US20210371462A1 - Cdca1-derived peptide and vaccine containing same - Google Patents
Cdca1-derived peptide and vaccine containing same Download PDFInfo
- Publication number
- US20210371462A1 US20210371462A1 US17/264,277 US201917264277A US2021371462A1 US 20210371462 A1 US20210371462 A1 US 20210371462A1 US 201917264277 A US201917264277 A US 201917264277A US 2021371462 A1 US2021371462 A1 US 2021371462A1
- Authority
- US
- United States
- Prior art keywords
- peptide
- present
- cancer
- peptides
- amino 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.)
- Abandoned
Links
- 108090000765 processed proteins & peptides Proteins 0.000 title claims abstract description 719
- 229960005486 vaccine Drugs 0.000 title description 21
- 101150033450 NUF2 gene Proteins 0.000 title description 3
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 claims abstract description 266
- 102000004196 processed proteins & peptides Human genes 0.000 claims abstract description 232
- 210000000612 antigen-presenting cell Anatomy 0.000 claims abstract description 216
- 238000000034 method Methods 0.000 claims abstract description 192
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 189
- 201000011510 cancer Diseases 0.000 claims abstract description 163
- 101000590482 Homo sapiens Kinetochore protein Nuf2 Proteins 0.000 claims abstract description 141
- 102100032431 Kinetochore protein Nuf2 Human genes 0.000 claims abstract description 134
- 102000040430 polynucleotide Human genes 0.000 claims abstract description 129
- 108091033319 polynucleotide Proteins 0.000 claims abstract description 129
- 239000002157 polynucleotide Substances 0.000 claims abstract description 129
- 230000001939 inductive effect Effects 0.000 claims abstract description 126
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 120
- 239000000203 mixture Substances 0.000 claims abstract description 60
- 230000028993 immune response Effects 0.000 claims abstract description 37
- 230000002980 postoperative effect Effects 0.000 claims abstract description 17
- 210000004027 cell Anatomy 0.000 claims description 221
- 108091007433 antigens Proteins 0.000 claims description 100
- 239000000427 antigen Substances 0.000 claims description 99
- 102000036639 antigens Human genes 0.000 claims description 99
- 150000001413 amino acids Chemical class 0.000 claims description 90
- 235000001014 amino acid Nutrition 0.000 claims description 87
- 210000001808 exosome Anatomy 0.000 claims description 78
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 49
- 108090000623 proteins and genes Proteins 0.000 claims description 38
- 108091008874 T cell receptors Proteins 0.000 claims description 36
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 claims description 36
- 230000027455 binding Effects 0.000 claims description 35
- 239000002671 adjuvant Substances 0.000 claims description 27
- 238000000338 in vitro Methods 0.000 claims description 25
- 238000011321 prophylaxis Methods 0.000 claims description 24
- 230000002265 prevention Effects 0.000 claims description 23
- 239000004615 ingredient Substances 0.000 claims description 21
- 238000011282 treatment Methods 0.000 claims description 21
- 238000006467 substitution reaction Methods 0.000 claims description 20
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 claims description 18
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 claims description 18
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 claims description 18
- 238000001727 in vivo Methods 0.000 claims description 17
- 125000000539 amino acid group Chemical group 0.000 claims description 14
- 238000012258 culturing Methods 0.000 claims description 14
- 125000001433 C-terminal amino-acid group Chemical group 0.000 claims description 11
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 claims description 11
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 claims description 11
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 claims description 11
- 235000003704 aspartic acid Nutrition 0.000 claims description 11
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 claims description 11
- 239000000839 emulsion Substances 0.000 claims description 11
- 235000013922 glutamic acid Nutrition 0.000 claims description 11
- 239000004220 glutamic acid Substances 0.000 claims description 11
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 claims description 11
- 206010005003 Bladder cancer Diseases 0.000 claims description 10
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 10
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims description 10
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 claims description 10
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 10
- 206010006187 Breast cancer Diseases 0.000 claims description 9
- 208000026310 Breast neoplasm Diseases 0.000 claims description 9
- 206010008342 Cervix carcinoma Diseases 0.000 claims description 9
- 206010009944 Colon cancer Diseases 0.000 claims description 9
- 208000000461 Esophageal Neoplasms Diseases 0.000 claims description 9
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 9
- 206010025323 Lymphomas Diseases 0.000 claims description 9
- 206010060862 Prostate cancer Diseases 0.000 claims description 9
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 9
- 206010038389 Renal cancer Diseases 0.000 claims description 9
- 206010041067 Small cell lung cancer Diseases 0.000 claims description 9
- 206010068771 Soft tissue neoplasm Diseases 0.000 claims description 9
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 9
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 claims description 9
- 201000010881 cervical cancer Diseases 0.000 claims description 9
- 208000029742 colonic neoplasm Diseases 0.000 claims description 9
- 201000004101 esophageal cancer Diseases 0.000 claims description 9
- 206010017758 gastric cancer Diseases 0.000 claims description 9
- 201000010536 head and neck cancer Diseases 0.000 claims description 9
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 9
- 201000010982 kidney cancer Diseases 0.000 claims description 9
- 201000008968 osteosarcoma Diseases 0.000 claims description 9
- 208000000587 small cell lung carcinoma Diseases 0.000 claims description 9
- 201000011549 stomach cancer Diseases 0.000 claims description 9
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 claims description 8
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 claims description 8
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 claims description 8
- 239000004473 Threonine Substances 0.000 claims description 8
- 125000001493 tyrosinyl group Chemical group [H]OC1=C([H])C([H])=C(C([H])=C1[H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 claims description 8
- 239000003937 drug carrier Substances 0.000 claims description 6
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 claims description 5
- 238000012216 screening Methods 0.000 claims description 4
- 239000004480 active ingredient Substances 0.000 abstract description 39
- 230000008685 targeting Effects 0.000 abstract description 11
- 125000003275 alpha amino acid group Chemical group 0.000 description 84
- 229940024606 amino acid Drugs 0.000 description 78
- 238000004519 manufacturing process Methods 0.000 description 52
- 102210024048 HLA-A*01:01 Human genes 0.000 description 47
- 239000013598 vector Substances 0.000 description 41
- 108010074328 Interferon-gamma Proteins 0.000 description 40
- 102100037850 Interferon gamma Human genes 0.000 description 39
- 230000014509 gene expression Effects 0.000 description 31
- 239000000243 solution Substances 0.000 description 20
- 210000004443 dendritic cell Anatomy 0.000 description 19
- 239000000126 substance Substances 0.000 description 19
- 230000001472 cytotoxic effect Effects 0.000 description 18
- 235000018102 proteins Nutrition 0.000 description 18
- 102000004169 proteins and genes Human genes 0.000 description 18
- 101001109518 Homo sapiens N-acetylneuraminate lyase Proteins 0.000 description 17
- 102100022686 N-acetylneuraminate lyase Human genes 0.000 description 17
- 239000013604 expression vector Substances 0.000 description 17
- 108091005601 modified peptides Proteins 0.000 description 17
- 238000009472 formulation Methods 0.000 description 16
- 230000006698 induction Effects 0.000 description 16
- 239000000463 material Substances 0.000 description 16
- 239000002609 medium Substances 0.000 description 16
- 230000004048 modification Effects 0.000 description 15
- 238000012986 modification Methods 0.000 description 15
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 14
- 230000008569 process Effects 0.000 description 14
- 201000010099 disease Diseases 0.000 description 13
- 239000003814 drug Substances 0.000 description 11
- 230000006870 function Effects 0.000 description 11
- 230000003053 immunization Effects 0.000 description 11
- 238000002360 preparation method Methods 0.000 description 11
- 150000003839 salts Chemical class 0.000 description 11
- 210000002966 serum Anatomy 0.000 description 11
- 238000002965 ELISA Methods 0.000 description 10
- 241000588724 Escherichia coli Species 0.000 description 10
- 241001465754 Metazoa Species 0.000 description 10
- 150000007523 nucleic acids Chemical class 0.000 description 10
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 10
- 239000000523 sample Substances 0.000 description 10
- 210000001519 tissue Anatomy 0.000 description 10
- 210000004369 blood Anatomy 0.000 description 9
- 239000008280 blood Substances 0.000 description 9
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 9
- 230000001404 mediated effect Effects 0.000 description 9
- 102000039446 nucleic acids Human genes 0.000 description 9
- 108020004707 nucleic acids Proteins 0.000 description 9
- 239000000047 product Substances 0.000 description 9
- 229940079593 drug Drugs 0.000 description 8
- 230000000694 effects Effects 0.000 description 8
- 230000035755 proliferation Effects 0.000 description 8
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 8
- 108020004705 Codon Proteins 0.000 description 7
- 238000011510 Elispot assay Methods 0.000 description 7
- 102000004190 Enzymes Human genes 0.000 description 7
- 108090000790 Enzymes Proteins 0.000 description 7
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 7
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 7
- 108010002350 Interleukin-2 Proteins 0.000 description 7
- 101100221606 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) COS7 gene Proteins 0.000 description 7
- 229940088598 enzyme Drugs 0.000 description 7
- 210000004408 hybridoma Anatomy 0.000 description 7
- 238000009169 immunotherapy Methods 0.000 description 7
- 210000004698 lymphocyte Anatomy 0.000 description 7
- 238000010647 peptide synthesis reaction Methods 0.000 description 7
- 229920001184 polypeptide Polymers 0.000 description 7
- 208000024891 symptom Diseases 0.000 description 7
- 238000003786 synthesis reaction Methods 0.000 description 7
- 108020004414 DNA Proteins 0.000 description 6
- 102100028972 HLA class I histocompatibility antigen, A alpha chain Human genes 0.000 description 6
- 108010075704 HLA-A Antigens Proteins 0.000 description 6
- 241000124008 Mammalia Species 0.000 description 6
- 239000012472 biological sample Substances 0.000 description 6
- 210000002865 immune cell Anatomy 0.000 description 6
- 238000002649 immunization Methods 0.000 description 6
- 238000002156 mixing Methods 0.000 description 6
- 229920000642 polymer Polymers 0.000 description 6
- 230000001954 sterilising effect Effects 0.000 description 6
- 230000001225 therapeutic effect Effects 0.000 description 6
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 5
- 238000004458 analytical method Methods 0.000 description 5
- 238000003556 assay Methods 0.000 description 5
- 230000008901 benefit Effects 0.000 description 5
- 238000002619 cancer immunotherapy Methods 0.000 description 5
- 230000001413 cellular effect Effects 0.000 description 5
- 150000001875 compounds Chemical class 0.000 description 5
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 5
- 239000012634 fragment Substances 0.000 description 5
- 230000002401 inhibitory effect Effects 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 230000000670 limiting effect Effects 0.000 description 5
- 150000002632 lipids Chemical class 0.000 description 5
- 239000002953 phosphate buffered saline Substances 0.000 description 5
- 239000002504 physiological saline solution Substances 0.000 description 5
- 230000001629 suppression Effects 0.000 description 5
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 4
- 108700028369 Alleles Proteins 0.000 description 4
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 4
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 4
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 4
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 4
- 206010035226 Plasma cell myeloma Diseases 0.000 description 4
- 230000002776 aggregation Effects 0.000 description 4
- 238000004220 aggregation Methods 0.000 description 4
- 230000000259 anti-tumor effect Effects 0.000 description 4
- 230000036765 blood level Effects 0.000 description 4
- 239000000969 carrier Substances 0.000 description 4
- 230000007910 cell fusion Effects 0.000 description 4
- 238000004587 chromatography analysis Methods 0.000 description 4
- 239000002299 complementary DNA Substances 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 238000001914 filtration Methods 0.000 description 4
- 238000004108 freeze drying Methods 0.000 description 4
- 229910052739 hydrogen Inorganic materials 0.000 description 4
- 230000001900 immune effect Effects 0.000 description 4
- 230000002998 immunogenetic effect Effects 0.000 description 4
- 230000005764 inhibitory process Effects 0.000 description 4
- 229930182817 methionine Natural products 0.000 description 4
- 210000001616 monocyte Anatomy 0.000 description 4
- 201000000050 myeloid neoplasm Diseases 0.000 description 4
- 239000013642 negative control Substances 0.000 description 4
- 239000008363 phosphate buffer Substances 0.000 description 4
- 229920003023 plastic Polymers 0.000 description 4
- 239000004033 plastic Substances 0.000 description 4
- 230000003389 potentiating effect Effects 0.000 description 4
- 238000003127 radioimmunoassay Methods 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 239000002904 solvent Substances 0.000 description 4
- 238000004659 sterilization and disinfection Methods 0.000 description 4
- 230000004936 stimulating effect Effects 0.000 description 4
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 150000008574 D-amino acids Chemical class 0.000 description 3
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 3
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 3
- 206010058467 Lung neoplasm malignant Diseases 0.000 description 3
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 3
- 108010076504 Protein Sorting Signals Proteins 0.000 description 3
- 108020004511 Recombinant DNA Proteins 0.000 description 3
- 101710120037 Toxin CcdB Proteins 0.000 description 3
- 235000004279 alanine Nutrition 0.000 description 3
- -1 antibody Proteins 0.000 description 3
- 230000030741 antigen processing and presentation Effects 0.000 description 3
- 238000013459 approach Methods 0.000 description 3
- 210000003719 b-lymphocyte Anatomy 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- KRKNYBCHXYNGOX-UHFFFAOYSA-N citric acid Chemical compound OC(=O)CC(O)(C(O)=O)CC(O)=O KRKNYBCHXYNGOX-UHFFFAOYSA-N 0.000 description 3
- 239000003085 diluting agent Substances 0.000 description 3
- 238000003113 dilution method Methods 0.000 description 3
- 238000001415 gene therapy Methods 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 238000010353 genetic engineering Methods 0.000 description 3
- 238000004128 high performance liquid chromatography Methods 0.000 description 3
- 230000036039 immunity Effects 0.000 description 3
- 238000010166 immunofluorescence Methods 0.000 description 3
- 230000002163 immunogen Effects 0.000 description 3
- 238000010324 immunological assay Methods 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 238000003780 insertion Methods 0.000 description 3
- 230000037431 insertion Effects 0.000 description 3
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 3
- 201000005202 lung cancer Diseases 0.000 description 3
- 208000020816 lung neoplasm Diseases 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 238000012544 monitoring process Methods 0.000 description 3
- 125000003729 nucleotide group Chemical group 0.000 description 3
- 239000000546 pharmaceutical excipient Substances 0.000 description 3
- 229910052700 potassium Inorganic materials 0.000 description 3
- 239000002243 precursor Substances 0.000 description 3
- 239000003755 preservative agent Substances 0.000 description 3
- 238000004393 prognosis Methods 0.000 description 3
- 230000000644 propagated effect Effects 0.000 description 3
- 238000000926 separation method Methods 0.000 description 3
- 239000003381 stabilizer Substances 0.000 description 3
- 238000010561 standard procedure Methods 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 238000010254 subcutaneous injection Methods 0.000 description 3
- 239000004094 surface-active agent Substances 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 125000000341 threoninyl group Chemical group [H]OC([H])(C([H])([H])[H])C([H])(N([H])[H])C(*)=O 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 239000003656 tris buffered saline Substances 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- 239000008215 water for injection Substances 0.000 description 3
- 241000282693 Cercopithecidae Species 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 2
- 206010059866 Drug resistance Diseases 0.000 description 2
- VZCYOOQTPOCHFL-OWOJBTEDSA-N Fumaric acid Chemical compound OC(=O)\C=C\C(O)=O VZCYOOQTPOCHFL-OWOJBTEDSA-N 0.000 description 2
- 239000004471 Glycine Substances 0.000 description 2
- 102000004457 Granulocyte-Macrophage Colony-Stimulating Factor Human genes 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- 150000008575 L-amino acids Chemical class 0.000 description 2
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 2
- 241000283953 Lagomorpha Species 0.000 description 2
- 241000282567 Macaca fascicularis Species 0.000 description 2
- 206010027476 Metastases Diseases 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- 108010046117 N-palmitoyl-5,6-dipalmitoyl-S-glycerylcysteinyl-seryl-serine Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 108010033276 Peptide Fragments Proteins 0.000 description 2
- 102000007079 Peptide Fragments Human genes 0.000 description 2
- NBIIXXVUZAFLBC-UHFFFAOYSA-N Phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 241000288906 Primates Species 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 2
- 241000700618 Vaccinia virus Species 0.000 description 2
- 206010046865 Vaccinia virus infection Diseases 0.000 description 2
- 238000001042 affinity chromatography Methods 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 239000002585 base Substances 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- 239000012620 biological material Substances 0.000 description 2
- 238000009566 cancer vaccine Methods 0.000 description 2
- 229940022399 cancer vaccine Drugs 0.000 description 2
- 229910052799 carbon Inorganic materials 0.000 description 2
- RUDATBOHQWOJDD-BSWAIDMHSA-N chenodeoxycholic acid Chemical compound C([C@H]1C[C@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(O)=O)C)[C@@]2(C)CC1 RUDATBOHQWOJDD-BSWAIDMHSA-N 0.000 description 2
- 239000012531 culture fluid Substances 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- XBDQKXXYIPTUBI-UHFFFAOYSA-N dimethylselenoniopropionate Natural products CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 238000005538 encapsulation Methods 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 229910052731 fluorine Inorganic materials 0.000 description 2
- 239000011888 foil Substances 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 230000013595 glycosylation Effects 0.000 description 2
- 238000006206 glycosylation reaction Methods 0.000 description 2
- 239000001963 growth medium Substances 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 238000003384 imaging method Methods 0.000 description 2
- 238000003018 immunoassay Methods 0.000 description 2
- 230000005847 immunogenicity Effects 0.000 description 2
- 230000001965 increasing effect Effects 0.000 description 2
- 238000010253 intravenous injection Methods 0.000 description 2
- 238000004255 ion exchange chromatography Methods 0.000 description 2
- 210000001821 langerhans cell Anatomy 0.000 description 2
- 210000000265 leukocyte Anatomy 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 2
- 238000002493 microarray Methods 0.000 description 2
- 229960004857 mitomycin Drugs 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 230000003647 oxidation Effects 0.000 description 2
- 238000007254 oxidation reaction Methods 0.000 description 2
- 239000002245 particle Substances 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 210000005259 peripheral blood Anatomy 0.000 description 2
- 239000011886 peripheral blood Substances 0.000 description 2
- 229910052698 phosphorus Inorganic materials 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 238000006116 polymerization reaction Methods 0.000 description 2
- 230000037452 priming Effects 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 230000006798 recombination Effects 0.000 description 2
- 238000005215 recombination Methods 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 239000007929 subcutaneous injection Substances 0.000 description 2
- 229910052717 sulfur Inorganic materials 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 238000007910 systemic administration Methods 0.000 description 2
- 239000003053 toxin Substances 0.000 description 2
- 231100000765 toxin Toxicity 0.000 description 2
- VZCYOOQTPOCHFL-UHFFFAOYSA-N trans-butenedioic acid Natural products OC(=O)C=CC(O)=O VZCYOOQTPOCHFL-UHFFFAOYSA-N 0.000 description 2
- 230000009261 transgenic effect Effects 0.000 description 2
- XVQKZSLOGHBCET-INVHGPFASA-N tripalmitoyl-S-glyceryl-cysteinyl-seryl-serine Chemical compound CCCCCCCCCCCCCCCC(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CO)C(O)=O)CSCC(OC(=O)CCCCCCCCCCCCCCC)COC(=O)CCCCCCCCCCCCCCC XVQKZSLOGHBCET-INVHGPFASA-N 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 229910052721 tungsten Inorganic materials 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- 208000007089 vaccinia Diseases 0.000 description 2
- 229910052727 yttrium Inorganic materials 0.000 description 2
- YYGNTYWPHWGJRM-UHFFFAOYSA-N (6E,10E,14E,18E)-2,6,10,15,19,23-hexamethyltetracosa-2,6,10,14,18,22-hexaene Chemical compound CC(C)=CCCC(C)=CCCC(C)=CCCC=C(C)CCC=C(C)CCC=C(C)C YYGNTYWPHWGJRM-UHFFFAOYSA-N 0.000 description 1
- BJEPYKJPYRNKOW-REOHCLBHSA-N (S)-malic acid Chemical compound OC(=O)[C@@H](O)CC(O)=O BJEPYKJPYRNKOW-REOHCLBHSA-N 0.000 description 1
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 1
- UKAUYVFTDYCKQA-UHFFFAOYSA-N -2-Amino-4-hydroxybutanoic acid Natural products OC(=O)C(N)CCO UKAUYVFTDYCKQA-UHFFFAOYSA-N 0.000 description 1
- VQFKFAKEUMHBLV-BYSUZVQFSA-N 1-O-(alpha-D-galactosyl)-N-hexacosanoylphytosphingosine Chemical compound CCCCCCCCCCCCCCCCCCCCCCCCCC(=O)N[C@H]([C@H](O)[C@H](O)CCCCCCCCCCCCCC)CO[C@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O VQFKFAKEUMHBLV-BYSUZVQFSA-N 0.000 description 1
- LEBVLXFERQHONN-UHFFFAOYSA-N 1-butyl-N-(2,6-dimethylphenyl)piperidine-2-carboxamide Chemical compound CCCCN1CCCCC1C(=O)NC1=C(C)C=CC=C1C LEBVLXFERQHONN-UHFFFAOYSA-N 0.000 description 1
- GOJUJUVQIVIZAV-UHFFFAOYSA-N 2-amino-4,6-dichloropyrimidine-5-carbaldehyde Chemical group NC1=NC(Cl)=C(C=O)C(Cl)=N1 GOJUJUVQIVIZAV-UHFFFAOYSA-N 0.000 description 1
- BFSVOASYOCHEOV-UHFFFAOYSA-N 2-diethylaminoethanol Chemical compound CCN(CC)CCO BFSVOASYOCHEOV-UHFFFAOYSA-N 0.000 description 1
- BMYNFMYTOJXKLE-UHFFFAOYSA-N 3-azaniumyl-2-hydroxypropanoate Chemical compound NCC(O)C(O)=O BMYNFMYTOJXKLE-UHFFFAOYSA-N 0.000 description 1
- RHKWIGHJGOEUSM-UHFFFAOYSA-N 3h-imidazo[4,5-h]quinoline Chemical class C1=CN=C2C(N=CN3)=C3C=CC2=C1 RHKWIGHJGOEUSM-UHFFFAOYSA-N 0.000 description 1
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 1
- XZKIHKMTEMTJQX-UHFFFAOYSA-N 4-Nitrophenyl Phosphate Chemical compound OP(O)(=O)OC1=CC=C([N+]([O-])=O)C=C1 XZKIHKMTEMTJQX-UHFFFAOYSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- 102100027715 4-hydroxy-2-oxoglutarate aldolase, mitochondrial Human genes 0.000 description 1
- 241001556567 Acanthamoeba polyphaga mimivirus Species 0.000 description 1
- 102100027241 Adenylyl cyclase-associated protein 1 Human genes 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 206010003445 Ascites Diseases 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 101100136076 Aspergillus oryzae (strain ATCC 42149 / RIB 40) pel1 gene Proteins 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 241000193738 Bacillus anthracis Species 0.000 description 1
- 244000063299 Bacillus subtilis Species 0.000 description 1
- 235000014469 Bacillus subtilis Nutrition 0.000 description 1
- 108020000946 Bacterial DNA Proteins 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 1
- 102000003930 C-Type Lectins Human genes 0.000 description 1
- 108090000342 C-Type Lectins Proteins 0.000 description 1
- 108010077333 CAP1-6D Proteins 0.000 description 1
- 101100314454 Caenorhabditis elegans tra-1 gene Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 241000282552 Chlorocebus aethiops Species 0.000 description 1
- 102000009016 Cholera Toxin Human genes 0.000 description 1
- 108010049048 Cholera Toxin Proteins 0.000 description 1
- VYZAMTAEIAYCRO-UHFFFAOYSA-N Chromium Chemical compound [Cr] VYZAMTAEIAYCRO-UHFFFAOYSA-N 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 108050006400 Cyclin Proteins 0.000 description 1
- 102000016736 Cyclin Human genes 0.000 description 1
- 102100024829 DNA polymerase delta catalytic subunit Human genes 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- FEWJPZIEWOKRBE-JCYAYHJZSA-N Dextrotartaric acid Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O FEWJPZIEWOKRBE-JCYAYHJZSA-N 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 102000004533 Endonucleases Human genes 0.000 description 1
- 108010042407 Endonucleases Proteins 0.000 description 1
- 108010040721 Flagellin Proteins 0.000 description 1
- 208000000666 Fowlpox Diseases 0.000 description 1
- 241000941423 Grom virus Species 0.000 description 1
- 108010026122 HLA-A*33 antigen Proteins 0.000 description 1
- 108010013476 HLA-A24 Antigen Proteins 0.000 description 1
- 241001554831 Hamadryas <angiosperm> Species 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 101001081225 Homo sapiens 4-hydroxy-2-oxoglutarate aldolase, mitochondrial Proteins 0.000 description 1
- 101000868333 Homo sapiens Cyclin-dependent kinase 1 Proteins 0.000 description 1
- 101000909198 Homo sapiens DNA polymerase delta catalytic subunit Proteins 0.000 description 1
- 101000604027 Homo sapiens Nuclear protein localization protein 4 homolog Proteins 0.000 description 1
- 101000974007 Homo sapiens Nucleosome assembly protein 1-like 3 Proteins 0.000 description 1
- 101001099181 Homo sapiens TATA-binding protein-associated factor 2N Proteins 0.000 description 1
- PMMYEEVYMWASQN-DMTCNVIQSA-N Hydroxyproline Chemical compound O[C@H]1CN[C@H](C(O)=O)C1 PMMYEEVYMWASQN-DMTCNVIQSA-N 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 1
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 108010002586 Interleukin-7 Proteins 0.000 description 1
- 108010063738 Interleukins Proteins 0.000 description 1
- 102000015696 Interleukins Human genes 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine Chemical compound OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-ZXPFJRLXSA-N L-methionine (R)-S-oxide Chemical compound C[S@@](=O)CC[C@H]([NH3+])C([O-])=O QEFRNWWLZKMPFJ-ZXPFJRLXSA-N 0.000 description 1
- QEFRNWWLZKMPFJ-UHFFFAOYSA-N L-methionine sulphoxide Natural products CS(=O)CCC(N)C(O)=O QEFRNWWLZKMPFJ-UHFFFAOYSA-N 0.000 description 1
- LRQKBLKVPFOOQJ-YFKPBYRVSA-N L-norleucine Chemical compound CCCC[C@H]([NH3+])C([O-])=O LRQKBLKVPFOOQJ-YFKPBYRVSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- 108010063045 Lactoferrin Proteins 0.000 description 1
- 102000010445 Lactoferrin Human genes 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 239000012097 Lipofectamine 2000 Substances 0.000 description 1
- 108010028921 Lipopeptides Proteins 0.000 description 1
- 108090001030 Lipoproteins Proteins 0.000 description 1
- 102000004895 Lipoproteins Human genes 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 102000043129 MHC class I family Human genes 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 241000282560 Macaca mulatta Species 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- PWHULOQIROXLJO-UHFFFAOYSA-N Manganese Chemical compound [Mn] PWHULOQIROXLJO-UHFFFAOYSA-N 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- GUVMFDICMFQHSZ-UHFFFAOYSA-N N-(1-aminoethenyl)-1-[4-[[5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[5-(4-amino-5-methyl-2-oxopyrimidin-1-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[hydroxy-[[3-[hydroxy-[[3-hydroxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy]phosphinothioyl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy]phosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(2-amino-6-oxo-1H-purin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(2-amino-6-oxo-1H-purin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxyoxolan-2-yl]methoxy-hydroxyphosphinothioyl]oxy-5-[[[2-[[[2-[[[5-(2-amino-6-oxo-1H-purin-9-yl)-2-[[[5-(4-amino-2-oxopyrimidin-1-yl)-2-[[hydroxy-[2-(hydroxymethyl)-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-3-yl]oxyphosphinothioyl]oxymethyl]oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-3-yl]oxy-hydroxyphosphinothioyl]oxymethyl]oxolan-2-yl]-5-methylimidazole-4-carboxamide Chemical compound CC1=C(C(=O)NC(N)=C)N=CN1C1OC(COP(O)(=S)OC2C(OC(C2)N2C(N=C(N)C=C2)=O)COP(O)(=S)OC2C(OC(C2)N2C(NC(=O)C(C)=C2)=O)COP(O)(=S)OC2C(OC(C2)N2C3=C(C(NC(N)=N3)=O)N=C2)COP(O)(=S)OC2C(OC(C2)N2C(N=C(N)C=C2)=O)COP(O)(=S)OC2C(OC(C2)N2C(NC(=O)C(C)=C2)=O)CO)C(OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(N=C(N)C=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C3=C(C(NC(N)=N3)=O)N=C2)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)OP(O)(=S)OCC2C(CC(O2)N2C(NC(=O)C(C)=C2)=O)O)C1 GUVMFDICMFQHSZ-UHFFFAOYSA-N 0.000 description 1
- 108091061960 Naked DNA Proteins 0.000 description 1
- 229930193140 Neomycin Natural products 0.000 description 1
- 108700019961 Neoplasm Genes Proteins 0.000 description 1
- 102000048850 Neoplasm Genes Human genes 0.000 description 1
- GRYLNZFGIOXLOG-UHFFFAOYSA-N Nitric acid Chemical compound O[N+]([O-])=O GRYLNZFGIOXLOG-UHFFFAOYSA-N 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 102100038438 Nuclear protein localization protein 4 homolog Human genes 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 241000282577 Pan troglodytes Species 0.000 description 1
- 108090000526 Papain Proteins 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 241000235648 Pichia Species 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- OFOBLEOULBTSOW-UHFFFAOYSA-N Propanedioic acid Natural products OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 241000700159 Rattus Species 0.000 description 1
- 208000006265 Renal cell carcinoma Diseases 0.000 description 1
- 108020005091 Replication Origin Proteins 0.000 description 1
- 101100094105 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) NPL6 gene Proteins 0.000 description 1
- 241000607142 Salmonella Species 0.000 description 1
- 241000293871 Salmonella enterica subsp. enterica serovar Typhi Species 0.000 description 1
- 229920002684 Sepharose Polymers 0.000 description 1
- 108020004459 Small interfering RNA Proteins 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- 101710137500 T7 RNA polymerase Proteins 0.000 description 1
- 102100038917 TATA-binding protein-associated factor 2N Human genes 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric acid Natural products [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- BHEOSNUKNHRBNM-UHFFFAOYSA-N Tetramethylsqualene Natural products CC(=C)C(C)CCC(=C)C(C)CCC(C)=CCCC=C(C)CCC(C)C(=C)CCC(C)C(C)=C BHEOSNUKNHRBNM-UHFFFAOYSA-N 0.000 description 1
- ZLJJDBSDZSZVTF-LXOQPCSCSA-N Trehalose-6,6'-dibehenate Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](COC(=O)CCCCCCCCCCCCCCCCCCCCC)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](COC(=O)CCCCCCCCCCCCCCCCCCCCC)O1 ZLJJDBSDZSZVTF-LXOQPCSCSA-N 0.000 description 1
- 102000007537 Type II DNA Topoisomerases Human genes 0.000 description 1
- 108010046308 Type II DNA Topoisomerases Proteins 0.000 description 1
- 229910052770 Uranium Inorganic materials 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 210000000683 abdominal cavity Anatomy 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 238000011481 absorbance measurement Methods 0.000 description 1
- 235000011054 acetic acid Nutrition 0.000 description 1
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 238000005377 adsorption chromatography Methods 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 229910052783 alkali metal Inorganic materials 0.000 description 1
- 150000001340 alkali metals Chemical class 0.000 description 1
- 229910052784 alkaline earth metal Inorganic materials 0.000 description 1
- 150000001342 alkaline earth metals Chemical class 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- BJEPYKJPYRNKOW-UHFFFAOYSA-N alpha-hydroxysuccinic acid Natural products OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 1
- 229940037003 alum Drugs 0.000 description 1
- AZDRQVAHHNSJOQ-UHFFFAOYSA-N alumane Chemical class [AlH3] AZDRQVAHHNSJOQ-UHFFFAOYSA-N 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- ILRRQNADMUWWFW-UHFFFAOYSA-K aluminium phosphate Chemical compound O1[Al]2OP1(=O)O2 ILRRQNADMUWWFW-UHFFFAOYSA-K 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 150000001412 amines Chemical class 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229960003896 aminopterin Drugs 0.000 description 1
- 238000012870 ammonium sulfate precipitation Methods 0.000 description 1
- 229960000723 ampicillin Drugs 0.000 description 1
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 1
- 239000003708 ampul Substances 0.000 description 1
- 229940035676 analgesics Drugs 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 239000000730 antalgic agent Substances 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 210000000628 antibody-producing cell Anatomy 0.000 description 1
- 230000005975 antitumor immune response Effects 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 238000003149 assay kit Methods 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000003190 augmentative effect Effects 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 238000001815 biotherapy Methods 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000004067 bulking agent Substances 0.000 description 1
- 229960003150 bupivacaine Drugs 0.000 description 1
- KDYFGRWQOYBRFD-NUQCWPJISA-N butanedioic acid Chemical compound O[14C](=O)CC[14C](O)=O KDYFGRWQOYBRFD-NUQCWPJISA-N 0.000 description 1
- 210000004900 c-terminal fragment Anatomy 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 230000005880 cancer cell killing Effects 0.000 description 1
- 230000009702 cancer cell proliferation Effects 0.000 description 1
- 239000003560 cancer drug Substances 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 150000001732 carboxylic acid derivatives Chemical class 0.000 description 1
- 108700021031 cdc Genes Proteins 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000007969 cellular immunity Effects 0.000 description 1
- 210000002230 centromere Anatomy 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 229960005091 chloramphenicol Drugs 0.000 description 1
- WIIZWVCIJKGZOK-RKDXNWHRSA-N chloramphenicol Chemical compound ClC(Cl)C(=O)N[C@H](CO)[C@H](O)C1=CC=C([N+]([O-])=O)C=C1 WIIZWVCIJKGZOK-RKDXNWHRSA-N 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 238000010668 complexation reaction Methods 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- NKLPQNGYXWVELD-UHFFFAOYSA-M coomassie brilliant blue Chemical compound [Na+].C1=CC(OCC)=CC=C1NC1=CC=C(C(=C2C=CC(C=C2)=[N+](CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=2C=CC(=CC=2)N(CC)CC=2C=C(C=CC=2)S([O-])(=O)=O)C=C1 NKLPQNGYXWVELD-UHFFFAOYSA-M 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 230000000254 damaging effect Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 239000010432 diamond Substances 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- PMMYEEVYMWASQN-UHFFFAOYSA-N dl-hydroxyproline Natural products OC1C[NH2+]C(C([O-])=O)C1 PMMYEEVYMWASQN-UHFFFAOYSA-N 0.000 description 1
- PRAKJMSDJKAYCZ-UHFFFAOYSA-N dodecahydrosqualene Natural products CC(C)CCCC(C)CCCC(C)CCCCC(C)CCCC(C)CCCC(C)C PRAKJMSDJKAYCZ-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 239000002158 endotoxin Substances 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 239000012997 ficoll-paque Substances 0.000 description 1
- 238000011049 filling Methods 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 235000011087 fumaric acid Nutrition 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 238000011223 gene expression profiling Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 230000005484 gravity Effects 0.000 description 1
- 210000004013 groin Anatomy 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- FAHBNUUHRFUEAI-UHFFFAOYSA-M hydroxidooxidoaluminium Chemical compound O[Al]=O FAHBNUUHRFUEAI-UHFFFAOYSA-M 0.000 description 1
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 229960002751 imiquimod Drugs 0.000 description 1
- DOUYETYNHWVLEO-UHFFFAOYSA-N imiquimod Chemical compound C1=CC=CC2=C3N(CC(C)C)C=NC3=C(N)N=C21 DOUYETYNHWVLEO-UHFFFAOYSA-N 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 238000003317 immunochromatography Methods 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 229940027941 immunoglobulin g Drugs 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 230000003308 immunostimulating effect 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
- 238000010348 incorporation Methods 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 229960000318 kanamycin Drugs 0.000 description 1
- 229930027917 kanamycin Natural products 0.000 description 1
- SBUJHOSQTJFQJX-NOAMYHISSA-N kanamycin Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CN)O[C@@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](N)[C@H](O)[C@@H](CO)O2)O)[C@H](N)C[C@@H]1N SBUJHOSQTJFQJX-NOAMYHISSA-N 0.000 description 1
- 229930182823 kanamycin A Natural products 0.000 description 1
- 210000003292 kidney cell Anatomy 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- CSSYQJWUGATIHM-IKGCZBKSSA-N l-phenylalanyl-l-lysyl-l-cysteinyl-l-arginyl-l-arginyl-l-tryptophyl-l-glutaminyl-l-tryptophyl-l-arginyl-l-methionyl-l-lysyl-l-lysyl-l-leucylglycyl-l-alanyl-l-prolyl-l-seryl-l-isoleucyl-l-threonyl-l-cysteinyl-l-valyl-l-arginyl-l-arginyl-l-alanyl-l-phenylal Chemical compound C([C@H](N)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CC(C)C)C(=O)NCC(=O)N[C@@H](C)C(=O)N1CCC[C@H]1C(=O)N[C@@H](CO)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CS)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)C1=CC=CC=C1 CSSYQJWUGATIHM-IKGCZBKSSA-N 0.000 description 1
- 101150066555 lacZ gene Proteins 0.000 description 1
- 229940078795 lactoferrin Drugs 0.000 description 1
- 235000021242 lactoferrin Nutrition 0.000 description 1
- GZQKNULLWNGMCW-PWQABINMSA-N lipid A (E. coli) Chemical compound O1[C@H](CO)[C@@H](OP(O)(O)=O)[C@H](OC(=O)C[C@@H](CCCCCCCCCCC)OC(=O)CCCCCCCCCCCCC)[C@@H](NC(=O)C[C@@H](CCCCCCCCCCC)OC(=O)CCCCCCCCCCC)[C@@H]1OC[C@@H]1[C@@H](O)[C@H](OC(=O)C[C@H](O)CCCCCCCCCCC)[C@@H](NC(=O)C[C@H](O)CCCCCCCCCCC)[C@@H](OP(O)(O)=O)O1 GZQKNULLWNGMCW-PWQABINMSA-N 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 229920006008 lipopolysaccharide Polymers 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 239000007791 liquid phase Substances 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- VZCYOOQTPOCHFL-UPHRSURJSA-N maleic acid Chemical compound OC(=O)\C=C/C(O)=O VZCYOOQTPOCHFL-UPHRSURJSA-N 0.000 description 1
- 239000011976 maleic acid Substances 0.000 description 1
- 239000001630 malic acid Substances 0.000 description 1
- 235000011090 malic acid Nutrition 0.000 description 1
- 229910052748 manganese Inorganic materials 0.000 description 1
- 239000011572 manganese Substances 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 150000002739 metals Chemical class 0.000 description 1
- 230000001394 metastastic effect Effects 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- 229940098779 methanesulfonic acid Drugs 0.000 description 1
- LSDPWZHWYPCBBB-UHFFFAOYSA-O methylsulfide anion Chemical compound [SH2+]C LSDPWZHWYPCBBB-UHFFFAOYSA-O 0.000 description 1
- 239000000693 micelle Substances 0.000 description 1
- 230000005486 microgravity Effects 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 230000000394 mitotic effect Effects 0.000 description 1
- 238000002715 modification method Methods 0.000 description 1
- 102000035118 modified proteins Human genes 0.000 description 1
- 108091005573 modified proteins Proteins 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 239000000178 monomer Substances 0.000 description 1
- 210000002433 mononuclear leukocyte Anatomy 0.000 description 1
- 210000004898 n-terminal fragment Anatomy 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 229910017604 nitric acid Inorganic materials 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 229940046166 oligodeoxynucleotide Drugs 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 235000006408 oxalic acid Nutrition 0.000 description 1
- IPCSVZSSVZVIGE-UHFFFAOYSA-N palmitic acid group Chemical group C(CCCCCCCCCCCCCCC)(=O)O IPCSVZSSVZVIGE-UHFFFAOYSA-N 0.000 description 1
- 229940055729 papain Drugs 0.000 description 1
- 235000019834 papain Nutrition 0.000 description 1
- 101150040383 pel2 gene Proteins 0.000 description 1
- 101150050446 pelB gene Proteins 0.000 description 1
- NRNCYVBFPDDJNE-UHFFFAOYSA-N pemoline Chemical compound O1C(N)=NC(=O)C1C1=CC=CC=C1 NRNCYVBFPDDJNE-UHFFFAOYSA-N 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 229940023041 peptide vaccine Drugs 0.000 description 1
- 210000001322 periplasm Anatomy 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- BZQFBWGGLXLEPQ-REOHCLBHSA-N phosphoserine Chemical compound OC(=O)[C@@H](N)COP(O)(O)=O BZQFBWGGLXLEPQ-REOHCLBHSA-N 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 229920001606 poly(lactic acid-co-glycolic acid) Polymers 0.000 description 1
- 238000007694 polyacrylamide gel isoelectric focusing Methods 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 238000009117 preventive therapy Methods 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000009862 primary prevention Effects 0.000 description 1
- 235000019260 propionic acid Nutrition 0.000 description 1
- 108010031970 prostasin Proteins 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 238000012514 protein characterization Methods 0.000 description 1
- 238000001742 protein purification Methods 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 239000001397 quillaja saponaria molina bark Substances 0.000 description 1
- IUVKMZGDUIUOCP-BTNSXGMBSA-N quinbolone Chemical compound O([C@H]1CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)C=C4CC3)C)CC[C@@]21C)C1=CCCC1 IUVKMZGDUIUOCP-BTNSXGMBSA-N 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 208000015347 renal cell adenocarcinoma Diseases 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 238000004366 reverse phase liquid chromatography Methods 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 238000004007 reversed phase HPLC Methods 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 238000005185 salting out Methods 0.000 description 1
- 229930182490 saponin Natural products 0.000 description 1
- 150000007949 saponins Chemical class 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 230000007480 spreading Effects 0.000 description 1
- 238000003892 spreading Methods 0.000 description 1
- 229940031439 squalene Drugs 0.000 description 1
- TUHBEKDERLKLEC-UHFFFAOYSA-N squalene Natural products CC(=CCCC(=CCCC(=CCCC=C(/C)CCC=C(/C)CC=C(C)C)C)C)C TUHBEKDERLKLEC-UHFFFAOYSA-N 0.000 description 1
- 238000003756 stirring Methods 0.000 description 1
- 238000005728 strengthening Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 125000004434 sulfur atom Chemical group 0.000 description 1
- 229920001059 synthetic polymer Polymers 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 210000001550 testis Anatomy 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 229940126622 therapeutic monoclonal antibody Drugs 0.000 description 1
- 229940104230 thymidine Drugs 0.000 description 1
- FGMPLJWBKKVCDB-UHFFFAOYSA-N trans-L-hydroxy-proline Natural products ON1CCCC1C(O)=O FGMPLJWBKKVCDB-UHFFFAOYSA-N 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000001131 transforming effect Effects 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 230000004614 tumor growth Effects 0.000 description 1
- 238000000108 ultra-filtration Methods 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 229910052720 vanadium Inorganic materials 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 230000003245 working effect Effects 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4748—Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/15—Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/17—Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/04—Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
- A61K38/08—Peptides having 5 to 11 amino acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
- A61K39/001148—Regulators of development
- A61K39/001149—Cell cycle regulated proteins, e.g. cyclin, CDC, CDK or INK-CCR
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- 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
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4615—Dendritic cells
-
- 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
- A61K39/46—Cellular immunotherapy
- A61K39/462—Cellular immunotherapy characterized by the effect or the function of the cells
- A61K39/4622—Antigen presenting cells
-
- 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
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464448—Regulators of development
- A61K39/464449—Cell cycle regulated proteins, e.g. cyclin, CDC, CDK or INK-CCR
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/04—Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/16—Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/08—Drugs for disorders of the urinary system of the prostate
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/10—Drugs for disorders of the urinary system of the bladder
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/12—Drugs for disorders of the urinary system of the kidneys
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P15/00—Drugs for genital or sexual disorders; Contraceptives
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P21/00—Drugs for disorders of the muscular or neuromuscular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/04—Immunostimulants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70539—MHC-molecules, e.g. HLA-molecules
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K7/00—Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
- C07K7/04—Linear peptides containing only normal peptide links
- C07K7/06—Linear peptides containing only normal peptide links having 5 to 11 amino acids
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K7/00—Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
- C07K7/04—Linear peptides containing only normal peptide links
- C07K7/08—Linear peptides containing only normal peptide links having 12 to 20 amino acids
-
- 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
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0639—Dendritic cells, e.g. Langherhans cells in the epidermis
-
- 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
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/10—Cells modified by introduction of foreign genetic material
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/02—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5044—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
- G01N33/5047—Cells of the immune system
- G01N33/505—Cells of the immune system involving T-cells
-
- 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
- C12N2502/00—Coculture with; Conditioned medium produced by
- C12N2502/11—Coculture with; Conditioned medium produced by blood or immune system cells
- C12N2502/1114—T cells
-
- 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
- C12N2510/00—Genetically modified cells
Definitions
- the present invention relates to the field of biological science, more specifically to the field of cancer therapy.
- the present invention relates to novel peptides that are effective as cancer vaccines, methods for either or both of treating and preventing cancers using the peptide(s), and pharmaceutical compositions comprising the peptide(s).
- TTLs Cytotoxic T lymphocytes
- TAAs tumor-associated antigens
- MHC major histocompatibility complex
- TAAs tumor-associated antigens
- MAGE major histocompatibility complex
- NPL3 Harris C C, J Natl Cancer Inst 1996, 88(20): 1442-55; NPL4: Butterfield L H et al., Cancer Res 1999, 59(13): 3134-42; NPLS: Vissers J L et al., Cancer Res 1999, 59(21): 5554-9; NPL6: van der Burg S H et al., J Immunol 1996, 156(9): 3308-14; NPL7: Tanaka F et al., Cancer Res 1997, 57(20): 4465-8; NPL8: Fujie T et al., Int J Cancer 1999, 80(2): 169-72; NPL9: Kikuchi M et al., Int J Cancer 1999, 81(3): 459-66; NPL10: Oiso M et al., Int J Cancer 1999,
- CDCA1 cell division cycle associated 1; also described as NUF2, NDC80 kinetochore complex component: Nuf2; reference sequence: GeneBank Accession Number NM_145697 (SEQ ID NO: 44) or GeneBank Accession Number NM_031423 (SEQ ID NO: 46)
- CDC2 reference sequence: GeneBank Accession Number NM_145697 (SEQ ID NO: 44) or GeneBank Accession Number NM_031423 (SEQ ID NO: 46)
- NPL14 Walker et al., Curr Cancer Drug Targets 2001, 1(1): 73-83.
- CDCA1 has been found to be related to the centromere of HeLa cells undergoing mitotic division, and is considered to be a functional homologue of yeast Nuf2 (NPL15: Wigge P A et al., J Cell Biol 2001, 152(2): 349-60). Meanwhile, CDCA1 has been identified as a gene showing elevated expression in non-small cell lung cancer by gene expression profiling based on a genome-wide cDNA microarray targeting 27648 genes (NPL16: Hayama et al., Cancer Res 2006, 66(21): 10339-48; PTL1: WO2007/013480; PTL2: WO2005/089735).
- the CDCA1 expression is observed in lung cancer tissues and lung cancer cell lines, but little expression is observed in 22 normal tissues except the testis (NPL16; PTL1). Further, as a result of siRNA-mediated suppression of the CDCA1 expression, suppression of cell proliferation in CDCA1-expressing lung cancer cell lines is caused (NPL16; PTLs 1-2). Moreover, elevated expression of CDCA1 is also observed in various cancers such as cholangiocellular cancer, bladder cancer and renal cell cancer (NPL17: Harao M et al., Int J Cancer 2008, 123(11): 2616-25).
- CDCA1-derived HLA-A02-restricted CTL epitope peptides (NPL17: Harao et al., Int J Cancer. 2008, 123(11): 2616-25; PTL3: WO2009/025117), HLA-A24-restricted CTL epitope peptides (PTL4: WO2009/153992), HLA-Al 1-restricted CTL epitope peptides (PTL5: WO2016/021508), HLA-A33-restricted CTL epitope peptides (PTL5: WO2016/021508) and HLA-A03-restricted CTL epitope peptides (PTL5: WO2016/021508) have been identified.
- Therapeutic effects by these peptides are expected in cancer patients having the HLA-A02 type, HLA-A24 type, HLA-Al 1 type, HLA-A33 type or HLA-A03 type, but for cancer patients having HLA types other than those, peptides corresponding to respective HLA type are desired.
- the present invention relates to peptides that can induce cytotoxic T cells (CTLs) specifically responding to CDCA1-expressing cells.
- CTLs cytotoxic T cells
- APCs antigen-presenting cells
- HLA human leukocyte antigen
- CD8-positive T cells CD8-positive T cells
- CDCA1-derived peptides that have been identified so far to have CTL-inducing ability (CTL inducibility) are HLA-A02-restricted, HLA-A24-restricted, HLA-A11-restricted, HLA-A33-restricted, or HLA-A03-restricted peptides, and when antigen-presenting cells do not express these HLAs, the peptides cannot induce CTLs. Therefore, conventional peptides are not suitable for performing immunotherapy on cancer patients (subjects) that do not have these HLAs.
- HLA-A01 is an HLA allele highly frequently observed in Caucasians (Cao K et al., Hum Immunol 2001, 62(9): 1009-30). It is desirable to administer HLA-A01-restricted peptides to HLA-A01-positive cancer patients.
- the present invention relates to CDCA1l-derived peptides with CTL-inducing ability that are restrictive to HLA-A01. Based on results disclosed herein, the peptides of the present invention have been proven to be epitope peptides that can induce a potent and specific immune response against cancer cells expressing CDCA1 and HLA A01.
- one of the objectives of the present invention is to provide CDCA1-derived peptides that can induce CTLs in an HLA-A01-restrictive manner. These peptides can be used to induce CTLs in vitro, ex vivo or in vivo, or can be used to administer to subjects for the purpose of inducing an immune response against CDCA1-expressing cancer cells.
- Preferable peptides are peptides comprising the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37; more preferable peptides are nonapeptides or decapeptides; and even more preferable peptides are peptides consisting of the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- the peptides of the present invention encompass peptides in which one, two, or more amino acid(s) is/are substituted, deleted, inserted and/or added, as long as the resultant modified peptides retain the CTL-inducing ability of the original peptide.
- the present invention further provides isolated polynucleotides encoding any one of the peptides of the present invention. Similar to the peptides of the present invention, these polynucleotides can be used for inducing APCs with CTL-inducing ability, and can be administered to subjects for inducing an immune response against CDCA1-expressing cancer cells.
- the present invention also provides compositions comprising one or more types of peptides of the present invention, one or more types of polynucleotides encoding one or more types of peptides of the present invention, APCs of the present invention, exosomes presenting peptides of the present invention, and/or CTLs of the present invention.
- the compositions of the present invention are preferably pharmaceutical compositions.
- the pharmaceutical compositions of the present invention can be used for treating and/or preventing cancer, as well as preventing postoperative recunence thereof. They can also be used for inducing an immune response against cancer. When administered to a subject, a peptide of the present invention is presented on the surface of an APC, and as a result CTLs targeting the peptide are induced.
- compositions for inducing CTLs wherein the compositions comprise one or more types of peptides of the present invention, one or more types of polynucleotides encoding one or more types of peptides of the present invention, APCs of the present invention, and/or exosomes presenting peptides of the present invention.
- a further objective of the present invention is to provide methods of inducing APCs having CTL-inducing ability, wherein the methods comprise a step of contacting one or more types of peptides of the present invention with an APC, or a step of introducing a polynucleotide encoding any one peptide of the present invention into an APC.
- the present invention further provides a method of inducing CTLs, comprising a step of co-culturing a CD8-positive T cell with an APC that presents on its surface a complex of an HLA antigen and a peptide of the present invention, a step of co-culturing a CDR-positive T cell with an exosome that presents on its surface a complex of an HLA antigen and a peptide of the present invention, or a step of introducing into a CD8-positive T cell a vector comprising a polynucleotide encoding each subunit of a T cell receptor (TCR) capable of binding to a peptide of the present invention presented by an HLA antigen on a cell surface.
- TCR T cell receptor
- a further objective of the present invention is to provide isolated APCs that present on their surface a complex of an HLA antigen and a peptide of the present invention.
- the present invention further provides isolated CTLs targeting a peptide of the present invention. These APCs and CTLs can be used in immunotherapy for CDCA1-expressing cancers.
- Another objective of the present invention is to provide methods of inducing an immune response against cancer in a subject, wherein the methods comprise a step of administering to the subject a composition(s) comprising a peptide(s) of the present invention or a polynucleotide(s) encoding the peptide(s), an APC(s) of the present invention, an exosome(s) presenting a peptide(s) of the present invention, and/or a CTL(s) of the present invention.
- Another objective of the present invention is to provide methods of treating and/or preventing cancer, as well as preventing postoperative recurrence thereof in a subject, wherein the methods comprise a step of administering to the subject a peptide(s) of the present invention, a polynucleotide(s) encoding the peptide(s), an APC(s) of the present invention, an exosome(s) presenting a peptide(s) of the present invention, and/or a CTL(s) of the present invention.
- FIG. 1 is comprised of photos (a) to (j) showing the results of the IFN-gamma enzyme-linked immunospot (ELISPOT) assay performed using cells induced using peptides derived from CDCA1.
- ELISPOT enzyme-linked immunospot
- “+” shows IFN-gamma production against target cells pulsed with a peptide of interest
- “ ⁇ ” shows IFN-gamma production against target cells that have not been pulsed with any peptides (negative target).
- peptide-specific IFN-gamma production was observed in:
- CDCA1-A01-10-66 SEQ ID NO: 9 (j) is shown as an example of typical negative data where peptide-specific IFN-gamma production was not observed.
- FIG. 2 is comprised of a series of line graphs (a) to (c) showing IFN-gamma production of CTL clones established by limiting dilution method following induction by CDCA1-A01-10-136 (SEQ ID NO: 8), CDCA1-A01-10-56 (SEQ ID NO: 10), or CDCA1-A01-10-48 (SEQ ID NO: 13). These results show peptide-specific IFN-gamma production of the CTL clones.
- FIG. 3 is a line graph showing IFN-gamma production of a CTL clone against target cells expressing both CDCA1 and HLA-A*01:01.
- COS7 cells expressing either HLA-A*01:01 or the full-length CDCA1 gene served as a negative control.
- the CTL clone established following induction by CDCA1-A01-10-136 (SEQ ID NO: 8) showed IFN-gamma production in COS7 cells into which both CDCA1 and HLA-A*01:01 genes were introduced (black diamond). Meanwhile, it did not show significant IFN-gamma production in COS7 cells into which either HLA-A*01:01 (triangle) or CDCA1 (white circle) was introduced.
- isolated and purified used in relation with a substance indicate that the substance does not substantially contain at least one substance that may else be included in a natural source.
- an isolated or purified peptide refers to a peptide that does not substantially contain another cellular material, for example, carbohydrate, lipid and other contaminating proteins from the cell or tissue source from which the peptide is derived.
- an isolated or purified peptide refers to a peptide that does not substantially contain a precursor substance or another chemical substance.
- a peptide that does not substantially contain a cellular material encompasses peptide preparations that contain less than about 30%, 20%, 10%, or 5%, 3%, 2% or 1% (dry weight basis) of other cellular materials.
- an isolated or purified peptide does not substantially contain culture medium, which encompasses peptide preparations that contain culture medium less than about 20%, 10%, or 5%, 3%, 2% or 1% (dry weight basis) of the volume of the peptide preparation.
- an isolated or purified peptide does not substantially contain a precursor substance or other chemical substances, which encompasses peptide preparations that contain a precursor substance or other chemical substances less than about 30%, 20%, 10%, 5%, 3%, 2% or 1% (dry weight basis) of the volume of the peptide preparation. That a particular peptide preparation is an isolated or purified peptide can be confirmed, for example, by the appearance of a single band following sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis and Coomassie Brilliant Blue staining or such of the gel.
- SDS sodium dodecyl sulfate
- the peptides and polynucleotides of the present invention are isolated or purified.
- polypeptide refers to polymers of amino acid residues. These terms are applied to also non-naturally occurring amino acid polymers comprising one or more non-naturally occurring amino acid residues, in addition to naturally occurring amino acid polymers.
- Non-naturally occurring amino acids include amino acid analogs, amino acid mimetics, and such.
- amino acid refers to naturally occurring amino acids, as well as amino acid analogs and amino acid mimetics that functions similarly to the naturally occurring amino acids.
- Naturally occurring amino acids are those encoded by the genetic code, as well as those modified after translation in cells (e.g., hydroxyproline, gamma-carboxygiutamate, and O-phosphoserine, etc.).
- amino acid analog refers to compounds that have the same basic chemical structure (an alpha carbon bound to a hydrogen, a carboxy group, an amino group, and an R group) as a naturally occurring amino acid but have a modified R group or modified backbones (e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, and such).
- modified R group or modified backbones e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, and such.
- amino acid mimetic refers to chemical compounds that have different structures but similar functions to general amino acids. Amino acids can be either L-amino acids or D-amino acids, and the peptides of the present invention are preferably L-amino acid polymers.
- polynucleotide refers to a polymer of nucleotides.
- composition used in the present specification is intended to encompass products that include specified ingredients in specified amounts, and any products generated directly or indirectly from combination of specified ingredients in the specified amounts.
- composition is intended to encompass products including active ingredient(s) and inert ingredient(s), as well as any products generated directly or indirectly from combination, complexation or aggregation of any two or more ingredients, from dissociation of one or more ingredients, or from other types of reactions or interactions of one or more ingredients.
- pharmaceutical compositions of the present invention encompass any compositions made by admixing compounds or cells of the present invention with a pharmaceutically or physiologically acceptable carrier.
- pharmaceutically acceptable carrier or “physiologically acceptable carrier” used in the present specification include liquid or solid bulking agents, diluents, excipients, solvents, and encapsulation materials; and mean pharmaceutically or physiologically acceptable materials, compositions, substances or media.
- the term “cancer” refers to a cancer that overexpresses the CDCA1 gene; and examples thereof include bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, colon cancer and such, without being limited thereto.
- the “cancer” is a cancer that expresses CDCA1 and HLA-A01.
- cytotoxic T lymphocyte and “cytotoxic T cell” and “CTL” are used interchangeably herein. Unless otherwise specifically indicated, they refer to a sub-group of T lymphocytes that can recognize non-self cells (for example, tumor/cancer cells, virus-infected cells) and induce the death of such cells.
- non-self cells for example, tumor/cancer cells, virus-infected cells
- HLA-A01 (HLA-A1) refers to the HLA-A01 type which includes subtypes such as HLA-A*01:01, HLA-A*01:02, HLA-A*01:03, and HLA-A*01:04.
- HLA antigen of a subject is HLA-A01
- HLA-A01 refers to that a subject or patient has the HLA-A01 antigen gene homozygously or heterozygously as the MHC (Major Histocompatibility Complex) Class I molecule, and that the HLA-A01 antigen is expressed in the cells of the subject or patient as the HLA antigen.
- the treatment is considered “efficacious” when it achieves clinical advantages, for example, reduction in the size, spreading or metastatic ability of cancer, retardation of cancer progression, alleviation of clinical symptoms of cancer, prolongation of survival period, suppression of postoperative recurrence in a subject.
- “efficacious” means that the treatment retards or prevents cancer formation, or prevents or alleviates clinical symptoms of cancer. Effectiveness is determined in relation to any publicly known method for diagnosing or treating a specific tumor type.
- prevention includes any work that eases the load of disease-associated mortality or morbidity.
- Prevention can be carried out at the “primary, secondary and tertiary prevention (prophylaxis) levels”. Whereas the primary prevention (prophylaxis) avoids the development of a disease, prevention (prophylaxis) at the secondary and tertiary levels encompasses prevention (prophylaxis) of disease progression and appearance of symptoms, as well as workings intended to reduce adverse effects of the existing disease by restoring functions and reducing disease-associated complications. Alternately, prevention (prophylaxis) can include alleviation of severity of a specific disorder, for example, extensive preventive therapy intended to reduce tumor growth and metastasis.
- the treatment and/or prevention (prophylaxis) of cancer and/or prevention (prophylaxis) of postoperative recurrence thereof include either of the events such as inhibition of cancer cell proliferation, tumor involution or regression, induction of remission and suppression of cancer development, tumor regression, as well as reduction or inhibition of metastasis, suppression of postoperative recurrence of cancer, and prolongation of survival period.
- Effective treatment and/or prevention (prophylaxis) of cancer reduce mortality, improve prognosis of an individual with cancer, reduce the blood levels of tumor markers, and alleviate detectable symptoms associated with cancer.
- alleviation or improvement of symptoms constitutes effective treatment and/or prevention (prophylaxis), and includes a condition in which the symptoms are alleviated or stable by 10%, 20%, 30% or more.
- an antibody refers to immunoglobulins and fragments thereof that are specifically reactive to a designated protein or peptide thereof.
- An antibody can include human antibodies, primatized antibodies, chimeric antibodies, bispecific antibodies, humanized antibodies, antibodies fused to other proteins or radiolabels, and antibody fragments.
- an “antibody” herein is used in the broadest sense and specifically covers intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies) formed from two or more intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
- An “antibody” may be antibodies of all classes (e.g., IgA, IgD, IgE, IgG and IgM).
- HLA-A01 is an HLA allele commonly seen in Caucasians (Cao et al., Hum Immunol 2001, 62(9): 1009-30).
- an effective method of treating CDCA1-expressing cancers for a great population of Caucasians can be provided by providing CDCA1-derived CTL-inducing peptides restricted to HLA-A01.
- the present invention provides CDCA1-derived peptides that are capable of inducing CTLs in an HLA-A01-restrictive manner.
- the peptides of the present invention are CDCA1-derived peptides that are capable of inducing CTLs in an HLA-A01-restrictive manner
- Peptides capable of inducing CTLs in an HLA-A01-restrictive manner include peptides having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- CTLs having a cytotoxic activity specific to these peptides can be established by in vitro stimulation of T cells by dendritic cells pulsed with these peptides.
- the established CTLs show a specific cytotoxic activity against target cells pulsed with each of the peptides.
- the CDCA1 gene is overexpressed in cancer cells such as cancer cells in, for example, bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, colon cancer and such, but is not expressed in most normal organs. It is thus an excellent target for immunotherapy. Therefore, the peptides of the present invention can be suitably used for cancer immunotherapy.
- cancer cells such as cancer cells in, for example, bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, colon cancer and such, but is not expressed in most normal organs. It
- a preferred peptide is a nonapeptide (a peptide consisting of 9 amino acid residues) or a decapeptide (a peptide consisting of 10 amino acid residues), and it is more preferably a peptide consisting of the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- a peptide having the amino acid sequence of SEQ ID NO: 8 is suitable for induction of CTLs that show a specific cytotoxic activity against cells expressing HLA-A01 and CDCA1, and can be suitably used for cancer immunotherapy in HLA-A01-positive patients.
- the peptide of the present invention is a peptide consisting of the amino acid sequence of SEQ ID NO: 8.
- an additional amino acid residue(s) can be made to adjoin the amino acid sequence of the peptide of the present invention, as long as the resultant peptides retain the CTL-inducing ability of the original peptide.
- the additional amino acid residue(s) may be composed of any types of amino acid(s), as long as they do not impair the CTL-inducing ability of the original peptide. Therefore, the peptides of the present invention encompass peptides having CTL-inducing ability, comprising the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- Such peptides are, for example, less than about 40 amino acids, in many cases less than about 20 amino acids, and usually less than about 15 amino acids. Therefore, if the original peptide is a nonapeptide, the peptide of the present invention encompasses peptides that are 10 amino-acid long or 11-40 amino-acid long, which are produced by adjoining additional amino acid(s) to the peptide. Furthermore, if the original peptide is a decapeptide, the peptide of the present invention encompasses peptides that are 11-40 amino-acid long. Such a peptide can be, for example, a peptide that is 11-20 amino-acid long or a peptide that is 11-15 amino-acid long.
- an additional amino acid residue(s) is an amino acid residue(s) adjacent to the amino acid sequence of the peptide of the present invention in the full-length amino acid sequence of CDCA1 (for example, SEQ ID NO: 64). Therefore, the peptides of the present invention encompass peptides comprising the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37, and wherein the peptides are peptide fragments of CDCA1 and have CTL-inducing ability.
- modified peptides i.e., peptides composed of the amino acid sequence in which one, two or several amino acid residues are modified (i.e., substituted, deleted, inserted, and/or added) compared to the original reference sequence
- modified peptides are known to retain the biological activity of the original peptide (Mark et al., Proc Natl Acad Sci USA 1984, 81: 5662-6; Zoller and Smith, Nucleic Acids Res 1982, 10: 6487-500; Dalbadie-McFarland et al., Proc Nati Acad Sci USA 1982, 79: 6409-13).
- the peptides of the present invention can be peptides comprising the amino acid sequence in which one, two or several amino acids are substituted, deleted, inserted and/or added to the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 and having CTL-inducing ability.
- amino acid side chain characteristics that functionally resemble include, for example, hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T), and side chains having the following functional groups or characteristics in common: an aliphatic side-chain (G, A, V, L, I, P); a hydroxyl group containing side-chain (S, T, Y); a sulfur atom containing side-chain (C, M); a carboxylic acid and amide containing side-chain (D, N, E, Q); a base containing side-chain (R, K, H); and an aromatic containing side-chain (H, F, Y, W).
- the following eight groups each contain amino acids that are accepted in the art as conservative substitutions for one another:
- peptides of the present invention are also encompassed in peptides of the present invention.
- peptides of the present invention are not restricted thereto and can include non-conservative modifications, so long as the modified peptide retains the CTL-inducing ability of the original peptide.
- modified peptides do not exclude CTL inducible peptides derived from polymorphic variants, interspecies homologues, and alleles of CDCA1.
- a small number for example, 1, 2 or several
- a small percentage of amino acids for example, 1, 2 or several
- the term “several” means 5 or fewer amino acids, for example, 4 or 3 or fewer.
- the percentage of amino acids to be modified is preferably 20% or less, more preferably 15% or less, even more preferably 10% or less or 1 to 5%.
- peptides of the present invention are presented on the surface of a cell or exosome, preferably as a complex with an HLA antigen. Therefore, it is preferable that the peptides of the present invention possess high binding affinity to the HLA antigen. To that end, the peptides can be modified by substitution, deletion, insertion, and/or addition of the amino acid residues to yield a modified peptide having improved binding affinity.
- the second amino acid from the N terminus and the C-terminal amino acid are generally anchor residues involved in the binding to HLA Class I (Rainmensee HG, et al., Immunogenetics. 1995, 41(4): 178-228.).
- aspartic acid and glutamic acid for the third amino acid from the N terminus and tyrosine for the C-terminal amino acid are known as anchor residues with high binding affinity for HLA-A01.
- HLA-A01 there is auxiliary anchor residues at position 2 from the N terminus; and it is known that threonine and serine are preferred as the second amino acid from the N terminus (Kubo, R. T Journal of Immunology 1994, 152: 3913-24; Gambacorti-Passerini, C. Clinical Cancer Research 1997, 3: 675-83; Falk, K. Immunogenetics 1994, 40: 238-41).
- threonine and serine are preferred as the second amino acid from the N terminus.
- peptides with CTL-inducing ability comprising an amino acid sequence in which, in the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37, the second amino acid from the N terminus is substituted with threonine or serine; the third amino acid from the N terminus is substituted with aspartic acid or glutamic acid; and/or the C-terminal amino acid is substituted with tyrosine are encompassed by the peptides of the present invention.
- the peptide of the present invention can be a peptide having CTL-inducing ability that consists of an amino acid sequence in which, in the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37, the second amino acid from the N terminus is substituted with threonine or serine; the third amino acid from the N terminus is substituted with aspartic acid or glutamic acid; and/or the C-terminal amino acid is substituted with tyrosine.
- the peptides of the present invention encompass peptides having CTL-inducing ability, which comprise an amino acid sequence with one or more substitutions selected from (a) to (c) below in the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37:
- the peptide of the present invention may be a peptide having CTL-inducing ability that consists of an amino acid sequence in which one or more substitutions selected from (a) to (c) above are introduced into the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- the preferred number of substitutions is 1, 2 or 3 substitutions selected from (a) to (c) above.
- the peptide of the present invention may be a peptide having CTL-inducing ability, which comprises an amino acid sequence in which, in the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37, the third amino acid from the N terminus is substituted with aspartic acid or glutamic acid, and/or the C-terminal amino acid is substituted with tyrosine.
- the peptide of the present invention may be a peptide having CTL-inducing ability, which consists of an amino acid sequence in which, in the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37, the third amino acid from the N terminus is substituted with aspartic acid or glutamic acid, and/or the C-terminal amino acid is substituted with tyrosine.
- the peptide of the present invention may be a peptide having CTL-inducing ability, which comprises an amino acid sequence in which one or more substitutions selected from (a) and (b) below are introduced into the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37:
- the peptide of the present invention may be a peptide having CTL-inducing ability, which consists of an amino acid sequence in which one or more substitutions selected from (a) to (b) above are introduced into the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- Substitution(s) may be introduced into amino acid(s) not only at the anchor site(s), but also at a position(s) of potential T cell receptor (TCR) recognition site(s) of the peptides.
- TCR T cell receptor
- a peptide that has amino acid substitutions such as CAP1, p53 (264-272) , Her-2/neu (369-377) or gp100 (209-217) , may have equal to or better activity than that of the original peptide (Zaremba et al. Cancer Res. 1997, 57, 4570-7; T. K. Hoffmann et al. J Immunol. 2002, 168(3): 1338-47; S. O. Dionne et al. Cancer Immunol immunother. 2003, 52: 199-206; and S. 0. Dionne et al. Cancer Immunology, Immunotherapy 2004, 53, 307-14).
- the present invention also contemplates that one, two or several amino acids can be added to the N terminus and/or C terminus of the peptides of the present invention (for example, peptides consisting of the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37).
- modified peptides that retain CTL-inducing ability are also included in the present invention.
- a peptide in which one, two or several amino acids are added to the N terminus and/or C terminus of a peptide consisting of the amino acid sequence of SEQ ID NO: 8 is contacted with an APC(s), it is incorporated into the APC(s) and processed to become a peptide consisting of the amino acid sequence of SEQ ID NO: 8.
- peptides of the present invention can be peptides in which one, two or several amino acids are added to either or both of the N terminus and C terminus.
- peptides consisting of an amino acid sequence which comprises one, two, or several amino acid substitutions in the amino acid sequence referenced by each SEQ ID NO and which further comprises one, two, or several amino acids added to either or both of the N-terminus and the C-terminus of the substitution-bearing amino acid sequence are provided.
- a desired position for the substitution can be, for example, one, two, or three selected from position 2 from the N-terminus, position 3 from the N-terminus, and the C-terminus in the amino acid sequence referenced by SEQ ID NO: 1, 8, 10, 13, 25, 33 to 35 or 37 which is comprised in said peptide of the present invention.
- the amino acid sequence of a peptide is identical to a portion of the amino acid sequence of an endogenous or exogenous protein having a different function, side effects such as autoimmune disorders and/or allergic symptoms against specific substances may be induced. Therefore, it is preferable to perform homology searches using available databases to avoid situations in which the amino acid sequence of the peptide matches the amino acid sequence of another protein.
- the objective peptide can be modified in order to increase its binding affinity with HLA antigens, and/or increase its CTL-inducing ability without danger of such side effects.
- Peptides in which one, two or several amino acids of a peptide of the present invention are modified are predicted to be able to retain CTL-inducing ability of the original peptide; however, it is preferable to verify the CTL-inducing ability of the modified peptides.
- the “peptide having CTL-inducing ability (CTL inducibility)” refers to a peptide that induces CTLs through APCs stimulated with the peptide.
- CTL induction includes induction of differentiation into CTLs, induction of CTL activation, induction of CTL proliferation, induction of CTL's cytotoxic activity, induction of CTL-mediated dissolution of target cells, and induction of increase of IFN-gamma production of CTLs.
- the CTL-inducing ability can be confirmed by inducing and stimulating APCs that retain an HLA antigen (for example, B lymphocytes, macrophages, and dendritic cells) with a peptide, and mixing them with CD8-positive T cells; and then measuring IFN-gamma released by CTLs against the target cells.
- HLA antigen for example, B lymphocytes, macrophages, and dendritic cells
- human peripheral blood mononuclear leukocyte-derived dendritic cells can be preferably used.
- transgenic animals generated to express an HLA antigen can be used.
- the target cells may be radio-labelled with 51 Cr or such, and the cytotoxic activity of the peptide-induced CTLs may be calculated from the radioactivity emitted from the target cells.
- the peptides of the present invention can be linked to other peptides as long as the resultant linked peptide retains the CTL-inducing ability.
- An example of an appropriate peptide to be linked with the peptides of the present invention includes a TAA-derived CTL-inducing peptide.
- the peptides of the present invention can also be linked with each other. Suitable linkers for use in linking peptides are known in the art, and for example, linkers such as AAY (P. M. Daftarian et al., J Trans Med. 2007, 5: 26), AAA, NKRK (SEQ ID NO: 48) (R. P. M. Sutmuller et al., J Immunol.
- Peptides can be linked in various arrangements (for example, catenulate, repeated, etc.), and one can also link three or more peptides.
- the peptides of the present invention can also be linked to other substances as long as the resultant linked peptide retains the CTL-inducing ability.
- Examples of an appropriate substance to be linked with a peptide of the present invention include, for example, a peptide, a lipid, a sugar or sugar chain, an acetyl group, and a naturally-occurring or synthetic polymer.
- the peptides of the present invention can be modified by glycosylation, side-chain oxidation, phosphorylation or such, as long as their CTL-inducing ability is not impaired. One can also perform such types of modifications to confer additional functions (for example, targeting function and delivery function) or to stabilize the peptide.
- Peptide stability can be assayed by several methods. For example, stability can be tested by using a peptidase as well as various biological media such as human plasma and serum (see, e.g., Verhoef et al., Eur J Drug Metab Phannacokin 1986, 11: 291-302).
- the present invention also provides methods of screening for or selecting modified peptides that have equal to or higher activity than that of the original peptide.
- the present invention provides a method of screening for a peptide having CTL-inducing ability, wherein the method comprises the steps of:
- the APCs in step (c) above are desirably cells having HLA-A01.
- the peptide of the present invention is also described as a “CDCA1 peptide(s)” or a “CDCA1 polypeptide(s)”.
- peptides of the present invention can be synthesized individually, or as longer polypeptides including two or more peptides.
- Peptides of the present invention can be isolated from host cells or synthesis reaction products after they are produced in the host cells using recombinant DNA technology or after they are chemically synthesized. That is, peptides of the present invention can be purified or isolated so as not to substantially contain other host-cell proteins and fragments thereof, or any other chemical substances.
- the peptides of the present invention may contain modifications, such as glycosylation, side chain oxidation, or phosphorylation provided such modifications do not destroy the biological activity of the original peptide.
- modifications such as glycosylation, side chain oxidation, or phosphorylation provided such modifications do not destroy the biological activity of the original peptide.
- Other illustrative modifications include incorporation of D-amino acids or other amino acid mimetics that may be used, for example, to increase the serum half life of the peptides.
- a peptide of the present invention can be obtained through chemical synthesis based on the selected amino acid sequence.
- Examples of conventional peptide synthesis methods that can be adapted to the synthesis include the methods described in the documents below:
- the peptides of the present invention can be obtained by adapting any known genetic engineering methods for producing peptides (e.g., Morrison J, J Bacteriology 1977, 132: 349-51; Clark-Curtiss & Curtiss, Wu et al., Methods in Enzymology 1983, 101: 347-62).
- a suitable vector harboring a polynucleotide encoding the peptide of the present invention in an expressible form e.g., downstream of a regulatory sequence corresponding to a promoter sequence
- the host cell is then cultured to produce the peptide of the present invention.
- the peptide of the present invention can also be produced in vitro using an in vitro translation system.
- the present invention also provides a polynucleotide which encodes any of the peptides of the present invention.
- polynucleotides which include polynucleotides derived from the naturally occurring CDCA1 gene (e.g., GenBank Accession No. NM_145697 (SEQ ID NO: 44) or GenBank Accession No. NM_031423 (SEQ ID NO: 46)) as well as those having a conservatively modified nucleotide sequence thereof.
- the phrase “conservatively modified nucleotide sequence” refers to sequences which encode identical or essentially identical amino acid sequences. Due to the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein.
- the codons GCA, GCC, GCG, and GCU all encode the amino acid alanine.
- the codon can be altered to any of the corresponding codons described above without altering the encoded polypeptide.
- Such nucleic acid variations are “silent variations”, which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a peptide also describes every possible silent variation of the nucleic acid.
- each codon in a nucleic acid can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid that encodes a peptide is implicitly described in each disclosed sequence.
- the polynucleotide of the present invention can be composed of DNA, RNA, and derivatives thereof
- a DNA is suitably composed of bases such as A, T, C, and G, and T is replaced by U in an RNA.
- the polynucleotide of the present invention can encode multiple peptides of the present invention with or without intervening amino acid sequences in between.
- the intervening amino acid sequence can provide a cleavage site (e.g., enzyme recognition sequence) of the polynucleotide or the translated peptides.
- the polynucleotide can include any additional sequences to the coding sequence encoding the peptide of the present invention.
- the polynucleotide can be a recombinant polynucleotide that includes regulatory sequences required for the expression of the peptide or can be an expression vector (e.g., plasmid) with marker genes and such.
- such recombinant polynucleotides can be prepared by the manipulation of polynucleotides through conventional recombinant techniques using, for example, polymerases and endonucleases.
- a polynucleotide can be produced by insertion into an appropriate vector, which can be expressed when transfected into a competent cell.
- a polynucleotide can be amplified using PCR techniques or expression in suitable hosts (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1989).
- a polynucleotide can be synthesized using the solid phase techniques, as described in Beaucage SL & Iyer RP, Tetrahedron 1992, 48: 2223-311; Matthes et al., EMBO J 1984, 3: 801-5.
- the present invention further provides intracellular vesicles, referred to as exosomes, that present complexes formed between the peptides of the present invention and HLA antigens on their surface.
- Exosomes can be prepared, for example, using the methods detailed in JPH11-510507 and WO99/03499, and can be prepared using APCs obtained from patients who are subject to treatment and/or prevention (prophylaxis).
- the exosomes of the present invention can be inoculated as vaccines, in a fashion similar to the peptides of the present invention.
- HLA-A01 for example, HLA-A*01:01
- HLA-A*01:01 HLA-A allele highly frequently observed in Caucasians
- this HLA antigen type is considered to be suitable for treatment in Caucasian patients.
- the exosomes of the present invention present on their surface a complex of a peptide of the present invention and HLA-A01.
- the peptide of the present invention is preferably a peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 or a modified peptide thereof, and more preferably a peptide consisting of the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 or a modified peptide thereof.
- the present invention further provides APCs that present on their surface complexes formed between HLA antigens and peptides of the present invention.
- the present invention provides APCs having on their cell surface complexes formed between HLA antigens and peptides of the present invention.
- the APCs of the present invention can be isolated APCs. When used in the context of cells (APCs, CTLs, etc.), the teliu “isolated” means that the cells are separated from another type of cells.
- the APCs of the present invention may be APCs induced from APCs derived from the patient to be subjected to treatment and/or prevention (prophylaxis), and can be administered as a vaccine by themselves or in combination with other drugs including a peptide(s), an exosome(s) or a CTL(s) of the present invention.
- the APCs of the present invention are not limited to specific types of cells, and include cells known to present proteinaceous antigens on their cell surface so as to be recognized by lymphocytes, for example, dendritic cells (DCs), Langerhans cells, macrophages, B cells, and activated T cells. Since DC is a representative APC that has the strongest CTL-inducing activity among APCs, DCs can be preferably used as the APCs of the present invention.
- DCs dendritic cells
- Langerhans cells Langerhans cells
- macrophages macrophages
- B cells activated T cells. Since DC is a representative APC that has the strongest CTL-inducing activity among APCs, DCs can be preferably used as the APCs of the present invention.
- APCs of the present invention can be obtained by inducing DCs from peripheral blood monocytes and then stimulating them in vitro, ex vivo or in vivo with the peptides of the present invention.
- APCs presenting the peptide of the present invention are induced in the body of the subject. Therefore, after the peptides of the present invention are administered to a subject, the APCs of the present invention can be obtained by collecting APCs from the subject.
- the APCs of the present invention can be obtained by contacting APCs collected from a subject with a peptide of the present invention.
- the APCs of the present invention can be administered to the subject by themselves or in combination with other drugs including peptide(s), exosome(s) or CTL(s) of the present invention.
- the ex vivo administration can comprise the following steps of:
- the first subject and the second subject may be the same individual, or may be different individuals. When the first subject and the second subject are different individuals, it is preferable that the HLAs of the first subject and the second subject are of the same type.
- the APC obtained in step (b) above can be a vaccine for cancer treatment and/or prevention (prophylaxis).
- the APCs of the present invention obtained by a method such as described above have CTL-inducing ability.
- CTL-inducing ability CTL inducibility
- the CTL(s) induced by the APC of the present invention is a CTL(s) specific to CDCA1 and demonstrates a specific cytotoxic activity against CDCA1-expressing cells.
- the APCs of the present invention can be prepared by introducing a polynucleotide encoding a peptide of the present invention into APCs in vitro.
- the polynucleotide to be introduced can be in the form of DNA or RNA.
- the method of introduction is not particularly limited, and examples thereof include various methods conventionally performed in the art such as lipofection, electroporation and the calcium phosphate method. More specifically, methods described in Cancer Res 1996, 56: 5672-7; J Immunol 1998, 161: 5607-13; J Exp Med 1996, 184: 465-72; and JP2000-509281 can be used.
- polynucleotide encoding a peptide of the present invention By introducing a polynucleotide encoding a peptide of the present invention into an APC, the polynucleotide is transcribed and translated in the cell, and then the produced peptide is processed by MHC Class I and proceeds through a presentation pathway to present the peptide of the present invention on the cell surface of the APC.
- the APC of the present invention is an APC that presents on its cell surface a complex formed between HLA-A01 (more preferably HLA-A*01:01) and a peptide of the present invention.
- HLA-A01 the HLA that forms a complex with a peptide of the present invention is HLA-A01
- the peptide of the present invention is preferably a peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 or a modified peptide thereof, and more preferably a peptide consisting of the amino sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- the APC(s) of the present invention is preferably an APC(s) induced by a method comprising a step described (a) or (b) below:
- the peptide of the present invention to be contacted with the HLA-A01-expressing APC(s) is preferably a peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 or a modified peptide thereof, and more preferably a peptide consisting of the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- the present invention provides use of a peptide of the present invention for the manufacture of a pharmaceutical composition that induces an APC(s) having CTL-inducing ability.
- the present invention provides a method or process of manufacturing a pharmaceutical composition that induces an APC(s) having CTL-inducing ability.
- the present invention provides a peptide of the present invention for inducing an APC(s) having CTL-inducing ability.
- CTLs Cytotoxic T Lymphocytes
- the CTL induced by a peptide of the present invention can be used as a vaccine in a similar manner to the peptide of the present invention since it enhances an immune response targeting CDCA I-expressing cancer cell in vivo.
- the present invention provides CTLs that are induced or activated by a peptide of the present invention.
- the CTLs of the present invention are CTLs that target a peptide of the present invention, and are capable of binding to a complex of a peptide of the present invention and an HLA antigen. Binding of a CTL to the complex is mediated via a T cell receptor (TCR) present on the cell surface of the CTL.
- TCR T cell receptor
- the CTLs of the present invention can be isolated CTLs.
- the CTLs of the present invention can be obtained by (1) administering a peptide of the present invention to a subject, (2) stimulating APCs and CD8-positive T cells, or peripheral blood mononuclear cells (PBMCs) derived from a subject with a peptide of the present invention in vitro, (3) contacting in vitro CD8-positive T cells or PBMCs with APCs or exosomes that present on their surface a complex of an HLA antigen and a peptide of the present invention, or (4) introducing into CD8-positive T cells a vector comprising a polynucleotide encoding each subunit of a T cell receptor (TCR) capable of binding to a peptide of the present invention presented on cell surface via an HLA antigen.
- TCR T cell receptor
- exosomes and APCs used in the method of (2) or (3) above can be prepared by methods described in the “V. Exosomes” and “VI. Antigen-presenting cells (APCs)” sections, respectively, and the details of the method of (4) above will be described in the “VIII. T cell receptors (TCRs)” section.
- the CTLs of the present invention can be administered by themselves to patients who are subject to treatment and/or prevention (prophylaxis), or in combination with other drugs including peptide(s), APC(s) or exosome(s) of the present invention for the purpose of regulating effects.
- the CTLs of the present invention can be CTLs induced from CD8-positive T cells derived from the patients who are subject to administration of the CTLs.
- the CTLs of the present invention act specifically on target cells that present the peptides of the present invention, for example, the same peptides used to induce the CTLs of the present invention.
- the target cells may be cells that endogenously express CDCA1, such as cancer cells, or cells transfected with the CDCA1 gene.
- Cells that present a peptide of the present invention on their cell surface due to stimulation by the peptide can become a target of attack by the CTLs of the present invention.
- the cells targeted by the CTLs of the present invention are preferably cells that are positive for HLA-A01 (more preferably HLA-A*01:01).
- the CTLs of the present invention target specifically cells that express both HLA-A01 (more preferably HLA-A*01:01) and CDCA1.
- the CTL “targets” cells refers to CTL recognition of cells that present on their cell surface a complex of HLA and a peptide of the present invention and demonstration of a cytotoxic activity against the cells.
- specifically target refers to that the CTLs demonstrate a cytotoxic activity against those cells, but do not show a damaging activity to other cells.
- the expression “recognize cells” used in the context of CTLs refers to binding to a complex of HLA and a peptide of the present invention presented on cell surface via its TCR, and demonstrating a specific cytotoxic activity against the cell. Therefore, the CTLs of the present invention are preferably CTLs that can bind via TCR to a complex formed between HLA-A01 (more preferably HLA-A*01:01) and a peptide of the present invention presented on cell surface.
- the CTLs of the present invention are preferably CTLs induced by a method comprising a step described in (a) or (b) below:
- TCRs T Cell Receptors
- the present invention also provides compositions comprising a polynucleotide encoding each subunit of a TCR capable of binding to a peptide of the present invention presented on cell surface by an HLA antigen, and methods of using the same.
- the polynucleotide confers CD8-positive T cells with specificity against CDCA1-expressing cancer cells through expression of a TCR on cell surface capable of binding to a peptide of the present invention presented on cell surface by an HLA antigen.
- Polynucleotides encoding an alpha chain(s) and a beta chain(s) can be identified as the TCR subunit of the CTL induced by a peptide of the present invention by using known methods in the art (WO2007/032255 and Morgan et al., J Immunol, 2003, 171, 3288). For example, PCR methods are preferred for TCR analysis.
- PCR primers for analysis may be, for example, 5′-R Primer (5′-gtctaccaggcattcgcttcat-3′) (SEQ ID NO: 40) as a 5′ side primer; and 3-TRa-C Primer (5′-tcagctggaccacagccgcagcgt-3′) (SEQ ID NO: 41) specific to TCR-alpha-chain C-region, 3-TRb-C1 Primer (5′-tcagaaatectttctcttgac-3′) (SEQ ID NO: 42) specific to TCR-beta-chain C1-region, or 3-TRb-C2 Primer (5′-ctagcctctggaatcctttctcttt-3′) (SEQ ID NO: 43) specific to TCR-beta-chain C2-region as 3′ side primers, but are not limited thereto.
- the TCRs formed by introducing the identified polynucleotides into CD8-positive T cells can bind with high binding affinity to the target cells that present a peptide of the present invention, and mediates efficient killing of the target cells presenting a peptide of the present invention in vivo and in vitro.
- a polynucleotide encoding each TCR subunit can be incorporated into an appropriate vector, for example, retrovirus vector. These vectors are well known in the art.
- the polynucleotide or a vector comprising thereof in an expressible form can be introduced into a CD8-positive T cell, for example, a CD8-positive T cell derived from a patient.
- the present invention provides off-the-shelf compositions that allow rapid and easy production of modified T cells that have superior cancer cell-killing properties by rapid modification of the patient's own T cells (or T cells derived from another subject).
- a specific TCR is a TCR that can confer a specific cytotoxic activity against target cells by specifically recognizing a complex of a peptide of the present invention and an HLA antigen presented on the surface of the target cell when the TCR is present on the surface of a CD8-positive T cell.
- Specific recognition of the above-described complex can be confirmed by any known method, and preferable examples thereof include HLA multimer staining analysis using HLA molecules and peptides of the present invention and ELISPOT assay methods.
- Specific TCR-mediated recognition of target cell by T cell introduced with the above-described polynucleotide and signal transduction in the cell can be confirmed by carrying out an ELISPOT assay.
- TCR When the above-described TCR is present on the surface of a CD8-positive T cell, whether the TCR can confer a target cell-specific cytotoxic activity against the CD8-positive T cell can also be confirmed by known methods. Preferable methods include, for example, measuring the cytotoxic activity against target cells by a chrome release assay method or such.
- the present invention provides, in the context of HLA-A01, CTLs prepared by transforming CD8-positive T cells with a polynucleotide encoding each subunit of TCR that binds to, for example, a peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- the transformed CTLs are capable of homing in vivo and may be propagated by a well-known in vitro culturing method (for example, Kawakami et al., J Immunol., 1989, 142, 3452-61).
- the CTLs of the present invention can be used to form an immunogenic composition useful for disease treatment or prevention (prophylaxis) in a patient in need of treatment or prevention (prophylaxis) (the contents are incorporated herein for reference WO2006/031221).
- compositions or pharmaceutical compositions comprising at least one active ingredient selected from below:
- compositions of the present invention can comprise as needed a carrier(s), an excipient(s) or such commonly used in pharmaceuticals without particular limitations, in addition to the active ingredient(s) described above.
- a carrier that can be used in a pharmaceutical composition of the present invention includes sterilized water, physiological saline, phosphate buffer, culture fluid and such. Therefore, the present invention also provides pharmaceutical compositions, comprising at least one active ingredient selected from (a) to (e) below and a pharmaceutically acceptable carrier:
- compositions of the present invention can comprise, as needed, stabilizers, suspensions, preservatives, surfactants, solubilizing agents, pH adjusters, aggregation inhibitors and such.
- a peptide of the present invention or a polynucleotide encoding the peptide can be used to treat and/or prevent cancer, and/or prevent postoperative recurrence thereof. Therefore, the present invention provides pharmaceutical compositions for treating and/or preventing cancer, and/or preventing postoperative recurrence thereof, comprising one or more types of peptides or polynucleotides of the present invention as an active ingredient.
- the peptides of the present invention can be made to be presented on the surface of exosomes or APCs for use as pharmaceutical compositions.
- CTLs of the present invention targeting any one of the peptides of the present invention can also be used as an active ingredient of the pharmaceutical compositions of the present invention.
- the pharmaceutical compositions of the present invention may comprise a therapeutically effective amount or a pharmaceutically effective amount of the above-described active ingredient.
- compositions of the present invention may also be used as a vaccine.
- the term “vaccine” also called “immunogenic composition” refers to a composition that has a function of inducing an immune response that leads to antitumor action when inoculated into an animal.
- a pharmaceutical composition of the present invention can be used to induce an immune response that leads to antitumor action.
- the immune response induced by a peptide, a polynucleotide, an APC, a CTL and a pharmaceutical composition of the present invention is not particularly limited as long as it is an immune response that leads to antitumor action, and examples include induction of cancer cell-specific CTLs and induction of cancer cell-specific cytotoxic activity.
- the pharmaceutical compositions of the present invention can be used to treat and/or prevent cancer, and/or prevent postoperative recurrence thereof in human subjects or patients.
- the pharmaceutical compositions of the present invention can be used preferably to a subject positive for HLA-A01. Further, the pharmaceutical compositions of the present invention can be used preferably to treat and/or prevent cancers expressing HLA A01 and CDCA1, and/or prevent postoperative recurrence thereof.
- the present invention provides use of an active ingredient selected from below in the manufacture of a pharmaceutical composition for treating or preventing cancer expressing HLA-A01 and CDCA1:
- the present invention further provides an active ingredient selected from below for use in treating or preventing cancer expressing HLA-A01 and CDCA1:
- the present invention further provides a method or process for manufacturing a pharmaceutical composition for treating or preventing cancer expressing HLA-A01 and CDCA1, wherein the method or process comprises a step of formulating at least one active ingredient selected from below with a pharmaceutically or physiologically acceptable carrier:
- the present invention further provides a method or process for manufacturing a pharmaceutical composition for treating or preventing cancer expressing HLA-A01 and CDCA1, wherein the method or process comprises a step of mixing an active ingredient selected from below with a pharmaceutically or physiologically acceptable carrier:
- the present invention further provides a method for treating or preventing cancer expressing HLA-A01 and CDCA1, which comprises a step of administering to a subject at least one active ingredient selected from below:
- compositions of the present invention comprising at least one peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 are suitable particularly for administration to a subject having HLA-A01 (for example, HLA-A*01:01) as an HLA antigen.
- compositions comprising a polynucleotide encoding any of these peptides (i.e., polynucleotides of the present invention), an APC or exosome that presents these peptides (i.e., APCs or exosomes of the present invention), or a CTL targeting these peptides (i.e., CTLs of the present invention).
- pharmaceutical compositions comprising an active ingredient in association with a peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 are suitable for administration to subjects having HLA-A01 (i.e., HLA-A01-positive subjects).
- the pharmaceutical composition of the present invention is a pharmaceutical composition that comprises a peptide having the amino acid sequence of SEQ ID NO: 8.
- Cancers to be treated and/or prevented by pharmaceutical compositions of the present invention are not particularly limited as long as they are cancers that express CDCA1, and include various cancers, bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, colon cancer and such.
- CML chronic myeloid leukemia
- compositions of the present invention can comprise the other peptides that have the ability to induce CTLs against cancer cells (for example, the other TAA-derived CTL-inducing peptides), the other polynucleotides encoding the other peptides, the other cells that present the other peptides, or such.
- the pharmaceutical compositions of the present invention may also optionally comprise the other therapeutic substances as an active ingredient, as long as they do not inhibit the anti-tumor effects of the above-described active ingredients such as peptides of the present invention.
- the pharmaceutical compositions of the present invention may optionally comprise anti-inflammatory compositions, analgesics, chemotherapeutics and the like.
- the dose of the pharmaceutical composition of the present invention and the other pharmaceutical compositions depend on, for example, the type of pharmaceutical composition used and the disease being treated, as well as the scheduling and routes of administration.
- the pharmaceutical composition of the present invention may include other components conventional in the art, in addition to the ingredients specifically mentioned herein.
- the present invention also provides articles of manufacture or kits that comprise a pharmaceutical composition of the present invention.
- the articles of manufacture or kits of the present invention can include a container that houses the pharmaceutical composition of the present invention.
- An example of an appropriate container includes a bottle, a vial or a test tube, but is not limited thereto.
- the container may be formed of various materials such as glass or plastic.
- a label may be attached to the container, and the disease or disease state to which the pharmaceutical composition of the present invention should be used may be described in the label.
- the label may also indicate directions for administration and such.
- the articles of manufacture or kits of the present invention may further comprise a second container that houses pharmaceutically acceptable diluents optionally, in addition to the container that houses the pharmaceutical composition of the present invention.
- the articles of manufacture or kits of the present invention may further comprise the other materials desirable from a commercial standpoint and the user's perspective, such as the other buffers, diluents, filters, injection needles, syringes, package inserts with instructions for use.
- the pharmaceutical composition of the present invention can be provided in a pack or dispenser device that can contain one or more units of dosage forms containing active ingredients.
- the pack can include, for example, a metallic foil or a plastic foil such as a blister pack. Instructions for administration can be attached to the pack or dispenser device.
- compositions comprising Peptide(s) as an Active Ingredient
- the pharmaceutical composition comprising a peptide of the present invention can be formulated by conventional formulation methods as needed.
- the pharmaceutical compositions of the present invention may comprise as needed in addition to the peptide of the present invention, carriers, excipients and such commonly used in pharmaceuticals without particular limitations.
- carriers that can be used in pharmaceutical compositions of the present invention include sterilized water (for example, water for injection), physiological saline, phosphate buffer, phosphate buffered saline, Tris buffered saline, 0.3% glycine, culture fluid, and such.
- the pharmaceutical compositions of the present invention may comprise as needed stabilizers, suspensions, preservatives, surfactants, solubilizing agents, pH adjusters, aggregation inhibitors, and such.
- the pharmaceutical compositions of the present invention can induce specific immunity against CDCA1-expressing cancer cells, and thus can be applied for the purpose of cancer treatment or prevention (prophylaxis).
- the pharmaceutical compositions of the present invention can be prepared by dissolving in pharmaceutically or physiologically acceptable water-soluble carriers such as sterilized water (for example, water for injection), physiological saline, phosphate buffer, phosphate buffered saline, and Tris buffered saline and adding, as needed, stabilizers, suspensions, preservatives, surfactants, solubilizing agents, pH adjusters, aggregation inhibitors and such, and then sterilizing the peptide solution.
- the method of sterilizing a peptide solution is not particularly limited, and is preferably carried out by filtration sterilization. Filtration sterilization can be performed using, for example, a filtration sterilization filter of 0.22 micro-m or less in pore diameter.
- the filtration-sterilized peptide solution can be administered to a subject, for example, as an injection, without being limited thereto.
- the pharmaceutical compositions of the present invention may be prepared as a freeze-dried formulation by freeze-drying the above-described peptide solution.
- the freeze-dried formulation can be prepared by filling the peptide solution prepared as described above into an appropriate container such as an ampule, a vial or a plastic container, followed by freeze drying and encapsulation into the container with a wash-sterilized rubber plug or such after pressure recovery.
- the freeze-dried formulation can be administered to a subject after it is re-dissolved in pharmaceutically or physiologically acceptable water-soluble carriers such as sterilized water (for example, water for injection), physiological saline, phosphate buffer, phosphate buffered saline, Tris buffered saline and such before administration.
- pharmaceutical compositions of the present invention include injections of such a filtration-sterilized peptide solution, and freeze-dried formulations that result from freeze-drying the peptide solution.
- the present invention further encompasses kits comprising such a freeze-dried formulation and re-dissolving solution.
- kits also encompasses kits comprising a container that houses the freeze-dried formulation, which is a pharmaceutical composition of the present invention, and a container that stores a re-dissolving solution thereof.
- compositions of the present invention can comprise a combination of two or more types of the peptides of the present invention.
- the combination of peptides can take a cocktail form of mixed peptides, or can be conjugated with each other using standard techniques.
- peptides can be chemically linked or expressed as single fusion polypeptide sequences.
- APCs for example, DCs
- APCs are removed from a subject, and subsequently stimulated with peptides of the present invention to obtain APCs that present any of the peptides of the present invention on their cell surface.
- APCs are re-administered to a subject to induce CTLs in the subject, and as a result, the aggressiveness towards CDCA1-expressing cancer cells can be increased.
- compositions of the present invention may also comprise an adjuvant known for effectively establishing cellular immunity.
- An adjuvant refers to a compound that enhances the immune response against an antigen that has immunological activity when administered together (or successively) with the antigen.
- a suitable adjuvant include aluminum salts (aluminum phosphate, aluminum hydroxide, aluminum oxyhydroxide and such), alum, cholera toxin, Salmonella toxin,
- IFA Incomplete Freund's adjuvant
- CFA Complete Freund's adjuvant
- ISCOMatrix GM-CSF and other immunostimulatory cytokines, oligodeoxynucleotide containing the CpG motif (CpG7909 and such), oil-in-water emulsions, Saponin or its derivatives (QS21 and such), lipopolysaccharide such as Lipid A or its derivatives (MPL, RC529, GLA, E6020 and such), lipopeptides, lactoferrin, flagellin, double-stranded RNA or its derivatives (poli IC and such), bacterial DNA, imidazoquinolines (Imiquimod, R848 and such), C-type lectin ligand (trehalose-6,6′-dibehenate (TDB) and such), CD1d ligand (alpha-galactosylceramide and such), squalene emulsions (MF59, AS03
- kits comprising the pharmaceutical composition of the present invention.
- the adjuvant and the pharmaceutical composition may be administered to a subject in succession, or mixed together immediately before administration to a subject.
- kits comprising a pharmaceutical composition comprising a peptide of the present invention and an adjuvant are also provided by the present invention.
- the pharmaceutical composition of the present invention is a freeze-dried formulation
- the kit can further comprise a re-dissolving solution.
- the present invention provides kits comprising a container that houses a pharmaceutical composition of the present invention and a container that stores an adjuvant.
- the kit can further comprise as needed a container that stores the re-dissolving solution.
- the pharmaceutical composition of the present invention may be prepared as an emulsion.
- Emulsions can be prepared, for example, by mixing and stirring the peptide solution prepared as described above and an oil adjuvant.
- the peptide solution may be one that has been re-dissolved after freeze-drying.
- the emulsion may be either of the W/O-type emulsion and O/W-type emulsion, and the W/O-type emulsion is preferred for obtaining a high immune response-enhancing effect.
- IFA can be preferably used as an oil adjuvant, without being limited thereto.
- the pharmaceutical composition of the present invention may be provided as a kit comprising the peptide solution of the present invention and an oil adjuvant.
- the kit can further comprise a re-dissolving solution.
- composition of the present invention may be a liposome formulation within which a peptide of the present invention is encapsulated, a granular formulation in which a peptide is bound to beads with several micrometers in diameter, or a formulation in which a lipid is bound to a peptide.
- the peptide of the present invention may also be administered in the form of a pharmaceutically acceptable salt.
- salts include salts with alkali metals (lithium, potassium, sodium and such), salts with alkaline-earth metals (calcium, magnesium and such), salts with other metals (copper, iron, zinc, manganese and such), salts with organic bases, salts with amines, salts with organic acids (acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid and such), and salts with inorganic acids (hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, nitric acid and such).
- compositions comprising a pharmaceutically acceptable salt of a peptide of the present invention are also encompassed by the present invention.
- the “peptide of the present invention” also encompasses, in addition to the free peptide, pharmaceutically acceptable salts thereof.
- the pharmaceutical compositions of the present invention may further include a component which primes CTLs.
- Lipids have been identified as substances capable of priming CTLs in vivo against viral antigens.
- palmitic acid residues can be attached to the epsilon- and alpha-amino groups of a lysine residue and then linked to a peptide of the present invention.
- the lipidated peptide can then be administered either directly in a micelle or particle, incorporated into a liposome, or emulsified in an adjuvant.
- lipid priming of CTL responses E.
- coil lipoproteins such as tripalmitoyl-S-glycerylcysteinyl-seryl-serine (P3CSS) can be used to prime CTLs when covalently attached to an appropriate peptide (see, e.g., Deres et al., Nature 1989, 342: 561-4).
- P3CSS tripalmitoyl-S-glycerylcysteinyl-seryl-serine
- Examples of methods for administering the peptides or pharmaceutical compositions of the present invention include oral, epidermal, subcutaneous, intramuscular, intraosseous, peritoneal, and intravenous injections, as well as systemic administration or local administration to the vicinity of the targeted sites, but are not limited thereto.
- a preferred administration method includes subcutaneous injection to the vicinity of lymph nodes such as the armpit or groin.
- the administration can be performed by single administration or boosted by multiple administrations.
- the peptides of the present invention can be administered to a subject in a therapeutically or pharmaceutically effective amount for treating cancer or in a therapeutically or pharmaceutically effective amount for inducing immunity (more specifically CTLs) against CDCA1-expressing cancer cells.
- the dose of the peptides of the present invention can be appropriately adjusted according to the disease to be treated, the patient's age and weight, the method of administration and such.
- the dose is usually 0.001 mg-1000 mg, for example, 0.01 mg-100 mg, for example, 0.1 mg-30 mg, for example, 0.1 mg-10 mg, for example, 0.5 mg-5 mg.
- the dosing interval can be once every several days to several months, and for example, the dosing can be done in a once-per-week interval. A skilled artisan can appropriately select a suitable dosage.
- the pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a peptide of the present invention and a pharmaceutically or physiologically acceptable carrier.
- the pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a peptide of the present invention, a pharmaceutically or physiologically acceptable cancer, and an adjuvant.
- the pharmaceutical compositions of the present invention can comprise 0.001 mg-1000 mg, preferably 0.01 mg-100 mg, more preferably 0.1 mg-30 mg, even more preferably 0.1 mg-10 mg, for example, 0.5 mg-5 mg of a peptide of the present invention.
- a pharmaceutical composition of the present invention when a pharmaceutical composition of the present invention is an injection, it can comprise a peptide of the present invention at a concentration of 0.001 mg/ml-1000 mg/ml, preferably 0.01 mg/ml-100 mg/ml, more preferably 0.1 mg/ml-30 mg/ml, even more preferably 0.1 mg/ml-10 mg/ml, for example, 0.5 mg/ml-5 mg/ml.
- 0.1 to 5 ml preferably 0.5 ml to 2 ml of the pharmaceutical composition of the present invention can be administered to a subject by injection.
- the present invention provides methods of treating and/or preventing cancer and/or preventing postoperative recurrence thereof, which comprise administering to a subject a therapeutically effective amount of a peptide of the present invention or a pharmaceutical composition of the present invention.
- the peptides of the present invention can be administered to a subject in a single dose of usually 0.001 mg-1000 mg, for example, 0.01 mg-100 mg, for example, 0.1 mg-30 mg, for example, 0.1 mg-10 mg, or for example, 0.5 mg-5 mg.
- the peptides of the present invention are administered to a subject together with an adjuvant.
- the dosing interval can be once every several days to several months, preferably once every several days to every month, for example, once every week or once every two weeks.
- the CDCA1 peptides of the invention carry an HLA-A01-restrictive CTL-inducing activity. Accordingly, their therapeutic effect is also effective in HLA-A01-positive subjects.
- an HLA-A01-positive subject can be selected in advance, before the administration of a CDCA1 peptide of the present invention.
- the therapeutic effect of the CDCA1 of the present invention is CDCA1-specific, it is a desirable condition that cancer in a subject is expressing CDCA1. That is, the methods of treating cancer of the present invention can comprise the step of selecting an HLA-A01-positive subject and the step of selecting a subject having cancer expressing CDCA1, before the administration of the CDCA1 peptide.
- compositions of the present invention can also contain polynucleotides encoding the peptides of the present invention in an expressible form.
- the phrase “in an expressible form” means that the polynucleotide, when introduced into a cell, will be expressed as a peptide of the present invention.
- the sequence of the polynucleotide of the present invention includes regulatory elements necessary for expression of the peptide of the present invention.
- the polynucleotide(s) of the present invention can be equipped with a sequence necessary to achieve stable insertion into the genome of the target cell (see, e.g., Thomas K R & Capecchi M R, Cell 1987, 51: 503-12 for a description of homologous recombination cassette vectors). See, e.g., Wolff et al., Science 1990, 247: 1465-8; U.S. Pat. Nos. 5,580,859, 5,589,466, 5,804,566, 5,739,118, 5,736,524, 5,679,647; and WO98/04720.
- DNA-based delivery technologies include “naked DNA”, facilitated (bupivacaine, polymers, peptide-mediated) delivery, cationic lipid complexes, and particle-mediated (“gene gun”) or pressure-mediated delivery (see, e.g., U.S. Pat. No. 5,922,687).
- the peptides of the present invention can also be expressed by viral or bacterial vectors.
- expression vectors include attenuated viral hosts, such as vaccinia or fowlpox.
- vaccinia virus can be used as a vector to express the peptide of the present invention.
- the recombinant vaccinia virus Upon introduction into a host, the recombinant vaccinia virus expresses the immunogenic peptide, and thereby elicits an immune response.
- Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848.
- Another vector is BCG (Bacille Calmette Guerin).
- BCG vectors are described in Stover et al., Nature 1991, 351: 456-60.
- a wide variety of other vectors useful for therapeutic administration or immunization e.g., adeno and adeno-associated virus vectors, retroviral vectors, Salmonella typhi vectors, detoxified anthrax toxin vectors, and the like, will be apparent. See, e.g., Shata et al., Mol Med Today 2000, 6: 66-71; Shedlock et al., J Leukoc Biol 2000, 68: 793-806; Hipp et al., In Vivo 2000, 14: 571-85.
- Delivery of a polynucleotide of the present invention into a patient can be either direct, in which case the patient can be directly exposed to a vector harboring the polynucleotide of the present invention, or indirect, in which case, cells are first transformed with the vector harboring the polynucleotide of the present invention in vitro, then the cells are transplanted into the patient.
- direct in which case the patient can be directly exposed to a vector harboring the polynucleotide of the present invention
- indirect in which case, cells are first transformed with the vector harboring the polynucleotide of the present invention in vitro, then the cells are transplanted into the patient.
- Administration methods may be oral, intradermal, subcutaneous, or intravenous injection, and such.
- a systemic administration or a local administration to the vicinity of the targeted sites is used.
- the administration can be performed by single administration or boosted by multiple administrations.
- the polynucleotides of the present invention can be administered to a subject in a therapeutically or pharmaceutically effective dose for inducing immunity (more specifically CTLs) against CDCA1-expressing cancer cells, or in a therapeutically or pharmaceutically effective dose for treating cancer.
- the dose of a polynucleotide in a suitable carrier or the dose of a polynucleotide in cells transformed with a polynucleotide encoding a peptide of the present invention can be appropriately adjusted according to the disease to be treated, the patient's age and weight, the method of administration and such, and this may be usually 0.001 mg-1000 mg, for example, 0.01 mg-100 mg, for example, 0.1 mg-30 mg, for example, 0.1 mg-10 mg, or for example, 0.5 mg-5 mg.
- the dosing interval can be once every several days to several months, and for example, the dosing can be done in a once-per-week interval. A skilled artisan can appropriately select a suitable dosage.
- the peptides and polynucleotides of the present invention can be used to induce APCs and CTLs. CTLs can also be induced using the exosomes and APCs of the present invention.
- the peptides, polynucleotides, exosomes, and APCs can be used in combination with any other compound(s) as long as their CTL-inducing ability is not inhibited. Therefore, CTLs of the present invention can be induced using a pharmaceutical composition comprising any of the peptides, polynucleotides, APCs and exosomes of the present invention. Further, APCs of the present invention can be induced using a pharmaceutical composition comprising a peptide or polynucleotide of the present invention.
- the present invention provides methods of inducing APCs having CTL-inducing ability, using a peptide(s) or polynucleotide(s) of the present invention.
- the methods of the present invention comprise a step of contacting an APC with a peptide of the present invention in vitro, ex vivo, or in vivo.
- a method of contacting APCs with the peptide ex vivo may comprise the steps below:
- APCs are not limited to a particular type of cell, and DCs, Langerhans cells, macrophages, B cells, and activated T cells, which are known to present a proteinaceous antigen on their cell surface to be recognized by lymphocytes, can be used.
- DCs have the most potent CTL-inducing ability among APCs, and thus it is preferable to use DCs.
- Any peptides of the present invention can be used by themselves or in combination with other peptides of the present invention. Further, peptides of the present invention can be used in combination with other CTL-inducing peptides (for example, other TAA-derived CTL-inducing peptides).
- the methods of the present invention may comprise a step of administering a peptide of the present invention to a subject.
- a polynucleotide of the present invention is administered to a subject in an expressible form, a peptide of the present invention is expressed in vivo, the expressed peptide is contacted with APCs in vivo, and as a result APCs having a high CTL-inducing ability are induced in the body of the subject. Therefore, the present invention may also comprise a step of administering a polynucleotide of the present invention to a subject.
- the present invention may comprise a step of introducing a polynucleotide of the present invention into APCs.
- the method may comprise the steps below:
- the present invention provides a method of inducing APCs having CTL-inducing ability, which comprises the step (a) or (b) below:
- the present invention provides a method of preparing APCs having CTL-inducing ability, which comprises the step (a) or (b) below:
- APCs used in the above-described methods can be derived from a subject scheduled for administration of the induced APCs, or they may be derived from a different subject.
- APCs derived from a subject (donor) different from the subject scheduled for administration are used, the subject of administration and the donor must have the identical HLA type.
- the HLA type is preferably HLA-A01 (more preferably HLA-A*01:01) in both the subject of administration and the donor.
- APCs used in the above-described methods are preferably APCs that express HLA-A01 (more preferably HLA-A*01:01).
- the APCs can be prepared using known methods from PBMCs after PBMCs are separated from blood collected from a donor by a specific gravity centrifugal method or such.
- the present invention also provides pharmaceutical compositions that comprise a peptide of the present invention or a polynucleotide encoding the peptide for inducing an APC(s) having CTL-inducing ability.
- the present invention further provides use of a peptide of the present invention or a polynucleotide encoding the peptide in the manufacture of a pharmaceutical composition for inducing an APC(s) having CTL-inducing ability.
- the present invention further provides peptides of the present invention or polynucleotides encoding the peptides for use in the induction of an APC(s) having CTL-inducing ability.
- the present invention further provides methods or processes of manufacturing a pharmaceutical composition for inducing an APC(s), wherein the method or process comprises a step of formulating a peptide of the present invention or a polynucleotide encoding the peptide with a pharmaceutically or physiologically acceptable carrier.
- the present invention further provides methods or processes of manufacturing a pharmaceutical composition for inducing an APC(s) having CTL-inducing ability, wherein the method or process comprises a step of mixing a peptide of the present invention or a polynucleotide encoding the peptide with a pharmaceutically or physiologically acceptable carrier.
- APCs induced by the methods of the present invention can induce CTLs specific to CDCA1 (i.e., CTLs of the present invention).
- the present invention also provides methods of inducing CTLs using peptides, polynucleotides, exosomes or APCs of the present invention.
- the methods of the present invention may comprise a step of administering a peptide(s), a polynucleotide(s), an APC(s) or an exosome(s) of the present invention to a subject.
- CTLs can be induced by using them in vitro or ex vivo.
- the methods of the present invention may include the following steps:
- the induced CTLs may be returned to the subject afterwards.
- the APCs to be co-cultured with the CD8-positive T cells in step (c) above can also be prepared by introducing into APCs a polynucleotide encoding a peptide of the present invention as described above in the “VI. Antigen-presenting cells (APCs)” section.
- APCs Antigen-presenting cells
- the APCs to be used in the methods of the present invention are not limited thereto, and any APCs that present on their surface a complex of an HLA antigen and a peptide of the present invention can be used.
- exosomes that present on their surface a complex of an HLA antigen and a peptide of the present invention can also be used. That is, the methods of the present invention can comprise a step of co-culturing with exosomes that present on their surface a complex of an HLA antigen and a peptide of the present invention.
- exosomes can be prepared by the above-described methods in the “V. Exosomes” section.
- CTLs can also be induced by introducing into a CD8-positive T cell a vector comprising a polynucleotide encoding each subunit of a TCR capable of binding to a peptide of the present invention presented by an HLA antigen on the cell surface.
- TCRs T cell receptors
- the present invention provides methods of inducing CTLs, comprising a step selected from below:
- APCs or exosomes and CD8-positive T cells used in the above-described methods may be derived from a subject scheduled for administration of the induced CTLs, or they may be derived from a different subject.
- APCs or exosomes and CD8-positive T cells derived from a subject (donor) different from the subject scheduled for administration are used, the subject of administration and the donor must have the identical HLA type.
- the HLA type in both the subject of administration and the donor is preferably HLA-A01 (more preferably HLA-A*01:01).
- APCs or exosomes used in the above-described methods are preferably APCs or exosomes that present on their surface a complex of HLA-A01 (more preferably HLA-A*01:01) and a peptide of the present invention (a peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37 or a modified peptide thereof).
- the induced CTLs show a specific cytotoxic activity against cells that present a complex of HLA-A01 and a peptide of the present invention (for example, CDCA1-expressing HLA-A01-positive cells).
- the present invention also provides compositions or pharmaceutical compositions for inducing CTLs, comprising at least one active ingredient selected from below:
- the present invention also provides use of an active ingredient selected from below in the manufacture of compositions or pharmaceutical compositions for inducing CTLs:
- the present invention further provides an active ingredient selected from below for use in inducing CTLs:
- the present invention further provides a method or process for manufacturing a composition or pharmaceutical composition for inducing CTLs, which is a method or process that comprises a step of formulating an active ingredient selected from below with a pharmaceutically or physiologically acceptable carrier:
- the present invention further provides a method or process for manufacturing a composition or pharmaceutical composition for inducing CTLs, which is a method or process that comprises a step of mixing an active ingredient selected from below with a pharmaceutically or physiologically acceptable carrier:
- the present invention further provides methods of inducing an immune response against CDCA1-expressing cancers.
- Applicable cancers include bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, colon cancer and such, but are not limited thereto. It is preferable that the cancer expresses HLA-A01.
- the present invention further provides methods of inducing an immune response against CDCA1-expressing cancer cells.
- CDCA1 is recognized to be overexpressed in various types of cancers described above.
- proliferation of the cancer cells is inhibited as a result.
- the present invention further provides methods of inhibiting proliferation of CDCA1-expressing cancer cells.
- the methods of the present invention are suitable, in particular, for inhibiting proliferation of cancer cells expressing CDCA1 and HLA-A01.
- the methods of the present invention may comprise a step of administering a composition comprising any of the peptides of the present invention or a polynucleotide(s) encoding the peptide(s).
- the methods of the present invention also contemplate administration of APCs or exosomes presenting any of the peptides of the present invention.
- the details can be referred to the “IX. Pharmaceutical compositions” section, particularly portions describing regarding use of the pharmaceutical compositions of the present invention as vaccines.
- exosomes and APCs that can be used in the methods of the present invention for inducing an immune response are described in detail in “V. Exosomes”, “VI. Antigen-presenting cells (APCs)” and in Items (1) and (2) of “X. Methods of using peptides, exosomes, APCs and CTLs” described above.
- the present invention provides pharmaceutical compositions or vaccines for inducing an immune response against cancers expressing CDCA1 and HLA-A01, wherein the pharmaceutical composition or vaccine comprises an active ingredient selected from below:
- the present invention also provides pharmaceutical compositions or vaccines for inducing an immune response against cancer cells expressing CDCA1 and HLA-A01, wherein the pharmaceutical composition or vaccine comprises an active ingredient selected from below:
- the present invention further provides pharmaceutical compositions or vaccines for inhibiting proliferation of cancer cells expressing CDCA1 and HLA-A01, wherein the pharmaceutical composition or vaccine comprises an active ingredient selected from below:
- the present invention provides use of an active ingredient selected from below in the manufacture of pharmaceutical compositions or vaccines for inducing an immune response against cancers expressing CDCA1 and HLA-A01:
- the present invention also provides use of an active ingredient selected from below in the manufacture of pharmaceutical compositions or vaccines for inducing an immune response against cancer cells expressing CDCA1 and HLA-A01:
- the present invention further provides use of an active ingredient selected from below in the manufacture of pharmaceutical compositions or vaccines for inhibiting proliferation of cancer cells expressing CDCA1 and HLA-A01:
- the present invention further provides methods or processes for manufacturing pharmaceutical compositions that induce an immune response against cancers expressing CDCA1 and HLA-A01, which is a method that may comprise a step of mixing or formulating a peptide of the present invention with a pharmaceutically acceptable carrier.
- the present invention provides methods for inhibiting proliferation of cancer cells expressing CDCA1 and HLA-A01 in diseased sites of diseases mediated by angiogenesis or methods of inducing an immune response against cancers, which comprises a step of administering to a subject vaccines or pharmaceutical compositions comprising an active ingredient selected from below:
- cancers expressing CDCA1 and HLA-A01 can be treated by administering a peptide, a polynucleotide, an APC, an exosome and/or a CTL of the present invention.
- an immune response against CDCA1-expressing cancers can be induced by administering a peptide, a polynucleotide, an APC, an exosome and/or a CTL of the present invention.
- cancers examples include bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, colon cancer and such, but are not limited thereto.
- CML chronic myeloid leukemia
- esophagus cancer gastric cancer
- non-small-cell lung cancer lymphoma
- osteosarcoma prostate cancer
- kidney cancer small-cell lung cancer
- head and neck cancer soft tissue tumor, colon cancer and such, but are not limited thereto.
- an immune response against cancer cells expressing CDCA1 and HLA-A01 can be induced by administering a peptide, a polynucleotide, an APC, an exosome and/or a CTL of the present invention.
- the present invention can select a subject who is HLA-A01-positive and who has cancer expressing CDCA1 and administer the vaccine or the pharmaceutical composition of the present invention to the selected subject.
- the present invention provides a method of treating a cancer expressing CDCA1 and HLA-A01 in a patient in need of the cancer treatment, wherein the method comprises the steps below:
- the present invention further provides vaccines or pharmaceutical compositions comprising at least one active ingredient selected from the group consisting of (a) to (e) above for administration to an HLA-A01-positive subject with CDCA1-expressing cancer.
- the present invention further provides a method of identifying or selecting a subject to be treated with at least one active ingredient selected from the group consisting of (a) to (e) above, wherein the method comprises the steps below:
- Biological samples collected from a subject for measuring the CDCA1 expression level in the above-described methods are not particularly limited, and for example, tissue samples containing cancer cells collected by biopsy or such can be preferably used.
- the CDCA1 expression level in a biological sample can be measured by known methods, and for example, methods that detect transcription products of the CDCA1 gene by probes or PCR methods (for example, cDNA microarray method, Northern blot method, RT-PCR method or such), methods that detect translation products of the CDCA1 gene by antibodies or such (for example, Western blot method, immunostaining method or such), and such can be used.
- biological samples may be blood samples, and in this case, the blood level of an antibody against CDCA1 is measured, and the CDCA1 expression level at a diseased site may be assessed based on the blood level.
- the blood level of an antibody against CDCA1 can be measured using known methods, and for example, enzyme immunoassay (EIA), enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RLA) and such using the CDCA1 protein or a peptide of the present invention as an antigen can be used.
- EIA enzyme immunoassay
- ELISA enzyme-linked immunosorbent assay
- RLA radioimmunoassay
- the CDCA1 expression level at a diseased site may be assessed by detecting CTLs specific to a peptide of the present invention.
- the CTL levels specific to a peptide of the present invention can be measured, for example, by separating PBMCs from the blood collected from a subject and measuring the cytotoxic activity against target cells pulsed with the peptide of the present invention.
- the cytotoxic activity can be measured, for example, by the amount of interferon-gamma release.
- complexes of the peptides of the present invention and HLA described below can also be used for measuring the CTL levels. Whether a subject's cancer expresses CDCA1 or not may be determined by comparing with the measurement results in a biological material of the same type collected from a subject without cancer.
- the level of a substance to be measured in a biological sample collected from a subject having cancer is elevated relative to the level in a biological material of the same type collected from another subject without cancer (normal control level), it can be determined that the cancer of the subject having cancer expresses CDCA1.
- the HLA type of a subject before administering at least one active ingredient selected from the group consisting of (a) to (e) above.
- an HLA-A01-positive subject as a subject to be administered with an active ingredient which is related to a peptide having the amino acid sequence selected from among SEQ ID NOs: 1, 8, 10, 13, 25, 33 to 35 and 37.
- the HLA of a subject can be confirmed by immunologically detecting an HLA on the surface of cells collected from the subject with an HLA-A01-specific antibody.
- the HLA of a subject can be determined by analyzing the genetic information of genomic DNA or mRNA obtained from cells collected from the subject. If a subject is HLA-A01-positive, then effective treatment can be expected in the present invention. In other words, a subject having homozygous or heterozygous HLA-A01 can be a target of administration.
- the present invention further provides complexes of a peptide of the present invention and HLA.
- the complexes of the present invention described above may be monomers or multimers.
- the number of polymerization is not particularly limited, and it can be a multimer of any number of polymerization. Examples include a tetramer, pentamer, hexamer and such, but are not limited thereto.
- the multimers of the present invention also encompass dextramers (WO2002/072631) and streptamers (Knabel M et al., Nat Med. 2002; 8(6): 631-7.).
- Complexes of a peptide of the present invention and HLA can be prepared according to known methods (for example, Altman J D et al., Science. 1996, 274(5284): 94-6; WO2002/072631; WO2009/003492; Knabel M et al., Nat Med. 2002; 8(6): 631-7, and such).
- the complexes of the present invention can be used in the quantification of CTLs specific to a peptide of the present invention.
- a blood sample is collected from a subject administered with a pharmaceutical composition of the present invention, and CD4-negative cells are adjusted after separation of PBMCs and contacted with a fluorescent dye-conjugated complex of the present invention.
- the percentage of CTLs specific to a peptide of the present invention can be measured by flow cytometry analysis.
- immune response-inducing effects by a pharmaceutical composition of the present invention can be monitored by measuring the specific CTLs against a peptide of the present invention before, during and/or after administration of the pharmaceutical composition of the present invention.
- the present invention further provides antibodies that bind to the peptide of the present invention.
- Preferable antibodies bind specifically to a peptide of the present invention, but do not bind (or weakly bind) to one that is not the peptide of the present invention.
- the binding specificity of an antibody can be confirmed by inhibition assay. That is, if the binding between an antibody to be analyzed and a full-length CDCA1 polypeptide is inhibited in the presence of a peptide of the present invention, this antibody is shown to specifically bind to the peptide of the present invention.
- Antibodies against peptides of the present invention can be used in assays of disease diagnosis and prognosis, as well as subject selection for administration of the pharmaceutical compositions of the present invention and monitoring of the pharmaceutical compositions of the present invention.
- the present invention also provides various immunological assays for detecting and/or quantifying peptides of the present invention or fragments thereof.
- immunological assays include radioimmunoassay, immunochromatography, enzyme-linked immunosorbent assay (ELISA), enzyme-linked immunofluorescence assay (ELIFA) and such, without being limited thereto, and are performed within the scope of the various immunological assay formats well known in the art.
- the antibodies of the present invention can be used in immunological imaging methods that can detect CDCA1-epxressing diseases, and examples thereof include radioactive scintigraphic imaging using a labelled antibody of the present invention, without being limited thereto.
- Such assay methods are used clinically in the detection, monitoring, and prognosis of CDCA1-expressing cancers; and examples of such cancer include bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, colon cancer and such, without being limited thereto.
- CML chronic myeloid leukemia
- the antibodies of the present invention can be used in any arbitrary form such as monoclonal antibodies or polyclonal antibodies, and may further include anti-sera obtained by immunizing an animal such as a rabbit with a peptide of the present invention, all classes of polyclonal antibodies and monoclonal antibodies, human antibodies, as well as chimeric antibodies and humanized antibodies generated through gene recombination.
- the peptide of the present invention or a fragment thereof used as an antigen for obtaining antibodies can be obtained by chemical synthesis or genetic engineering techniques based on the amino acid sequences disclosed herein.
- the peptide used as an immunizing antigen may be a peptide of the present invention or a fragment of a peptide of the present invention. Further, the peptide may be bound to or conjugated with a carrier for increasing immunogenicity.
- KLH Keyhole limpet hemocyanin
- Methods for binding KLH to a peptide are also well known in the art.
- animals of the order Rodentia, Lagomorpha or Primate can be used.
- Animals of the order Rodentia include, for example, mice, rats and hamsters.
- Animals of the order Lagomorpha include, for example, rabbits.
- Animals of the order Primate include, for example, Catarrhini monkeys (old world monkeys) such as cynomolgus monkey ( Macaca fascicularis ), rhesus monkeys, hamadryas, and chimpanzee.
- an antigen is diluted and suspended in an appropriate amount of phosphate buffered saline (PBS), physiological saline, or such.
- PBS phosphate buffered saline
- an antigen suspension solution can be administered to mammals after being mixed with an appropriate amount of a standard adjuvant such as Freund's complete adjuvant and emulsified. Then, it is preferable to administer the antigen mixed with an appropriate amount of a Freund's incomplete adjuvant several times every 4 to 21 days.
- a suitable carrier may be used for immunization. After the above immunization, the serum can be examined by standard method with respect to increase in the quantity of the desired antibody.
- Polyclonal antibodies against a peptide of the present invention can be prepared by collecting blood from mammals that have been confirmed with an increase in the serum level of the desired antibody after immunization, and separating the serum from blood by any conventional method.
- a polyclonal antibody may be a polyclonal antibody-containing serum, or a polyclonal antibody-containing fraction may be isolated from the serum.
- Immunoglobulin G or M can be prepared from fractions that recognize only a peptide of the present invention by, for example, using an affinity column conjugated with the peptide of the present invention, and then further purifying the fractions using a protein A or protein G column
- immune cells are collected from the mammals and subjected to cell fusion.
- Immune cells used for cell fusion may be preferably obtained from the spleen.
- a mammalian myeloma cell preferably a myeloma cell that has acquired a property for drug selection of fusion cells can be used.
- the above immune cells can be fused with myeloma cells by following known methods, for example, the method of Milstein et al. (Galfre and Milstein, Methods Enzymol 73: 1981, 3-46).
- Hybridomas obtained by cell fusion can be selected by culturing them in a standard selection medium such as the HAT medium (a medium containing hypoxanthine, aminopterin and thymidine). Cell culturing is typically continued in the HAT medium for a sufficient period of time (for example, several days to several weeks) to allow death of all other cells (non-fused cells) besides the desired hybridomas. Then, hybridoma cells producing the desired antibody can be screened and cloned by performing a standard limiting dilution.
- HAT medium a medium containing hypoxanthine, aminopterin and thymidine
- human lymphocytes such as EB vines-infected lymphocytes can be immunized in vitro with a peptide, cells expressing the peptide, or lysates thereof. Then, the immunized lymphocytes can be fused with immortalized human-derived myeloma cells such as U266 to obtain hybridomas producing a desired human antibody capable of binding to the peptide (JPS63-17688).
- the obtained hybridoma is transplanted into the abdominal cavity of a mouse, and the ascites is extracted.
- the obtained monoclonal antibody can be purified by, for example, ammonium sulfate precipitation, protein A or protein G column, DEAE ion-exchange chromatography, or affinity column conjugated with the peptide of the present invention.
- antibody-producing immune cells such as the immunized lymphocytes can be immortalized by a cancer gene and used for the preparation of monoclonal antibodies.
- the monoclonal antibodies obtained as such can also be prepared by recombination using genetic engineering techniques (see, e.g., Borrebaeck and Larrick, Therapeutic Monoclonal Antibodies published in United Kingdom by MacMillan Publishers LTD (1990)).
- an antibody-encoding DNA can be cloned from immune cells such as antibody-producing hybridoma or immunized lymphocytes and inserted into a suitable vector, and then this is introduced into host cells to prepare a recombinant antibody.
- the present invention also provides recombinant antibodies prepared as described above.
- the antibodies of the present invention may be antibody fragments or modified antibodies, as long as they bind to the peptides of the present invention.
- the antibody fragments may be Fab, F(ab′) 2 , Fv, or a single chain Fv (scFv) in which Fv fragments derived from an H chain and an L chain are linked with a suitable linker (Huston et al., Proc Natl Acad Sci USA 1988, 85: 5879-83). More specifically, antibody fragments may be generated by treating an antibody with an enzyme such as papain or pepsin.
- a gene encoding an antibody fragment may be constructed, inserted into an expression vector, and expressed in an appropriate host cell (see, e.g., Co et al., J Immunol 1994, 152: 2968-76; Better and Horwitz,
- Antibodies may be modified by conjugation with various molecules such as polyethyleneglycol (PEG).
- PEG polyethyleneglycol
- the present invention provides such modified antibodies.
- Modified antibodies can be obtained by chemically modifying the antibodies. These modification methods are conventional in the art.
- the antibodies of the present invention can be obtained as chimeric antibodies of a non-human antibody-derived variable region and a human antibody-derived constant region, or as humanized antibodies comprising a non-human antibody-derived complementarity determining region (CDR) and a human antibody-derived framework region (FR) and constant region.
- CDR complementarity determining region
- FR human antibody-derived framework region
- Such antibodies can be prepared according to known techniques. Humanization can be carried out by substituting a human antibody sequence(s) with a corresponding non-human antibody CDR sequence(s) (see, e.g., Verhoeyen et al., Science 1988, 239: 1534-6).
- humanized antibodies are chimeric antibodies in which the substantially less than an intact human variable domain has been substituted with a corresponding sequence from a non-human species.
- Intact human antibodies comprising a human variable region in addition to the human framework and constant regions can also be used.
- Such antibodies can be generated using various techniques known in the art. For example, in vitro methods include use of recombinant libraries of human antibody fragments presented on bacteriophages (for example, Hoogenboom & Winter, J. Mol. Biol. 1991, 227: 381).
- human antibodies can also be generated by introducing human immunoglobulin gene loci into transgenic animals, for example, mice, in which the endogenous immunoglobulin genes have been partially or completely inactivated. This approach is described in, for example, U.S. Pat. Nos. 6,150,584, 5,545,807, 5,545,806, 5,569,825, 5,625,126, 5,633,425 and 5,661,016.
- Antibodies obtained as described above may be purified to homogeneity.
- antibody separation and purification can be performed according to separation methods and purification methods used for general proteins.
- an antibody can be separated and isolated by appropriately selecting and combining use of column chromatographies such as affinity chromatography, filter, ultrafiltration, salting-out, dialysis, SDS-polyacrylamide gel electrophoresis and isoelectric focusing electrophoresis (Antibodies: A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory (1988)), but are not limited thereto.
- Protein A column and protein G column can be used as the affinity column.
- Exemplary protein A columns to be used include, for example, Hyper D, POROS and Sepharose F. F. (Pharmacia).
- exemplary chromatography includes, for example, ion-exchange chromatography, hydrophobic chromatography, gel filtration, reversed-phase chromatography, adsorption chromatography and such (Strategies for Protein Purification and Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press (1996)).
- the chromatography procedures can be carried out by liquid-phase chromatography such as HPLC and FPLC.
- the antigen-binding activity of an antibody of the present invention can be measured, for example, by using absorbance measurement, enzyme-linked immunosorbent assay (ELISA), enzyme immunoassay (EIA), radioimmunoassay (RIA), and/or immunofluorescence (IF).
- ELISA enzyme-linked immunosorbent assay
- EIA enzyme immunoassay
- RIA radioimmunoassay
- IF immunofluorescence
- an antibody of the present invention is immobilized onto a plate, a peptide of the present invention is applied to the plate, and then a sample containing the desired antibody, such as culture supernatant of antibody-producing cells or purified antibodies, is applied. Next, a secondary antibody that recognizes the primary antibody and is labelled with an enzyme such as alkaline phosphatase is applied and the plate is incubated.
- an enzyme substrate such as p-nitrophenyl phosphate is applied to the plate, and the antigen-binding activity of the sample is evaluated by measuring absorbance.
- an enzyme substrate such as p-nitrophenyl phosphate
- peptide fragments such as C-terminal or N-terminal fragments may be used as an antigen.
- BIAcore Pharmacia
- an antibody of the present invention can be used to detect a peptide of the present invention present in the blood sample (for example, serum sample) of a subject.
- an antibody of the present invention present in the blood sample (for example, serum sample) of a subject can also be detected using a peptide of the present invention.
- the result of measuring a peptide of the present invention or an antibody of the present invention in the blood sample of a subject can be utilized to the subject selection for administration of the pharmaceutical compositions of the present invention or monitoring of the efficacy of the pharmaceutical compositions of the present invention.
- the present invention provides vectors comprising a polynucleotide encoding a peptide of the present invention and host cells introduced with the vectors.
- a vector of the present invention may be used to keep a polynucleotide of the present invention in a host cell, to express a peptide of the present invention in a host cell, or to administer a polynucleotide of the present invention for gene therapy.
- E. coli When E. coli is a host cell and a vector is amplified and produced in a large amount in E. coli (for example, JM109, DH5-alpha, HB101 or XL1-Blue), the vector needs to have a “replication origin” for amplification in E. coli and a marker gene for selection of transformed E. coli (for example, a drug resistance gene selected by a drug such as ampicillin, tetracycline, kanamycin, chloramphenicol).
- a marker gene for selection of transformed E. coli for example, a drug resistance gene selected by a drug such as ampicillin, tetracycline, kanamycin, chloramphenicol.
- the M13-series vectors, pUC-series vectors, pBR322, pBluescript, pCR-Script and such can be used.
- an expression vector can be used.
- an expression vector for expression in E. coli needs to have the above features for amplification in E. coli.
- E. coli such as JM109, DHS-alpha, HB101 or XL1-Blue are used as a host cell, the vector needs to have a promoter, for example, lacZ promoter (Ward et al., Nature 1989, 341: 544-6; FASEB J.
- the vector may contain a signal sequence for peptide secretion.
- An exemplary signal sequence that directs the peptide to be secreted to the periplasm of the E. coli is the pelB signal sequence (Lei et al., J Bacteriol. 1987, 169: 4379).
- Means for introducing the vectors into the target host cells include, for example, the calcium chloride method and the electroporation method.
- expression vectors derived from mammals for example, pcDNA3 (Invitrogen) and pEGF-BOS (Nucleic Acids Res 1990, 18(17): 5322), pEF, pCDM8), expression vectors derived from insect cells (for example, “Bac-to-BAC baculovirus expression system” (GIBCO BRL), pBacPAK8), expression vectors derived from plants (e.g., pMH1, pMH2), expression vectors derived from animal viruses (e.g., pHSV, pMV, pAdexLcw), expression vectors derived from retroviruses (e.g., pZIpneo), expression vectors derived from yeast (e.g., “Pichia Expression Kit” (Invitrogen), pNV11, SP-Q01) and expression vectors derived from Bacillus subtilis (e.g., pPL608, pKTH50)
- mammals for example, pcDNA3
- the vector In order to express the vector in animal cells such as CHO, COS or NIH3T3 cells, the vector needs to carry a promoter necessary for expression in such cells, for example, the SV40 promoter (Mulligan et al., Nature 1979, 277: 108), the MMLV-LTR promoter, the EF1-alpha promoter (Mizushima et al., Nucleic Acids Res. 1990, 18: 5322), the CMV promoter and the like, and preferably a marker gene for selecting transformants (for example, a drug resistance gene selected by a drug (e.g., neomycin, G418)).
- a promoter necessary for expression in such cells for example, the SV40 promoter (Mulligan et al., Nature 1979, 277: 108), the MMLV-LTR promoter, the EF1-alpha promoter (Mizushima et al., Nucleic Acids Res. 1990, 18: 5322
- the kit of the present invention further comprises an HLA-A01 detection reagent additionally.
- C1R an HLA-A- and HLA-B-negative human B lymphoblastoid cell line
- COS7 an African green monkey kidney cell line
- C1R cells that constitutively express HLA-A*01:01 was used as cells for stimulating CTLs.
- a cDNA encoding the HLA-A*01:01 gene was amplified by PCR and introduced into an expression vector.
- C1R cells incorporated with HLA-A*01:01 gene expression vector were cultured for two weeks under drug selection with a medium containing G418 (Invitrogen). After diluting G418-resistant C1R cell suspension, the cells were seeded into a 96-well plate, and subjected to selective culture for another 30 days with a medium containing G418.
- the HLA-A*01:01 expression in C1R cells was verified by flow cytometric analysis.
- CDCA1-derived 8 mer, 9 mer and 10 mer peptides that are expected to bind to HLA-A*01:01 were determined using the binding prediction server “NetMHC 3.2” (www.cbs.dtu.dk/services/NetMHC-3.20 (Buus et al., Tissue Antigens. 2003, 62(5): 378-84; Nielsen et al., Protein Sci. 2003, 12(5): 1007-17; Lundegaard C et al., Bioinformatics.
- the peptides were chemically synthesized by American Peptide Company (Sunnyvale, Calif.) according to a standard solid-phase synthesis method, and purified by reversed phase high-performance liquid chromatography (HPLC). The quality (purity of 90% or higher) of the peptides was guaranteed by HPLC and mass spectrometry.
- the peptides were dissolved in dimethylsulfoxide (final concentration: 20 mg/ml) and stored at ⁇ 80 degrees C.
- DCs Monocyte-derived dendritic cells
- HLAs human leukocyte antigens
- PBMCs peripheral blood mononuclear cells
- HLA-A*01:01-positive Ficoll-Paque plus solution
- Pharmacia Ficoll-Paque plus solution
- the cells were cultured in the presence of 1000 IU/ml granulocyte macrophage colony-stimulating factor (R&D System) and 1000 IU/ml interleukin(IL)-4 (R&D System) for seven days.
- R&D System granulocyte macrophage colony-stimulating factor
- IL interleukin
- As medium an AIM-V medium (Invitrogen) containing inactivated AB type serum (MP Biomedicals) was used (2% ABS/AIM-V medium).
- DCs induced to differentiate from monocytes by cytokines were pulsed with 20 micro-g/ml each of the synthesized peptides (37 degrees C., for three hours). Peptide pulsing was performed in the AIM-V medium. These peptide-pulsed DCs were inactivated by X-ray irradiation (20 Gy), and mixed in a 1:20 ratio with autologous CD8-positive T cells obtained by using the CD8 Positive Isolation Kit (Invitrogen) (1.5 ⁇ 10 4 DCs and 3 ⁇ 10 5 CD8-positive T cells), and cultured in a 48-well plate (Coming).
- the vokmie of 2% ABS/AIM-V medium was 0.5 ml per 1 well, and IL-7 (R&D System) was added (final concentration: 10 ng/ml) to the well.
- IL-7 R&D System
- IL-2 Novartis
- IFN-gamma production of CD8-positive T cells against peptide-pulsed C1R-A01 was confirmed by enzyme-linked immunospot (ELISPOT) assay (Tanaka H et al., Br J Cancer 2001, 84(1): 94-9; Umano Y et al., Br J Cancer 2001, 84(8): 1052-7; Uchida N et al., Clin Cancer Res 2004, 10(24): 8577-86; Suda T et al., Cancer Sci 2006, 97(5): 411-9; Watanabe T et al., Cancer Sci 2005, 96(8): 498-506).
- ELISPOT enzyme-linked immunospot
- CTLs were propagated using a method similar to that reported by Riddell et al. (Walter E A et al., N Engl J Med 1995, 333(16): 1038-44; Riddell SR et al., Nat Med 1996, 2(2): 216-23).
- FALCON tissue culture
- the CTLs were cultured in 5% ABS/AIM-V medium (culture solution volume: 25 ml/flask) with two types of human B lymphoblastoid cell lines (5 ⁇ 10 6 cells each) treated with mitomycin C and anti-CD3 antibody (BD biosciences, final concentration: 40 ng/ml).
- IL-2 was added to the culture (IL-2 final concentration: 120 IU/ml).
- the medium was changed to 5% ABS/AIM-V medium containing 60 IU/ml IL-2 (IL-2 final concentration: 30 IU/ml) (Tanaka H et al., Br J Cancer 2001, 84(1): 94-9; Umano Y et al., Br J Cancer 2001, 84(8): 1052-7; Uchida N et al., Clin Cancer Res 2004, 10(24): 8577-86; Suda T et al., Cancer Sci 2006, 97(5): 411-9; Watanabe T et al., Cancer Sci 2005, 96(8): 498-506).
- the CTLs were seeded at 1 cell/well or 10 cells/well in a 96-well round-bottomed microplate (Corning).
- the CTLs were cultured in 5% ABS/AIM-V medium (culture solution volume: 150 ⁇ l/well) with two types of human B lymphoblastoid cell lines (1 ⁇ 10 4 cells each) treated with mitomycin C, anti-CD3 antibody (final concentration: 30 ng/ml), and IL-2 (final concentration: 125 Mimi).
- 50 ⁇ l of 5% ABS/AIM-V medium containing 500 IU/ml IL-2 was added to the culture.
- the IFN-gamma ELISPOT assay and IFN-gamma ELISA were performed to confirm peptide-specific IFN-gamma production of CTLs induced by a peptide.
- C1R-A01 pulsed with a peptide was prepared as target cells.
- the IFN-gamma ELISPOT assay and IFN-gamma ELISA were performed according to the procedures recommended by the assay kit manufacturer.
- the cDNA encoding the CDCA1 or HLA-A*01:01 gene were amplified by PCR. Each PCR amplified product was incorporated into an expression vector. Using Lipofectamine 2000 (Invitrogen), either or both of the CDCA1 gene expression vector and the HLA-A*01:01 gene expression vector were introduced into COST cells, an HLA-negative cell line, according to the procedure recommended by the manufacturer. On the next day of the gene introduction, the COST cells were separated using Versene (Invitrogen) and used as target cells.
- Table 1a, Table 1 b, and Table 1c show 8 mer, 9 mer, and 10 mer peptides derived from CDCA1 that were predicted to bind to HLA-A*01:01 by “NetMHC 3.2” in descending order of binding affinity, respectively.
- Table 2a and Table 2b show 9 mer and 10 mer peptides derived from CDCA1 that were predicted to bind to HLA-A*01:01 by “BIMAS” in descending order of binding score.
- Table 3a and Table 3b show 9 mer and 10 mer peptides derived from CDCA1 that were predicted to bind to HLA-A*01:01 by “SYFPEITHI” in descending order of binding score.
- the number of Start Position in each table indicates the position of the first amino acid of the peptide when counted from the N-terminus of the CDCA1 protein. Thirty-nine peptides in total potentially having HLA-A*01:01 binding ability became epitope peptide candidates.
- CTL clones were established from cells in Well #2 which showed IFN-gamma production specific to CDCA1-A01-10-136 (SEQ ID NO: 8) ( FIG. 1 g ), Well #6 which showed IFN-gamma production specific to CDCA1-A01-10-56 (SEQ ID NO: 10) ( FIG. 1 h ), and Well #6 which showed IFN-gamma production specific to CDCA1-A01-10-48 (SEQ ID NO: 13) ( FIG. 1 i ) in the IFN-gamma ELISPOT assay.
- CTL clones stimulated by CDCA1-A01-10-136 (SEQ ID NO: 8), CDCA1-A01-10-56 (SEQ ID NO: 10), or CDCA1-A01-10-48 (SEQ ID NO: 13) showed peptide-specific IFN-gamma production ( FIG. 2 ).
- CTL clones specific to CDCA1-A01-10-136 (SEQ ID NO: 8) against target cells expressing CDCA1 and HLA-A*01:01 was verified.
- COS7 cells expressing both CDCA1 and HLA-A*01:01 were prepared as target cells.
- COS7 cells expressing either CDCA1 or HLA-A*01:01 were prepared as negative control cells.
- CTL clones specific to CDCA1-A01-10-136 (SEQ ID NO: 8) showed IFN-gamma production against COS7 cells expressing both CDCA1 and HLA-A*01:01 ( FIG. 3 ). By contrast, no significant IFN-gamma production was observed against negative control cells.
- CDCA1-A01-10-136 (SEQ ID NO: 8) is a peptide produced by antigen processing and is presented on the cell surface together with the HLA-A01 molecule and recognized by CTLs.
- CDCA1-A01-10-136 (SEQ ID NO: 8) is useful as a cancer vaccine targeting cancer patients with elevated CDCA1 expression.
- CDCA1-A01-8-138 SEQ ID NO: 1
- CDCA1-A01-9-290 SEQ ID NO: 25
- CDCA1-A01-9-130 SEQ ID NO: 33
- CDCA1-A01-9-246 SEQ ID NO: 34
- CDCA1-A01-9-268 SEQ ID NO: 35
- CDCA1-A01-9-288 SEQ ID NO: 37
- CDCA1-A01-10-136 SEQ ID NO: 8
- CDCA1-A01-10-56 SEQ ID NO: 10
- CDCA1-A01-10-48 SEQ ID NO: 13
- CDCA1-A01-8-138 SEQ ID NO: 1
- CDCA1-A01-9-290 SEQ ID NO: 25
- CDCA1-A01-9-130 SEQ ID NO: 33
- CDCA1-A01-9-246 SEQ ID NO: 34
- CDCA1-A01-9-268 SEQ ID NO: 35
- CDCA1-A01-9-288 SEQ ID NO: 37
- CDCA1-A01-10-136 SEQ ID NO: 8
- CDCA1-A01-10-56 SEQ ID NO: 10
- CDCA1-A01-10-48 SEQ ID NO: 13
- the homology analysis of the peptide sequences was performed using the BLAST algorithm (http://blast.ncbi.nlm.nih.gov/Blast.cgi).
- CDCA1-A01-8-138 SEQ ID NO: 1
- CDCA1-A01-9-290 SEQ ID NO: 25
- CDCA1 A01-9-130 SEQ ID NO: 33
- CDCA1-A01-9-246 SEQ ID NO: 34
- CDCA1-A01-9-268 SEQ ID NO: 35
- CDCA1-A01-9-288 SEQ ID NO: 37
- CDCA1-A01-10-136 SEQ ID NO: 8
- CDCA1 A01-10-56 SEQ ID NO: 10
- CDCA1-A01-10-48 SEQ ID NO: 13
- the present invention provides CDCA 1-derived novel HLA-A01-restricted epitope peptides that induce a potent and specific anti-tumor immune response and thus have applicability for a wide range of cancer types.
- the peptides, compositions, APCs. and CTLs in the present invention can be used as a peptide vaccine for cancer expressing CDCA1, for example, bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer, small-cell lung cancer, head and neck cancer, soft tissue tumor, and colon cancer.
- CDCA1 for example, bladder cancer, breast cancer, cervical cancer, cholangiocellular cancer, chronic myeloid leukemia (CML), esophagus cancer, gastric cancer, non-small-cell lung cancer, lymphoma, osteosarcoma, prostate cancer, kidney cancer,
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Immunology (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Cell Biology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biomedical Technology (AREA)
- Mycology (AREA)
- Zoology (AREA)
- Hematology (AREA)
- Genetics & Genomics (AREA)
- Biotechnology (AREA)
- Biochemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Wood Science & Technology (AREA)
- Oncology (AREA)
- Biophysics (AREA)
- Virology (AREA)
- Developmental Biology & Embryology (AREA)
- Urology & Nephrology (AREA)
- General Engineering & Computer Science (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Endocrinology (AREA)
- Pulmonology (AREA)
- Physics & Mathematics (AREA)
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2018145607 | 2018-08-02 | ||
JP2018-145607 | 2018-08-02 | ||
PCT/JP2019/030117 WO2020027239A1 (ja) | 2018-08-02 | 2019-08-01 | Cdca1由来ペプチドおよびそれを含むワクチン |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2019/030117 A-371-Of-International WO2020027239A1 (ja) | 2018-08-02 | 2019-08-01 | Cdca1由来ペプチドおよびそれを含むワクチン |
Related Child Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US18/094,167 Continuation US20230159588A1 (en) | 2018-08-02 | 2023-01-06 | Cdca1-derived peptide and vaccine containing same |
Publications (1)
Publication Number | Publication Date |
---|---|
US20210371462A1 true US20210371462A1 (en) | 2021-12-02 |
Family
ID=69232532
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US17/264,277 Abandoned US20210371462A1 (en) | 2018-08-02 | 2019-08-01 | Cdca1-derived peptide and vaccine containing same |
US18/094,167 Pending US20230159588A1 (en) | 2018-08-02 | 2023-01-06 | Cdca1-derived peptide and vaccine containing same |
Family Applications After (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US18/094,167 Pending US20230159588A1 (en) | 2018-08-02 | 2023-01-06 | Cdca1-derived peptide and vaccine containing same |
Country Status (13)
Country | Link |
---|---|
US (2) | US20210371462A1 (de) |
EP (1) | EP3831943A4 (de) |
JP (1) | JP7448124B2 (de) |
KR (1) | KR20210040106A (de) |
CN (1) | CN112839950A (de) |
AU (1) | AU2019313948A1 (de) |
BR (1) | BR112021001664A2 (de) |
CA (1) | CA3108403A1 (de) |
IL (1) | IL280450A (de) |
MX (1) | MX2021001357A (de) |
SG (1) | SG11202101090RA (de) |
TW (1) | TW202023581A (de) |
WO (1) | WO2020027239A1 (de) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN111363009B (zh) * | 2020-03-18 | 2020-10-30 | 北京鼎成肽源生物技术有限公司 | 一种直肠癌靶标抗原及其刺激培养的ctl细胞及其应用 |
Family Cites Families (34)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4722848A (en) | 1982-12-08 | 1988-02-02 | Health Research, Incorporated | Method for immunizing animals with synthetically modified vaccinia virus |
JPS60217905A (ja) | 1984-04-13 | 1985-10-31 | のむら産業株式会社 | 自動包装機 |
GB8823869D0 (en) | 1988-10-12 | 1988-11-16 | Medical Res Council | Production of antibodies |
US5703055A (en) | 1989-03-21 | 1997-12-30 | Wisconsin Alumni Research Foundation | Generation of antibodies through lipid mediated DNA delivery |
US6150584A (en) | 1990-01-12 | 2000-11-21 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
US5545806A (en) | 1990-08-29 | 1996-08-13 | Genpharm International, Inc. | Ransgenic non-human animals for producing heterologous antibodies |
US5633425A (en) | 1990-08-29 | 1997-05-27 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
ATE158021T1 (de) | 1990-08-29 | 1997-09-15 | Genpharm Int | Produktion und nützung nicht-menschliche transgentiere zur produktion heterologe antikörper |
US5661016A (en) | 1990-08-29 | 1997-08-26 | Genpharm International Inc. | Transgenic non-human animals capable of producing heterologous antibodies of various isotypes |
US5625126A (en) | 1990-08-29 | 1997-04-29 | Genpharm International, Inc. | Transgenic non-human animals for producing heterologous antibodies |
US5679647A (en) | 1993-08-26 | 1997-10-21 | The Regents Of The University Of California | Methods and devices for immunizing a host against tumor-associated antigens through administration of naked polynucleotides which encode tumor-associated antigenic peptides |
US5804566A (en) | 1993-08-26 | 1998-09-08 | The Regents Of The University Of California | Methods and devices for immunizing a host through administration of naked polynucleotides with encode allergenic peptides |
US5739118A (en) | 1994-04-01 | 1998-04-14 | Apollon, Inc. | Compositions and methods for delivery of genetic material |
US5736524A (en) | 1994-11-14 | 1998-04-07 | Merck & Co.,. Inc. | Polynucleotide tuberculosis vaccine |
US5922687A (en) | 1995-05-04 | 1999-07-13 | Board Of Trustees Of The Leland Stanford Junior University | Intracellular delivery of nucleic acids using pressure |
US5853719A (en) | 1996-04-30 | 1998-12-29 | Duke University | Methods for treating cancers and pathogen infections using antigen-presenting cells loaded with RNA |
WO1998004720A1 (en) | 1996-07-26 | 1998-02-05 | Sloan-Kettering Institute For Cancer Research | Method and reagents for genetic immunization |
FR2766205B1 (fr) | 1997-07-16 | 2002-08-30 | Inst Nat Sante Rech Med | Nouveau procede de sensibilisation de cellules presentatrices d'antigene et nouveaux moyens pour la mise en oeuvre du procede |
NZ508996A (en) | 1998-06-25 | 2003-09-26 | Kyogo Itoh | Tumor antigen peptides derived from cyclophilin B |
AU2002226030A1 (en) * | 2000-12-04 | 2002-06-18 | Argonex Pharmaceuticals | Cytotoxic t-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer |
WO2002072631A2 (en) | 2001-03-14 | 2002-09-19 | Dakocytomation Denmark A/S | Mhc molecule constructs and their usesfor diagnosis and therapy |
WO2009003492A1 (en) | 2007-07-03 | 2009-01-08 | Dako Denmark A/S | Mhc multimers, methods for their generation, labeling and use |
EP1730534A2 (de) | 2004-03-24 | 2006-12-13 | Oncotherapy Science, Inc. | Zusammensetzung und verfahren zur behandlung von lungenkrebs |
WO2006031221A1 (en) | 2004-09-13 | 2006-03-23 | Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services | Compositions comprising t cell receptors and methods of use thereof |
US7943295B2 (en) | 2005-07-29 | 2011-05-17 | Oncotherapy Science, Inc. | Screening and therapeutic method for NSCLC targeting CDCA1-KNTC2 complex |
US8003770B2 (en) | 2005-09-13 | 2011-08-23 | Mie University | T-cell receptor and nucleic acid encoding the receptor |
JP5493076B2 (ja) | 2007-08-20 | 2014-05-14 | オンコセラピー・サイエンス株式会社 | Cdca1ペプチド及びこれを含む薬剤 |
TWI526219B (zh) * | 2008-06-19 | 2016-03-21 | 腫瘤療法 科學股份有限公司 | Cdca1抗原決定位胜肽及含此胜肽的疫苗 |
GB201004575D0 (en) * | 2010-03-19 | 2010-05-05 | Immatics Biotechnologies Gmbh | Composition of tumor associated peptides and related anti cancer vaccine for the treatment of gastric cancer and other cancers |
JP6255593B2 (ja) * | 2012-07-10 | 2018-01-10 | オンコセラピー・サイエンス株式会社 | Th1細胞のCDCA1エピトープペプチドおよびこれを含有するワクチン |
CN111925431A (zh) * | 2014-08-04 | 2020-11-13 | 肿瘤疗法科学股份有限公司 | Koc1衍生的肽和包含它们的疫苗 |
US10676514B2 (en) * | 2014-08-04 | 2020-06-09 | Oncotherapy Science, Inc. | CDCA1-derived peptide and vaccine containing same |
IL257335B (en) * | 2015-08-12 | 2022-08-01 | Oncotherapy Science Inc | A depdc1-derived peptide and a component containing it |
MX2018004308A (es) * | 2015-10-08 | 2018-05-01 | Oncotherapy Science Inc | Peptido derivado de foxm1 y vacuna que lo incluye. |
-
2019
- 2019-07-31 TW TW108127154A patent/TW202023581A/zh unknown
- 2019-08-01 CA CA3108403A patent/CA3108403A1/en active Pending
- 2019-08-01 AU AU2019313948A patent/AU2019313948A1/en not_active Withdrawn
- 2019-08-01 BR BR112021001664-3A patent/BR112021001664A2/pt not_active Application Discontinuation
- 2019-08-01 KR KR1020217006095A patent/KR20210040106A/ko unknown
- 2019-08-01 US US17/264,277 patent/US20210371462A1/en not_active Abandoned
- 2019-08-01 JP JP2020534728A patent/JP7448124B2/ja active Active
- 2019-08-01 WO PCT/JP2019/030117 patent/WO2020027239A1/ja unknown
- 2019-08-01 SG SG11202101090RA patent/SG11202101090RA/en unknown
- 2019-08-01 EP EP19844090.1A patent/EP3831943A4/de active Pending
- 2019-08-01 CN CN201980064092.0A patent/CN112839950A/zh active Pending
- 2019-08-01 MX MX2021001357A patent/MX2021001357A/es unknown
-
2021
- 2021-01-27 IL IL280450A patent/IL280450A/en unknown
-
2023
- 2023-01-06 US US18/094,167 patent/US20230159588A1/en active Pending
Non-Patent Citations (3)
Title |
---|
Biosynth entry (retrieved from https://www.biosynth.com/p/MM30170/9049-98-3-mannide-monooleate on 9/20/22, 2 pages) (Year: 2022) * |
NCBI entry (retrieved from https://www.ncbi.nlm.nih.gov/books/NBK304428/ on 9/20/22, 15 pages) (Year: 2022) * |
Stills HF (‘Adjuvants and antibody production:dispelling the myths associated with Freund’s complete and other adjuvants’ ILAR Journal v46(3) 2005 pages 280-293) (Year: 2005) * |
Also Published As
Publication number | Publication date |
---|---|
EP3831943A1 (de) | 2021-06-09 |
BR112021001664A2 (pt) | 2021-05-04 |
CN112839950A (zh) | 2021-05-25 |
MX2021001357A (es) | 2021-06-23 |
TW202023581A (zh) | 2020-07-01 |
CA3108403A1 (en) | 2020-02-06 |
WO2020027239A1 (ja) | 2020-02-06 |
US20230159588A1 (en) | 2023-05-25 |
KR20210040106A (ko) | 2021-04-12 |
EP3831943A4 (de) | 2022-12-21 |
AU2019313948A1 (en) | 2021-03-04 |
JP7448124B2 (ja) | 2024-03-12 |
SG11202101090RA (en) | 2021-03-30 |
IL280450A (en) | 2021-03-25 |
JPWO2020027239A1 (ja) | 2021-08-10 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20220112241A1 (en) | Foxm1-derived peptide, and vaccine including same | |
US11673915B2 (en) | DEPDC1-derived peptide and vaccine containing same | |
US11547723B2 (en) | KOC1-derived peptide and vaccine including same | |
AU2020202681B2 (en) | CDCA1-derived peptide and vaccine containing same | |
US20230159588A1 (en) | Cdca1-derived peptide and vaccine containing same | |
AU2020201216B2 (en) | URLC10-derived peptide and vaccine containing same | |
US10793599B2 (en) | MPHOSPH1-derived peptide, and vaccine including same |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AS | Assignment |
Owner name: ONCOTHERAPY SCIENCE, INC., JAPAN Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:YAMASHITA, SACHIKO;HIKICHI, TETSURO;SIGNING DATES FROM 20210412 TO 20210420;REEL/FRAME:056754/0603 |
|
AS | Assignment |
Owner name: ONCOTHERAPY SCIENCE, INC., JAPAN Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:YAMASHITA, SACHIKO;HIKICHI, TETSURO;SIGNING DATES FROM 20210410 TO 20210412;REEL/FRAME:056884/0739 |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: NON FINAL ACTION MAILED |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: FINAL REJECTION MAILED |
|
STCB | Information on status: application discontinuation |
Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION |