genes 0.000 description 1
- 108010062466 Enzyme Precursors Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- JOYRKODLDBILNP-UHFFFAOYSA-N Ethyl urethane Chemical compound CCOC(N)=O JOYRKODLDBILNP-UHFFFAOYSA-N 0.000 description 1
- 206010015993 Eyelid oedema Diseases 0.000 description 1
- 208000010412 Glaucoma Diseases 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- 206010019233 Headaches Diseases 0.000 description 1
- 102000008949 Histocompatibility Antigens Class I Human genes 0.000 description 1
- 108010088652 Histocompatibility Antigens Class I Proteins 0.000 description 1
- 101000881168 Homo sapiens SPARC Proteins 0.000 description 1
- 101150106931 IFNG gene Proteins 0.000 description 1
- 108010054738 Immunologic Receptors Proteins 0.000 description 1
- 102000001749 Immunologic Receptors Human genes 0.000 description 1
- 108700001097 Insect Genes Proteins 0.000 description 1
- 108090000176 Interleukin-13 Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 102000029749 Microtubule Human genes 0.000 description 1
- 108091022875 Microtubule Proteins 0.000 description 1
- MSFSPUZXLOGKHJ-UHFFFAOYSA-N Muraminsaeure Natural products OC(=O)C(C)OC1C(N)C(O)OC(CO)C1O MSFSPUZXLOGKHJ-UHFFFAOYSA-N 0.000 description 1
- 102000005604 Myosin Heavy Chains Human genes 0.000 description 1
- 108010084498 Myosin Heavy Chains Proteins 0.000 description 1
- 238000005481 NMR spectroscopy Methods 0.000 description 1
- 108091061960 Naked DNA Proteins 0.000 description 1
- 208000031662 Noncommunicable disease Diseases 0.000 description 1
- 206010030113 Oedema Diseases 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 206010033078 Otitis media Diseases 0.000 description 1
- 101710116435 Outer membrane protein Proteins 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
- 108010013639 Peptidoglycan Proteins 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 208000037062 Polyps Diseases 0.000 description 1
- 108010013381 Porins Proteins 0.000 description 1
- 101000621511 Potato virus M (strain German) RNA silencing suppressor Proteins 0.000 description 1
- 101710194807 Protective antigen Proteins 0.000 description 1
- 102000009516 Protein Serine-Threonine Kinases Human genes 0.000 description 1
- 108010009341 Protein Serine-Threonine Kinases Proteins 0.000 description 1
- 101710188315 Protein X Proteins 0.000 description 1
- 208000003251 Pruritus Diseases 0.000 description 1
- 239000012980 RPMI-1640 medium Substances 0.000 description 1
- 208000033464 Reiter syndrome Diseases 0.000 description 1
- 208000036071 Rhinorrhea Diseases 0.000 description 1
- 206010039101 Rhinorrhoea Diseases 0.000 description 1
- 102100037599 SPARC Human genes 0.000 description 1
- 241000607142 Salmonella Species 0.000 description 1
- 241000607768 Shigella Species 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 239000000150 Sympathomimetic Substances 0.000 description 1
- 210000001744 T-lymphocyte Anatomy 0.000 description 1
- 208000007536 Thrombosis Diseases 0.000 description 1
- 206010062910 Vascular infections Diseases 0.000 description 1
- 206010047141 Vasodilatation Diseases 0.000 description 1
- 241000607734 Yersinia <bacteria> Species 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 238000011481 absorbance measurement Methods 0.000 description 1
- 239000002250 absorbent Substances 0.000 description 1
- 230000002745 absorbent Effects 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 239000002390 adhesive tape Substances 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 239000011543 agarose gel Substances 0.000 description 1
- 208000002205 allergic conjunctivitis Diseases 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 208000003455 anaphylaxis Diseases 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 235000003484 annual ragweed Nutrition 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 230000002590 anti-leukotriene effect Effects 0.000 description 1
- 230000001775 anti-pathogenic effect Effects 0.000 description 1
- 210000000612 antigen-presenting cell Anatomy 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 208000024998 atopic conjunctivitis Diseases 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- 244000052616 bacterial pathogen Species 0.000 description 1
- 229920002988 biodegradable polymer Polymers 0.000 description 1
- 239000004621 biodegradable polymer Substances 0.000 description 1
- 229960000074 biopharmaceutical Drugs 0.000 description 1
- 229920001400 block copolymer Polymers 0.000 description 1
- 229910000085 borane Inorganic materials 0.000 description 1
- 206010006451 bronchitis Diseases 0.000 description 1
- 229940124630 bronchodilator Drugs 0.000 description 1
- 239000000168 bronchodilator agent Substances 0.000 description 1
- 235000006263 bur ragweed Nutrition 0.000 description 1
- 238000010805 cDNA synthesis kit Methods 0.000 description 1
- 229910001424 calcium ion Inorganic materials 0.000 description 1
- 230000000711 cancerogenic effect Effects 0.000 description 1
- 150000001735 carboxylic acids Chemical class 0.000 description 1
- 231100000357 carcinogen Toxicity 0.000 description 1
- 239000003183 carcinogenic agent Substances 0.000 description 1
- 231100000260 carcinogenicity Toxicity 0.000 description 1
- 230000007670 carcinogenicity Effects 0.000 description 1
- 238000012656 cationic ring opening polymerization Methods 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000006800 cellular catabolic process Effects 0.000 description 1
- 230000008859 change Effects 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 239000003638 chemical reducing agent Substances 0.000 description 1
- 229910052801 chlorine Inorganic materials 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 208000037976 chronic inflammation Diseases 0.000 description 1
- 208000037893 chronic inflammatory disorder Diseases 0.000 description 1
- 230000012085 chronic inflammatory response Effects 0.000 description 1
- 208000021930 chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids Diseases 0.000 description 1
- 208000027157 chronic rhinosinusitis Diseases 0.000 description 1
- 235000003488 common ragweed Nutrition 0.000 description 1
- 229940052810 complex b Drugs 0.000 description 1
- 230000009918 complex formation Effects 0.000 description 1
- 230000000536 complexating effect Effects 0.000 description 1
- 238000006482 condensation reaction Methods 0.000 description 1
- 230000001276 controlling 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
- 238000004132 cross linking Methods 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 230000016396 cytokine production Effects 0.000 description 1
- KWGRBVOPPLSCSI-UHFFFAOYSA-N d-ephedrine Natural products CNC(C)C(O)C1=CC=CC=C1 KWGRBVOPPLSCSI-UHFFFAOYSA-N 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 238000000354 decomposition reaction Methods 0.000 description 1
- 230000006735 deficit Effects 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 201000010860 egg allergy Diseases 0.000 description 1
- 235000013601 eggs Nutrition 0.000 description 1
- 230000009881 electrostatic interaction Effects 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 230000000021 endosomolytic effect Effects 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 210000003979 eosinophil Anatomy 0.000 description 1
- 230000036566 epidermal hyperplasia Effects 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 210000001508 eye Anatomy 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000013020 final formulation Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000011888 foil Substances 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000002695 general anesthesia Methods 0.000 description 1
- 229960002743 glutamine Drugs 0.000 description 1
- 239000005090 green fluorescent protein Substances 0.000 description 1
- 231100001261 hazardous Toxicity 0.000 description 1
- 231100000869 headache Toxicity 0.000 description 1
- 229960001340 histamine Drugs 0.000 description 1
- 210000003630 histaminocyte Anatomy 0.000 description 1
- 230000007440 host cell apoptosis Effects 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 244000052637 human pathogen Species 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 229940124452 immunizing agent Drugs 0.000 description 1
- 238000003119 immunoblot Methods 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 230000001976 improved effect Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000001095 inductively coupled plasma mass spectrometry Methods 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 208000030603 inherited susceptibility to asthma Diseases 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 239000003999 initiator Substances 0.000 description 1
- 239000002054 inoculum Substances 0.000 description 1
- 239000002919 insect venom Substances 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 244000000056 intracellular parasite Species 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 230000001788 irregular Effects 0.000 description 1
- 230000007803 itching Effects 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 231100001231 less toxic Toxicity 0.000 description 1
- 230000021633 leukocyte mediated immunity Effects 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 230000037356 lipid metabolism Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 229920006008 lipopolysaccharide Polymers 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 239000003120 macrolide antibiotic agent Substances 0.000 description 1
- 229940041033 macrolides Drugs 0.000 description 1
- 125000000317 mannobiose group Chemical group 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 238000010197 meta-analysis Methods 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000003641 microbiacidal effect Effects 0.000 description 1
- 244000000010 microbial pathogen Species 0.000 description 1
- 229940124561 microbicide Drugs 0.000 description 1
- 239000002855 microbicide agent Substances 0.000 description 1
- 238000000386 microscopy Methods 0.000 description 1
- 210000004688 microtubule Anatomy 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 239000003068 molecular probe Substances 0.000 description 1
- 239000002808 molecular sieve Substances 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 210000004877 mucosa Anatomy 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 231100000219 mutagenic Toxicity 0.000 description 1
- 230000003505 mutagenic effect Effects 0.000 description 1
- 210000002850 nasal mucosa Anatomy 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 238000011369 optimal treatment Methods 0.000 description 1
- 238000012261 overproduction Methods 0.000 description 1
- 150000002918 oxazolines Chemical class 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 210000003254 palate Anatomy 0.000 description 1
- 244000045947 parasite Species 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 208000022719 perennial allergic rhinitis Diseases 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 238000001050 pharmacotherapy Methods 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 210000004180 plasmocyte Anatomy 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920002851 polycationic polymer Polymers 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 102000007739 porin activity proteins Human genes 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- 239000000843 powder Substances 0.000 description 1
- 230000000207 pro-atherogenic effect Effects 0.000 description 1
- 239000003805 procoagulant Substances 0.000 description 1
- 230000009465 prokaryotic expression Effects 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 230000001737 promoting effect Effects 0.000 description 1
- 230000002633 protecting effect Effects 0.000 description 1
- 125000006239 protecting group Chemical group 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- KWGRBVOPPLSCSI-WCBMZHEXSA-N pseudoephedrine Chemical compound CN[C@@H](C)[C@@H](O)C1=CC=CC=C1 KWGRBVOPPLSCSI-WCBMZHEXSA-N 0.000 description 1
- 229960003908 pseudoephedrine Drugs 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 235000009736 ragweed Nutrition 0.000 description 1
- 229920005604 random copolymer Polymers 0.000 description 1
- 239000002994 raw material Substances 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 208000036273 reactive airway disease Diseases 0.000 description 1
- 208000002574 reactive arthritis Diseases 0.000 description 1
- 230000009257 reactivity Effects 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 229940126583 recombinant protein vaccine Drugs 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 210000002345 respiratory system Anatomy 0.000 description 1
- 208000023504 respiratory system disease Diseases 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 239000012266 salt solution Substances 0.000 description 1
- 201000000306 sarcoidosis Diseases 0.000 description 1
- 102000014452 scavenger receptors Human genes 0.000 description 1
- 108010078070 scavenger receptors Proteins 0.000 description 1
- 208000017022 seasonal allergic rhinitis Diseases 0.000 description 1
- 230000001932 seasonal effect Effects 0.000 description 1
- 230000008786 sensory perception of smell Effects 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 230000000405 serological effect Effects 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 201000009890 sinusitis Diseases 0.000 description 1
- 230000008591 skin barrier function Effects 0.000 description 1
- URGAHOPLAPQHLN-UHFFFAOYSA-N sodium aluminosilicate Chemical compound [Na+].[Al+3].[O-][Si]([O-])=O.[O-][Si]([O-])=O URGAHOPLAPQHLN-UHFFFAOYSA-N 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 230000008961 swelling Effects 0.000 description 1
- 230000001975 sympathomimetic effect Effects 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 150000003512 tertiary amines Chemical class 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- 210000001578 tight junction Anatomy 0.000 description 1
- 230000024664 tolerance induction Effects 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 230000037317 transdermal delivery Effects 0.000 description 1
- 150000004043 trisaccharides Chemical class 0.000 description 1
- 210000004026 tunica intima Anatomy 0.000 description 1
- 238000002211 ultraviolet spectrum Methods 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 230000024883 vasodilation Effects 0.000 description 1
- 231100000611 venom Toxicity 0.000 description 1
- 229940023147 viral vector vaccine Drugs 0.000 description 1
- 230000001018 virulence Effects 0.000 description 1
- 239000000304 virulence factor Substances 0.000 description 1
- 230000007923 virulence factor Effects 0.000 description 1
Classifications
-
- B—PERFORMING OPERATIONS; TRANSPORTING
- B82—NANOTECHNOLOGY
- B82Y—SPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
- B82Y5/00—Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
- A61K39/21—Retroviridae, e.g. equine infectious anemia virus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/08—Antiallergic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/525—Virus
- A61K2039/5258—Virus-like particles
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/53—DNA (RNA) vaccination
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/545—Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/60—Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
- A61K2039/6093—Synthetic polymers, e.g. polyethyleneglycol [PEG], Polymers or copolymers of (D) glutamate and (D) lysine
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16034—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- the invention belongs to the field of immunotherapy and vaccines. More particularly, the invention relates to a pharmaceutical composition comprising nanoparticles being composed of macromolecules and linear polyethyleneimine (1-PEI) or 1-PEI derivative in a pharmaceutically acceptable liquid formulation, as well as a method for the preparation thereof. Uses of said compositions are also provided.
- Viral vectors have been developed to facilitate the delivery of therapeutic genes and immunogenic antigens because viruses evolved to efficiently enter into the cells, to overcome the cellular degradations and to express their genetic materials (Lungwitz et al., 2005; Gore, 2003).
- viral vector- based vaccines present significant manufacturing and safety challenges compared to plasmid DNA, and their repeated administration is restricted due to vector immunity (Lehman, 1999; Gore, 2003).
- Cationic polymers are particularly attractive for the delivery of nucleic acids, since they allow synthetic modification of their structure, enabling the enhancement of transfection efficiency and reduction in toxicity.
- the interaction between the positively charged polymer backbone and negatively charged DNA leads to the spontaneous formation of nano-size complexes (polyplexes, nanoparticles) in aqueous milieu.
- the compact structure of the polyplex core prevents the access of nucleases and other degradation factors to the enclosed nucleic acid drugs.
- the polyplexes should maintain their stability before cellular uptake by endocytosis and should escape from the endosomal compartment inside the cell.
- PEI polyethylenimine
- the branched form of PEI (b-PEI; CAS/RN: 9002-98- 6; synonyms: aziridine homopolymer, ethyleneimine homopolymer) can be synthesized by acid-catalysed polymerization of aziridine monomers but technically the overall reaction is hardly controlable and dangerous (von Harpe et al, 2000).
- the other drawback in using this compound in biologicals and in vivo experiments is that the monomer (ethyleneimine or aziridine) is a cytotoxic and carcinogen material (RTECS, Toxline). It is very reactive, reacts with all kind of nucleophiles (amines, thiols, alcohols, carboxylic acids, etc.) to the ring-opened product.
- Branched PEI has a high density and variety of amine groups (primary, secondary and tertiary) which have different reactivity in physiological environment. Due to this fact and to the branched morphology, in the non-viral gene delivery studies the branched form of PEI is used at a higher N/P ratio (10-50) in the condensation reaction of pDNA into nanoparticles - which is cytotoxic - than in case of linear PEI (Jeong et al, 2007).
- Linear PEI (1-PEI; CAS/RN: 26913-06-04; synonym: polyamine) can be obtained by cationic ring opening polymerization of 2-substituted 2-oxazoline monomers, followed by acid-catalysed hydrolysis of the N- substituted polymer.
- Oxazoline is a cyclic 5 member ring with much less crowding and much less reactive than aziridine. It will form ring-opened products only with strong nucleophiles (like primary amines) and its reaction can be modified to allow different lengths of PEI chains.
- Oxazolines aren't usually considered mutagenic and neither is 1-PEI (Goddard et al, 1989; Zalipski et al, 1996). Neither one gives a positive Ames test. There are no enzymatic or chemical breakdown products from PEI that are reactive. Linear PEI contains only secondary amines (less reactive than primary amines) which are mostly protonated under physiological conditions.
- the relative gene expression can be strongly influenced by the particle size, which, in turn, can be moderated by the molecular weight of the PEI derivative, the method of particle preparation, and the N/P ratio.
- the polyplex size decreases with increasing molecular weight, an excess of the polyamine and in low ionic strength media, most likely due to improved DNA compaction (Ogris et al., 1998).
- These polyplexes are most commonly formulated in either glucose solutions or physiological salt.
- the physiological salt solution due to its high ionic strength, induces the aggregation of the polyplexes, thus decreasing their cellular uptake. After intravenal injection, the aggregation of these polyplexes represents a safety challenge for these formulations.
- Targeted gene delivery capitalizes on the presence of specific cell surface receptors for DNA uptake into cells by receptor mediated endocytosis. Therefore receptor-binding ligands, like sugars, are coupled to polycationic compounds like PEI that bind and condense DNA and effectively enhance gene transfer efficiency (US2001/0031498A1; Diebold et al., 1999).
- linear PEI for gene transfer was investigated and compared to random copolymers of poly(2-ethyl-2-oxazoline)-co-poly(ethylenimine) and N-alkylethylenimine (Jeong et al., 2001; Brissault et al., 2003), or biodegradable, cross-linked cationic multi-block copolymers of PEI with esters, amides (PCT/US2005/039779), or PEG molecules (US 6,652,886).
- Allergic rhinitis (hay fever) is the most frequent allergic disease affecting 10-20% of the population. The symptoms last only for a few months in some patients (seasonal rhinitis), while in others they last the whole year (perennial rhinitis). Some of the rhinitis symptoms are sneezing, watery rhinorrhea, itching of the nose, palate, throat and eyes, nose blocking, edema of eyelids and occasionally sense of smell impairment. Throat clearing and coughing can often accompany the symptoms.
- Medications are only available for treating allergic symptoms (e.g. decongestants, bronchodilators, antihistamines, and corticosteroids).
- immunotherapy is prescribed that includes a series of immunizations with extracts of various allergens in gradually increasing doses.
- allergen an abnormally high reaction (hypersensitivity) to a specific antigen (called allergen) which results in the release of mediators of the immediate type hypersensitivity.
- Allergic diseases are caused by complex interactions between environmental (e.g. diet, pollution, exposure to everyday chemicals, stress, etc.) and genetic factors.
- An allergen can be any particular substance, but some of the most common allergies include allergens from house dust mites, pollen, insect venoms and certain foods (e.g. peanuts, milk, egg). After sensitization (first contact with a given allergen), immunoglobulins E (IgE) are produced by B cells specific for the allergen.
- IgE immunoglobulins E
- IgE elicits hypersensitivity reaction trough binding Fc ⁇ RJ receptors on mast cells, one of the major effector cells of the immediate type allergic reaction. Their main preformed mediator is histamine which induces vasodilatation, increased vessel permeability, edema and mucous hypersecretion in the mucosa, thereby causing clinical symptoms.
- Allergic rhinitis which is an IgE mediated inflammatory disease of the nasal mucosa, characterized by T H 2 type (IL-4, IL-5, IL- 13) cytokine response.
- T H 2 type IL-4, IL-5, IL- 13
- the World Health Organization has stated that the optimal treatment of allergy consists of four actions: avoiding the substance that causes the allergy, taking symptomatic medication, undergoing an allergy vaccination program and education. Presently, only this combination of actions makes it possible for some allergic people to live a symptom-free life.
- many sufferers use prescription and over-the-counter pharmacotherapies such as antihistamines, nasal corticosteroids, anti-leukotriene agents and decongestants.
- these medications may provide temporary symptomatic relief, they can be inconvenient to use, cause unpleasant side effects and, as a consequence, may be unsuitable for certain disease populations.
- pseudoephedrine a sympathomimetic amine commonly used as a decongestant
- can cause hypertension thus is unsuitable for patients suffering with high blood pressure, diabetes and glaucoma. None of these treatments modifies the underlying disease state.
- Allergen immunotherapy (specific immunotherapy, SIT) is currently used as the standard method for treatment of seasonal and perennial allergic rhinitis caused by a variety of allergens (ragweed, house dust mite, animal dander). Increasing doses of the allergen extract administered by weekly injections over a period of several years result in decreased symptoms during the allergy season. Moreover, de sensitization with total allergen extracts can cause severe anaphylactic reactions during the treatment. - A -
- SCIT Subcutaneous immunotherapy
- IL-4, IL-5, IL-13 T H 2 type cytokine induction
- IgE IgE production from plasma cells
- eosinophil recruitment in the tissue SCIT is associated with immune deviation in favor of T H 1 responses with an overproduction of IFN ⁇ , and induction of regulatory T (Treg) lymphocytes that produce mainly IL-10 and/or TGF ⁇ .
- Treg cells suppress the allergen-specific T H 2 responses. These effects are accompanied by an increase in allergen-specific IgG antibodies, particularly in the IgG4 subclass in human (IgG2a and IgG2b in mice). The biologic relevance of increases in IgG4 after SCIT has been questioned, because there is a poor correlation with the improvement of clinical symptoms.
- Chlamydiaceae consists of one genus Chlamydia with three species (C. trachomatis, C. pneumoniae and C. psittaci) that cause human diseases.
- Chlamydia small obligate intracellular parasites, are similar to viruses, however, they contain DNA, RNA and ribosomes and since they make their own proteins and nucleic acids and are now considered to be true bacteria. They possess an inner and outer membrane similar to gram-negative bacteria and a lipopolysaccharide but do not have a peptidoglycan layer. Although they synthesize most of their metabolic intermediates, they are unable to make their own ATP and thus are energy parasites.
- C pneumoniae is a human respiratory pathogen that causes acute respiratory disease and accounts for 5-10% of the cases of community-acquired pneumonia, bronchitis and sinusitis.
- C pneumoniae has been associated with other acute and chronic respiratory diseases such as otitis media and chronic obstructive pulmonary disease.
- the clinical spectrum of C pneumoniae infection has been extended to atherosclerosis, a major cause of cardiovascular disease and death in the Western world. There are effective antibiotics, but eradication is often unsuccessful because the intracellularly persisting inactive bacteria are not accessible for antibiotics.
- the possibilities of vaccination for C pneumoniae include the activation of the humoral or cellular immune system.
- Humoral immune response frequently recognizes epitopes present on the surface of C pneumoniae, thus specific B-cell mediated immunity develops with antibody production, enabling vaccination possibilities to induce a humoral immune response with C pneumoniae proteins or peptides.
- the metabolically active life cycle of C pneumoniae lasts 72 hours, and antibodies are effective only against the extracellular form (elementary body, EB) of the bacteria.
- EB extracellular form
- CD8+ cytotoxic T lymphocytes have been shown to be crucial for protection during experimental infection (Penttila et al., 1999).
- Cytotoxic T lymphocytes are potentially capable to eliminate infected cells, based on the presentation of bacterial proteins associated with MHC I molecules, enabling vaccination possibilities with products that capable to induce a cellular immune response (e.g. plasmid DNA immunization).
- AIl Chlamydias species share a unique triphasic developmental cycle. The infectious form binds to the host cell and triggers its uptake. Once internalized, bacteria reside in a membrane -bound compartment, called the inclusion. Throughout their developmental cycle, Chlamydiae utilize the host cell to establish an environment favorable for their proliferation. The molecular mechanisms by which Chlamydiae elicit various host responses are mostly unknown.
- Type III secretion systems are part of the virulence machinery of major human pathogens, including Yersinia, Shigella and Salmonella. These highly sophisticated secretion devices are able to secrete virulence factors into the host cell cytoplasm or membrane. The components of the secretion apparatus are conserved between species, whereas type III effector proteins are not. All genes necessary for the assembly of a complete type III secretion apparatus are present in the Chlamydial genome.
- LcrE was identified as protective antigen against C pneumoniae infection.
- Thorpe et al. (Thorpe et al., 2007) showed that vaccination with an LcrE fusion protein enhanced CD4+ and CD8+ T cells in both the lung and airways during an intranasal C pneumoniae challenge in a murine model.
- the challenge inoculum was absent within 7 days in treated mice while permanent in untreated mice.
- Atherosclerosis a multifactorial process which commences as early as childhood but is clinically manifested later in life, is increasingly considered as an immune -mediated process of the vascular system.
- Several mechanisms have been proposed by which infection could contribute to atherosclerosis. These include infection as an initiator and perpetuator of initial damage to endothelium, an increase in the risk of thrombus formation through expression of pro coagulant factor, effects on serum lipids, and an increase in the chronic inflammatory response through cytokine production.
- ox-LDL low density lipoprotein
- Ox-LDL is taken up via the scavenger receptor, a process not controlled by intracellular cholesterol content and its accumulation in macrophages leads to foam-cell formation, an early event in the formation of atheromatous lesions.
- the current concept of atherosclerosis as a chronic inflammatory disease has thus renewed the interest in a potential infectious component.
- the pathogenic burden hypothesis postulates induction of an increasingly proatherogenic state by infection with one or more persistent microbial pathogens.
- the obligate intracellular bacterium C pneumoniae has been associated with atherosclerosis by seroepidemiology and a variety of direct detection methods including the cultural recovery of the viable pathogen from the plaque.
- DNA vaccination techniques have also been disclosed in US patent documents. These include the application of different DNA constructs expressing different proteins of the bacteria.
- the most commonly used protein is the bacterial major outer membrane protein (MOMP) or its fragments (see e.g. US 6,235,290; US 6,696,421; US 2002 c /0142001, US 7,220,423, etc.).
- MOMP major outer membrane protein
- nucleic acid and antigen delivery that is pharmaceutically acceptable, protects the macromolecule from degradation and is biologically and chemically stable in selected storage temperature.
- the present inventors surprisingly found a pharmaceutically acceptable liquid formulation suitable for stable complexation of antigens (a peptide, protein, plasmid DNA) with linear polyethylenimine (1-PEI) or 1-PEI derivative in a well-defined ratio.
- the present solution provides compacted nanoparticles with a diameter within the range from about 50 nm to about 500 nm, which provides protection for the antigens from degradations and supports their cellular delivery and its presentation to the host's immune system.
- the compact nanoparticles according to the present invention are termed herein as "NanoComp".
- WO96/02655 describes compositions containing cationic polymers, e.g. PEI for the delivery nucleic acids into cells.
- PEI cationic polymers
- Several factors that were deemed to be important for the preservation of biological activity of the nucleic acid were investigated.
- Complexes of PEI with nucleic acids are claimed at a ratio between 0,5 and 50, more preferably between 5 and 30, with the size of PEI in the range of 50 kDa and 800 kDa. Only complexes of the nucleic acids and cationic polymers are disclosed, formation of nanoparticles are neither taught nor suggested. Accordingly, the particular relevance of the nanoparticle formation for the biological efficiency, especially relevant for antigen delivery, was not revealed.
- US2003/0027784A1 describes compositions containing low molecular weight PEI of 5 kDa to 50 kDa. These polymers are synthesized by polymerization of aziridine monomer which leads to the formation of branched three-dimensional PEI molecules.
- the described molecular weight range overlaps with molecular weight of the PEI polymers claimed in present invention, there is major difference in the structure of polymers.
- the topology of the PEI polymers is an important factor for their safety and efficiency during in vitro or in vivo use.
- complexes formed between 1-PEI and DNA have different physico-chemical behavior from b-PEI/DNA complexes (Jeong et al, 2007).
- the imunogenic nanomedicine composition according to the present invention comprises pathogen-like intelligent nanoparticles generated from antigens (e.g. DNA, proteins, peptides) to mimic a pathogen exposure.
- a NanoComp resembles pathogens because it has condensed antigens in the core that enveloped with a polymer with sugar residues on the surface (Fig. IA-B).
- the NanoComp enters into antigen-presenting cells similarly to pathogens, primarily to the endosomes via receptor-mediated endocytosis (Fig. 2). Inside the cell, the polymer envelop of the NanoComp protects the antigen core from endosomal degradation and support antigen presentation. Importantly for pharmaceutical use, the NanoComp formulation improves the stability and immunogenicity of the antigens incorporated.
- NanoComp plasmid DNA formulation combines the advantages of viral vector- based and plasmid DNA vaccines. It utilizes chemical excipients to form a pathogen-like nanoparticle with features similar to viruses.
- PEIm mannobiosylated polymer
- NanoComp and its ingredients can be manufactured consistently at large scale and administered repeatedly since it only contains formulation excipients beside the disease-specific antigens.
- NanoComp formulation is particularly attractive for the delivery of nucleic acids due its excellent safety features and transfection efficiency, and can be used to induce immune responses for the prevention and treatment of diseases (e.g. chronic infectious pathogens, including intracellular viruses and bacteria, and neoplastic diseases, allergy).
- diseases e.g. chronic infectious pathogens, including intracellular viruses and bacteria, and neoplastic diseases, allergy).
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising nanoparticles being composed of macromolecules and linear polyethylenimine (1-PEI) or 1-PEI derivative in a pharmaceutically acceptable liquid or lyophilized formulation, wherein the diameter of said nanoparticles is within the range from about 50 nm to about 500 nm, and wherein the molar ratio of the PEI or PEI derivative and the macromolecule is from about 2 to about 10, preferably from about 2 to about 6, and more preferably about 3 to about 4.5.
- said 1-PEI or 1-PEI-derivative is in a protonated salt form, wherein the ratio of the protonable N atoms / total N atoms is from about 0.1 to about 0.8, preferably about 0.4.
- said 1-PEI or 1-PEI-derivative has a molecular weight less than 5OkDa, preferably between IkDa and 35kDa, more preferably between 1OkDa and 25kDa.
- the macromolecule is a nucleic acid, preferably plasmid DNA or RNA, and preferably said nucleic acid encodes one or more antigens, more preferably is a virus-like particle (VLP).
- VLP virus-like particle
- the size of said nucleic acid is from about 3 kbp to about 20 kbp, preferably from about 6 kbp to about 13 kbp.
- the macromolecule is a polypeptide, and preferably the size of said polypeptide is from about 1 kDa to about 400 kDa, preferably from about 20 kDa to about 100 kDa.
- liquid formulation of the pharmaceutical composition according to the invention is an aqueous solution having a pH value from about 5 to about 10, preferably from about 7 to about 8.
- the ionic strength of said formulation is from about 0.2 mM to about 50 mM, preferably from about 2 mM to about 5 mM.
- said pharmaceutically acceptable liquid formulation further comprises at least one sugar alcohol according to the general formula (I): H(HCHO)nH (I) wherein n is an integer from 2 to 12, and wherein said sugar alcohols include all enantiomers and epimers.
- n is an integer from 2 to 12, and wherein said sugar alcohols include all enantiomers and epimers.
- two, three or more different sugar alcohols are used.
- n is 6, and said sugar alcohol is preferably selected from the group consisting of mannitol, sorbitol, xylitol and combinations thereof, more preferably said sugar alcohol is D/L- mannitol.
- the concentration of said sugar alcohols is in the range from 0 to about 50% (w/v), preferably from about 10% to about 30%.
- the pharmaceutical composition according to the invention is stable at 0-40
- the relevant biological activity of the macromolecule is maintained for a time period of at least 4 weeks, more preferably at least 8 weeks.
- said macromolecule is a plasmid DNA, and wherein the physico-chemical properties (characterizable by the oc/ccc ratio) of said plasmid DNA are maintained at 0-40 0 C for a time period of at least 1 week, more preferably at least 4 weeks, thereby said plasmid DNA is protected from degradation.
- the invention provides a pharmaceutical composition, for use in inducing immune responses.
- the pharmaceutical composition is provided for prevention or for treatment of chronic diseases.
- the pharmaceutical composition is provided for use as a pharmaceutical to be introduced by needle injection or by mucosal, oral, vaginal, rectal or transdermal administration.
- the present invention provides a method for the preparation of a pharmaceutical composition comprising nanoparticles being composed of macromolecules and linear PEI or PEI derivative, said method comprising mixing macromolecules with 1-PEI or 1-PEI derivative in a pharmaceutically acceptable solvent, wherein the molar ratio of the PEI or PEI derivative and the macromolecule is from about 2 to about 10, preferably from about 2 to about 6, and more preferably about 3 to about 4,5, thereby forming nanoparticles with a diameter from about 50 nm to about 500 nm.
- said PEI or PEI-derivative is in a protonated salt form, wherein the ratio of the protonable N atoms / total N atoms is from about 0.1 to about 0.8, preferably about 0.4.
- said 1-PEI or 1-PEI-derivative has a molecular weight less than 5OkDa, preferably between IkDa and 35kDa, more preferably between 1OkDa and 25kDa.
- said macromolecule is a nucleic acid, preferably plasmid DNA or RNA, and preferably said nucleic acid encodes one or more antigens, more preferably is a virus-like particle (VLP).
- VLP virus-like particle
- the size of said nucleic acid is from about 3 kbp to about 20 kbp, preferably from about 6 kb to about 13 kbp.
- the macromolecule is a polypeptide, and preferably the size of said polypeptide is from about 1 kDa to about 400 kDa, preferably from about 20 kDa to about 100 kDa.
- the liquid formulation of the pharmaceutical composition according to the invention is an aqueous solution having a pH value from about 5 to about 10, preferably from about 7 to about 8.
- the ionic strength of said pharmaceutical composition is from about 0.2 mM to about 50 mM, preferably from about 2 mM to about 5 mM.
- said pharmaceutically acceptable liquid formulation further comprises at least one sugar alcohol according to the general formula (I): H(HCHO)nH (I) wherein n is an integer from 2 to 12, and said sugar alcohols include all enantiomers and epimers. In a specific embodiment, two, three or more different sugar alcohols are used.
- n 6
- said sugar alcohol is preferably selected from the group consisting of mannitol, sorbitol, xylitol and combinations thereof, more preferably said sugar alcohol is D/L- mannitol.
- the concentration of said sugar alcohols is in the range from 0 to about 50%, preferably from about 10% to about 30%.
- the ratio of the volume of solvent and the volume of macro molecule solution is from 0 to about 20, preferably about 6.
- said sugar alcohols are added in a solution prepared in a pH buffer (6.5 ⁇ pH ⁇ 8.1), with a weaker base than the polymer, having the general formula (II):
- the base according to general formula (II) is preferably triethylamine or triethanolamine.
- said pharmaceutical composition is reconstituted from a lyophilized state extemporaneously with sterile water.
- said pharmaceutical composition is reconstituted from frozen state after thawing.
- said macromolecule and said linear PEI or linear PEI derivative are separately frozen in buffered solution, and mixed together extemporaneously after thawing.
- said macromolecule is reconstituted from a lyophilized state extemporaneously with a buffered linear PEI or linear PEI derivative solution.
- PEI has been widely used for non-viral transfection in vitro and in vivo and has an advantage over other polycationes in that it combines strong DNA compaction capacity whith an intrinsic endosomolytic activity (Lungwitz et al., 2005).
- pyridine borane (BH 3 -pyridine complex, abbr.: BH 3 - pyridine) can be applied effectively in the preparation of derivatives of carbohydrates with N-atom containing compounds (e.g. polymers, like PEI).
- N-atom containing compounds e.g. polymers, like PEI.
- This catalyst is less toxic and hazardous than the CN-containing ,,state-of- art" catalyst (NaBH 3 CN).
- the present invention provides a process for the reductive amination of a carbohydrate with an amino-containing compound or acid addition salt thereof by using pyridine borane (BH 3 - pyridine complex) as catalyst.
- pyridine borane BH 3 - pyridine complex
- the said carbohydrate is a mono- or disaccharide.
- the said amino-containing compound is an amino-containing polymer, preferably polyethyleneimine (PEI).
- said carbohydrate is 4-O-( ⁇ -D-mannopyranosyl)- ⁇ -D-mannopyranose and the said amino-containing compound is polyethyleneimine (PEI).
- PEIm polyethyleneimine
- said PEI is applied in the form of an acid addition salt, more preferably as HCl salt.
- the pharmaceutical composition according to the invention is especially suited for the transdermal introduction of a nucleic acid encoding at least one allergenic epitope of a protein, thereby inducing tolerance and desensitizating or suppressing allergic symptoms to the introduced allergenic epitope.
- SCIT SCIT
- Conventional immunotherapy involves a gradual exposure to the natural allergen to enhance tolerance.
- SCIT SCIT is usually administered via a series of weekly injections over a period of six months to a year, during which the dose is steadily increased to a therapeutic level so as not to induce a severe allergic reaction.
- a therapeutic dosing level Once a therapeutic dosing level is reached, patients receive bi-weekly or monthly injections to build and maintain immunity over another two to four years.
- a course of treatment may require between 60 and 90 injections. Consequently, due to the inconvenience of requiring the treatment to be administered by a healthcare professional and the time period for the treatment to be completed, patient compliance is a significant issue.
- adverse side effects are relatively common, ranging from minor swelling at the injection site to systemic reactions, which in extremely rare circumstances include death.
- the aim of the present inventors was to develop an immune therapeutic DNA-based nanomedicine for allergy that can be administered transdermally instead of the presently available administration routes involving subcutaneous or intradermal injection.
- the invention provides a pharmaceutical composition as defined above, comprising a nucleic acid encoding at least one allergenic epitope of a protein for use in desensitization or suppressing allergic symptoms, wherein said use comprises the transdermal introduction of said nucleic acid into a subject.
- said allergenic epitope of a protein is at least one allergenic epitope of an airborne allergen.
- said airborne allergen originates from dust mite and advantageously is Derpl.
- said Derpl is mutated, wherein said mutation advantageously eliminates the protease activity of Derpl.
- the above nucleic acid of the invention is preferably comprised in a plasmid and still more preferably has the nucleotide sequence which is depicted in SEQ ID NO: 1.
- said nucleic acid is an ovalbumin encoding plasmid - DNA called pcOVA.
- composition according to the invention further comprises pharmaceutically acceptable excipients and/or additives
- a preferred composition comprises the following i) Derpl:pVAXl or pcOVA plasmid DNA construct, ii) PEIm, and iii) pharmaceutically acceptable excipient(s).
- the invention relates to a nucleic acid encoding at least one allergenic epitope of a protein for use in desensitization or suppressing allergic symptoms, wherein said use comprises the transdermal introduction of said nucleic acid into a subject.
- said allergenic epitope of a protein is at least one allergenic epitope of an airborne allergen.
- said airborne allergen originates from dust mite and advantageously is Derpl.
- said Derpl is mutated, wherein said mutation advantageously eliminates the protease activity of Derpl.
- the above nucleic acid of the invention is preferably comprised in a plasmid and still more preferably has the nucleotide sequence which is depicted in SEQ ID NO: 1.
- said nucleic acid is an ovalbumin encoding plasmid- DNA called pcOVA.
- the invention furthermore concerns a method of treating a patient against an allergic condition or allergic symptoms or preventing the onset of said condition or symptoms, wherein said treatment comprises the transdermal introduction of said nucleic acid according to the invention or said composition according to the invention.
- said allergic condition is caused by an airborne allergen and/or said allergic condition is allergic rhinitis and/or said airborne allergen originates from dust mite.
- said allergic condition is food allergy, preferably egg white allergy.
- plasmid DNA-based transdermal nanoparticle vaccination with Chlamydia-speci&c antigens i.e. the ChlamyDerm nanomedicine product candidate
- ChlamyDerm nanomedicine product candidate i.e. the ChlamyDerm nanomedicine product candidate
- the specific immunizing agent we have selected is a plasmid DNA enabling the expression of a fusion protein composed of two Chlamydial membrane proteins, namely CopN and MOMP.
- the present invention is not limited to these specific antigenic proteins, other Chlamydia antigens may also be included in the nanoparticle based composition according to the invention.
- transdermal vaccination might be effective against Chlamydial infections and/or any associated condition (e.g. atherosclerosis).
- DNA expression for immunization is an approach for generating immunity against various infectious and noninfectious diseases.
- DNA immunization provides protective immunity through expression of foreign proteins by host cells, thus allowing the presentation of antigens to the immune system in a manner more analogous to that which occurs during infection with viruses or intracellular pathogens.
- the present invention provides a pharmaceutical composition as defined above, comprising a nucleic acid encoding at least one antigenic epitope of a protein originating from a Chlamydia species, for use in the prevention or treatment of a disease associated with Chlamydia infection, wherein said use comprises the transdermal introduction of said nucleic acid into a subject.
- the nucleic acid encodes at least one antigenic epitope of the CopN or MOMP protein.
- the nucleic acid comprises a humanized coding sequence and encodes a fusion protein created from the CopN and MOMP proteins.
- the fusion protein has the humanized nucleotide sequence depicted as SEQ ID NO: 4.
- the nucleic acid is comprised in a plasmid DNA, enabling the expression of said at least one antigen in cells of said subject.
- composition according to the invention further comprises pharmaceutically acceptable excipients and/or additives
- the invention provides a nucleic acid encoding at least one antigenic epitope of a protein originating from a Chlamydia species, for use in the prevention or treatment of a condition associated with Chlamydia infection, wherein said use comprises the transdermal introduction of said nucleic acid into a subject.
- the nucleic acid encodes at least one antigenic epitope of the CopN or MOMP protein.
- the nucleic acid comprises a humanized coding sequence and encodes a fusion protein created from the CopN and MOMP proteins.
- the fusion protein has the humanized nucleotide sequence depicted as SEQ ID NO: 4.
- the nucleic acid is comprised in a plasmid DNA, enabling the expression of said at least one antigen in cells of said subject.
- the invention provides the nucleic acid or pharmaceutical composition as defined above, for use in the prevention or treatment of Chlamydia pneumoniae infection.
- the invention provides the nucleic acid or pharmaceutical composition as defined above, for use in the prevention or treatment of pneumonia caused by Chlamydia infection.
- the invention provides the nucleic acid or pharmaceutical composition as defined above, for use in the prevention or treatment of Chlamydia pneumoniae infection-induced atherosclerosis.
- the invention also provides a method of treating a patient against a disease associated with Chlamydia infection or preventing the onset of the symptoms of said disease, wherein said treatment comprises the transdermal introduction of the nucleic acid or the composition according to the invention to said patient.
- the disease is associated with Chlamydia pneumoniae infection.
- the disease is pneumonia caused by Chlamydia infection.
- the invention provides the disease is Chlamydia pneumoniae infection- induced atherosclerosis.
- the present invention provides a pharmaceutical composition, comprising nanoparticles ("NanoComp”) formed by an antigen (peptide, polypeptide, protein, DNA, RNA) and a linear PEI (1-PEI) or 1-PEI derivative in a pharmaceutically acceptable liquid formulation, which is stable at 0-40 0 C for at least one month.
- NanoComp nanoparticles
- an antigen peptide, polypeptide, protein, DNA, RNA
- 1-PEI linear PEI
- 1-PEI derivative 1-PEI derivative
- the antigen, polyanionic in a selected solvent, and the partially protonated, polycationic 1-PEI form by electrostatic interaction nanoparticles with effective diameter (D eff ) between 50 and 500 nm, preferably between about 100 and about 300 nm.
- D eff effective diameter
- the formation of stable nanoparticles require a slight excess of the 1-PEI (calculated as N concentration) added to the antigen, above a minimum molar ratio of about 2 of polycation added to antigen.
- the particle size of the nanoparticles is generally not affected by the excess of the polycation.
- the size of the formed nanoparticles, within the given range, is generally independent from the size of the antigen or 1-PEI derivative used (Fig. 3A - C).
- the 1-PEI derivative formulated in nanoparticles can be a 1-PEI or 1-PEI derivative with molecular weight less than 50 kDa, preferably between IkDa and 35 kDa, more preferably between 1OkDa and 25 kDa (Fig. 3C). Therefore, the statements and examples presented for the plasmid DNA (pDNA) and the linear, 3% mannobiose grafted PEI (which may be used interchangeably within the description with PEIm or other 1-PEI-derivatives, in particular in the context of nanoparticles formation) during the description are applicable for any antigen and 1-PEI or 1-PEI derivative.
- nanoparticles are proven by the fact that after complexation of the macromolecule (antigen) by the polymer, the size of the free antigen is considerably reduced resulting in a discrete particle size (Fig. 3B).
- Fig. 3B For the polymer, having a linear topology, can not be measured any value for effective diameter by dynamic light scattering method (DLS), nor for the plasmids.
- the 1-PEI or 1-PEI derivative used in the present invention can preferably be prepared as follows.
- Pyridine borane (BH 3 -pyridine complex) is a known agent and it has been found to be effective catalyst in great number of the reductive amination processes where the applied compounds are different from carbohydrates.
- Pyridine borane is commercially available, e.g. in pyridine solution in a concentration of ⁇ 8M.
- the present invention is disclosed in details through examples where D-mannose [Example 1, see formula
- the amino -containing compound should have at least one primary or secondary amino group. In some cases it is advantageous if the amino-containing compound is in partially protonated acid addition salt, e.g. in HCl salt form. It is preferred if the amino-containing compound is a polymer having primary or secondary amino groups. The amino group can be a part of the group which is formed during the coupling of the monomers of the polymer (e.g. the part of a peptide, urethane etc. bond). In a more preferred embodiment, the polymer is PEL PEIs are well known compounds in the field of polymer chemistry. In the most preferred embodiment of the invention 1-PEI is applied, preferably as partially protonated HCl salt.
- the antigen used in the nanoparticles according to the invention may be either a polynucleotide or a polypeptide.
- the term "polynucleotide” includes all kinds of nucleic acids irrespectively of origin, type, composition, substitutions, etc.
- the nucleic acid may be both DNA and RNA, it may be synthetic or isolated from living organisms, constructed to include genes or markers of interest, may comprise irregular nucleotides, etc.
- the antigen formulated in nanoparticles can be a nucleic acid (DNA or RNA) with 3-20 kbp length, more preferably between 6 and 13 kbp (Fig. 3A).
- polypeptide means molecules composed of any number of amino acids. Accordingly, the term includes shorter peptides of about 6-15 amino acids, up to long proteins of even 400 kDa in molecular weight.
- the polypeptide of the invention may comprise any kind of amino acids, may be synthetic or isolated from nature, and may have specific biological activity or serve as an immunogen.
- the antigen formulated in nanoparticles can be a polypeptide with a size of about 1 to about 400 kDa, preferably between 20-100 kDa.
- the NanoComps according to the present invention are antigen-containing mannosylated nanoparticles with a size in the range of about a few hundred nanometers, in aqueous solution resembling the size and appearance of viruses.
- the nucleic acids and/or other antigens are condensed in the core (Fig. IA-B) and on the positively charged polymer surface they may wear mannobiose residues to enhance their receptor-mediated uptake (Fig. 2).
- the NanoComp of the present invention may also wear alternative sugar residues on the surface and may also incorporate adjuvants (e.g. cytokines) to improve or redirect immune responses. These nanoparticles maintain their stability during storage before cellular uptake and escape from the endosomal compartment inside the cell.
- the NanoComp of the present invention provides a solution to the formulation, manufacturing and regulatory challenges of biologic and nanomedicine pharmaceuticals.
- the present invention not only gives evidence that the structure (degree of association, compactness) of the nanoparticles is also a key parameter for the efficient gene delivery, but found out the tool for measuring it (see Example 5).
- the compactness (degree of association) of the nanoparticles is given by the type and number of interactions between the polyanionic pDNA and polycationic PEI(m), which can be measured spectrophotometrically as the absorbance enhancement at 260 nm of the nanoparticles compared to the absorbance of pDNA at same concentrations.
- the cationic character of the polymer has an optimum value regarding the efficiency of the NanoComp which means that other efficiency parameter(s) require the opposite degree of the cationic character.
- the ionic strength of the formulation according to the invention is between about 1 mM and about 50 mM, preferably between 2 and 5 mM. This also applies to the ionic strength of the antigen (macromolecule) solution.
- the term “about”, as used with reference to numerical values means a deviation of ⁇ 15% from said value.
- the term “about”, as used with reference to pH, refers to ⁇ 0.2 pH value.
- the very low number of protonated (cationic) nitrogen atoms which is preferable for endosomal escape, can make less number of ionic interaction with the more or less number of anionic phosphate of pDNA (depends on the ionic strength of the pDNA solution), and the complex formed has a loose structure, which is not stable enough in the cytosol or in the liquid formulation and cannot protect the pDNA.
- These processes require PEIm with an optimum degree of cationic character and pDNA solution with not too high ionic strength (Fig. 4 and Fig. 5).
- the cationic character of the polymer may be preferably influenced by the selection of said polymer,
- the commercial PEI preparation Polyplus-transfection, France
- the commercial PEI preparation has a molecular weight of 25 kDa, accompanied with a ratio of the protonable N / Total N of about 0.1.
- the results presented herein ascertain that this ratio is not optimal for the optimal biological activity of NanoComps, the preferred range is being from about 0.1 to about 0.8, preferably about 0.4.
- Example 6 further decreasing the cationic character of PEI resulted in better characteristics for the nanoparticles (Fig. 5).
- the molar ratio of PEIm to the macromolecules — such as pDNA, which gives the abbreviated name for this ratio, i.e. N/P ratio for nitrogen/phosphate ratio) or alternatively a protein or peptide — used for the construction of the nanoparticles should have the same effect on the biological activity of the NanoComp, making loose (N/P ⁇ 3) or compact structures.
- the effect of the N/P ratio on the biological activity of NanoComp prepared and tested freshly or after one-week storage in pH buffered solution is shown on Fig. 6.
- a very good example of the importance of the relatively compact structure of the nanoparticles is shown in the case of the nanoparticles formed at N/P ratio of 2.
- the person skilled in the art will be able to determine which activity of the macromolecule is considered to be relevant for the purposes of the specific application. In preferred embodiments, this relevant biological activity is preserved or at least is not significantly decreased with the use of the NanoComp of the invention for elongated periods of time. It is also possible that the macromolecule has several quantifiable biological activities, and any of those may be maintained by the NanoComp. As long as at least one of these activities is the activity to be maintained, the NanoComp of the invention will fulfill its purpose as specified herein, but this is not limiting with respect to the preservation of any of the other activities of the macromolecule.
- this relevant biological activity can be its oc/ccc ratio, which characterizes its physico-chemical properties, and by maintaining this ratio at the original value it helps to keep the compact structure of the NanoComp substantially unchanged and thus protects the macromolecule (antigen) from degradation.
- the pH of the solution has effect on the stability of the nanoparticles.
- the low pH of the solution can induce the aggregation and subsequent precipitation of the nanoparticles and also the degradation of the pDNA (the pharmaceutically active ingredient).
- the stability of the pDNA inside the NanoComp proved to be dependent on the pH of the formulation solvent, at higher, pH > 6.5 the active pharmaceutical ingredient (API) did not show degradation (Fig. 8A).
- the type of the solvent used should further stabilize the nanoparticles, the sugar-like compounds have improving effect on the biological activity of the NanoComp compared to sterile water (Fig. 7A).
- sugar-like compounds such as sugar-alcohols, like mannitol, proved to have the same improving effect on the biological activity, but without side reactions with PEIm (Fig. 7A-B). Since the formulations with the aqueous solution of the sugars have pH only between 5 and 6, for stability concerns the sugar-alcohol solution is made in pharmaceutically acceptable pH buffer, which is weaker base than the secondary amine containing linear PEIm (to outpace the side reaction with the acidic pDNA).
- bases containing tertiary amines are pharmaceutically approved, non- reactive substances for buffering nucleic acid based compositions, in contrast to primary amine containing Tris [tris(hydroxymethyl)aminomethane] buffer, which during time increases the particle size of the NanoComp (Fig. 7C).
- the pH of the buffers should be between 6.5 and 8.1. The lower pH was shown to have degrading effect on the pDNA (Fig. 8A), at the higher pH the PEIm precipitates.
- the biological activity of the NanoComp mixed using pDNA solution with high ionic strength (3OmM, pDNA-A) and PEIm of high degree of cationic character (90% of the amine N were protonated, PEIm-A) stored at 37 0 C was much lower compared to the control than the potency of the NanoComp mixed with PEIm with low degree of cationic character (60% of the amine N were protonated, PEIm-B), at the same temperature and compared to its own control.
- the potency of the NanoComp mixed with pDNA-A and PEIm-B stored at 4 0 C and 37 0 C was the same, their potency during 3 week storage dropped significantly compared to the control (Fig. 10A).
- the major event occurred during storage was the aggregation and precipitation of the nanoparticles, which explain the reduced biological activity.
- NanoComp liquid formulation proved to be pDNA in low ionic strength solution, PEIm with low degree of cationic character, using at N/P ratio of 3 and formulated in triethanolamine buffered (pH 7.5) mannitol solution, which was stable at 4 0 C for 8 weeks without aggregation or decomplexation and without loss of biological activity (Fig. 10C).
- composition complemented with glycerol allowed the storage of the NanoComp formulation in frozen or lyophilized state for 4 weeks. After reconstitution the biological activity of the stored NanoComp formulations doesn't dropped significantly compared to the control formulation (Fig. 12A-C).
- the composition according to the present invention proved to be more efficient than the compositions commonly used (e.g. in water or glucose solution).
- the compositions commonly used e.g. in water or glucose solution.
- the NanoComps according to the present invention are pathogen-like nanoparticles generated from antigens (e.g. DNA, proteins, peptides) to mimic a pathogen exposure.
- antigens e.g. DNA, proteins, peptides
- the most common routes by which antigens are introduced into the body are subcutaneous, intradermal, transdermal, intramuscular, intravenous or intraperitoneal administration.
- the pharmaceutical composition according to the present invention may be administered vaginally or rectally for the prevention of infectious diseases as microbicide.
- Oral administration provides antigens into the gastrointestinal tract, while intranasal administration or inhalation brings antigens into the respiratory tract.
- the preferred mode of administration according to the present invention is the use of the transdermal route.
- pDNA/PEIm NanoComp formulation is not influenced by the sequence of the pDNA-encoded antigens or the size of the pDNA. Most of the above experiments, however, were performed with HIV pDNA.
- immunogenic nanomedicines comprises different pDNA and demonstrate their use to induce immune responses that is effective for the prevention and treatment of intracellular bacteria and allergy, more specifically Chlamydia infection and dust mite allergy.
- Vaccination using the DermAU and ChlamyDerm nanomedicines were performed in accordance with the nanoparticle based transdermal vaccination methodology as described in patent applications US 2002/0022033 Al, US 2002/0022034 Al and HU patent application No. P0900117, in conjunction to using the nanoparticle composition according to the present invention.
- An advantageous composition of the applied formulations comprised the herein described pDNA constructs and a polymer, advantageously poly(ethyleneimin) (PEI) or a derivative thereof, advantageously poly(ethyleneimin)-mannose (PEIm).
- Said formulations advantageously also comprised further pharmaceutically acceptable excipients and/or carriers e. g. dextrose and advantageously comprised a sugar alcohol.
- the preventive DermAU immunotherapy significantly decreased the nasal symptoms of allergic rhinitis in experimental animal model. Based on the results presented herein DermAU vaccination is expected to balance the ongoing immune response (Th2-type hypersensitivity), inhibit the allergic reaction and increase the tolerance to specific allergens in humans.
- the inventors have developed a new DNA construct, encoding the major allergenic epitope of house dust mite Dermatophagoides pteronys sinus, called Der p 1.
- Der p 1 is a cystein protease with proteolytic activity, therefore we modified the cDNA sequence to destroy its protease activity.
- the Der p 1 encoding modified cDNA sequence was cloned to a pVAXl (Invitrogen) expression vector.
- This newly developed Derpl: pVAXl construct was included in the above described nanoparticle composition and topically administered based on the DermaVir- platform technology, to naive BALB/c mice.
- This new topical administration and composition of DNA-based immunotherapy is called DermAU immunotherapy.
- the preventive Derpl -DermAU transdermal therapy significantly decreased the nasal symptoms of allergic rhinitis in this experimental animal model. It is anticipated that DermAU immunotherapy will be effective in preventive and therapeutic use for allergic patients
- House dust mites are major indoor triggers for atopic diseases: allergic rhinitis, asthma, conjunctivitis and dermatitis.
- atopic diseases allergic rhinitis, asthma, conjunctivitis and dermatitis.
- Dust mites are tiny 0.3 mm insects.
- the most common house dust mite species are Dermatophagoides pteronyssinus (Dp) and Dermatophagoides farinae (Df).
- the major allergens of Dermatophagoides pteronyssinus are the group 1 (Der p 1) and group 2 (Der p 2) allergens. It was reported in 1987 by Platt-Mills that the major allergen Der p 1 of house dust mite Dermatophagoides pteronyssinus is a major cause of allergic disorders such as bronchial asthma, atopic dermatitis and allergic rhinitis. From the serological analysis about 50-70% of the IgE and IgG antibodies bind to the group 1 and 2 allergens and 30% bind to the group 4, 5 and 7 allergens (HuangFu et al., 2006; Wayne et al., 2007).
- Der p 1 is a cystein protease. Mice injected with enzymatically active Der p 1 produce more IgE antibody than mice injected with inactivated Der p 1 providing evidence for a Th2 promoting effect of the protease activity (Shakib et al., 1998; Schulz et al., 1998). The ability of the proteolytic activity to cleave and release immunological receptors, activate dendritic cells and permeabilise tight junctions suggests several plausible mechanisms.
- Der p 1 cDNA encodes a precursor of 320 amino acid residues, comprising of a signal peptid (18 amino acids), a proenzyme region (80 amino acids), and a mature enzyme (222 amino acids) (Chua et al., 1988; Chua et al, 1993). Since Der p 1 is a cystein protease, DNA encoding enzymatically inactive immunogenic forms of the allergen may represent a feasible and safer Der p 1 vaccine.
- the proteins of various organisms are built of 20 genetically encoded amino acids.
- a possible way to overcome this problem is to analyze the sequence to be expressed in a heterologous system and redesign it in a way that exchanges all the codons that are suboptimal for the mammalian expression.
- OVA Ovalbumin
- OVA is a widely used allergen in experimental allergy models to induce allergic symptoms in mice and test the possible effect of various treatments.
- a person skilled in the art would understand that a composition or a method that alleviates or suppresses the symptoms caused by this allergen would achieve the same result with all of the airborne allergens. Therefore we have developed an OVA-specific allergic mouse model and tested OVA- encoding DNA-construct in DermAU immunotherapy system.
- the route of environmental exposure has a direct impact in determining the immunological response induced by allergens and intracellular bacteria.
- Repeated barrier-disruption stimulates signaling cascades that lead to inflammation and epidermal hyperplasia. Skin barrier dysfunction may also allow entry of allergens and pathogens, which can lead to primary systemic sensitization.
- T H 1 cells and regulatory T cells are involved in the mechanism of action of specific immunotherapy (Passalacqua et al., 2007).
- CD8+ T cells play a significant role in the allergen specific tolerance induction.
- Previous data of DermaVir immunization showed an increase in CD8+ and CD4+ T cell-mediated immune responses (Lisziewicz et al., 2005).
- the expression of both transdermal nanomedicines the Derpl- and the OVA allergens reduced the allergic symptoms in our experimental animal models. Therefore it is evident for a person skilled in the pertinent art that the used vaccination approach is not limited to DNA sequences encoding the herein applied allergenic epitopes, but other DNA sequences encoding other allergenic epitopes of an allergen may also be used according to the invention.
- An ordinary person skilled in the art based on the teaching given in this application can substitute the described plasmid DNA sequences with other plasmid DNA sequences encoding allergenic epitopes of another protein and can also make the desired nanoparticle for transdermal vaccination into a subject.
- the present invention provides a biological nanomedicine composition and a vaccination method for the treatment and prevention of C. pneumoniae infection.
- the inventors have developed a plasmid DNA construct, encoding two bacterial proteins, CopN and the major outer membrane protein (MOMP). Both proteins are expressed early and throughout the life cycle of the intracellularly persisting bacteria.
- the plasmid DNA of the invention was formulated according to the present invention to obtain the ChlamyDerm nanomedicine vaccine product.
- CopN is a member of a family of proteins common to obligate intracellular pathogenic organisms. It is a putative substrate of the C. pneumoniae type III system, a specialized secretion system that directly translocates proteins from the bacterial cytosol into host cells. During infection, CopN is detected on the inclusion membrane. CopN has been implicated to regulate type III secretion by controlling access to the secretory channel and modification of the host microtubule network. Following the contact of the bacteria with a eukaryotic cell, CopN is translocated along with other effectors into the cytoplasm of the host cell. The inhibitors of CopN inhibit infection with C. pneumoniae.
- the Chlamydial outer membrane complex is composed primarily of three proteins; MOMP and two cysteine - rich proteins, the outer membrane complex B protein (OmcB) and the outer membrane complex A protein (OmcA). It has been shown that MOMP is structurally and immunologically the dominant protein in the Chlamydial outer membrane complex. In artificial laboratory membranes MOMP functioned as a porin (or pore) protein, though it was not entirely typical of known outer envelope proteins. Exposure to thiol reducing agents lead to a reduction in -S-S- intramolecular bonding of MOMP, opening up its pore structure for nutrient ingress and perhaps partly triggering the early differentiation of elementary bodies (Ebs) after entry into the cell.
- Ebs elementary bodies
- the present invention concerns a nucleic acid encoding at least one antigenic epitope of a protein originating from a Chlamydia species, for use in the prevention or treatment of a disease associated with Chlamydia infection, wherein said nucleic acid is capable of enabling the expression of said at least one antigenic epitope in cells of a subject to be treated and wherein said use comprises the transdermal introduction of said nucleic acid into said subject and.
- CopN or MOMP protein of a Chlamydia species The immune therapeutic materials and uses according to the invention are advantageously applied in the prevention or treatment of conditions manifested in humans.
- nucleic acid constructs according to the invention for use in human therapy advantageously comprise humanized coding sequences encoding the suitable immunogenic epitopes of the pathogen to be fought against.
- the specific transdermal vaccination method employed in accordance with the present invention includes the expression of the pathogenic epitopes by specific human immune cells, wherein the humanized codone usage supposedly adds to the effectiveness of said expression.
- a nucleic acid comprising a humanized sequence encoding a fusion protein created from the
- CopN and MOMP protein of Chlamydia pneumonia is used, having advantageously the humanized nucleotide sequence depicted as SEQ ID NO: 4.
- the nucleic acid according to the invention in accordance with an advantageous embodiment, is a plasmid, enabling the expression of the above-referred at least one antigenic epitope in specific immune cells of the subject to be treated, advantageously in the Langerhans cells of said subject.
- This is advantageous because the employed nanoparticle based transdermal vaccination technology is thought to be effective via the uptake of the antigen encoding nanoparticles by the subject's Langerhans cells where the immunogenic epitopes should be expressed and consequently presented towards other cells of the immune system.
- the nucleic acid constructs of the invention are advantageously used in the form of pharmaceutical compositions according to the present invention as discussed above.
- the nucleic acid constructs or pharmaceutical compositions according to the invention are advantageously useful for the prevention or treatment of conditions associated with Chlamydia infection, advantageously with C. pneumoniae infection.
- the nucleic acid constructs or pharmaceutical compositions according to the invention are advantageously useful use in the prevention or treatment of pneumonia caused by Chlamydia infection and in the prevention or treatment of C pneumoniae infection-induced atherosclerosis.
- the invention further concerns methods for treating a patient against a disease associated with Chlamydia infection or preventing the onset of the symptoms of said disease in said patient, wherein said treatment comprises the transdermal introduction of a nucleic acid construct or a composition according to the invention.
- the treatment or prevention methods of the invention are advantageously useful against diseases associated with
- Chlamydia infection advantageously with C pneumoniae infection and, advantageously, against pneumonia caused by Chlamydia infection and against C pneumoniae infection-induced atherosclerosis.
- Fig. 1 Illustration of the nanoparticle formulation of antigens: A: NanoComp according to the invention. B: Molecular model of the NanoComp formulation calculated using Gaussian modeling method on 8 bp double stranded DNA at N/P ratio of 4. Fig. 2. Illustration of the pathogen-like mechanism of entry, antigen expression and antigen presentation with the NanoComp according to the invention.
- FIG. 3 Illustration of the particle size distribution of the NanoComp formulations prepared using different macromolecules and polymers: A: Formulations of 4, 6.5 and 12.5 kbp length pDNA-s with linear PEIm. B:
- Fig. 4 Illustration for the effect of the ionic strength of the pDNA solution on the compactness of the nanoparticles (measured as hyperchromicity) and on their biological activity.
- Fig. 5 Illustration for the effect of the cationic character (protonable N/Total N) of the PEIm on the biological activity of the nanoparticles.
- Fig. 6 Illustration for the effect of the N/P ratio on the biological activity and stability of the NanoComp stored in liquid formulation for 1 week at room temperature (approx. 25 0 C).
- Fig. 7 Illustration for the effect of the formulation solvent.
- A Biological activity of the NanoComp formulations prepared with water, physiological saline, sugars, sugar alcohols, buffer (pH 7.5) and sugar alcohol in buffer (pH 7.5).
- B Stability of the NanoComp formulations prepared with D-(+)-Glucose and D-(+)-Mannitol, with and without side reactions.
- C Particle size of the NanoComp formulations prepared with triethanolamine buffer (pH 7.5) and Tris buffer (pH 7.5) and stored for 6 days at room temperature (approx. 25 0 C).
- Fig. 8 Illustration for the effect of the formulation pH on the pDNA stability (oc/ccc ratio) inside the NanoComp.
- Fig. 9 Illustration on the protecting effect of the NanoComp formulation on the pDNA homogeneity, when stored at room temperature (approx. 25 0 C) for 18 days.
- Fig. 10 Illustration on the stability of 4 NanoComp formulations prepared using 2 lots of pDNA and 2 lots of PEIm.
- PEIm-A high degree of cationic character
- PEIm-B low degree of cationic character
- B Stability of the NanoComp formulations prepared from pDNA-B (in low ionic strength solution) with PEIm-A (high degree of cationic character) and PEIm-B (low degree of cationic character) stored for 3 weeks at 4 0 C and 37 0 C.
- C Stability of the NanoComp final formulation prepared from pDNA-B (in low ionic strength solution) with PEIm-B (low degree of cationic character) stored for 8 weeks at 4 0 C.
- Fig. 11 Illustration on the manufacturing of NanoComp liquid formulation from frozen or lyophilized materials, stability and biological activity compared to the freshly prepared NanoComp liquid formulation.
- Fig. 13. The modified Der p 1 cDNA sequence comprised in the DermAll vaccine. The conserved cysteine amino acid was mutated to alanine at the core catalytic center of the protein (A). Codon optimization (B), EcoRJ and Xhol restriction enzyme recognition sequences were added at the 5' and 3' end of the new construct, respectively.
- the generated final construct (Derpl :pV AXl) is capable of expressing the modified Der p 1 protein in mammalian HEK293 cells (C).
- Fig. 15 Gene expression study analyzing the expression changes of the immune -response related gene IFN ⁇ (interferon-gamma) after preventive Derpl -DermAll immunotherapy.
- OVA-DermAU preventive immunotherapy is associated with increased level of IL-IO and does not induce immunoglobulins (Ig) that inversly effects disease progression and part of the pathomechanism of allergic diseases.
- Ig immunoglobulins
- OVA-DermAU therapeutic immunization is associated with increased level of IFN ⁇ which is the hallmark cytokine of activated ThI lymphocytes.
- Fig. 20 pVAXl vector (Invitrogen, USA)
- Fig. 21 Active pharmaceutical ingredient comprised in ChlamyDerm: CMV-(CopN-MOMP) pDNA for the expression of CopN-MOMP fusion protein antigen in cells.
- Fig. 22 The quality control of plasmid.
- Fig. 25 The effect of ChlamyDerm on serum CRP (A) and on serum amyloid A (B) in normal and C. pneumoniae infected mice.
- Fig. 26 C. pneumoniae infection induced inflammation in histological samples obtained from ChlamyDerm-vaccinated animals, (mean ⁇ SEM, * p ⁇ 0,05 vs control nanomedicine non-infected animals, Kruskall-Wallis test).
- Fig. 27 Effect of ChlamyDerm on bacterial protein content (MOMP) of lung tissue in C57B1/6 and
- the PEIm/pDNA complexes were prepared at different ratios of nitrogen/phosphate (N/P). The actual N/P ratio and the type of the solvent are labeled separately at every formulation. Table 1.
- the in vitro biological activity test of the NanoComp is performed by transfection of 293T human cells. 50,000 cells are plated in 96 well plate 19 hours before transfection, using DMEM culture medium (DMEM, 1% FBS, 1% L-glutamine, 1% Penicillin- Streptomycin). 0.5 ⁇ g pDNA/NanoComp is added to each well followed by 21 hours incubation at 37°C in 5% CO 2 air condition. During the incubation period, the HIV-specific protein antigens encoded by the pDNA are expressed and secreted into the culture medium. After 21 hours, the supernatant of the cells are collected, and HIV-I p24 antigen ELISA is performed to quantify p24 protein produced by the pDNA.
- Example 4 NanoComp formation using different antigens and polymer a)
- Four NanoComp formulations were prepared according to Example 3 using PEIm for the formulation of different length of plasmid DNA (pLWXul 12.5 kbp, pGL2 plasmid 6.5 kbp, pJET 1.2 plasmid containing 700 bp insert) at N/P ratio of 3 and with 10% D-Glucose solvent.
- the complexation of pLWXul plasmid was performed also with linear PEI (22 kDa) at N/P ratio of 3 in 10% D-Glucose solution.
- the particle size measurements were performed by dynamic light scattering method (DLS) using Brookhaven ZetaPals Instrument. Results are shown in Fig. 3A.
- NanoComp formulations were prepared according to Example 3 using PEIm for the formulation of different size of polypeptides (ACP6574 standard, IkDa peptide; Green fluorescens protein (GFP), 27kDa protein, Millipore; and Albumin protein, 66 kDa, Sigma) at PEInxpolypeptide molar ratio of 3, 4, 6 or 8 and with triethanolamine -buffered (pH 7.5) mannitol solution.
- the size of the polypeptides without complexation is also measured.
- the particle size measurements were performed by DLS with Brookhaven ZetaPals Instrument. Results are shown in Fig. 3B.
- NanoComp formulations were prepared according to Example 3 using different molecular weight of PEIm (linear polyethyleneimine containing 3% grafted mannobiose of Mn 3700, 5600, 7200, 14200, 22000 and 29300 g/mol determined by GPC, Genetic Immunity) for the formulation of plasmid DNA (pLWXul 12.5 kbp), at N/P ratio of 3 and with 10% Mannitol solvent. The particle size measurements were performed by DLS using Brookhaven ZetaPals Instrument. Results are shown in Fig. 3C.
- Example 5 Effect of the ionic strength of the pDNA solution
- NanoComp formulations were prepared using three lots of pLWXul plasmid DNA.
- the formulations were prepared according to Example 3, using the same PEIm at N/P ratio of 4.22 and 10% D-Glucose as solvent.
- the supercoiled content (ccc form) of the plasmids determined by agarose gel electrophoresis was the same: 78 ⁇ 10%, but the ionic strength of the pDNAl, pDNA2 and pDNA3 plasmid solutions (determined by ICP-MS) were 1.5, 30 and 44mM, respectively.
- the absorbance measurements at 260 nm of the plasmid DNA solutions (1 mg/ml) and the prepared NanoComp formulations were performed in the same dilution using Jasco V-630 spectrophotometer. The calculated compactness was given by the percent increase (hyperchromicity) of the
- NanoComp formulations were made according to Example 3 using the same pDNA and PEIm containing different amount of protonable N, at N/P ratio of 3 and using Mannitol (triethanolamine buffered, pH 7.5) as solvent.
- Example 8 Effect of the formulation solvent a) NanoComp formulations were prepared according to Example 3, using the same PEIm and pDNA at N/P ratio of 3, and different solvents: sterile water, physiological NaCl (0.9%), D(+)-Mannitol (10 mg/ml), Sorbitol (10 mg/ml), D(+)-Glucose (10 mg/ml), Mannose (10 mg/ml), Sucrose (10 mg/ml), Triethanolamine buffer (TEA) (pH 7.5), D(+)-Mannitol (10 mg/ml) in triethanolamine (TEA) buffer (pH 7.5), Sorbitol (10 mg/ml) in triethanolamine buffer (pH 7.5) and D(+)-Mannitol (10 mg/ml) and Glycerol (10 mg/ml) in triethanolamine buffer (pH 7.5).
- sterile water physiological NaCl (0.9%)
- D(+)-Mannitol 10 mg/ml
- the biological activity is givenmual related to the biological activity of the formulation in sterile water. Results are shown in Fig. 7A.
- the prepared solutions were stored at room temperature (approx. 25 0 C) for 2 month. UV spectra were recorded without dilution. Results are shown in Fig. 7B.
- Triethanolamine buffer 10 mM, pH 7.5 and Tris buffer 10 mM, pH 7.5.
- the particle size of the formed complexes was measured using ZetaPALS, Brookehaven instrument freshly and after 6 days storage at room temperature. Results are shown in Fig. 1C.
- Example 9 Effect of the formulation pH on pDNA stability
- NanoComp formulations were prepared according to Example 3, using the same PEIm and pDNA at N/P ratio of 3, and different solvents using D-Mannitol (10 mg/ml in buffer) prepared in the same buffer with different pH.
- the effect of the formulation pH was measured on the degradation of the pDNA inside the NanoComp during storage at room temperature for 18 days.
- the oc/ccc ratio of the pDNA inside the nanoparticles was measured by agarose gel electrophoresis (AGE) after decomplexation with 2% SDS solution (0.8% agarose gel, 100 V, 120 min running). Results are shown in Fig. 8.
- Example 10 Stability of the pDNA inside the NanoComp formulation
- NanoComp formulations were prepared according to Example 3, using the same PEIm and pDNA at N/P ratio of 3, using D(+)-Mannitol (10 mg/ml) in Triethanolamine buffer (pH 7.5) as solvent.
- the NanoComp formulations were stored for 1 week at 37 0 C.
- plasmid DNA (1 mg/ml aqueous solution) was also stored at the same temperature and time period.
- NanoComp formulations were prepared according to Example 3, using two lots of pDNA and two lots of PEIm at N/P ratio of 3 and Mannitol (in Triethanolamine buffer, pH 7.5) as solvent.
- the specific parameters of the components and the abbreviation of the formulation prepared are listed in
- NanoComp formulation was prepared from frozen starting materials.
- the pDNA solution mixed with 3 volume of triethanolamine buffered mannitol solution, and PEIm solution mixed with 3 volume of triethanolamine buffered mannitol solution were separately stored at -20 0 C for 2 weeks.
- the two starting materials were allowed to defrost then the two solutions were mixed at N/P ratio of 3.
- the biological activity of the formulation was measured and compared to the freshly prepared liquid formulation according Example 3 (Fig. 11).
- Example 13 Preparation and stability of the NanoComp formulation prepared from lyophilized materials
- NanoComp formulations were prepared from lyophilized pDNA.
- the pDNA solution was mixed with 6 volume of 10% mannitol solution (pDNA-10Man) or 10% mannitol+30% glycerol (pDNA-10Man30Glyc) or 10% Mannitol+50% Glycerol (pDNA-10Man50Glyc) in Triethanolamine buffer pH 7.5 and was lyophilized and stored for 30 days at 4 0 C.
- the lyophilized pDNA powder was reconstituted with triethanolamine -buffered PEIm solution (pDNA-10Man) or PEIm aqueous solutions (pDNA-10Man30Glyc and pDNA-10Man50Glyc) respectively at N/P ratio of 3.
- the biological activity of the formulations was measured and compared to the freshly prepared liquid formulations according Example 3 (Fig. 11).
- NanoComp formulations were frozen at -20 0 C or lyophilized.
- the NanoComp formulations were prepared according to Example 3, using pDNA and PEIm at N/P ratio of 4 and 10% Mannitol (FrozenA and LyophA, respectively), 10% Mannitol + 30% Glycerol (FrozenB and LyophB, respectively) or 50% Glycerol (FrozenC and LyophC, respectively) in Triethanolamine buffer, pH 7.5 as solvent.
- the lyophilized formulations were stored for 30 days at -20 0 C and 4°C.
- the lyophilized powders were reconstituted with water, the frozen formulations were defrosted. The degree of reconstitution of the frozen and lyophilized formulations was measured at 260 nm (Fig.12A) along with the particle sizes (Fig. 12B).
- Example 15 Biological activity and stability of the NanoComp frozen and refrigerated formulation
- the biological activity of the Frozen C NanoComp formulation prepared in Example 14 was measured. This is a representative measurement for all frozen and lyophilized formulations prepared in Example 14 which could be 100 % reconstituted and their particle size were maintained. The biological activity of the formulation was measured and compared to the freshly prepared liquid formulation (Fig. 12B).
- Example 16 The modified Der p 1 cDNA sequence
- ELISA analysis was performed in order to demonstrate that the final Derpl :pV AXl construct is capable of synthesizing the Der p 1 protein.
- Human embryonic kidney (HEK 293) cells were plated to 6-well plates in 300.000 cells/well density. 48 hours later they were transfected using as a Derpl-DermAU nanomedicne vaccine containing endotoxin- free Derpl:pVAXl plasmid DNA + PEIm + excipient [10% dextrose] / well).
- the cells were incubated for 48 hours then lysed, and the cell lysate was used for ELISA analysis (Indoor Biotechnology, United Kingdom).
- the empty pVAXl vector and also a plasmid capable of expressing an unrelated cDNA green fluorescent protein - eGFP was used.
- Our results clearly show that the Derp LpVAXl plasmid is capable of expressing the Derpl protein.
- mice Male, 8-week-old, lOmice/ group, purchased from Charles River Laboratories, Germany
- Derpl-DermAU vaccine 6 times, every second week ( Figure 14. a).
- the transdermal immunizations were performed based on the DermaVir- platform technology.
- Derpl-DermAll vaccine was 25 ⁇ g Derpl:pVAXl construct+PEIm+excipient [10% dextrose].
- the control vaccine was composed as: 25 ⁇ g pVAXl vector+PEIm+excipient [10% dextrose].
- the mice were sensitized with intraperitoneal natural Der p 1 allergen extract (NA-DP 1-2, Indoor Biotechnology, United Kingdom) 4 times, once a week (single dose: 5 ⁇ g Der p 1/ mouse+ 1 mg Aluminium hydroxide (Sigma-Aldrich, St. Louise, MO) as an adjuvant).
- mice were intranasally (i.n.) challenged with 15 ⁇ g Der p 1 extract/mouse/day (NA-DP 1-2, Indoor Biotechnology, United Kingdom) for 5 consecutive days.
- the nasal symptoms of allergic rhinitis were assessed by counting the sneezing + nasal rubbing for 10 minutes after the challenge. The nasal symptoms were measured at day 3 and day 5 ( Figure 14. a).
- Example 18 Gene expression analysis to define immune response-related genes of transcutaneous Derpl-DermAll immunization Our preliminary experiments showed that the expression of IFN-gamma specifically increases in Der p 1 protein activated splenocytes in the Derpl-DermAU treated group (Fig. 15).
- Derpl-DermAll vaccinated and control mice (detailed description is in the legend of Figure 14. a) were sacrificed, and single cell suspensions of splenocytes were prepared at the end of the experiments. Each sample of individual mouse was handled separately; 2 million cells were used in 1 ml final volume in each experiment. The activated cells were treated with 20 ⁇ g low endotoxin purified Der p 1 protein (LoToxTM nDer p 1 ; Indoor Biotechnology, United Kingdom), and the control cells were cultured in complete medium (RPMI 1640 medium supplemented with 10% fetal bovine serum, L-glutamine, penicillin, streptomycin and 2-Mercapto-ethanol).
- complete medium RPMI 1640 medium supplemented with 10% fetal bovine serum, L-glutamine, penicillin, streptomycin and 2-Mercapto-ethanol.
- RNA isolation was performed using TRIzol® reagent (Invitrogen, 15596-018) as it is suggested by the manufacturer. The concentration of the samples and their purity was determined by spectrophotometry.
- Complementary DNA was generated from 0.5 ⁇ g total RNA using the iScriptTM cDNA Synthesis Kit (Bio-Rad, Hercules, California, 170-8897) in a 20 ⁇ l final volume.
- Real-time RT-PCR experiments were performed to quantify the amount of Ifng mRNA in the various samples.
- the amplification protocol contained one cycle of initial denaturation at 94°C for 5 min followed by varying number of cycles of denaturation at 94°C for 1 min, annealing at 55°C for 1 min, extension at 72°C for 2 min, and one cycle of terminal extension at 72°C for 10 min.
- the gene specific primer set gave a unique product.
- the IFN-gamma target gene expression was normalized between different samples based on the values of the expression of the internal positive control b-Act (beta-actin), and are presented as fold increases over the control vaccinated (p V AXl) untreated values.
- the chart shows the mean ⁇ SEM of tree to five independent samples per each group ( Figure 15.).
- the IFN-gamma mRNA expression in the Derpl-DermAll vaccinated group showed a marked increase after in vitro Der p 1 activation compared to control splenocytes.
- Ovalbumin (egg white, OVA) is the most frequently used allergen to induce allergic rhinitis in experimental animal models.
- the OVA encoding plasmid-DNA called pcOVA was kindly provided by Dr. A. Heit (Spies et al., 2003).
- Na ⁇ ve BALB/c mice female, 11 -week-old, 8 mice/ group, purchased from Charles River Laboratories, Germany
- OVA-DermAll vaccine 3 times, every second week ( Figure 16. a).
- the transdermal immunizations were performed based on the DermaVir- platform technology.
- the composition of OVA-DermAll vaccine was 25 ⁇ g pcOVA+ PEIm+ 10% dextrose as excipient.
- the control vaccine was composed as: PEIm+ 10% dextrose.
- mice Two weeks after the prophylactic DermAU immunotherapy the mice were sensitized with intraperitoneal ovalbumin (grade V, Sigma-Aldrich, St. Louis, MO) injection 3 times, every week (single dose: 25 ⁇ g OVA/ mouse + 2 mg Aluminium hydroxide as an adjuvant). Two weeks later the mice were intranasally (i.n.) challenged with 500 ⁇ g OVA/ day/mouse (grade V, Sigma-Aldrich) for 3 consecutive days. The nasal symptoms were measured on day 3 ( Figure 16. a).
- Serum samples were taken before the third i.p. OVA sensitization (week 8) and one week after the last sensitization (week 9).
- OVA-specific IgE levels were measured from the sera by a sandwich ELISA method
- Example 20 Preventive effect of OVA-DermAll immunotherapy Na ⁇ ve BALB/c mice (female, 8-week-old, 10 mice/ group, purchased from Charles River Laboratories,
- OVA-DermAll vaccine 6 times, every week ( Figure 17. a).
- the transdermal immunizations were performed based on the DermaVir- platform technology.
- the composition of OVA- DermAll vaccine was 25 ⁇ g pcOVA+ PEIm+ 10% dextrose as excipient.
- the control vaccine was composed as: PEIm+ 10% dextrose.
- One week after the last prophylactic DermAU immunotherapy the mice were sensitized with intraperitoneal ovalbumin (grade V, Sigma-Aldrich, St. Louis, MO) injection 2 times, every second week (single dose: 25 ⁇ g OVA/ mouse + 2 mg Aluminium hydroxide as an adjuvant).
- mice Two weeks later the mice were intranasally (i.n.) challenged with 500 ⁇ g OVA/ day/mouse (grade V, Sigma-Aldrich) for 3 consecutive days.
- OVA/ day/mouse grade V, Sigma-Aldrich
- the nasal symptoms of allergic rhinitis were assessed by counting the sneezing+ nasal rubbing for 10 minutes after the challenge. The nasal symptoms were measured on day 3 ( Figure 17. a).
- Example 21 OVA-DermAll preventive immunotherapy is associated with increased level of IL-IO and does not induce immunoglobulins (Ig) that intensify allergic diseases
- Na ⁇ ve BALB/c mice female, 8-week-old, 4 mice/group were immunized with OVA-DermAll vaccine 3 times, every second week (Figure 18. a).
- the transdermal immunizations were performed based on the DermaVir- platform technology.
- the composition of pcOVA-DermAll vaccine was pcOVA+ PEIm+ excipient [10% dextrose].
- the control vaccine was composed as: PEIm+ excipient [10% dextrose].
- mice Two weeks later the mice were sacrificed, and OVA-specific IgE, IgGl and IgG2a levels were measured from the sera by a sandwich ELISA method (all were purchased from Bethyl Laboratories, Germany).
- IgE and IgGl are known to be important in the allergy induction, and IgG2a is thought to be a blocking antibody.
- IgG2a is thought to be a blocking antibody.
- pcOVA-DermAll immunization did not induce immunoglobulins such as OVA-specific IgE, IgGl or IgG2a that are known to be important in the pathomechanism of allergic diseases.
- Na ⁇ ve BALB/c mice female, 8-week-old, 4 mice/group
- pcOVA-DermAll vaccine 3 times, every second week (Figure 12. a). Two weeks later the mice were sacrificed and single cell suspensions of splenocytes were prepared.
- Example 22 OVA-DermAll therapeutic immunization is associated with increased level of IFN ⁇ which is the hallmark cytokine of activated ThI cells
- Na ⁇ ve BALB/c mice female, 8-week-old, 8 mice/group, purchased from Charles River Laboratories, Germany
- mice were sensitized with intraperitoneal ovalbumin (grade V, Sigma-Aldrich, St. Louis, MO) injection 4 times, every week (single dose: 25 ⁇ g OVA/ mouse + 2 mg Aluminium hydroxide as an adjuvant) (Figure 12. a).
- Each mouse were intranasally (i.n.) challenged with 500 ⁇ g OV A/day/mouse (grade V, Sigma) for 3 consecutive days.
- the nasal symptoms of allergic rhinitis were assessed by counting the sneezing + nasal rubbing for 10 minutes after the last challenge on day 3.
- mice were treated with DermAll immunization and it was repeated 2 times, every second week (Figure 19. a).
- the transdermal immunizations were performed based on the DermaVir platform technology.
- the composition of OVA-DermAll nanomedicine vaccine was 25 ⁇ g pcOVA+ PEIm+ excipient [10% dextrose].
- the control nanomedicine vaccine was composed as: PEIm+ excipient [10% dextrose].
- mice were challenged again (week 12) in both groups with 500 ⁇ g OV A/day/mouse for 3 consecutive days.
- the nasal symptoms were measured on day 3.
- the mice were challenged again (week 12) to assess the effect of OVA-DermAll vaccination. We did not observed significant difference in the nasal symptoms between the OVA-DermAll group and the control group at week 12.
- the mice were sacrificed (week 13) and single cell suspensions of splenocytes were prepared (Figure 19. a). Each sample of individual mouse was handled separately; 2 million cells were activated in 1 ml complete medium in each experiment (2 ml final volume). The splenocytes were activated with 50 ⁇ g/ml ovalbumin (grade V., Sigma-Aldrich, St.
- ELISA test for IFN ⁇ detection was purchased from BD Biosciences Pharmingen (San-Diego, CA) and the tests were performed according to the manufacturer's instruction.
- Example 23 Plasmid constructions The following components were engineered, using standard techniques available in the art and/or commercially available, to create the constructions used for the preparation of nanomedicine compositions:
- CopN 1200 bp
- locus tag CpB0334 (C. pneumoniae TWl 83 genome, SEQ ID NO: 2) and Major Outer Membrane Protein (MOMP): 1170 bp
- locus tag CpB0722 (C. pneumoniae TW183 genome, SEQ ID NO: 3) was inserted into the vector pVAXl (Invitrogen, USA, Figure 20).
- pVAXl Invitrogen, USA, Figure 20.
- the prokaryotic codons were changed to eukaryotic ones (see SEQ ID NO: 4).
- Modified fusion protein CopN/MOMP is expressed as antigen in the ChlamyDerm nanomedicine.
- ChlamyDerm nanomedicine was formulated according to the invention with the plasmid DNA expressing the CopN-MOMP fusion protein antigen (SEQ ID NO: 4, Fig. 21).
- the control nanomedicine was formulated with the pVax vector (not expressing Chlamidia antigens).
- the strength of the nanomedicine products was measured by UV photometer at 260 nm.
- Fig. 22.A The formation of nanoparticle was tested by gel shift methods.
- Fig. 22.B. The expression of the Chlamydia antigens was tested by Western blot analyses after transfection of control and ChlamyDerm products using Hep-2 cultures cells.
- Fig. 22.C. The expression of the Chlamydia antigens was tested by Western blot analyses after transfection of control and ChlamyDerm products using Hep-2 cultures cells.
- mice were used for testing the effectiveness of ChlamyDerm nanomedicine for preventive and therapeutic use against C. pneumoniae infection.
- DermaPrep transdermal administration (HU patent application No. P0900117) was used for the administration of the ChlamyDerm and control nanomedicines to mice. DermaPrep administration targets the nanomedicine vaccines to the lymph node dendritic cells.
- the human administration methods described in the patent application was adopted for small animals: animals were anesthetized and the hair on the back of mouse from nape of neck to base of tail (7 x 5 cm) was removed by an electric clipper (ModeLOster Golden A5).
- mice 16 weeks old mice were infected intranasally with C. pneumoniae (TWAR183, ATC-VR-2282) and therapeutic vaccination started as noted on Fig. 23.A. ChlamyDerm vaccination was repeated every week for 5 weeks. After that animals were sacrificed for blood and tissue sample isolation. To test the preventive use of vaccine mice were treated with ChlamyDerm once a week for 6 weeks. At the end animals were infected intranasally as mentioned above. (Fig. 23.B.) One week later animals were sacrificed. Blood and tissue samples were obtained to check the effect of vaccination on the immune system and the C. pneumoniae content of the lung.
- ChlamyDerm vaccination-induced specific immune response was measured by IFN- ⁇ ELISPOT.
- Spleen cells from ChlamyDerm and Control nanomedicine vaccinated mice were isolated in sterile environment and stimulated using five different overlapping peptides derived from the CopN-MOMP fusion protein for 24 hours.
- Phytohaemagglutinin (PHA) was used to test the responsiveness of the spleen cells.
- ChlamyDerm vaccination induced specific immune response in both C57B16 and ApoE " ' " knock out mice. (Fig. 24).
- Example 25 Effect of ChlamyDerm on C. pneumoniae infection induced inflammation and histological changes
- ChlamyDerm vaccination significantly decreased bacterial infection-induced elevation of serum C-reactive protein (CRP) and amyloid-A levels compared to control nanomedicine (Fig. 25).
- CRP serum C-reactive protein
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Immunology (AREA)
- Virology (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Nanotechnology (AREA)
- Pulmonology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Hematology (AREA)
- General Engineering & Computer Science (AREA)
- Medical Informatics (AREA)
- Molecular Biology (AREA)
- Crystallography & Structural Chemistry (AREA)
- Communicable Diseases (AREA)
- Biotechnology (AREA)
- Biophysics (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicinal Preparation (AREA)
Abstract
L'invention appartient au domaine de l'immunothérapie et des vaccins. Plus particulièrement, l'invention porte sur une composition pharmaceutique comprenant des nanoparticules composées de macromolécules et de polyéthylèneimine linéaire (1-PEI) ou d'un dérivé de 1-PEI dans une formulation liquide pharmaceutiquement acceptable, ainsi que sur un procédé pour la préparation de celle-ci. L'invention porte également sur des utilisations desdites compositions.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP13165968.2A EP2623117A1 (fr) | 2009-04-30 | 2010-04-30 | Composition de nanomédicament immunogène, préparation et utilisations associées |
Applications Claiming Priority (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
HU0900269A HU0900269D0 (en) | 2009-04-30 | 2009-04-30 | Synhesis of carbohydrate n-conjugates with bh3-pyridine complex catalyst |
HU0900300A HU0900300D0 (en) | 2009-05-14 | 2009-05-14 | Imminogenic nanomedicine composition |
HU0900299A HU0900299D0 (en) | 2009-05-14 | 2009-05-14 | Immunogenic nanomedicine for the prevention and treatment of chlamidia infection |
HU0900298A HU0900298D0 (en) | 2009-05-14 | 2009-05-14 | Immungenic nanomedicine for the prevention and treatment of allergy |
PCT/IB2010/051909 WO2010125544A1 (fr) | 2009-04-30 | 2010-04-30 | Composition de nanomédicament immunogène, préparation et utilisations de cette composition |
Publications (1)
Publication Number | Publication Date |
---|---|
EP2453911A1 true EP2453911A1 (fr) | 2012-05-23 |
Family
ID=89988975
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP13165968.2A Withdrawn EP2623117A1 (fr) | 2009-04-30 | 2010-04-30 | Composition de nanomédicament immunogène, préparation et utilisations associées |
EP10719414A Withdrawn EP2453911A1 (fr) | 2009-04-30 | 2010-04-30 | Composition de nanomédicament immunogène, préparation et utilisations de cette composition |
Family Applications Before (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
EP13165968.2A Withdrawn EP2623117A1 (fr) | 2009-04-30 | 2010-04-30 | Composition de nanomédicament immunogène, préparation et utilisations associées |
Country Status (2)
Country | Link |
---|---|
EP (2) | EP2623117A1 (fr) |
WO (1) | WO2010125544A1 (fr) |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
AU2012217685B2 (en) * | 2011-02-15 | 2017-05-18 | Merrimack Pharmaceuticals, Inc. | Compositions and methods for delivering nucleic acid to a cell |
JP2016521727A (ja) * | 2013-06-10 | 2016-07-25 | ポリヴァロール ソシエテ アン コマンディト | 寸法および生物活性を維持するフリーズドライ高分子電解質複合体 |
KR101809795B1 (ko) | 2014-10-30 | 2017-12-18 | 서울대학교산학협력단 | 폴리올계 삼투압적 폴리디자일리톨 폴리머 유전자 전달체 및 이의 용도 |
Family Cites Families (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
FR2722506B1 (fr) | 1994-07-13 | 1996-08-14 | Rhone Poulenc Rorer Sa | Composition contenant des acides nucleiques, preparation et utilisations |
EP1024836B1 (fr) * | 1997-09-15 | 2006-12-13 | Genetic Immunity, LLC | Compositions pour l'administration de gènes a des cellules présentatrices d'antigènes de la peau |
DE19743135A1 (de) | 1997-09-30 | 1999-04-01 | Hoechst Marion Roussel De Gmbh | Biologisch verträgliche niedermolekular Polyethylenimine |
CA2703852A1 (fr) * | 2007-11-09 | 2009-05-14 | Northeastern University | Nanoparticules de type micelles auto-assemblantes pour une administration systemique de gene |
US8513206B2 (en) * | 2008-07-29 | 2013-08-20 | Council Of Scientific And Industrial Research | Crosslinked PEI nanoparticle transfection agents for delivery of biomolecules with increased efficiency |
-
2010
- 2010-04-30 EP EP13165968.2A patent/EP2623117A1/fr not_active Withdrawn
- 2010-04-30 EP EP10719414A patent/EP2453911A1/fr not_active Withdrawn
- 2010-04-30 WO PCT/IB2010/051909 patent/WO2010125544A1/fr active Application Filing
Non-Patent Citations (1)
Title |
---|
See references of WO2010125544A1 * |
Also Published As
Publication number | Publication date |
---|---|
WO2010125544A1 (fr) | 2010-11-04 |
EP2623117A1 (fr) | 2013-08-07 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Jain et al. | Messenger RNA-based vaccines: Past, present, and future directions in the context of the COVID-19 pandemic | |
Donnelly et al. | Technical and regulatory hurdles for DNA vaccines | |
JP6357487B2 (ja) | 安定化ヒト免疫不全ウイルス(hiv)エンベロープ(env)トリマーワクチン及びそれを使用する方法 | |
US7939318B2 (en) | Flexible vaccine assembly and vaccine delivery platform | |
Nguyen et al. | Enhanced cancer DNA vaccine via direct transfection to host dendritic cells recruited in injectable scaffolds | |
EP1108035B1 (fr) | Traitement du cancer du col ut rin | |
Trovato et al. | Novel antigen delivery systems | |
Sokolova et al. | The potential of nanoparticles for the immunization against viral infections | |
US20050112139A1 (en) | Immunogenic composition and method of developing a vaccine based on factor H binding sites | |
JP2009511636A (ja) | アルファウイルスレプリコン粒子による粘膜免疫および全身免疫 | |
Rodriguez-Gascon et al. | Vaginal gene therapy | |
ES2339762T3 (es) | Vacuna contra el vih y procedimiento de uso. | |
US20230090311A1 (en) | Self-assembling, self-adjuvanting system for delivery of vaccines | |
JP2019505567A (ja) | 治療用免疫調節組成物 | |
Knight et al. | Engineering Vaccines for Tissue‐Resident Memory T Cells | |
Uddin et al. | A novel formulation strategy to deliver combined DNA and VLP based HPV vaccine | |
ES2902787T3 (es) | Vacunas de ADNi y procedimientos para utilizar las mismas | |
Abedi et al. | Rabies vaccine: Recent update and comprehensive review of in vitro and in vivo studies | |
WO2010125544A1 (fr) | Composition de nanomédicament immunogène, préparation et utilisations de cette composition | |
US20050282263A1 (en) | Flexible vaccine assembly and vaccine delivery platform | |
Alpar et al. | Current status of DNA vaccines and their route of administration | |
WO2005091753A2 (fr) | Ensemble vaccin modulable et plate-forme d'administration de vaccins | |
Alpar et al. | Strategies for DNA vaccine delivery | |
JP2013545733A (ja) | ヒト免疫不全ウィルス(hiv)の組換えエンベロープ蛋白質及びそれを含むワクチン | |
KR102211077B1 (ko) | 바이러스 유사 입자를 이용한 슈도-타입 광견병 백신 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PUAJ | Public notification under rule 129 epc |
Free format text: ORIGINAL CODE: 0009425 |
|
PUAI | Public reference made under article 153(3) epc to a published international application that has entered the european phase |
Free format text: ORIGINAL CODE: 0009012 |
|
17P | Request for examination filed |
Effective date: 20120323 |
|
AK | Designated contracting states |
Kind code of ref document: A1 Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR |
|
DAX | Request for extension of the european patent (deleted) | ||
17Q | First examination report despatched |
Effective date: 20130128 |
|
STAA | Information on the status of an ep patent application or granted ep patent |
Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN |
|
18D | Application deemed to be withdrawn |
Effective date: 20130608 |