WO2025064470A1 - Viral peptides and uses thereof - Google Patents
Viral peptides and uses thereof Download PDFInfo
- Publication number
- WO2025064470A1 WO2025064470A1 PCT/US2024/047158 US2024047158W WO2025064470A1 WO 2025064470 A1 WO2025064470 A1 WO 2025064470A1 US 2024047158 W US2024047158 W US 2024047158W WO 2025064470 A1 WO2025064470 A1 WO 2025064470A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- peptide
- isolated
- cell
- molecule
- peptides
- Prior art date
Links
- 108090000765 processed proteins & peptides Proteins 0.000 title claims abstract description 607
- 102000004196 processed proteins & peptides Human genes 0.000 title claims abstract description 271
- 230000003612 virological effect Effects 0.000 title description 24
- 230000027455 binding Effects 0.000 claims abstract description 153
- 238000000034 method Methods 0.000 claims abstract description 103
- 241000714260 Human T-lymphotropic virus 1 Species 0.000 claims abstract description 94
- 239000013598 vector Substances 0.000 claims abstract description 75
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 67
- 102000040430 polynucleotide Human genes 0.000 claims abstract description 48
- 108091033319 polynucleotide Proteins 0.000 claims abstract description 48
- 239000002157 polynucleotide Substances 0.000 claims abstract description 48
- 239000008194 pharmaceutical composition Substances 0.000 claims abstract description 42
- 201000010099 disease Diseases 0.000 claims abstract description 39
- 208000015181 infectious disease Diseases 0.000 claims abstract description 23
- 210000004027 cell Anatomy 0.000 claims description 192
- 108700018351 Major Histocompatibility Complex Proteins 0.000 claims description 163
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 claims description 161
- 108091008874 T cell receptors Proteins 0.000 claims description 157
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 claims description 157
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 96
- 150000001413 amino acids Chemical class 0.000 claims description 96
- 239000000427 antigen Substances 0.000 claims description 82
- 108091007433 antigens Proteins 0.000 claims description 80
- 102000036639 antigens Human genes 0.000 claims description 80
- 239000012634 fragment Substances 0.000 claims description 79
- 108020001507 fusion proteins Proteins 0.000 claims description 52
- 102000037865 fusion proteins Human genes 0.000 claims description 52
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 39
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 claims description 39
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 39
- 102000008949 Histocompatibility Antigens Class I Human genes 0.000 claims description 34
- 239000002105 nanoparticle Substances 0.000 claims description 31
- 230000028993 immune response Effects 0.000 claims description 30
- 108010088652 Histocompatibility Antigens Class I Proteins 0.000 claims description 29
- 208000035475 disorder Diseases 0.000 claims description 28
- 208000016683 Adult T-cell leukemia/lymphoma Diseases 0.000 claims description 27
- 201000006966 adult T-cell leukemia Diseases 0.000 claims description 27
- 210000000612 antigen-presenting cell Anatomy 0.000 claims description 27
- 150000003839 salts Chemical class 0.000 claims description 24
- 239000002245 particle Substances 0.000 claims description 20
- 210000002865 immune cell Anatomy 0.000 claims description 18
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 15
- 208000006961 tropical spastic paraparesis Diseases 0.000 claims description 15
- 125000003729 nucleotide group Chemical group 0.000 claims description 14
- 239000002773 nucleotide Substances 0.000 claims description 13
- 230000001939 inductive effect Effects 0.000 claims description 12
- 239000002671 adjuvant Substances 0.000 claims description 11
- 210000000265 leukocyte Anatomy 0.000 claims description 11
- 239000013603 viral vector Substances 0.000 claims description 10
- 239000003937 drug carrier Substances 0.000 claims description 9
- 238000007385 chemical modification Methods 0.000 claims description 8
- 108020004999 messenger RNA Proteins 0.000 claims description 8
- 239000013604 expression vector Substances 0.000 claims description 7
- 210000000822 natural killer cell Anatomy 0.000 claims description 7
- 210000002540 macrophage Anatomy 0.000 claims description 6
- 230000002441 reversible effect Effects 0.000 claims description 6
- 206010028570 Myelopathy Diseases 0.000 claims description 5
- 206010044696 Tropical spastic paresis Diseases 0.000 claims description 5
- 238000004806 packaging method and process Methods 0.000 claims description 3
- 210000003527 eukaryotic cell Anatomy 0.000 claims description 2
- 125000003473 lipid group Chemical group 0.000 claims description 2
- 210000001236 prokaryotic cell Anatomy 0.000 claims description 2
- 239000000203 mixture Substances 0.000 abstract description 73
- 238000011282 treatment Methods 0.000 abstract description 23
- 241000700605 Viruses Species 0.000 abstract description 19
- 229960005486 vaccine Drugs 0.000 abstract description 16
- 230000009467 reduction Effects 0.000 abstract description 5
- 230000002265 prevention Effects 0.000 abstract description 4
- 230000002163 immunogen Effects 0.000 abstract description 3
- 235000001014 amino acid Nutrition 0.000 description 102
- 229940024606 amino acid Drugs 0.000 description 98
- 108090000623 proteins and genes Proteins 0.000 description 77
- 229920001184 polypeptide Polymers 0.000 description 70
- 102000004169 proteins and genes Human genes 0.000 description 57
- -1 HLA-A02 Proteins 0.000 description 55
- 235000018102 proteins Nutrition 0.000 description 52
- 125000005647 linker group Chemical group 0.000 description 42
- 150000007523 nucleic acids Chemical group 0.000 description 35
- 102000015736 beta 2-Microglobulin Human genes 0.000 description 32
- 108010081355 beta 2-Microglobulin Proteins 0.000 description 32
- 206010028980 Neoplasm Diseases 0.000 description 30
- 108020004414 DNA Proteins 0.000 description 29
- 102000043131 MHC class II family Human genes 0.000 description 29
- 108091054438 MHC class II family Proteins 0.000 description 29
- 102000039446 nucleic acids Human genes 0.000 description 26
- 108020004707 nucleic acids Proteins 0.000 description 26
- 230000003211 malignant effect Effects 0.000 description 25
- 102000004127 Cytokines Human genes 0.000 description 23
- 108090000695 Cytokines Proteins 0.000 description 23
- 208000009746 Adult T-Cell Leukemia-Lymphoma Diseases 0.000 description 22
- 230000004048 modification Effects 0.000 description 22
- 238000012986 modification Methods 0.000 description 22
- 101001100327 Homo sapiens RNA-binding protein 45 Proteins 0.000 description 21
- 108091054437 MHC class I family Proteins 0.000 description 21
- 102100038823 RNA-binding protein 45 Human genes 0.000 description 21
- 230000000694 effects Effects 0.000 description 21
- 230000015572 biosynthetic process Effects 0.000 description 20
- 125000000539 amino acid group Chemical group 0.000 description 18
- 230000000890 antigenic effect Effects 0.000 description 18
- 238000009472 formulation Methods 0.000 description 18
- 239000000243 solution Substances 0.000 description 18
- 241000598436 Human T-cell lymphotropic virus Species 0.000 description 17
- 108060003951 Immunoglobulin Proteins 0.000 description 17
- 102000018358 immunoglobulin Human genes 0.000 description 17
- 239000003446 ligand Substances 0.000 description 17
- 102100028972 HLA class I histocompatibility antigen, A alpha chain Human genes 0.000 description 16
- 108010075704 HLA-A Antigens Proteins 0.000 description 16
- 102000043129 MHC class I family Human genes 0.000 description 16
- 239000002253 acid Substances 0.000 description 15
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 14
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 14
- 230000006870 function Effects 0.000 description 14
- 239000000178 monomer Substances 0.000 description 14
- 238000006467 substitution reaction Methods 0.000 description 14
- 101000716102 Homo sapiens T-cell surface glycoprotein CD4 Proteins 0.000 description 13
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 13
- 230000003993 interaction Effects 0.000 description 13
- 230000000670 limiting effect Effects 0.000 description 13
- 102000014150 Interferons Human genes 0.000 description 12
- 108010050904 Interferons Proteins 0.000 description 12
- 230000004913 activation Effects 0.000 description 12
- 239000002585 base Substances 0.000 description 12
- 201000011510 cancer Diseases 0.000 description 12
- 239000003795 chemical substances by application Substances 0.000 description 12
- 238000001727 in vivo Methods 0.000 description 12
- 229940079322 interferon Drugs 0.000 description 12
- 210000004698 lymphocyte Anatomy 0.000 description 12
- 230000035772 mutation Effects 0.000 description 12
- 108700028369 Alleles Proteins 0.000 description 11
- 239000003153 chemical reaction reagent Substances 0.000 description 11
- 150000001875 compounds Chemical class 0.000 description 11
- 235000018417 cysteine Nutrition 0.000 description 11
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 11
- 230000004927 fusion Effects 0.000 description 11
- 230000001404 mediated effect Effects 0.000 description 11
- 229920001223 polyethylene glycol Polymers 0.000 description 11
- 229920000642 polymer Polymers 0.000 description 11
- 239000007787 solid Substances 0.000 description 11
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 10
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 10
- 108010002350 Interleukin-2 Proteins 0.000 description 10
- 102000000588 Interleukin-2 Human genes 0.000 description 10
- 239000011230 binding agent Substances 0.000 description 10
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 10
- 239000010931 gold Substances 0.000 description 10
- 238000003786 synthesis reaction Methods 0.000 description 10
- 201000009030 Carcinoma Diseases 0.000 description 9
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 9
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 9
- 108091028043 Nucleic acid sequence Proteins 0.000 description 9
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 9
- 125000000217 alkyl group Chemical group 0.000 description 9
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 9
- 239000003814 drug Substances 0.000 description 9
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 8
- 102100028976 HLA class I histocompatibility antigen, B alpha chain Human genes 0.000 description 8
- 108010058607 HLA-B Antigens Proteins 0.000 description 8
- 101000946843 Homo sapiens T-cell surface glycoprotein CD8 alpha chain Proteins 0.000 description 8
- 108010090804 Streptavidin Proteins 0.000 description 8
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 8
- 238000007792 addition Methods 0.000 description 8
- 150000001408 amides Chemical class 0.000 description 8
- 238000013459 approach Methods 0.000 description 8
- 239000002775 capsule Substances 0.000 description 8
- 230000008878 coupling Effects 0.000 description 8
- 238000005859 coupling reaction Methods 0.000 description 8
- 210000004443 dendritic cell Anatomy 0.000 description 8
- 229910052737 gold Inorganic materials 0.000 description 8
- 238000002347 injection Methods 0.000 description 8
- 239000007924 injection Substances 0.000 description 8
- 230000003834 intracellular effect Effects 0.000 description 8
- 238000002360 preparation method Methods 0.000 description 8
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 8
- 239000003826 tablet Substances 0.000 description 8
- 230000008685 targeting Effects 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 241001430294 unidentified retrovirus Species 0.000 description 8
- 108020004635 Complementary DNA Proteins 0.000 description 7
- 102100034349 Integrase Human genes 0.000 description 7
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 7
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 7
- 102100029215 Signaling lymphocytic activation molecule Human genes 0.000 description 7
- 208000009956 adenocarcinoma Diseases 0.000 description 7
- 230000001580 bacterial effect Effects 0.000 description 7
- 229960002685 biotin Drugs 0.000 description 7
- 235000020958 biotin Nutrition 0.000 description 7
- 239000011616 biotin Substances 0.000 description 7
- 238000010804 cDNA synthesis Methods 0.000 description 7
- 238000006243 chemical reaction Methods 0.000 description 7
- 239000002299 complementary DNA Substances 0.000 description 7
- 238000010168 coupling process Methods 0.000 description 7
- 230000001086 cytosolic effect Effects 0.000 description 7
- 238000012217 deletion Methods 0.000 description 7
- 230000037430 deletion Effects 0.000 description 7
- 238000006471 dimerization reaction Methods 0.000 description 7
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 7
- 150000002632 lipids Chemical group 0.000 description 7
- 239000002502 liposome Substances 0.000 description 7
- 238000004519 manufacturing process Methods 0.000 description 7
- 238000004949 mass spectrometry Methods 0.000 description 7
- 230000007935 neutral effect Effects 0.000 description 7
- 238000003752 polymerase chain reaction Methods 0.000 description 7
- 102000005962 receptors Human genes 0.000 description 7
- 108020003175 receptors Proteins 0.000 description 7
- 239000002904 solvent Substances 0.000 description 7
- 239000000126 substance Substances 0.000 description 7
- 235000000346 sugar Nutrition 0.000 description 7
- 230000001225 therapeutic effect Effects 0.000 description 7
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 6
- 241000894006 Bacteria Species 0.000 description 6
- 241000588724 Escherichia coli Species 0.000 description 6
- 241000238631 Hexapoda Species 0.000 description 6
- 101000662909 Homo sapiens T cell receptor beta constant 1 Proteins 0.000 description 6
- 101000662902 Homo sapiens T cell receptor beta constant 2 Proteins 0.000 description 6
- 102100037850 Interferon gamma Human genes 0.000 description 6
- 108010074328 Interferon-gamma Proteins 0.000 description 6
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 6
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 6
- 241000699666 Mus <mouse, genus> Species 0.000 description 6
- 108091034117 Oligonucleotide Proteins 0.000 description 6
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 6
- 102100037272 T cell receptor beta constant 1 Human genes 0.000 description 6
- 102100037298 T cell receptor beta constant 2 Human genes 0.000 description 6
- DTQVDTLACAAQTR-UHFFFAOYSA-N Trifluoroacetic acid Chemical compound OC(=O)C(F)(F)F DTQVDTLACAAQTR-UHFFFAOYSA-N 0.000 description 6
- 102100028785 Tumor necrosis factor receptor superfamily member 14 Human genes 0.000 description 6
- 238000011467 adoptive cell therapy Methods 0.000 description 6
- 210000003719 b-lymphocyte Anatomy 0.000 description 6
- 210000004899 c-terminal region Anatomy 0.000 description 6
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 description 6
- 210000000805 cytoplasm Anatomy 0.000 description 6
- 210000000987 immune system Anatomy 0.000 description 6
- 238000001990 intravenous administration Methods 0.000 description 6
- 239000007788 liquid Substances 0.000 description 6
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 6
- 239000004005 microsphere Substances 0.000 description 6
- 230000003647 oxidation Effects 0.000 description 6
- 238000007254 oxidation reaction Methods 0.000 description 6
- 125000006239 protecting group Chemical group 0.000 description 6
- 241000894007 species Species 0.000 description 6
- 238000007920 subcutaneous administration Methods 0.000 description 6
- 238000013518 transcription Methods 0.000 description 6
- 230000035897 transcription Effects 0.000 description 6
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 6
- 108010078791 Carrier Proteins Proteins 0.000 description 5
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 5
- 102000053602 DNA Human genes 0.000 description 5
- 102000004190 Enzymes Human genes 0.000 description 5
- 108090000790 Enzymes Proteins 0.000 description 5
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 5
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 5
- 102000003886 Glycoproteins Human genes 0.000 description 5
- 108090000288 Glycoproteins Proteins 0.000 description 5
- 102100028971 HLA class I histocompatibility antigen, C alpha chain Human genes 0.000 description 5
- 108010052199 HLA-C Antigens Proteins 0.000 description 5
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 5
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 5
- 206010061218 Inflammation Diseases 0.000 description 5
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 5
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 241001529936 Murinae Species 0.000 description 5
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 5
- 241000700618 Vaccinia virus Species 0.000 description 5
- 238000004458 analytical method Methods 0.000 description 5
- 239000011324 bead Substances 0.000 description 5
- 239000010949 copper Substances 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 239000000539 dimer Substances 0.000 description 5
- 229940079593 drug Drugs 0.000 description 5
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 5
- 229940088598 enzyme Drugs 0.000 description 5
- 230000002998 immunogenetic effect Effects 0.000 description 5
- 230000001024 immunotherapeutic effect Effects 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 230000004054 inflammatory process Effects 0.000 description 5
- 238000003780 insertion Methods 0.000 description 5
- 230000037431 insertion Effects 0.000 description 5
- 238000007918 intramuscular administration Methods 0.000 description 5
- 210000004962 mammalian cell Anatomy 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 238000002515 oligonucleotide synthesis Methods 0.000 description 5
- 230000035755 proliferation Effects 0.000 description 5
- 230000004936 stimulating effect Effects 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 238000002560 therapeutic procedure Methods 0.000 description 5
- 239000003053 toxin Substances 0.000 description 5
- 231100000765 toxin Toxicity 0.000 description 5
- 108700012359 toxins Proteins 0.000 description 5
- 210000002845 virion Anatomy 0.000 description 5
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 4
- 102100038077 CD226 antigen Human genes 0.000 description 4
- 101150013553 CD40 gene Proteins 0.000 description 4
- 102000014914 Carrier Proteins Human genes 0.000 description 4
- 108091026890 Coding region Proteins 0.000 description 4
- 150000008574 D-amino acids Chemical class 0.000 description 4
- 201000004624 Dermatitis Diseases 0.000 description 4
- 238000002965 ELISA Methods 0.000 description 4
- 239000004471 Glycine Substances 0.000 description 4
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 4
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 4
- 102100028967 HLA class I histocompatibility antigen, alpha chain G Human genes 0.000 description 4
- 108010050568 HLA-DM antigens Proteins 0.000 description 4
- 108010024164 HLA-G Antigens Proteins 0.000 description 4
- 102000018713 Histocompatibility Antigens Class II Human genes 0.000 description 4
- 108010027412 Histocompatibility Antigens Class II Proteins 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 101000884298 Homo sapiens CD226 antigen Proteins 0.000 description 4
- 101000889276 Homo sapiens Cytotoxic T-lymphocyte protein 4 Proteins 0.000 description 4
- 101000633780 Homo sapiens Signaling lymphocytic activation molecule Proteins 0.000 description 4
- 101000934346 Homo sapiens T-cell surface antigen CD2 Proteins 0.000 description 4
- 241000713310 Human T-cell lymphotropic virus type 4 Species 0.000 description 4
- 241000714259 Human T-lymphotropic virus 2 Species 0.000 description 4
- 241001136003 Human T-lymphotropic virus 3 Species 0.000 description 4
- 108090000172 Interleukin-15 Proteins 0.000 description 4
- 102000003812 Interleukin-15 Human genes 0.000 description 4
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 4
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 4
- 239000004472 Lysine Substances 0.000 description 4
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 4
- 241000699670 Mus sp. Species 0.000 description 4
- 108091005804 Peptidases Proteins 0.000 description 4
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 4
- 230000006044 T cell activation Effects 0.000 description 4
- 102100025237 T-cell surface antigen CD2 Human genes 0.000 description 4
- 102100040247 Tumor necrosis factor Human genes 0.000 description 4
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 4
- 230000001154 acute effect Effects 0.000 description 4
- 239000007864 aqueous solution Substances 0.000 description 4
- 230000008901 benefit Effects 0.000 description 4
- 239000001506 calcium phosphate Substances 0.000 description 4
- 210000000349 chromosome Anatomy 0.000 description 4
- 238000004132 cross linking Methods 0.000 description 4
- 239000002552 dosage form Substances 0.000 description 4
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 4
- 238000004108 freeze drying Methods 0.000 description 4
- 230000013595 glycosylation Effects 0.000 description 4
- 238000006206 glycosylation reaction Methods 0.000 description 4
- 210000002443 helper t lymphocyte Anatomy 0.000 description 4
- 230000007062 hydrolysis Effects 0.000 description 4
- 238000006460 hydrolysis reaction Methods 0.000 description 4
- 238000009169 immunotherapy Methods 0.000 description 4
- 238000001802 infusion Methods 0.000 description 4
- 230000002401 inhibitory effect Effects 0.000 description 4
- 238000007912 intraperitoneal administration Methods 0.000 description 4
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 4
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 4
- 239000002777 nucleoside Substances 0.000 description 4
- 150000003833 nucleoside derivatives Chemical class 0.000 description 4
- 210000004940 nucleus Anatomy 0.000 description 4
- 210000000056 organ Anatomy 0.000 description 4
- 210000005259 peripheral blood Anatomy 0.000 description 4
- 239000011886 peripheral blood Substances 0.000 description 4
- 239000003755 preservative agent Substances 0.000 description 4
- 238000012545 processing Methods 0.000 description 4
- 230000010076 replication Effects 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- 230000011664 signaling Effects 0.000 description 4
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 4
- 230000004083 survival effect Effects 0.000 description 4
- 239000000725 suspension Substances 0.000 description 4
- FPGGTKZVZWFYPV-UHFFFAOYSA-M tetrabutylammonium fluoride Chemical compound [F-].CCCC[N+](CCCC)(CCCC)CCCC FPGGTKZVZWFYPV-UHFFFAOYSA-M 0.000 description 4
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- 241001515965 unidentified phage Species 0.000 description 4
- 239000003981 vehicle Substances 0.000 description 4
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 3
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 3
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 3
- VHUUQVKOLVNVRT-UHFFFAOYSA-N Ammonium hydroxide Chemical compound [NH4+].[OH-] VHUUQVKOLVNVRT-UHFFFAOYSA-N 0.000 description 3
- 206010003571 Astrocytoma Diseases 0.000 description 3
- 241000283690 Bos taurus Species 0.000 description 3
- 102100027207 CD27 antigen Human genes 0.000 description 3
- 102100038078 CD276 antigen Human genes 0.000 description 3
- 101710185679 CD276 antigen Proteins 0.000 description 3
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 3
- 102100035793 CD83 antigen Human genes 0.000 description 3
- 108010067225 Cell Adhesion Molecules Proteins 0.000 description 3
- 102000016289 Cell Adhesion Molecules Human genes 0.000 description 3
- 229920002261 Corn starch Polymers 0.000 description 3
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 3
- 101710177611 DNA polymerase II large subunit Proteins 0.000 description 3
- 101710184669 DNA polymerase II small subunit Proteins 0.000 description 3
- 206010011968 Decreased immune responsiveness Diseases 0.000 description 3
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 3
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical group CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- 238000011510 Elispot assay Methods 0.000 description 3
- 108010067306 Fibronectins Proteins 0.000 description 3
- 102000016359 Fibronectins Human genes 0.000 description 3
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 102100028966 HLA class I histocompatibility antigen, alpha chain F Human genes 0.000 description 3
- 102100036243 HLA class II histocompatibility antigen, DQ alpha 1 chain Human genes 0.000 description 3
- 108010058597 HLA-DR Antigens Proteins 0.000 description 3
- 102000006354 HLA-DR Antigens Human genes 0.000 description 3
- 101000914511 Homo sapiens CD27 antigen Proteins 0.000 description 3
- 101000946856 Homo sapiens CD83 antigen Proteins 0.000 description 3
- 101000986080 Homo sapiens HLA class I histocompatibility antigen, alpha chain F Proteins 0.000 description 3
- 101001078158 Homo sapiens Integrin alpha-1 Proteins 0.000 description 3
- 101000935040 Homo sapiens Integrin beta-2 Proteins 0.000 description 3
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 3
- 101000633786 Homo sapiens SLAM family member 6 Proteins 0.000 description 3
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 3
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 3
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 3
- 102100025323 Integrin alpha-1 Human genes 0.000 description 3
- 102100032816 Integrin alpha-6 Human genes 0.000 description 3
- 102100025390 Integrin beta-2 Human genes 0.000 description 3
- 102000013462 Interleukin-12 Human genes 0.000 description 3
- 108010065805 Interleukin-12 Proteins 0.000 description 3
- 108010002586 Interleukin-7 Proteins 0.000 description 3
- 102100021592 Interleukin-7 Human genes 0.000 description 3
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 3
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 3
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 3
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 3
- 206010025323 Lymphomas Diseases 0.000 description 3
- OFOBLEOULBTSOW-UHFFFAOYSA-N Malonic acid Chemical compound OC(=O)CC(O)=O OFOBLEOULBTSOW-UHFFFAOYSA-N 0.000 description 3
- 229930195725 Mannitol Natural products 0.000 description 3
- 102000018697 Membrane Proteins Human genes 0.000 description 3
- 108010052285 Membrane Proteins Proteins 0.000 description 3
- 102100038082 Natural killer cell receptor 2B4 Human genes 0.000 description 3
- 108700026244 Open Reading Frames Proteins 0.000 description 3
- MUBZPKHOEPUJKR-UHFFFAOYSA-N Oxalic acid Chemical compound OC(=O)C(O)=O MUBZPKHOEPUJKR-UHFFFAOYSA-N 0.000 description 3
- 241000282577 Pan troglodytes Species 0.000 description 3
- 102000035195 Peptidases Human genes 0.000 description 3
- 239000002202 Polyethylene glycol Substances 0.000 description 3
- KWYUFKZDYYNOTN-UHFFFAOYSA-M Potassium hydroxide Chemical compound [OH-].[K+] KWYUFKZDYYNOTN-UHFFFAOYSA-M 0.000 description 3
- 108010076504 Protein Sorting Signals Proteins 0.000 description 3
- 229940022005 RNA vaccine Drugs 0.000 description 3
- 238000003559 RNA-seq method Methods 0.000 description 3
- 102100029197 SLAM family member 6 Human genes 0.000 description 3
- 108010074687 Signaling Lymphocytic Activation Molecule Family Member 1 Proteins 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 230000005867 T cell response Effects 0.000 description 3
- 102100027208 T-cell antigen CD7 Human genes 0.000 description 3
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 3
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 3
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 3
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 3
- 102100032101 Tumor necrosis factor ligand superfamily member 9 Human genes 0.000 description 3
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 3
- 206010046851 Uveitis Diseases 0.000 description 3
- 230000021736 acetylation Effects 0.000 description 3
- 238000006640 acetylation reaction Methods 0.000 description 3
- 150000007513 acids Chemical class 0.000 description 3
- 238000001261 affinity purification Methods 0.000 description 3
- 125000003545 alkoxy group Chemical group 0.000 description 3
- 125000003277 amino group Chemical group 0.000 description 3
- 239000000908 ammonium hydroxide Substances 0.000 description 3
- 206010003246 arthritis Diseases 0.000 description 3
- 125000003118 aryl group Chemical group 0.000 description 3
- 229960000190 bacillus calmette–guérin vaccine Drugs 0.000 description 3
- 230000004071 biological effect Effects 0.000 description 3
- 230000006287 biotinylation Effects 0.000 description 3
- 238000007413 biotinylation Methods 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 229910000389 calcium phosphate Inorganic materials 0.000 description 3
- 235000011010 calcium phosphates Nutrition 0.000 description 3
- 230000001413 cellular effect Effects 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
- 238000003776 cleavage reaction Methods 0.000 description 3
- 230000021615 conjugation Effects 0.000 description 3
- 229910052802 copper Inorganic materials 0.000 description 3
- 230000009260 cross reactivity Effects 0.000 description 3
- 125000004122 cyclic group Chemical group 0.000 description 3
- 102000003675 cytokine receptors Human genes 0.000 description 3
- 108010057085 cytokine receptors Proteins 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 238000010511 deprotection reaction Methods 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 239000006185 dispersion Substances 0.000 description 3
- 239000000839 emulsion Substances 0.000 description 3
- 150000002148 esters Chemical class 0.000 description 3
- 238000000684 flow cytometry Methods 0.000 description 3
- NBVXSUQYWXRMNV-UHFFFAOYSA-N fluoromethane Chemical compound FC NBVXSUQYWXRMNV-UHFFFAOYSA-N 0.000 description 3
- 235000003599 food sweetener Nutrition 0.000 description 3
- 229920000159 gelatin Polymers 0.000 description 3
- 239000008273 gelatin Substances 0.000 description 3
- 238000012239 gene modification Methods 0.000 description 3
- 230000005017 genetic modification Effects 0.000 description 3
- 235000013617 genetically modified food Nutrition 0.000 description 3
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 3
- 239000000833 heterodimer Substances 0.000 description 3
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 3
- 239000000710 homodimer Substances 0.000 description 3
- 210000005260 human cell Anatomy 0.000 description 3
- 239000000017 hydrogel Substances 0.000 description 3
- 230000002209 hydrophobic effect Effects 0.000 description 3
- 125000001165 hydrophobic group Chemical group 0.000 description 3
- 230000001900 immune effect Effects 0.000 description 3
- 210000000428 immunological synapse Anatomy 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 229940117681 interleukin-12 Drugs 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 238000005304 joining Methods 0.000 description 3
- 239000008101 lactose Substances 0.000 description 3
- 108700021021 mRNA Vaccine Proteins 0.000 description 3
- 235000019359 magnesium stearate Nutrition 0.000 description 3
- 239000000594 mannitol Substances 0.000 description 3
- 235000010355 mannitol Nutrition 0.000 description 3
- 201000001441 melanoma Diseases 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 210000003071 memory t lymphocyte Anatomy 0.000 description 3
- 239000000693 micelle Substances 0.000 description 3
- 239000003921 oil Substances 0.000 description 3
- 235000019198 oils Nutrition 0.000 description 3
- 238000007911 parenteral administration Methods 0.000 description 3
- 230000006320 pegylation Effects 0.000 description 3
- 239000012071 phase Substances 0.000 description 3
- 150000008300 phosphoramidites Chemical class 0.000 description 3
- 239000006187 pill Substances 0.000 description 3
- 229920000747 poly(lactic acid) Polymers 0.000 description 3
- 229920001282 polysaccharide Polymers 0.000 description 3
- 239000005017 polysaccharide Substances 0.000 description 3
- 150000004804 polysaccharides Chemical class 0.000 description 3
- 230000001566 pro-viral effect Effects 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 230000009257 reactivity Effects 0.000 description 3
- 238000006722 reduction reaction Methods 0.000 description 3
- 230000001105 regulatory effect Effects 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 230000000717 retained effect Effects 0.000 description 3
- 230000007017 scission Effects 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 239000011780 sodium chloride Substances 0.000 description 3
- 239000007790 solid phase Substances 0.000 description 3
- 125000006850 spacer group Chemical group 0.000 description 3
- 230000009870 specific binding Effects 0.000 description 3
- 239000003765 sweetening agent Substances 0.000 description 3
- 239000006188 syrup Substances 0.000 description 3
- 235000020357 syrup Nutrition 0.000 description 3
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 3
- 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 3
- 230000009278 visceral effect Effects 0.000 description 3
- DRHZYJAUECRAJM-DWSYSWFDSA-N (2s,3s,4s,5r,6r)-6-[[(3s,4s,4ar,6ar,6bs,8r,8ar,12as,14ar,14br)-8a-[(2s,3r,4s,5r,6r)-3-[(2s,3r,4s,5r,6s)-5-[(2s,3r,4s,5r)-4-[(2s,3r,4r)-3,4-dihydroxy-4-(hydroxymethyl)oxolan-2-yl]oxy-3,5-dihydroxyoxan-2-yl]oxy-3,4-dihydroxy-6-methyloxan-2-yl]oxy-5-[(3s,5s, Chemical compound O([C@H]1[C@H](O)[C@H](O[C@H]([C@@H]1O[C@H]1[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O1)O)O[C@H]1CC[C@]2(C)[C@H]3CC=C4[C@@H]5CC(C)(C)CC[C@@]5([C@@H](C[C@@]4(C)[C@]3(C)CC[C@H]2[C@@]1(C=O)C)O)C(=O)O[C@@H]1O[C@H](C)[C@@H]([C@@H]([C@H]1O[C@H]1[C@@H]([C@H](O)[C@@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@](O)(CO)CO3)O)[C@H](O)CO2)O)[C@H](C)O1)O)O)OC(=O)C[C@@H](O)C[C@H](OC(=O)C[C@@H](O)C[C@@H]([C@@H](C)CC)O[C@H]1[C@@H]([C@@H](O)[C@H](CO)O1)O)[C@@H](C)CC)C(O)=O)[C@@H]1OC[C@@H](O)[C@H](O)[C@H]1O DRHZYJAUECRAJM-DWSYSWFDSA-N 0.000 description 2
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 2
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 2
- VPFUWHKTPYPNGT-UHFFFAOYSA-N 3-(3,4-dihydroxyphenyl)-1-(5-hydroxy-2,2-dimethylchromen-6-yl)propan-1-one Chemical compound OC1=C2C=CC(C)(C)OC2=CC=C1C(=O)CCC1=CC=C(O)C(O)=C1 VPFUWHKTPYPNGT-UHFFFAOYSA-N 0.000 description 2
- 108010082808 4-1BB Ligand Proteins 0.000 description 2
- JVJFIQYAHPMBBX-UHFFFAOYSA-N 4-hydroxynonenal Chemical compound CCCCCC(O)C=CC=O JVJFIQYAHPMBBX-UHFFFAOYSA-N 0.000 description 2
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 2
- 235000002198 Annona diversifolia Nutrition 0.000 description 2
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 2
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 2
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 2
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 2
- BPYKTIZUTYGOLE-IFADSCNNSA-N Bilirubin Chemical compound N1C(=O)C(C)=C(C=C)\C1=C\C1=C(C)C(CCC(O)=O)=C(CC2=C(C(C)=C(\C=C/3C(=C(C=C)C(=O)N\3)C)N2)CCC(O)=O)N1 BPYKTIZUTYGOLE-IFADSCNNSA-N 0.000 description 2
- 241000167854 Bourreria succulenta Species 0.000 description 2
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 2
- 125000001433 C-terminal amino-acid group Chemical group 0.000 description 2
- 102100024263 CD160 antigen Human genes 0.000 description 2
- 102100032937 CD40 ligand Human genes 0.000 description 2
- 102100025221 CD70 antigen Human genes 0.000 description 2
- 101100112922 Candida albicans CDR3 gene Proteins 0.000 description 2
- 201000000274 Carcinosarcoma Diseases 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 229920001661 Chitosan Polymers 0.000 description 2
- 208000005243 Chondrosarcoma Diseases 0.000 description 2
- 108020004705 Codon Proteins 0.000 description 2
- 108010045171 Cyclic AMP Response Element-Binding Protein Proteins 0.000 description 2
- 102000005636 Cyclic AMP Response Element-Binding Protein Human genes 0.000 description 2
- 108010014303 DNA-directed DNA polymerase Proteins 0.000 description 2
- 102000016928 DNA-directed DNA polymerase Human genes 0.000 description 2
- 241000702421 Dependoparvovirus Species 0.000 description 2
- 229920002307 Dextran Polymers 0.000 description 2
- QSJXEFYPDANLFS-UHFFFAOYSA-N Diacetyl Chemical compound CC(=O)C(C)=O QSJXEFYPDANLFS-UHFFFAOYSA-N 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 201000008808 Fibrosarcoma 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
- 241000233866 Fungi Species 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- BCCRXDTUTZHDEU-VKHMYHEASA-N Gly-Ser Chemical compound NCC(=O)N[C@@H](CO)C(O)=O BCCRXDTUTZHDEU-VKHMYHEASA-N 0.000 description 2
- AEMRFAOFKBGASW-UHFFFAOYSA-N Glycolic acid Chemical compound OCC(O)=O AEMRFAOFKBGASW-UHFFFAOYSA-N 0.000 description 2
- 241000282575 Gorilla Species 0.000 description 2
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 2
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 2
- 102000009465 Growth Factor Receptors Human genes 0.000 description 2
- 108010009202 Growth Factor Receptors Proteins 0.000 description 2
- HVLSXIKZNLPZJJ-TXZCQADKSA-N HA peptide Chemical compound C([C@@H](C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](C)C(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=C(O)C=C1 HVLSXIKZNLPZJJ-TXZCQADKSA-N 0.000 description 2
- 102100028970 HLA class I histocompatibility antigen, alpha chain E Human genes 0.000 description 2
- 102100031258 HLA class II histocompatibility antigen, DM beta chain Human genes 0.000 description 2
- 102100031547 HLA class II histocompatibility antigen, DO alpha chain Human genes 0.000 description 2
- 102100036241 HLA class II histocompatibility antigen, DQ beta 1 chain Human genes 0.000 description 2
- 102100040505 HLA class II histocompatibility antigen, DR alpha chain Human genes 0.000 description 2
- 108010036972 HLA-A11 Antigen Proteins 0.000 description 2
- 108010041384 HLA-DPA antigen Proteins 0.000 description 2
- 108010086786 HLA-DQA1 antigen Proteins 0.000 description 2
- 108010067148 HLA-DQbeta antigen Proteins 0.000 description 2
- 108010067802 HLA-DR alpha-Chains Proteins 0.000 description 2
- 102100029360 Hematopoietic cell signal transducer Human genes 0.000 description 2
- 208000017604 Hodgkin disease Diseases 0.000 description 2
- 208000010747 Hodgkins lymphoma Diseases 0.000 description 2
- 101000864344 Homo sapiens B- and T-lymphocyte attenuator Proteins 0.000 description 2
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 2
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 2
- 101000761938 Homo sapiens CD160 antigen Proteins 0.000 description 2
- 101000868215 Homo sapiens CD40 ligand Proteins 0.000 description 2
- 101000934356 Homo sapiens CD70 antigen Proteins 0.000 description 2
- 101000986085 Homo sapiens HLA class I histocompatibility antigen, alpha chain E Proteins 0.000 description 2
- 101000866278 Homo sapiens HLA class II histocompatibility antigen, DO alpha chain Proteins 0.000 description 2
- 101000990188 Homo sapiens Hematopoietic cell signal transducer Proteins 0.000 description 2
- 101000994375 Homo sapiens Integrin alpha-4 Proteins 0.000 description 2
- 101000994365 Homo sapiens Integrin alpha-6 Proteins 0.000 description 2
- 101001035237 Homo sapiens Integrin alpha-D Proteins 0.000 description 2
- 101001046687 Homo sapiens Integrin alpha-E Proteins 0.000 description 2
- 101000935043 Homo sapiens Integrin beta-1 Proteins 0.000 description 2
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 description 2
- 101001109503 Homo sapiens NKG2-C type II integral membrane protein Proteins 0.000 description 2
- 101000884270 Homo sapiens Natural killer cell receptor 2B4 Proteins 0.000 description 2
- 101000934341 Homo sapiens T-cell surface glycoprotein CD5 Proteins 0.000 description 2
- 101001018021 Homo sapiens T-lymphocyte surface antigen Ly-9 Proteins 0.000 description 2
- 101000648507 Homo sapiens Tumor necrosis factor receptor superfamily member 14 Proteins 0.000 description 2
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 description 2
- 101000679851 Homo sapiens Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 2
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 2
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 description 2
- 108090000144 Human Proteins Proteins 0.000 description 2
- 102000003839 Human Proteins Human genes 0.000 description 2
- VEXZGXHMUGYJMC-UHFFFAOYSA-N Hydrochloric acid Chemical compound Cl VEXZGXHMUGYJMC-UHFFFAOYSA-N 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- 206010020751 Hypersensitivity Diseases 0.000 description 2
- 102100034980 ICOS ligand Human genes 0.000 description 2
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 2
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 2
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 2
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 2
- 102100034353 Integrase Human genes 0.000 description 2
- 108010061833 Integrases Proteins 0.000 description 2
- 102100032818 Integrin alpha-4 Human genes 0.000 description 2
- 102100039904 Integrin alpha-D Human genes 0.000 description 2
- 102100022341 Integrin alpha-E Human genes 0.000 description 2
- 102100022338 Integrin alpha-M Human genes 0.000 description 2
- 102100022297 Integrin alpha-X Human genes 0.000 description 2
- 102100025304 Integrin beta-1 Human genes 0.000 description 2
- 102100037877 Intercellular adhesion molecule 1 Human genes 0.000 description 2
- 102000006992 Interferon-alpha Human genes 0.000 description 2
- 108010047761 Interferon-alpha Proteins 0.000 description 2
- 101800003050 Interleukin-16 Proteins 0.000 description 2
- 102000049772 Interleukin-16 Human genes 0.000 description 2
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 2
- 102000004388 Interleukin-4 Human genes 0.000 description 2
- 108090000978 Interleukin-4 Proteins 0.000 description 2
- 102000004889 Interleukin-6 Human genes 0.000 description 2
- 108090001005 Interleukin-6 Proteins 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- 102000002698 KIR Receptors Human genes 0.000 description 2
- 108010043610 KIR Receptors Proteins 0.000 description 2
- AHLPHDHHMVZTML-BYPYZUCNSA-N L-Ornithine Chemical compound NCCC[C@H](N)C(O)=O AHLPHDHHMVZTML-BYPYZUCNSA-N 0.000 description 2
- RHGKLRLOHDJJDR-BYPYZUCNSA-N L-citrulline Chemical compound NC(=O)NCCC[C@H]([NH3+])C([O-])=O RHGKLRLOHDJJDR-BYPYZUCNSA-N 0.000 description 2
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine Chemical compound OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 2
- LRQKBLKVPFOOQJ-YFKPBYRVSA-N L-norleucine Chemical compound CCCC[C@H]([NH3+])C([O-])=O LRQKBLKVPFOOQJ-YFKPBYRVSA-N 0.000 description 2
- 125000000174 L-prolyl group Chemical group [H]N1C([H])([H])C([H])([H])C([H])([H])[C@@]1([H])C(*)=O 0.000 description 2
- ZKZBPNGNEQAJSX-REOHCLBHSA-N L-selenocysteine Chemical compound [SeH]C[C@H](N)C(O)=O ZKZBPNGNEQAJSX-REOHCLBHSA-N 0.000 description 2
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 2
- 102000017578 LAG3 Human genes 0.000 description 2
- 240000007472 Leucaena leucocephala Species 0.000 description 2
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 2
- 102000019298 Lipocalin Human genes 0.000 description 2
- 108050006654 Lipocalin Proteins 0.000 description 2
- 241000712899 Lymphocytic choriomeningitis mammarenavirus Species 0.000 description 2
- 102100026894 Lymphotoxin-beta Human genes 0.000 description 2
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 2
- 241000218605 Macacine betaherpesvirus 3 Species 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 206010027145 Melanocytic naevus Diseases 0.000 description 2
- AFVFQIVMOAPDHO-UHFFFAOYSA-N Methanesulfonic acid Chemical compound CS(O)(=O)=O AFVFQIVMOAPDHO-UHFFFAOYSA-N 0.000 description 2
- AIJULSRZWUXGPQ-UHFFFAOYSA-N Methylglyoxal Chemical compound CC(=O)C=O AIJULSRZWUXGPQ-UHFFFAOYSA-N 0.000 description 2
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 description 2
- PCLIMKBDDGJMGD-UHFFFAOYSA-N N-bromosuccinimide Chemical compound BrN1C(=O)CCC1=O PCLIMKBDDGJMGD-UHFFFAOYSA-N 0.000 description 2
- 108010057466 NF-kappa B Proteins 0.000 description 2
- 102000003945 NF-kappa B Human genes 0.000 description 2
- 102100022683 NKG2-C type II integral membrane protein Human genes 0.000 description 2
- 108010042215 OX40 Ligand Proteins 0.000 description 2
- 102000004473 OX40 Ligand Human genes 0.000 description 2
- 201000010133 Oligodendroglioma Diseases 0.000 description 2
- 108010058846 Ovalbumin Proteins 0.000 description 2
- 241000282576 Pan paniscus Species 0.000 description 2
- 206010061332 Paraganglion neoplasm Diseases 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 229920002873 Polyethylenimine Polymers 0.000 description 2
- 108010039918 Polylysine Proteins 0.000 description 2
- 241000700625 Poxviridae Species 0.000 description 2
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 description 2
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 2
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 2
- 239000004365 Protease Substances 0.000 description 2
- LCTONWCANYUPML-UHFFFAOYSA-N Pyruvic acid Chemical compound CC(=O)C(O)=O LCTONWCANYUPML-UHFFFAOYSA-N 0.000 description 2
- 102000009572 RNA Polymerase II Human genes 0.000 description 2
- 108010009460 RNA Polymerase II Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 108020004511 Recombinant DNA Proteins 0.000 description 2
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 2
- 241000283984 Rodentia Species 0.000 description 2
- 206010039491 Sarcoma Diseases 0.000 description 2
- 102100027744 Semaphorin-4D Human genes 0.000 description 2
- 241000710961 Semliki Forest virus Species 0.000 description 2
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 2
- 241000700584 Simplexvirus Species 0.000 description 2
- 241000710960 Sindbis virus Species 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 241000282887 Suidae Species 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 2
- 102100025244 T-cell surface glycoprotein CD5 Human genes 0.000 description 2
- 102100033447 T-lymphocyte surface antigen Ly-9 Human genes 0.000 description 2
- 102100036407 Thioredoxin Human genes 0.000 description 2
- 108010034949 Thyroglobulin Proteins 0.000 description 2
- 102000009843 Thyroglobulin Human genes 0.000 description 2
- GWEVSGVZZGPLCZ-UHFFFAOYSA-N Titan oxide Chemical compound O=[Ti]=O GWEVSGVZZGPLCZ-UHFFFAOYSA-N 0.000 description 2
- 108010018242 Transcription Factor AP-1 Proteins 0.000 description 2
- 102100023132 Transcription factor Jun Human genes 0.000 description 2
- 206010052779 Transplant rejections Diseases 0.000 description 2
- 102100032100 Tumor necrosis factor ligand superfamily member 8 Human genes 0.000 description 2
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 description 2
- 102100022156 Tumor necrosis factor receptor superfamily member 3 Human genes 0.000 description 2
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 2
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 description 2
- 206010046865 Vaccinia virus infection Diseases 0.000 description 2
- 241000710959 Venezuelan equine encephalitis virus Species 0.000 description 2
- 241000711975 Vesicular stomatitis virus Species 0.000 description 2
- 208000036142 Viral infection Diseases 0.000 description 2
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- SAZUGELZHZOXHB-UHFFFAOYSA-N acecarbromal Chemical compound CCC(Br)(CC)C(=O)NC(=O)NC(C)=O SAZUGELZHZOXHB-UHFFFAOYSA-N 0.000 description 2
- 235000011054 acetic acid Nutrition 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 230000010933 acylation Effects 0.000 description 2
- 238000005917 acylation reaction Methods 0.000 description 2
- 239000000654 additive Substances 0.000 description 2
- 229960003767 alanine Drugs 0.000 description 2
- 235000010443 alginic acid Nutrition 0.000 description 2
- 229920000615 alginic acid Polymers 0.000 description 2
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 2
- BJEPYKJPYRNKOW-UHFFFAOYSA-N alpha-hydroxysuccinic acid Natural products OC(=O)C(O)CC(O)=O BJEPYKJPYRNKOW-UHFFFAOYSA-N 0.000 description 2
- AZDRQVAHHNSJOQ-UHFFFAOYSA-N alumane Chemical class [AlH3] AZDRQVAHHNSJOQ-UHFFFAOYSA-N 0.000 description 2
- 230000002707 ameloblastic effect Effects 0.000 description 2
- 230000009435 amidation Effects 0.000 description 2
- 238000007112 amidation reaction Methods 0.000 description 2
- 125000003368 amide group Chemical group 0.000 description 2
- 210000004102 animal cell Anatomy 0.000 description 2
- RDOXTESZEPMUJZ-UHFFFAOYSA-N anisole Chemical compound COC1=CC=CC=C1 RDOXTESZEPMUJZ-UHFFFAOYSA-N 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 230000001745 anti-biotin effect Effects 0.000 description 2
- 230000001093 anti-cancer Effects 0.000 description 2
- 230000002924 anti-infective effect Effects 0.000 description 2
- 230000006023 anti-tumor response Effects 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 125000000637 arginyl group Chemical group N[C@@H](CCCNC(N)=N)C(=O)* 0.000 description 2
- 229960001230 asparagine Drugs 0.000 description 2
- 235000009582 asparagine Nutrition 0.000 description 2
- 125000004429 atom Chemical group 0.000 description 2
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 2
- 230000002238 attenuated effect Effects 0.000 description 2
- 230000001363 autoimmune Effects 0.000 description 2
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid Chemical compound OC(=O)C1=CC=CC=C1 WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 2
- UCMIRNVEIXFBKS-UHFFFAOYSA-N beta-alanine Chemical compound NCCC(O)=O UCMIRNVEIXFBKS-UHFFFAOYSA-N 0.000 description 2
- PFYXSUNOLOJMDX-UHFFFAOYSA-N bis(2,5-dioxopyrrolidin-1-yl) carbonate Chemical compound O=C1CCC(=O)N1OC(=O)ON1C(=O)CCC1=O PFYXSUNOLOJMDX-UHFFFAOYSA-N 0.000 description 2
- 230000037396 body weight Effects 0.000 description 2
- 229940098773 bovine serum albumin Drugs 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- FUFJGUQYACFECW-UHFFFAOYSA-L calcium hydrogenphosphate Chemical compound [Ca+2].OP([O-])([O-])=O FUFJGUQYACFECW-UHFFFAOYSA-L 0.000 description 2
- 229940023860 canarypox virus HIV vaccine Drugs 0.000 description 2
- 230000021235 carbamoylation Effects 0.000 description 2
- 230000015556 catabolic process Effects 0.000 description 2
- 125000002091 cationic group Chemical group 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 210000000170 cell membrane Anatomy 0.000 description 2
- 238000002659 cell therapy Methods 0.000 description 2
- 210000004671 cell-free system Anatomy 0.000 description 2
- 230000036755 cellular response Effects 0.000 description 2
- 239000001913 cellulose Substances 0.000 description 2
- 235000010980 cellulose Nutrition 0.000 description 2
- 229920002678 cellulose Polymers 0.000 description 2
- 235000019693 cherries Nutrition 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical compound C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 238000004587 chromatography analysis Methods 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- 208000009060 clear cell adenocarcinoma Diseases 0.000 description 2
- 238000000576 coating method Methods 0.000 description 2
- 230000000295 complement effect Effects 0.000 description 2
- 239000008120 corn starch Substances 0.000 description 2
- 229940099112 cornstarch Drugs 0.000 description 2
- 230000000139 costimulatory effect Effects 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- XVOYSCVBGLVSOL-UHFFFAOYSA-N cysteic acid Chemical compound OC(=O)C(N)CS(O)(=O)=O XVOYSCVBGLVSOL-UHFFFAOYSA-N 0.000 description 2
- 230000016396 cytokine production Effects 0.000 description 2
- 210000005220 cytoplasmic tail Anatomy 0.000 description 2
- 229940127089 cytotoxic agent Drugs 0.000 description 2
- 239000002254 cytotoxic agent Substances 0.000 description 2
- 231100000599 cytotoxic agent Toxicity 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- XBDQKXXYIPTUBI-UHFFFAOYSA-N dimethylselenoniopropionate Natural products CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 2
- 230000003292 diminished effect Effects 0.000 description 2
- 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 2
- 238000009826 distribution Methods 0.000 description 2
- OQALFHMKVSJFRR-UHFFFAOYSA-N dityrosine Chemical compound OC(=O)C(N)CC1=CC=C(O)C(C=2C(=CC=C(CC(N)C(O)=O)C=2)O)=C1 OQALFHMKVSJFRR-UHFFFAOYSA-N 0.000 description 2
- 239000012636 effector Substances 0.000 description 2
- 238000001962 electrophoresis Methods 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 238000005538 encapsulation Methods 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 108010078428 env Gene Products Proteins 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 2
- 235000019441 ethanol Nutrition 0.000 description 2
- 238000011156 evaluation Methods 0.000 description 2
- 238000000605 extraction Methods 0.000 description 2
- 230000002349 favourable effect Effects 0.000 description 2
- 239000000796 flavoring agent Substances 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 239000004220 glutamic acid Substances 0.000 description 2
- 125000000291 glutamic acid group Chemical group N[C@@H](CCC(O)=O)C(=O)* 0.000 description 2
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 2
- 239000005090 green fluorescent protein Substances 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 238000010438 heat treatment Methods 0.000 description 2
- 125000000623 heterocyclic group Chemical group 0.000 description 2
- 210000003917 human chromosome Anatomy 0.000 description 2
- 210000004408 hybridoma Anatomy 0.000 description 2
- 235000011167 hydrochloric acid Nutrition 0.000 description 2
- 150000004679 hydroxides Chemical class 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 230000005847 immunogenicity Effects 0.000 description 2
- 230000016784 immunoglobulin production Effects 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 230000001965 increasing effect Effects 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000002458 infectious effect Effects 0.000 description 2
- 230000001524 infective effect Effects 0.000 description 2
- 229940100601 interleukin-6 Drugs 0.000 description 2
- 239000000543 intermediate Substances 0.000 description 2
- 230000004068 intracellular signaling Effects 0.000 description 2
- 238000010255 intramuscular injection Methods 0.000 description 2
- 239000007927 intramuscular injection Substances 0.000 description 2
- PGLTVOMIXTUURA-UHFFFAOYSA-N iodoacetamide Chemical compound NC(=O)CI PGLTVOMIXTUURA-UHFFFAOYSA-N 0.000 description 2
- 230000002147 killing effect Effects 0.000 description 2
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 2
- 239000000787 lecithin Substances 0.000 description 2
- 229940067606 lecithin Drugs 0.000 description 2
- 235000010445 lecithin Nutrition 0.000 description 2
- 230000003902 lesion Effects 0.000 description 2
- 206010024378 leukocytosis Diseases 0.000 description 2
- 238000011068 loading method Methods 0.000 description 2
- 239000000314 lubricant Substances 0.000 description 2
- 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 2
- 239000001630 malic acid Substances 0.000 description 2
- 235000011090 malic acid Nutrition 0.000 description 2
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 description 2
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 2
- 244000005700 microbiome Species 0.000 description 2
- 239000011859 microparticle Substances 0.000 description 2
- 238000000386 microscopy Methods 0.000 description 2
- 150000007522 mineralic acids Chemical class 0.000 description 2
- 102000035118 modified proteins Human genes 0.000 description 2
- 108091005573 modified proteins Proteins 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 229940035032 monophosphoryl lipid a Drugs 0.000 description 2
- 229920005615 natural polymer Polymers 0.000 description 2
- 229910052757 nitrogen Inorganic materials 0.000 description 2
- 238000001668 nucleic acid synthesis Methods 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- 238000006384 oligomerization reaction Methods 0.000 description 2
- 150000007524 organic acids Chemical class 0.000 description 2
- 235000005985 organic acids Nutrition 0.000 description 2
- 229940092253 ovalbumin Drugs 0.000 description 2
- 239000007800 oxidant agent Substances 0.000 description 2
- 229910052760 oxygen Inorganic materials 0.000 description 2
- 239000001301 oxygen Substances 0.000 description 2
- 238000004091 panning Methods 0.000 description 2
- 208000007312 paraganglioma Diseases 0.000 description 2
- 230000001717 pathogenic effect Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 239000013610 patient sample Substances 0.000 description 2
- 238000010647 peptide synthesis reaction Methods 0.000 description 2
- 230000010412 perfusion Effects 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- OJUGVDODNPJEEC-UHFFFAOYSA-N phenylglyoxal Chemical compound O=CC(=O)C1=CC=CC=C1 OJUGVDODNPJEEC-UHFFFAOYSA-N 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 230000004962 physiological condition Effects 0.000 description 2
- 229920000656 polylysine Polymers 0.000 description 2
- 229920001592 potato starch Polymers 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000001737 promoting effect Effects 0.000 description 2
- 238000011321 prophylaxis Methods 0.000 description 2
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 2
- 230000017854 proteolysis Effects 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 210000003289 regulatory T cell Anatomy 0.000 description 2
- 230000003362 replicative effect Effects 0.000 description 2
- 239000011347 resin Substances 0.000 description 2
- 229920005989 resin Polymers 0.000 description 2
- 229950010550 resiquimod Drugs 0.000 description 2
- BXNMTOQRYBFHNZ-UHFFFAOYSA-N resiquimod Chemical compound C1=CC=CC2=C(N(C(COCC)=N3)CC(C)(C)O)C3=C(N)N=C21 BXNMTOQRYBFHNZ-UHFFFAOYSA-N 0.000 description 2
- 230000001177 retroviral effect Effects 0.000 description 2
- 238000004007 reversed phase HPLC Methods 0.000 description 2
- CVHZOJJKTDOEJC-UHFFFAOYSA-N saccharin Chemical compound C1=CC=C2C(=O)NS(=O)(=O)C2=C1 CVHZOJJKTDOEJC-UHFFFAOYSA-N 0.000 description 2
- YGSDEFSMJLZEOE-UHFFFAOYSA-N salicylic acid Chemical compound OC(=O)C1=CC=CC=C1O YGSDEFSMJLZEOE-UHFFFAOYSA-N 0.000 description 2
- FSYKKLYZXJSNPZ-UHFFFAOYSA-N sarcosine Chemical compound C[NH2+]CC([O-])=O FSYKKLYZXJSNPZ-UHFFFAOYSA-N 0.000 description 2
- 238000012216 screening Methods 0.000 description 2
- 229910052709 silver Inorganic materials 0.000 description 2
- 238000002741 site-directed mutagenesis Methods 0.000 description 2
- 239000007921 spray Substances 0.000 description 2
- 239000003381 stabilizer Substances 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 150000008163 sugars Chemical class 0.000 description 2
- 125000000472 sulfonyl group Chemical group *S(*)(=O)=O 0.000 description 2
- 231100000617 superantigen Toxicity 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 229920001059 synthetic polymer Polymers 0.000 description 2
- 238000007910 systemic administration Methods 0.000 description 2
- 238000011191 terminal modification Methods 0.000 description 2
- 229940124597 therapeutic agent Drugs 0.000 description 2
- 108060008226 thioredoxin Proteins 0.000 description 2
- 229940094937 thioredoxin Drugs 0.000 description 2
- 229960002175 thyroglobulin Drugs 0.000 description 2
- JOXIMZWYDAKGHI-UHFFFAOYSA-N toluene-4-sulfonic acid Chemical compound CC1=CC=C(S(O)(=O)=O)C=C1 JOXIMZWYDAKGHI-UHFFFAOYSA-N 0.000 description 2
- 230000000699 topical effect Effects 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
- 230000005030 transcription termination Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000012546 transfer Methods 0.000 description 2
- 238000013519 translation Methods 0.000 description 2
- 102000027257 transmembrane receptors Human genes 0.000 description 2
- 108091008578 transmembrane receptors Proteins 0.000 description 2
- YNJBWRMUSHSURL-UHFFFAOYSA-N trichloroacetic acid Chemical compound OC(=O)C(Cl)(Cl)Cl YNJBWRMUSHSURL-UHFFFAOYSA-N 0.000 description 2
- 125000002221 trityl group Chemical group [H]C1=C([H])C([H])=C([H])C([H])=C1C([*])(C1=C(C(=C(C(=C1[H])[H])[H])[H])[H])C1=C([H])C([H])=C([H])C([H])=C1[H] 0.000 description 2
- 210000004881 tumor cell Anatomy 0.000 description 2
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 2
- 208000007089 vaccinia Diseases 0.000 description 2
- 229950008737 vadimezan Drugs 0.000 description 2
- XGOYIMQSIKSOBS-UHFFFAOYSA-N vadimezan Chemical compound C1=CC=C2C(=O)C3=CC=C(C)C(C)=C3OC2=C1CC(O)=O XGOYIMQSIKSOBS-UHFFFAOYSA-N 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- 230000009385 viral infection Effects 0.000 description 2
- QBYIENPQHBMVBV-HFEGYEGKSA-N (2R)-2-hydroxy-2-phenylacetic acid Chemical compound O[C@@H](C(O)=O)c1ccccc1.O[C@@H](C(O)=O)c1ccccc1 QBYIENPQHBMVBV-HFEGYEGKSA-N 0.000 description 1
- LZOIGVDSAMDBIO-LXWJMTKESA-N (2S)-2-[[(2S,3R)-2-[[(2S)-2-[[(2S,3S)-2-[[(2S)-4-amino-2-[[(2S,3S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-amino-4-methylsulfanylbutanoyl]amino]-3-(4-hydroxyphenyl)propanoyl]amino]-3-phenylpropanoyl]amino]-3-methylpentanoyl]amino]-4-oxobutanoyl]amino]-3-methylpentanoyl]amino]-4-methylpentanoyl]amino]-3-hydroxybutanoyl]amino]-4-methylpentanoic acid Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(C)C)C(O)=O)[C@@H](C)CC)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@@H](N)CCSC)C1=CC=CC=C1 LZOIGVDSAMDBIO-LXWJMTKESA-N 0.000 description 1
- WTKYBFQVZPCGAO-LURJTMIESA-N (2s)-2-(pyridin-3-ylamino)propanoic acid Chemical compound OC(=O)[C@H](C)NC1=CC=CN=C1 WTKYBFQVZPCGAO-LURJTMIESA-N 0.000 description 1
- KUHSEZKIEJYEHN-BXRBKJIMSA-N (2s)-2-amino-3-hydroxypropanoic acid;(2s)-2-aminopropanoic acid Chemical compound C[C@H](N)C(O)=O.OC[C@H](N)C(O)=O KUHSEZKIEJYEHN-BXRBKJIMSA-N 0.000 description 1
- MJGBOFOZSAEULI-RUCXOUQFSA-N (2s)-5-oxopyrrolidine-2-carboxylic acid Chemical compound OC(=O)[C@@H]1CCC(=O)N1.OC(=O)[C@@H]1CCC(=O)N1 MJGBOFOZSAEULI-RUCXOUQFSA-N 0.000 description 1
- YUXKOWPNKJSTPQ-AXWWPMSFSA-N (2s,3r)-2-amino-3-hydroxybutanoic acid;(2s)-2-amino-3-hydroxypropanoic acid Chemical compound OC[C@H](N)C(O)=O.C[C@@H](O)[C@H](N)C(O)=O YUXKOWPNKJSTPQ-AXWWPMSFSA-N 0.000 description 1
- FYGDTMLNYKFZSV-URKRLVJHSA-N (2s,3r,4s,5s,6r)-2-[(2r,4r,5r,6s)-4,5-dihydroxy-2-(hydroxymethyl)-6-[(2r,4r,5r,6s)-4,5,6-trihydroxy-2-(hydroxymethyl)oxan-3-yl]oxyoxan-3-yl]oxy-6-(hydroxymethyl)oxane-3,4,5-triol Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1OC1[C@@H](CO)O[C@@H](OC2[C@H](O[C@H](O)[C@H](O)[C@H]2O)CO)[C@H](O)[C@H]1O FYGDTMLNYKFZSV-URKRLVJHSA-N 0.000 description 1
- LJRDOKAZOAKLDU-UDXJMMFXSA-N (2s,3s,4r,5r,6r)-5-amino-2-(aminomethyl)-6-[(2r,3s,4r,5s)-5-[(1r,2r,3s,5r,6s)-3,5-diamino-2-[(2s,3r,4r,5s,6r)-3-amino-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl]oxy-6-hydroxycyclohexyl]oxy-4-hydroxy-2-(hydroxymethyl)oxolan-3-yl]oxyoxane-3,4-diol;sulfuric ac Chemical compound OS(O)(=O)=O.N[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[C@@H](N)[C@@H]2O)O[C@@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)N)O[C@@H]1CO LJRDOKAZOAKLDU-UDXJMMFXSA-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
- WBYWAXJHAXSJNI-VOTSOKGWSA-M .beta-Phenylacrylic acid Natural products [O-]C(=O)\C=C\C1=CC=CC=C1 WBYWAXJHAXSJNI-VOTSOKGWSA-M 0.000 description 1
- DHBXNPKRAUYBTH-UHFFFAOYSA-N 1,1-ethanedithiol Chemical compound CC(S)S DHBXNPKRAUYBTH-UHFFFAOYSA-N 0.000 description 1
- ASOKPJOREAFHNY-UHFFFAOYSA-N 1-Hydroxybenzotriazole Chemical compound C1=CC=C2N(O)N=NC2=C1 ASOKPJOREAFHNY-UHFFFAOYSA-N 0.000 description 1
- DURPTKYDGMDSBL-UHFFFAOYSA-N 1-butoxybutane Chemical class CCCCOCCCC DURPTKYDGMDSBL-UHFFFAOYSA-N 0.000 description 1
- LMDZBCPBFSXMTL-UHFFFAOYSA-N 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide Chemical compound CCN=C=NCCCN(C)C LMDZBCPBFSXMTL-UHFFFAOYSA-N 0.000 description 1
- MCTWTZJPVLRJOU-UHFFFAOYSA-N 1-methyl-1H-imidazole Chemical compound CN1C=CN=C1 MCTWTZJPVLRJOU-UHFFFAOYSA-N 0.000 description 1
- 125000004214 1-pyrrolidinyl group Chemical group [H]C1([H])N(*)C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- NHJVRSWLHSJWIN-UHFFFAOYSA-N 2,4,6-trinitrobenzenesulfonic acid Chemical compound OS(=O)(=O)C1=C([N+]([O-])=O)C=C([N+]([O-])=O)C=C1[N+]([O-])=O NHJVRSWLHSJWIN-UHFFFAOYSA-N 0.000 description 1
- KFDPCYZHENQOBV-UHFFFAOYSA-N 2-(bromomethyl)-4-nitrophenol Chemical compound OC1=CC=C([N+]([O-])=O)C=C1CBr KFDPCYZHENQOBV-UHFFFAOYSA-N 0.000 description 1
- VUCNQOPCYRJCGQ-UHFFFAOYSA-N 2-[4-(hydroxymethyl)phenoxy]acetic acid Chemical class OCC1=CC=C(OCC(O)=O)C=C1 VUCNQOPCYRJCGQ-UHFFFAOYSA-N 0.000 description 1
- FBUTXZSKZCQABC-UHFFFAOYSA-N 2-amino-1-methyl-7h-purine-6-thione Chemical compound S=C1N(C)C(N)=NC2=C1NC=N2 FBUTXZSKZCQABC-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
- IZQAUUVBKYXMET-UHFFFAOYSA-N 2-bromoethanamine Chemical compound NCCBr IZQAUUVBKYXMET-UHFFFAOYSA-N 0.000 description 1
- BFSVOASYOCHEOV-UHFFFAOYSA-N 2-diethylaminoethanol Chemical compound CCN(CC)CCO BFSVOASYOCHEOV-UHFFFAOYSA-N 0.000 description 1
- JQPFYXFVUKHERX-UHFFFAOYSA-N 2-hydroxy-2-cyclohexen-1-one Natural products OC1=CCCCC1=O JQPFYXFVUKHERX-UHFFFAOYSA-N 0.000 description 1
- NYPYHUZRZVSYKL-ZETCQYMHSA-N 3,5-diiodo-L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC(I)=C(O)C(I)=C1 NYPYHUZRZVSYKL-ZETCQYMHSA-N 0.000 description 1
- MWOOKDULMBMMPN-UHFFFAOYSA-N 3-(2-ethyl-1,2-oxazol-2-ium-5-yl)benzenesulfonate Chemical compound O1[N+](CC)=CC=C1C1=CC=CC(S([O-])(=O)=O)=C1 MWOOKDULMBMMPN-UHFFFAOYSA-N 0.000 description 1
- LJGHYPLBDBRCRZ-UHFFFAOYSA-N 3-(3-aminophenyl)sulfonylaniline Chemical compound NC1=CC=CC(S(=O)(=O)C=2C=C(N)C=CC=2)=C1 LJGHYPLBDBRCRZ-UHFFFAOYSA-N 0.000 description 1
- BRMWTNUJHUMWMS-UHFFFAOYSA-N 3-Methylhistidine Natural products CN1C=NC(CC(N)C(O)=O)=C1 BRMWTNUJHUMWMS-UHFFFAOYSA-N 0.000 description 1
- HVCOBJNICQPDBP-UHFFFAOYSA-N 3-[3-[3,5-dihydroxy-6-methyl-4-(3,4,5-trihydroxy-6-methyloxan-2-yl)oxyoxan-2-yl]oxydecanoyloxy]decanoic acid;hydrate Chemical compound O.OC1C(OC(CC(=O)OC(CCCCCCC)CC(O)=O)CCCCCCC)OC(C)C(O)C1OC1C(O)C(O)C(O)C(C)O1 HVCOBJNICQPDBP-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
- UQTZMGFTRHFAAM-ZETCQYMHSA-N 3-iodo-L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C(I)=C1 UQTZMGFTRHFAAM-ZETCQYMHSA-N 0.000 description 1
- WHNPOQXWAMXPTA-UHFFFAOYSA-N 3-methylbut-2-enamide Chemical compound CC(C)=CC(N)=O WHNPOQXWAMXPTA-UHFFFAOYSA-N 0.000 description 1
- ADVPTQAUNPRNPO-REOHCLBHSA-N 3-sulfino-L-alanine Chemical compound OC(=O)[C@@H](N)C[S@@](O)=O ADVPTQAUNPRNPO-REOHCLBHSA-N 0.000 description 1
- 238000010600 3H thymidine incorporation assay Methods 0.000 description 1
- 101710134681 40 kDa protein Proteins 0.000 description 1
- 229940117976 5-hydroxylysine Drugs 0.000 description 1
- FHVDTGUDJYJELY-UHFFFAOYSA-N 6-{[2-carboxy-4,5-dihydroxy-6-(phosphanyloxy)oxan-3-yl]oxy}-4,5-dihydroxy-3-phosphanyloxane-2-carboxylic acid Chemical compound O1C(C(O)=O)C(P)C(O)C(O)C1OC1C(C(O)=O)OC(OP)C(O)C1O FHVDTGUDJYJELY-UHFFFAOYSA-N 0.000 description 1
- 102100023990 60S ribosomal protein L17 Human genes 0.000 description 1
- 125000003341 7 membered heterocyclic group Chemical group 0.000 description 1
- ZCYVEMRRCGMTRW-UHFFFAOYSA-N 7553-56-2 Chemical compound [I] ZCYVEMRRCGMTRW-UHFFFAOYSA-N 0.000 description 1
- 230000005730 ADP ribosylation Effects 0.000 description 1
- 108010066676 Abrin Proteins 0.000 description 1
- 101710159080 Aconitate hydratase A Proteins 0.000 description 1
- 101710159078 Aconitate hydratase B Proteins 0.000 description 1
- 108700027273 Acyl Carrier Proteins 0.000 description 1
- 241001655883 Adeno-associated virus - 1 Species 0.000 description 1
- 241000702423 Adeno-associated virus - 2 Species 0.000 description 1
- 241000972680 Adeno-associated virus - 6 Species 0.000 description 1
- 208000004804 Adenomatous Polyps Diseases 0.000 description 1
- 241000701242 Adenoviridae Species 0.000 description 1
- 102100036775 Afadin Human genes 0.000 description 1
- 102000054930 Agouti-Related Human genes 0.000 description 1
- 108010011170 Ala-Trp-Arg-His-Pro-Gln-Phe-Gly-Gly Proteins 0.000 description 1
- 102000009027 Albumins Human genes 0.000 description 1
- 108010088751 Albumins Proteins 0.000 description 1
- 241000710929 Alphavirus Species 0.000 description 1
- 206010001889 Alveolitis Diseases 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 102100022704 Amyloid-beta precursor protein Human genes 0.000 description 1
- 201000003076 Angiosarcoma Diseases 0.000 description 1
- 108010049777 Ankyrins Proteins 0.000 description 1
- 102000008102 Ankyrins Human genes 0.000 description 1
- 244000303258 Annona diversifolia Species 0.000 description 1
- 102100021569 Apoptosis regulator Bcl-2 Human genes 0.000 description 1
- 241000272478 Aquila Species 0.000 description 1
- 101001007348 Arachis hypogaea Galactose-binding lectin Proteins 0.000 description 1
- 241000712891 Arenavirus Species 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- KDZOASGQNOPSCU-WDSKDSINSA-N Argininosuccinic acid Chemical compound OC(=O)[C@@H](N)CCC\N=C(/N)N[C@H](C(O)=O)CC(O)=O KDZOASGQNOPSCU-WDSKDSINSA-N 0.000 description 1
- 241000228212 Aspergillus Species 0.000 description 1
- 101000669426 Aspergillus restrictus Ribonuclease mitogillin Proteins 0.000 description 1
- 241000416162 Astragalus gummifer Species 0.000 description 1
- 206010065869 Astrocytoma, low grade Diseases 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 102100027205 B-cell antigen receptor complex-associated protein alpha chain Human genes 0.000 description 1
- 102100027203 B-cell antigen receptor complex-associated protein beta chain Human genes 0.000 description 1
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 1
- 108010074708 B7-H1 Antigen Proteins 0.000 description 1
- 108091012583 BCL2 Proteins 0.000 description 1
- BXTVQNYQYUTQAZ-UHFFFAOYSA-N BNPS-skatole Chemical compound N=1C2=CC=CC=C2C(C)(Br)C=1SC1=CC=CC=C1[N+]([O-])=O BXTVQNYQYUTQAZ-UHFFFAOYSA-N 0.000 description 1
- 244000063299 Bacillus subtilis Species 0.000 description 1
- 235000014469 Bacillus subtilis Nutrition 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 102100036597 Basement membrane-specific heparan sulfate proteoglycan core protein Human genes 0.000 description 1
- 102000049979 Basic-leucine zipper domains Human genes 0.000 description 1
- 108700039137 Basic-leucine zipper domains Proteins 0.000 description 1
- 208000035821 Benign schwannoma Diseases 0.000 description 1
- 239000005711 Benzoic acid Substances 0.000 description 1
- 102100026189 Beta-galactosidase Human genes 0.000 description 1
- 229920002498 Beta-glucan Polymers 0.000 description 1
- 101000984722 Bos taurus Pancreatic trypsin inhibitor Proteins 0.000 description 1
- 241000701106 Bovine adenovirus 3 Species 0.000 description 1
- 241000701822 Bovine papillomavirus Species 0.000 description 1
- 208000007690 Brenner tumor Diseases 0.000 description 1
- 206010073258 Brenner tumour Diseases 0.000 description 1
- 208000003170 Bronchiolo-Alveolar Adenocarcinoma Diseases 0.000 description 1
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 description 1
- 108010074051 C-Reactive Protein Proteins 0.000 description 1
- 102100032752 C-reactive protein Human genes 0.000 description 1
- 108010056102 CD100 antigen Proteins 0.000 description 1
- 108010017009 CD11b Antigen Proteins 0.000 description 1
- 102100036008 CD48 antigen Human genes 0.000 description 1
- 108010062802 CD66 antigens Proteins 0.000 description 1
- 102100027217 CD82 antigen Human genes 0.000 description 1
- 101710139831 CD82 antigen Proteins 0.000 description 1
- 102100037904 CD9 antigen Human genes 0.000 description 1
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 1
- 229940045513 CTLA4 antagonist Drugs 0.000 description 1
- 102000000905 Cadherin Human genes 0.000 description 1
- 108050007957 Cadherin Proteins 0.000 description 1
- 101100505076 Caenorhabditis elegans gly-2 gene Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 102000005701 Calcium-Binding Proteins Human genes 0.000 description 1
- 108010045403 Calcium-Binding Proteins Proteins 0.000 description 1
- 102000000584 Calmodulin Human genes 0.000 description 1
- 108010041952 Calmodulin Proteins 0.000 description 1
- 241000282832 Camelidae Species 0.000 description 1
- 241000282836 Camelus dromedarius Species 0.000 description 1
- 241000701157 Canine mastadenovirus A Species 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 101710158575 Cap-specific mRNA (nucleoside-2'-O-)-methyltransferase Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 101710132601 Capsid protein Proteins 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 102000005367 Carboxypeptidases Human genes 0.000 description 1
- 108010006303 Carboxypeptidases Proteins 0.000 description 1
- 102100024533 Carcinoembryonic antigen-related cell adhesion molecule 1 Human genes 0.000 description 1
- 102100025466 Carcinoembryonic antigen-related cell adhesion molecule 3 Human genes 0.000 description 1
- 206010007275 Carcinoid tumour Diseases 0.000 description 1
- 102100026550 Caspase-9 Human genes 0.000 description 1
- 108090000566 Caspase-9 Proteins 0.000 description 1
- 108010039939 Cell Wall Skeleton Proteins 0.000 description 1
- 108010051109 Cell-Penetrating Peptides Proteins 0.000 description 1
- 102000020313 Cell-Penetrating Peptides Human genes 0.000 description 1
- 102100035361 Cerebellar degeneration-related protein 2 Human genes 0.000 description 1
- 208000013641 Cerebrofacial arteriovenous metameric syndrome Diseases 0.000 description 1
- 108010075016 Ceruloplasmin Proteins 0.000 description 1
- 102100023321 Ceruloplasmin Human genes 0.000 description 1
- 108010023798 Charybdotoxin Proteins 0.000 description 1
- 102000009410 Chemokine receptor Human genes 0.000 description 1
- 108050000299 Chemokine receptor Proteins 0.000 description 1
- 102000019034 Chemokines Human genes 0.000 description 1
- 108010012236 Chemokines Proteins 0.000 description 1
- 102000012336 Cholesterol Ester Transfer Proteins Human genes 0.000 description 1
- 108010061846 Cholesterol Ester Transfer Proteins Proteins 0.000 description 1
- 241000251730 Chondrichthyes Species 0.000 description 1
- 201000009047 Chordoma Diseases 0.000 description 1
- 208000006332 Choriocarcinoma Diseases 0.000 description 1
- WBYWAXJHAXSJNI-SREVYHEPSA-N Cinnamic acid Chemical compound OC(=O)\C=C/C1=CC=CC=C1 WBYWAXJHAXSJNI-SREVYHEPSA-N 0.000 description 1
- 101100007328 Cocos nucifera COS-1 gene Proteins 0.000 description 1
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 1
- 108010026206 Conalbumin Proteins 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 241000700626 Cowpox virus Species 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- 239000004971 Cross linker Substances 0.000 description 1
- 108700032819 Croton tiglium crotin II Proteins 0.000 description 1
- 208000006081 Cryptococcal meningitis Diseases 0.000 description 1
- JPVYNHNXODAKFH-UHFFFAOYSA-N Cu2+ Chemical compound [Cu+2] JPVYNHNXODAKFH-UHFFFAOYSA-N 0.000 description 1
- 108010025905 Cystine-Knot Miniproteins Proteins 0.000 description 1
- 108010020076 Cytochrome P-450 CYP2B1 Proteins 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 206010011831 Cytomegalovirus infection Diseases 0.000 description 1
- 102000000311 Cytosine Deaminase Human genes 0.000 description 1
- 108010080611 Cytosine Deaminase Proteins 0.000 description 1
- FDKWRPBBCBCIGA-UWTATZPHSA-N D-Selenocysteine Natural products [Se]C[C@@H](N)C(O)=O FDKWRPBBCBCIGA-UWTATZPHSA-N 0.000 description 1
- VVNCNSJFMMFHPL-VKHMYHEASA-N D-penicillamine Chemical compound CC(C)(S)[C@@H](N)C(O)=O VVNCNSJFMMFHPL-VKHMYHEASA-N 0.000 description 1
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 1
- YVGGHNCTFXOJCH-UHFFFAOYSA-N DDT Chemical compound C1=CC(Cl)=CC=C1C(C(Cl)(Cl)Cl)C1=CC=C(Cl)C=C1 YVGGHNCTFXOJCH-UHFFFAOYSA-N 0.000 description 1
- 108010041986 DNA Vaccines Proteins 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- 229940021995 DNA vaccine Drugs 0.000 description 1
- 241000450599 DNA viruses Species 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 241001663879 Deltaretrovirus Species 0.000 description 1
- 108700022150 Designed Ankyrin Repeat Proteins Proteins 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
- 235000019739 Dicalciumphosphate Nutrition 0.000 description 1
- QOSSAOTZNIDXMA-UHFFFAOYSA-N Dicylcohexylcarbodiimide Chemical compound C1CCCCC1N=C=NC1CCCCC1 QOSSAOTZNIDXMA-UHFFFAOYSA-N 0.000 description 1
- 108010053187 Diphtheria Toxin Proteins 0.000 description 1
- 102000016607 Diphtheria Toxin Human genes 0.000 description 1
- 101150075508 Dr gene Proteins 0.000 description 1
- 208000003556 Dry Eye Syndromes Diseases 0.000 description 1
- 206010013774 Dry eye Diseases 0.000 description 1
- 208000037162 Ductal Breast Carcinoma Diseases 0.000 description 1
- 208000007033 Dysgerminoma Diseases 0.000 description 1
- 101000609473 Ecballium elaterium Trypsin inhibitor 2 Proteins 0.000 description 1
- 102100029722 Ectonucleoside triphosphate diphosphohydrolase 1 Human genes 0.000 description 1
- 201000009051 Embryonal Carcinoma Diseases 0.000 description 1
- 101710121417 Envelope glycoprotein Proteins 0.000 description 1
- 206010014967 Ependymoma Diseases 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 101000998556 Escherichia coli Colicin-E7 immunity protein Proteins 0.000 description 1
- CTKXFMQHOOWWEB-UHFFFAOYSA-N Ethylene oxide/propylene oxide copolymer Chemical compound CCCOC(C)COCCO CTKXFMQHOOWWEB-UHFFFAOYSA-N 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 208000006168 Ewing Sarcoma Diseases 0.000 description 1
- 101710082714 Exotoxin A Proteins 0.000 description 1
- 102000018700 F-Box Proteins Human genes 0.000 description 1
- 108010066805 F-Box Proteins Proteins 0.000 description 1
- 101150089023 FASLG gene Proteins 0.000 description 1
- 201000006107 Familial adenomatous polyposis Diseases 0.000 description 1
- 102000015212 Fas Ligand Protein Human genes 0.000 description 1
- 108010039471 Fas Ligand Protein Proteins 0.000 description 1
- 102000030914 Fatty Acid-Binding Human genes 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 102000002090 Fibronectin type III Human genes 0.000 description 1
- 108050009401 Fibronectin type III Proteins 0.000 description 1
- 206010053717 Fibrous histiocytoma Diseases 0.000 description 1
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 1
- KRHYYFGTRYWZRS-UHFFFAOYSA-M Fluoride anion Chemical compound [F-] KRHYYFGTRYWZRS-UHFFFAOYSA-M 0.000 description 1
- 208000004463 Follicular Adenocarcinoma Diseases 0.000 description 1
- 102000016970 Follistatin Human genes 0.000 description 1
- 108010014612 Follistatin Proteins 0.000 description 1
- 102100029379 Follistatin-related protein 3 Human genes 0.000 description 1
- 102100027581 Forkhead box protein P3 Human genes 0.000 description 1
- 208000000666 Fowlpox Diseases 0.000 description 1
- 102000030782 GTP binding Human genes 0.000 description 1
- 108091000058 GTP-Binding Proteins 0.000 description 1
- 101710177291 Gag polyprotein Proteins 0.000 description 1
- 101000812705 Gallus gallus Endoplasmin Proteins 0.000 description 1
- 206010017708 Ganglioneuroblastoma Diseases 0.000 description 1
- 108700004714 Gelonium multiflorum GEL Proteins 0.000 description 1
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 1
- 208000008999 Giant Cell Carcinoma Diseases 0.000 description 1
- 208000002966 Giant Cell Tumor of Bone Diseases 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 102000058063 Glucose Transporter Type 1 Human genes 0.000 description 1
- SXRSQZLOMIGNAQ-UHFFFAOYSA-N Glutaraldehyde Chemical compound O=CCCCC=O SXRSQZLOMIGNAQ-UHFFFAOYSA-N 0.000 description 1
- 244000068988 Glycine max Species 0.000 description 1
- 235000010469 Glycine max Nutrition 0.000 description 1
- 229930186217 Glycolipid Natural products 0.000 description 1
- 102100039619 Granulocyte colony-stimulating factor Human genes 0.000 description 1
- 208000005234 Granulosa Cell Tumor Diseases 0.000 description 1
- 102100035943 HERV-H LTR-associating protein 2 Human genes 0.000 description 1
- 102100033079 HLA class II histocompatibility antigen, DM alpha chain Human genes 0.000 description 1
- 102100031546 HLA class II histocompatibility antigen, DO beta chain Human genes 0.000 description 1
- 108010088729 HLA-A*02:01 antigen Proteins 0.000 description 1
- 108010074032 HLA-A2 Antigen Proteins 0.000 description 1
- 102000025850 HLA-A2 Antigen Human genes 0.000 description 1
- 108010013476 HLA-A24 Antigen Proteins 0.000 description 1
- 108010008553 HLA-B*07 antigen Proteins 0.000 description 1
- 108010010378 HLA-DP Antigens Proteins 0.000 description 1
- 102000015789 HLA-DP Antigens Human genes 0.000 description 1
- 108010062347 HLA-DQ Antigens Proteins 0.000 description 1
- 208000033855 HTLV-1 carrier Diseases 0.000 description 1
- 239000012981 Hank's balanced salt solution Substances 0.000 description 1
- 241000713858 Harvey murine sarcoma virus Species 0.000 description 1
- 208000002125 Hemangioendothelioma Diseases 0.000 description 1
- 208000006050 Hemangiopericytoma Diseases 0.000 description 1
- 208000001258 Hemangiosarcoma Diseases 0.000 description 1
- 241000700739 Hepadnaviridae Species 0.000 description 1
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 1
- 241000700586 Herpesviridae Species 0.000 description 1
- 102100026122 High affinity immunoglobulin gamma Fc receptor I Human genes 0.000 description 1
- 208000021519 Hodgkin lymphoma Diseases 0.000 description 1
- 241000282418 Hominidae Species 0.000 description 1
- 101000823051 Homo sapiens Amyloid-beta precursor protein Proteins 0.000 description 1
- 101000914489 Homo sapiens B-cell antigen receptor complex-associated protein alpha chain Proteins 0.000 description 1
- 101000914491 Homo sapiens B-cell antigen receptor complex-associated protein beta chain Proteins 0.000 description 1
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 1
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 description 1
- 101000716130 Homo sapiens CD48 antigen Proteins 0.000 description 1
- 101000738354 Homo sapiens CD9 antigen Proteins 0.000 description 1
- 101000914337 Homo sapiens Carcinoembryonic antigen-related cell adhesion molecule 3 Proteins 0.000 description 1
- 101000737796 Homo sapiens Cerebellar degeneration-related protein 2 Proteins 0.000 description 1
- 101001012447 Homo sapiens Ectonucleoside triphosphate diphosphohydrolase 1 Proteins 0.000 description 1
- 101001062529 Homo sapiens Follistatin-related protein 3 Proteins 0.000 description 1
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 description 1
- 101000746367 Homo sapiens Granulocyte colony-stimulating factor Proteins 0.000 description 1
- 101001021491 Homo sapiens HERV-H LTR-associating protein 2 Proteins 0.000 description 1
- 101000986086 Homo sapiens HLA class I histocompatibility antigen, A alpha chain Proteins 0.000 description 1
- 101000866281 Homo sapiens HLA class II histocompatibility antigen, DO beta chain Proteins 0.000 description 1
- 101000864089 Homo sapiens HLA class II histocompatibility antigen, DP alpha 1 chain Proteins 0.000 description 1
- 101000930802 Homo sapiens HLA class II histocompatibility antigen, DQ alpha 1 chain Proteins 0.000 description 1
- 101000968032 Homo sapiens HLA class II histocompatibility antigen, DR beta 3 chain Proteins 0.000 description 1
- 101001068133 Homo sapiens Hepatitis A virus cellular receptor 2 Proteins 0.000 description 1
- 101000913074 Homo sapiens High affinity immunoglobulin gamma Fc receptor I Proteins 0.000 description 1
- 101001019455 Homo sapiens ICOS ligand Proteins 0.000 description 1
- 101001046683 Homo sapiens Integrin alpha-L Proteins 0.000 description 1
- 101001046686 Homo sapiens Integrin alpha-M Proteins 0.000 description 1
- 101001046668 Homo sapiens Integrin alpha-X Proteins 0.000 description 1
- 101001015037 Homo sapiens Integrin beta-7 Proteins 0.000 description 1
- 101000599852 Homo sapiens Intercellular adhesion molecule 1 Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101000971538 Homo sapiens Killer cell lectin-like receptor subfamily F member 1 Proteins 0.000 description 1
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 1
- 101000777628 Homo sapiens Leukocyte antigen CD37 Proteins 0.000 description 1
- 101000984189 Homo sapiens Leukocyte immunoglobulin-like receptor subfamily B member 2 Proteins 0.000 description 1
- 101000984186 Homo sapiens Leukocyte immunoglobulin-like receptor subfamily B member 4 Proteins 0.000 description 1
- 101000868279 Homo sapiens Leukocyte surface antigen CD47 Proteins 0.000 description 1
- 101000917858 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-A Proteins 0.000 description 1
- 101000917839 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor III-B Proteins 0.000 description 1
- 101001063392 Homo sapiens Lymphocyte function-associated antigen 3 Proteins 0.000 description 1
- 101000764535 Homo sapiens Lymphotoxin-alpha Proteins 0.000 description 1
- 101000764294 Homo sapiens Lymphotoxin-beta Proteins 0.000 description 1
- 101000991061 Homo sapiens MHC class I polypeptide-related sequence B Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101001109501 Homo sapiens NKG2-D type II integral membrane protein Proteins 0.000 description 1
- 101000589305 Homo sapiens Natural cytotoxicity triggering receptor 2 Proteins 0.000 description 1
- 101000873418 Homo sapiens P-selectin glycoprotein ligand 1 Proteins 0.000 description 1
- 101001074571 Homo sapiens PIN2/TERF1-interacting telomerase inhibitor 1 Proteins 0.000 description 1
- 101001073025 Homo sapiens Peroxisomal targeting signal 1 receptor Proteins 0.000 description 1
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 1
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 1
- 101000633778 Homo sapiens SLAM family member 5 Proteins 0.000 description 1
- 101000633784 Homo sapiens SLAM family member 7 Proteins 0.000 description 1
- 101000914496 Homo sapiens T-cell antigen CD7 Proteins 0.000 description 1
- 101000596234 Homo sapiens T-cell surface protein tactile Proteins 0.000 description 1
- 101100369992 Homo sapiens TNFSF10 gene Proteins 0.000 description 1
- 101100207070 Homo sapiens TNFSF8 gene Proteins 0.000 description 1
- 101000809875 Homo sapiens TYRO protein tyrosine kinase-binding protein Proteins 0.000 description 1
- 101000830600 Homo sapiens Tumor necrosis factor ligand superfamily member 13 Proteins 0.000 description 1
- 101000830596 Homo sapiens Tumor necrosis factor ligand superfamily member 15 Proteins 0.000 description 1
- 101000597779 Homo sapiens Tumor necrosis factor ligand superfamily member 18 Proteins 0.000 description 1
- 101000638255 Homo sapiens Tumor necrosis factor ligand superfamily member 8 Proteins 0.000 description 1
- 101000638251 Homo sapiens Tumor necrosis factor ligand superfamily member 9 Proteins 0.000 description 1
- 101000795169 Homo sapiens Tumor necrosis factor receptor superfamily member 13C Proteins 0.000 description 1
- 101000679857 Homo sapiens Tumor necrosis factor receptor superfamily member 3 Proteins 0.000 description 1
- 101000597785 Homo sapiens Tumor necrosis factor receptor superfamily member 6B Proteins 0.000 description 1
- 101000818543 Homo sapiens Tyrosine-protein kinase ZAP-70 Proteins 0.000 description 1
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 1
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 1
- 206010020460 Human T-cell lymphotropic virus type I infection Diseases 0.000 description 1
- 241000598171 Human adenovirus sp. Species 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 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
- 208000037147 Hypercalcaemia Diseases 0.000 description 1
- 101710093458 ICOS ligand Proteins 0.000 description 1
- 102000009438 IgE Receptors Human genes 0.000 description 1
- 108010073816 IgE Receptors Proteins 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
- 101710116034 Immunity protein Proteins 0.000 description 1
- 108010009817 Immunoglobulin Constant Regions Proteins 0.000 description 1
- 102000009786 Immunoglobulin Constant Regions Human genes 0.000 description 1
- 102000013463 Immunoglobulin Light Chains Human genes 0.000 description 1
- 108010065825 Immunoglobulin Light Chains Proteins 0.000 description 1
- 206010062016 Immunosuppression Diseases 0.000 description 1
- 108090000723 Insulin-Like Growth Factor I Proteins 0.000 description 1
- 102000004218 Insulin-Like Growth Factor I Human genes 0.000 description 1
- 108090001117 Insulin-Like Growth Factor II Proteins 0.000 description 1
- 102000048143 Insulin-Like Growth Factor II Human genes 0.000 description 1
- 102100022339 Integrin alpha-L Human genes 0.000 description 1
- 108010041100 Integrin alpha6 Proteins 0.000 description 1
- 108010030465 Integrin alpha6beta1 Proteins 0.000 description 1
- 102100033016 Integrin beta-7 Human genes 0.000 description 1
- 108010064593 Intercellular Adhesion Molecule-1 Proteins 0.000 description 1
- 102100026720 Interferon beta Human genes 0.000 description 1
- 108090000467 Interferon-beta Proteins 0.000 description 1
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 1
- 102000003810 Interleukin-18 Human genes 0.000 description 1
- 108090000171 Interleukin-18 Proteins 0.000 description 1
- 102100030704 Interleukin-21 Human genes 0.000 description 1
- 102000015696 Interleukins Human genes 0.000 description 1
- 108010063738 Interleukins Proteins 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 229920001202 Inulin Polymers 0.000 description 1
- 241000701377 Iridoviridae Species 0.000 description 1
- 102000008133 Iron-Binding Proteins Human genes 0.000 description 1
- 108010035210 Iron-Binding Proteins Proteins 0.000 description 1
- 108020003285 Isocitrate lyase Proteins 0.000 description 1
- 201000008869 Juxtacortical Osteosarcoma Diseases 0.000 description 1
- 208000007766 Kaposi sarcoma Diseases 0.000 description 1
- 208000009319 Keratoconjunctivitis Sicca Diseases 0.000 description 1
- 102100021458 Killer cell lectin-like receptor subfamily F member 1 Human genes 0.000 description 1
- 150000007649 L alpha amino acids Chemical class 0.000 description 1
- WTDRDQBEARUVNC-LURJTMIESA-N L-DOPA Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C(O)=C1 WTDRDQBEARUVNC-LURJTMIESA-N 0.000 description 1
- WTDRDQBEARUVNC-UHFFFAOYSA-N L-Dopa Natural products OC(=O)C(N)CC1=CC=C(O)C(O)=C1 WTDRDQBEARUVNC-UHFFFAOYSA-N 0.000 description 1
- FFFHZYDWPBMWHY-UHFFFAOYSA-N L-Homocysteine Natural products OC(=O)C(N)CCS FFFHZYDWPBMWHY-UHFFFAOYSA-N 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- 150000008575 L-amino acids Chemical class 0.000 description 1
- FFFHZYDWPBMWHY-VKHMYHEASA-N L-homocysteine Chemical compound OC(=O)[C@@H](N)CCS FFFHZYDWPBMWHY-VKHMYHEASA-N 0.000 description 1
- 102000003855 L-lactate dehydrogenase Human genes 0.000 description 1
- 108700023483 L-lactate dehydrogenases Proteins 0.000 description 1
- 102100033467 L-selectin Human genes 0.000 description 1
- 229930182853 L-selenocysteine Natural products 0.000 description 1
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- XUIIKFGFIJCVMT-LBPRGKRZSA-N L-thyroxine Chemical compound IC1=CC(C[C@H]([NH3+])C([O-])=O)=CC(I)=C1OC1=CC(I)=C(O)C(I)=C1 XUIIKFGFIJCVMT-LBPRGKRZSA-N 0.000 description 1
- 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 1
- 108090000942 Lactalbumin Proteins 0.000 description 1
- 102000004407 Lactalbumin Human genes 0.000 description 1
- 241000282838 Lama Species 0.000 description 1
- 102000008121 Latent TGF-beta Binding Proteins Human genes 0.000 description 1
- 108010049807 Latent TGF-beta Binding Proteins Proteins 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 208000018142 Leiomyosarcoma Diseases 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 102100031586 Leukocyte antigen CD37 Human genes 0.000 description 1
- 102100025583 Leukocyte immunoglobulin-like receptor subfamily B member 2 Human genes 0.000 description 1
- 102100025578 Leukocyte immunoglobulin-like receptor subfamily B member 4 Human genes 0.000 description 1
- 102100032913 Leukocyte surface antigen CD47 Human genes 0.000 description 1
- 201000004462 Leydig Cell Tumor Diseases 0.000 description 1
- 208000000265 Lobular Carcinoma Diseases 0.000 description 1
- 101001089108 Lotus tetragonolobus Anti-H(O) lectin Proteins 0.000 description 1
- 102100029185 Low affinity immunoglobulin gamma Fc region receptor III-B Human genes 0.000 description 1
- 101000680845 Luffa aegyptiaca Ribosome-inactivating protein luffin P1 Proteins 0.000 description 1
- 208000008771 Lymphadenopathy Diseases 0.000 description 1
- 102100034709 Lymphocyte cytosolic protein 2 Human genes 0.000 description 1
- 101710195102 Lymphocyte cytosolic protein 2 Proteins 0.000 description 1
- 102100030984 Lymphocyte function-associated antigen 3 Human genes 0.000 description 1
- 108010091221 Lymphotoxin beta Receptor Proteins 0.000 description 1
- 102000004083 Lymphotoxin-alpha Human genes 0.000 description 1
- 108090000542 Lymphotoxin-alpha Proteins 0.000 description 1
- 102100026238 Lymphotoxin-alpha Human genes 0.000 description 1
- 108010066345 MHC binding peptide Proteins 0.000 description 1
- 102100030301 MHC class I polypeptide-related sequence A Human genes 0.000 description 1
- 102100030300 MHC class I polypeptide-related sequence B Human genes 0.000 description 1
- 101001043810 Macaca fascicularis Interleukin-7 receptor subunit alpha Proteins 0.000 description 1
- 102100028123 Macrophage colony-stimulating factor 1 Human genes 0.000 description 1
- 101710127797 Macrophage colony-stimulating factor 1 Proteins 0.000 description 1
- 235000019759 Maize starch Nutrition 0.000 description 1
- 101710125418 Major capsid protein Proteins 0.000 description 1
- PEEHTFAAVSWFBL-UHFFFAOYSA-N Maleimide Chemical compound O=C1NC(=O)C=C1 PEEHTFAAVSWFBL-UHFFFAOYSA-N 0.000 description 1
- 208000035771 Malignant Sertoli-Leydig cell tumor of the ovary Diseases 0.000 description 1
- 229920000057 Mannan Polymers 0.000 description 1
- 241000712079 Measles morbillivirus Species 0.000 description 1
- 208000007054 Medullary Carcinoma Diseases 0.000 description 1
- 102000003939 Membrane transport proteins Human genes 0.000 description 1
- 108090000301 Membrane transport proteins Proteins 0.000 description 1
- 206010027209 Meningitis cryptococcal Diseases 0.000 description 1
- 244000246386 Mentha pulegium Species 0.000 description 1
- 235000016257 Mentha pulegium Nutrition 0.000 description 1
- 235000004357 Mentha x piperita Nutrition 0.000 description 1
- 208000002030 Merkel cell carcinoma Diseases 0.000 description 1
- 201000009574 Mesenchymal Chondrosarcoma Diseases 0.000 description 1
- 206010027406 Mesothelioma Diseases 0.000 description 1
- 206010054949 Metaplasia Diseases 0.000 description 1
- 206010027480 Metastatic malignant melanoma Diseases 0.000 description 1
- 108060004795 Methyltransferase Proteins 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 102000029749 Microtubule Human genes 0.000 description 1
- 108091022875 Microtubule Proteins 0.000 description 1
- 241001183012 Modified Vaccinia Ankara virus Species 0.000 description 1
- 241000713869 Moloney murine leukemia virus Species 0.000 description 1
- 244000302512 Momordica charantia Species 0.000 description 1
- 235000009811 Momordica charantia Nutrition 0.000 description 1
- 241000700627 Monkeypox virus Species 0.000 description 1
- 101000686985 Mouse mammary tumor virus (strain C3H) Protein PR73 Proteins 0.000 description 1
- 206010057269 Mucoepidermoid carcinoma Diseases 0.000 description 1
- 208000010357 Mullerian Mixed Tumor Diseases 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 101100207071 Mus musculus Tnfsf8 gene Proteins 0.000 description 1
- 208000007101 Muscle Cramp Diseases 0.000 description 1
- 206010052904 Musculoskeletal stiffness Diseases 0.000 description 1
- 241000238367 Mya arenaria Species 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 101710198035 Myosin light chain kinase, smooth muscle Proteins 0.000 description 1
- 102100035044 Myosin light chain kinase, smooth muscle Human genes 0.000 description 1
- 201000002481 Myositis Diseases 0.000 description 1
- JDHILDINMRGULE-LURJTMIESA-N N(pros)-methyl-L-histidine Chemical compound CN1C=NC=C1C[C@H](N)C(O)=O JDHILDINMRGULE-LURJTMIESA-N 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
- JJIHLJJYMXLCOY-BYPYZUCNSA-N N-acetyl-L-serine Chemical compound CC(=O)N[C@@H](CO)C(O)=O JJIHLJJYMXLCOY-BYPYZUCNSA-N 0.000 description 1
- VIHYIVKEECZGOU-UHFFFAOYSA-N N-acetylimidazole Chemical compound CC(=O)N1C=CN=C1 VIHYIVKEECZGOU-UHFFFAOYSA-N 0.000 description 1
- PYUSHNKNPOHWEZ-YFKPBYRVSA-N N-formyl-L-methionine Chemical compound CSCC[C@@H](C(O)=O)NC=O PYUSHNKNPOHWEZ-YFKPBYRVSA-N 0.000 description 1
- 108010084333 N-palmitoyl-S-(2,3-bis(palmitoyloxy)propyl)cysteinyl-seryl-lysyl-lysyl-lysyl-lysine Proteins 0.000 description 1
- 125000001429 N-terminal alpha-amino-acid group Chemical group 0.000 description 1
- DNSKDVWSTPVFCT-LURJTMIESA-N NC(=N)NCCC[C@@H](C(O)=O)NNCCS(O)(=O)=O Chemical compound NC(=N)NCCC[C@@H](C(O)=O)NNCCS(O)(=O)=O DNSKDVWSTPVFCT-LURJTMIESA-N 0.000 description 1
- 108010018525 NFATC Transcription Factors Proteins 0.000 description 1
- 102100022680 NKG2-D type II integral membrane protein Human genes 0.000 description 1
- 108091061960 Naked DNA Proteins 0.000 description 1
- 108010004217 Natural Cytotoxicity Triggering Receptor 1 Proteins 0.000 description 1
- 108010004222 Natural Cytotoxicity Triggering Receptor 3 Proteins 0.000 description 1
- 102100032870 Natural cytotoxicity triggering receptor 1 Human genes 0.000 description 1
- 102100032851 Natural cytotoxicity triggering receptor 2 Human genes 0.000 description 1
- 102100032852 Natural cytotoxicity triggering receptor 3 Human genes 0.000 description 1
- 101710141230 Natural killer cell receptor 2B4 Proteins 0.000 description 1
- 206010029260 Neuroblastoma Diseases 0.000 description 1
- 206010060860 Neurological symptom Diseases 0.000 description 1
- 102000002710 Neurophysins Human genes 0.000 description 1
- 108010018674 Neurophysins Proteins 0.000 description 1
- 102000004459 Nitroreductase Human genes 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 102000007999 Nuclear Proteins Human genes 0.000 description 1
- 108010089610 Nuclear Proteins Proteins 0.000 description 1
- 102100035413 Nuclear factor of activated T-cells 5 Human genes 0.000 description 1
- 102100034404 Nuclear factor of activated T-cells, cytoplasmic 1 Human genes 0.000 description 1
- 101710151542 Nuclear factor of activated T-cells, cytoplasmic 1 Proteins 0.000 description 1
- 102100034400 Nuclear factor of activated T-cells, cytoplasmic 2 Human genes 0.000 description 1
- 101710151538 Nuclear factor of activated T-cells, cytoplasmic 2 Proteins 0.000 description 1
- 102100034399 Nuclear factor of activated T-cells, cytoplasmic 3 Human genes 0.000 description 1
- 101710151545 Nuclear factor of activated T-cells, cytoplasmic 3 Proteins 0.000 description 1
- 102100034398 Nuclear factor of activated T-cells, cytoplasmic 4 Human genes 0.000 description 1
- 101710151215 Nuclear factor of activated T-cells, cytoplasmic 4 Proteins 0.000 description 1
- 108020004711 Nucleic Acid Probes Proteins 0.000 description 1
- 108091005461 Nucleic proteins Proteins 0.000 description 1
- GOWLTLODGKPXMN-MEKRSRHXSA-N OM-174 Chemical compound O1[C@H](OP(O)(O)=O)[C@H](NC(=O)C[C@H](O)CCCCCCCCCCC)[C@@H](O)[C@H](O)[C@H]1CO[C@H]1[C@H](NC(=O)C[C@H](CCCCCCCCCCC)OC(=O)CCCCCCCCCCC)[C@@H](O)[C@H](OP(O)(O)=O)[C@@H](CO)O1 GOWLTLODGKPXMN-MEKRSRHXSA-N 0.000 description 1
- 101150007521 ORF III gene Proteins 0.000 description 1
- 208000007871 Odontogenic Tumors Diseases 0.000 description 1
- 206010030154 Oesophageal candidiasis Diseases 0.000 description 1
- AHLPHDHHMVZTML-UHFFFAOYSA-N Orn-delta-NH2 Natural products NCCCC(N)C(O)=O AHLPHDHHMVZTML-UHFFFAOYSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 208000010191 Osteitis Deformans Diseases 0.000 description 1
- 206010073261 Ovarian theca cell tumour Diseases 0.000 description 1
- 240000007019 Oxalis corniculata Species 0.000 description 1
- 102100034925 P-selectin glycoprotein ligand 1 Human genes 0.000 description 1
- 102000000470 PDZ domains Human genes 0.000 description 1
- 108050008994 PDZ domains Proteins 0.000 description 1
- 102100036257 PIN2/TERF1-interacting telomerase inhibitor 1 Human genes 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 239000012648 POLY-ICLC Substances 0.000 description 1
- 208000027868 Paget disease Diseases 0.000 description 1
- 241000701945 Parvoviridae Species 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- 108010092494 Periplasmic binding proteins Proteins 0.000 description 1
- 102000006335 Phosphate-Binding Proteins Human genes 0.000 description 1
- 108010058514 Phosphate-Binding Proteins Proteins 0.000 description 1
- ABLZXFCXXLZCGV-UHFFFAOYSA-N Phosphorous acid Chemical class OP(O)=O ABLZXFCXXLZCGV-UHFFFAOYSA-N 0.000 description 1
- 101100413173 Phytolacca americana PAP2 gene Proteins 0.000 description 1
- 208000009077 Pigmented Nevus Diseases 0.000 description 1
- 208000019262 Pilomatrix carcinoma Diseases 0.000 description 1
- 208000007641 Pinealoma Diseases 0.000 description 1
- 208000005384 Pneumocystis Pneumonia Diseases 0.000 description 1
- 206010073755 Pneumocystis jirovecii pneumonia Diseases 0.000 description 1
- 239000004952 Polyamide Substances 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- 241000282569 Pongo Species 0.000 description 1
- 241000282405 Pongo abelii Species 0.000 description 1
- 241000188845 Porcine adenovirus Species 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 101710101148 Probable 6-oxopurine nucleoside phosphorylase Proteins 0.000 description 1
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 1
- 108010007568 Protamines Proteins 0.000 description 1
- 102000007327 Protamines Human genes 0.000 description 1
- 241000589517 Pseudomonas aeruginosa Species 0.000 description 1
- 239000004373 Pullulan Substances 0.000 description 1
- 229920001218 Pullulan Polymers 0.000 description 1
- 102000030764 Purine-nucleoside phosphorylase Human genes 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 102000012751 Pyruvate Dehydrogenase Complex Human genes 0.000 description 1
- 108010090051 Pyruvate Dehydrogenase Complex Proteins 0.000 description 1
- IWYDHOAUDWTVEP-UHFFFAOYSA-N R-2-phenyl-2-hydroxyacetic acid Natural products OC(=O)C(O)C1=CC=CC=C1 IWYDHOAUDWTVEP-UHFFFAOYSA-N 0.000 description 1
- 102000014128 RANK Ligand Human genes 0.000 description 1
- 108010025832 RANK Ligand Proteins 0.000 description 1
- 102000044126 RNA-Binding Proteins Human genes 0.000 description 1
- 101710105008 RNA-binding protein Proteins 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 1
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 1
- 108700005075 Regulator Genes Proteins 0.000 description 1
- 201000000582 Retinoblastoma Diseases 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 108010039491 Ricin Proteins 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 102100029216 SLAM family member 5 Human genes 0.000 description 1
- 102100029198 SLAM family member 7 Human genes 0.000 description 1
- 108091006296 SLC2A1 Proteins 0.000 description 1
- 108010077895 Sarcosine Proteins 0.000 description 1
- 108090000184 Selectins Proteins 0.000 description 1
- 102000003800 Selectins Human genes 0.000 description 1
- 229920002684 Sepharose Polymers 0.000 description 1
- 208000000097 Sertoli-Leydig cell tumor Diseases 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- 244000000231 Sesamum indicum Species 0.000 description 1
- 102000034755 Sex Hormone-Binding Globulin Human genes 0.000 description 1
- 108010089417 Sex Hormone-Binding Globulin Proteins 0.000 description 1
- 229920001800 Shellac Polymers 0.000 description 1
- 208000003252 Signet Ring Cell Carcinoma Diseases 0.000 description 1
- BQCADISMDOOEFD-UHFFFAOYSA-N Silver Chemical compound [Ag] BQCADISMDOOEFD-UHFFFAOYSA-N 0.000 description 1
- 208000009574 Skin Appendage Carcinoma Diseases 0.000 description 1
- 101710087759 Sliding-clamp-loader small subunit Proteins 0.000 description 1
- 208000001203 Smallpox Diseases 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 208000005392 Spasm Diseases 0.000 description 1
- 108010088160 Staphylococcal Protein A Proteins 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 108091027544 Subgenomic mRNA Proteins 0.000 description 1
- KDYFGRWQOYBRFD-UHFFFAOYSA-N Succinic acid Natural products OC(=O)CCC(O)=O KDYFGRWQOYBRFD-UHFFFAOYSA-N 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical group [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 206010042553 Superficial spreading melanoma stage unspecified Diseases 0.000 description 1
- 108090000054 Syndecan-2 Proteins 0.000 description 1
- 101000946719 Synechococcus elongatus (strain PCC 7942 / FACHB-805) Carboxysome assembly protein CcmN Proteins 0.000 description 1
- 230000024932 T cell mediated immunity Effects 0.000 description 1
- 208000000389 T-cell leukemia Diseases 0.000 description 1
- 208000028530 T-cell lymphoblastic leukemia/lymphoma Diseases 0.000 description 1
- 206010042971 T-cell lymphoma Diseases 0.000 description 1
- 208000027585 T-cell non-Hodgkin lymphoma Diseases 0.000 description 1
- 102100035268 T-cell surface protein tactile Human genes 0.000 description 1
- 102000046283 TNF-Related Apoptosis-Inducing Ligand Human genes 0.000 description 1
- 108700012411 TNFSF10 Proteins 0.000 description 1
- 102100038717 TYRO protein tyrosine kinase-binding protein 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
- 108010008125 Tenascin Proteins 0.000 description 1
- 102100038126 Tenascin Human genes 0.000 description 1
- 206010043276 Teratoma Diseases 0.000 description 1
- 102100024554 Tetranectin Human genes 0.000 description 1
- NYTOUQBROMCLBJ-UHFFFAOYSA-N Tetranitromethane Chemical compound [O-][N+](=O)C([N+]([O-])=O)([N+]([O-])=O)[N+]([O-])=O NYTOUQBROMCLBJ-UHFFFAOYSA-N 0.000 description 1
- 210000000447 Th1 cell Anatomy 0.000 description 1
- 210000000068 Th17 cell Anatomy 0.000 description 1
- 210000004241 Th2 cell Anatomy 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical group OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- 201000009365 Thymic carcinoma Diseases 0.000 description 1
- 102000006601 Thymidine Kinase Human genes 0.000 description 1
- 108020004440 Thymidine kinase Proteins 0.000 description 1
- AUYYCJSJGJYCDS-LBPRGKRZSA-N Thyrolar Chemical compound IC1=CC(C[C@H](N)C(O)=O)=CC(I)=C1OC1=CC=C(O)C(I)=C1 AUYYCJSJGJYCDS-LBPRGKRZSA-N 0.000 description 1
- 102000009488 Thyroxine-Binding Proteins Human genes 0.000 description 1
- 108010048889 Thyroxine-Binding Proteins Proteins 0.000 description 1
- 229920001615 Tragacanth Polymers 0.000 description 1
- 102000011409 Transcobalamins Human genes 0.000 description 1
- 108010023603 Transcobalamins Proteins 0.000 description 1
- 102000014034 Transcortin Human genes 0.000 description 1
- 108010011095 Transcortin Proteins 0.000 description 1
- 102000010912 Transferrin-Binding Proteins Human genes 0.000 description 1
- 101800004564 Transforming growth factor alpha Proteins 0.000 description 1
- 102400001320 Transforming growth factor alpha Human genes 0.000 description 1
- UATJOMSPNYCXIX-UHFFFAOYSA-N Trinitrobenzene Chemical compound [O-][N+](=O)C1=CC([N+]([O-])=O)=CC([N+]([O-])=O)=C1 UATJOMSPNYCXIX-UHFFFAOYSA-N 0.000 description 1
- 102000011408 Tripartite Motif Proteins Human genes 0.000 description 1
- 108010023649 Tripartite Motif Proteins Proteins 0.000 description 1
- 229940122618 Trypsin inhibitor Drugs 0.000 description 1
- 101710162629 Trypsin inhibitor Proteins 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 102000012883 Tumor Necrosis Factor Ligand Superfamily Member 14 Human genes 0.000 description 1
- 108010065158 Tumor Necrosis Factor Ligand Superfamily Member 14 Proteins 0.000 description 1
- 102100024584 Tumor necrosis factor ligand superfamily member 12 Human genes 0.000 description 1
- 101710097155 Tumor necrosis factor ligand superfamily member 12 Proteins 0.000 description 1
- 102100024585 Tumor necrosis factor ligand superfamily member 13 Human genes 0.000 description 1
- 102100024587 Tumor necrosis factor ligand superfamily member 15 Human genes 0.000 description 1
- 102100035283 Tumor necrosis factor ligand superfamily member 18 Human genes 0.000 description 1
- 102100031988 Tumor necrosis factor ligand superfamily member 6 Human genes 0.000 description 1
- 102100029690 Tumor necrosis factor receptor superfamily member 13C Human genes 0.000 description 1
- 101710187830 Tumor necrosis factor receptor superfamily member 1B Proteins 0.000 description 1
- 102100033733 Tumor necrosis factor receptor superfamily member 1B Human genes 0.000 description 1
- 102100035284 Tumor necrosis factor receptor superfamily member 6B Human genes 0.000 description 1
- 206010054094 Tumour necrosis Diseases 0.000 description 1
- 102100021125 Tyrosine-protein kinase ZAP-70 Human genes 0.000 description 1
- 108090000848 Ubiquitin Proteins 0.000 description 1
- 102000044159 Ubiquitin Human genes 0.000 description 1
- 108010079206 V-Set Domain-Containing T-Cell Activation Inhibitor 1 Proteins 0.000 description 1
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 1
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 1
- 241000870995 Variola Species 0.000 description 1
- 241000700647 Variola virus Species 0.000 description 1
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 1
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 1
- 240000001866 Vernicia fordii Species 0.000 description 1
- 241001416177 Vicugna pacos Species 0.000 description 1
- 108020005202 Viral DNA Proteins 0.000 description 1
- 108010003533 Viral Envelope Proteins Proteins 0.000 description 1
- 108010067390 Viral Proteins Proteins 0.000 description 1
- 102000050760 Vitamin D-binding protein Human genes 0.000 description 1
- 101710179590 Vitamin D-binding protein Proteins 0.000 description 1
- 208000008383 Wilms tumor Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- GANNOFFDYMSBSZ-UHFFFAOYSA-N [AlH3].[Mg] Chemical compound [AlH3].[Mg] GANNOFFDYMSBSZ-UHFFFAOYSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 description 1
- 238000005903 acid hydrolysis reaction Methods 0.000 description 1
- 208000006336 acinar cell carcinoma Diseases 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000003044 adaptive effect Effects 0.000 description 1
- 102000035181 adaptor proteins Human genes 0.000 description 1
- 108091005764 adaptor proteins Proteins 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 208000002517 adenoid cystic carcinoma Diseases 0.000 description 1
- 201000008395 adenosquamous carcinoma Diseases 0.000 description 1
- 108700010877 adenoviridae proteins Proteins 0.000 description 1
- 238000009098 adjuvant therapy Methods 0.000 description 1
- 208000020990 adrenal cortex carcinoma Diseases 0.000 description 1
- 208000007128 adrenocortical carcinoma Diseases 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 229940072056 alginate Drugs 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- 150000001447 alkali salts Chemical class 0.000 description 1
- 125000002877 alkyl aryl group Chemical group 0.000 description 1
- 239000013566 allergen Substances 0.000 description 1
- 230000000961 alloantigen Effects 0.000 description 1
- 230000000735 allogeneic effect Effects 0.000 description 1
- 150000001371 alpha-amino acids Chemical class 0.000 description 1
- 235000008206 alpha-amino acids Nutrition 0.000 description 1
- 108010001818 alpha-sarcin Proteins 0.000 description 1
- 206010065867 alveolar rhabdomyosarcoma Diseases 0.000 description 1
- 208000006431 amelanotic melanoma Diseases 0.000 description 1
- 208000010029 ameloblastoma Diseases 0.000 description 1
- 150000003862 amino acid derivatives Chemical class 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- DZHSAHHDTRWUTF-SIQRNXPUSA-N amyloid-beta polypeptide 42 Chemical compound C([C@@H](C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@H](C(=O)NCC(=O)N[C@@H](CO)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)NCC(=O)N[C@@H](C(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H]([C@@H](C)CC)C(=O)N[C@@H](C)C(O)=O)[C@@H](C)CC)C(C)C)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@@H](NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](N)CC(O)=O)C(C)C)C(C)C)C1=CC=CC=C1 DZHSAHHDTRWUTF-SIQRNXPUSA-N 0.000 description 1
- 102000010640 androgen binding protein Human genes 0.000 description 1
- 108010077825 androgen binding protein Proteins 0.000 description 1
- 150000008064 anhydrides Chemical class 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000003042 antagnostic effect Effects 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 230000000845 anti-microbial effect Effects 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 230000030741 antigen processing and presentation Effects 0.000 description 1
- 229940125715 antihistaminic agent Drugs 0.000 description 1
- 239000000739 antihistaminic agent Substances 0.000 description 1
- 201000007436 apocrine adenocarcinoma Diseases 0.000 description 1
- 239000012736 aqueous medium Substances 0.000 description 1
- 239000008346 aqueous phase Substances 0.000 description 1
- 239000008135 aqueous vehicle Substances 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 201000005476 astroblastoma Diseases 0.000 description 1
- 210000004666 bacterial spore Anatomy 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 201000007551 basophilic adenocarcinoma Diseases 0.000 description 1
- 208000001119 benign fibrous histiocytoma Diseases 0.000 description 1
- 235000010233 benzoic acid Nutrition 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- 229940000635 beta-alanine Drugs 0.000 description 1
- 150000001576 beta-amino acids Chemical class 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 239000003833 bile salt Substances 0.000 description 1
- 229940093761 bile salts Drugs 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- 229960000074 biopharmaceutical Drugs 0.000 description 1
- 238000005460 biophysical method Methods 0.000 description 1
- 230000002051 biphasic effect Effects 0.000 description 1
- 239000010836 blood and blood product Substances 0.000 description 1
- 229940125691 blood product Drugs 0.000 description 1
- 208000007047 blue nevus Diseases 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- 201000011143 bone giant cell tumor Diseases 0.000 description 1
- 201000003714 breast lobular carcinoma Diseases 0.000 description 1
- 201000011054 breast malignant phyllodes tumor Diseases 0.000 description 1
- 239000006189 buccal tablet Substances 0.000 description 1
- 239000000337 buffer salt Substances 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
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 125000004744 butyloxycarbonyl group Chemical group 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- AXCZMVOFGPJBDE-UHFFFAOYSA-L calcium dihydroxide Chemical compound [OH-].[OH-].[Ca+2] AXCZMVOFGPJBDE-UHFFFAOYSA-L 0.000 description 1
- 239000000920 calcium hydroxide Substances 0.000 description 1
- 229910001861 calcium hydroxide Inorganic materials 0.000 description 1
- 102000028861 calmodulin binding Human genes 0.000 description 1
- 108091000084 calmodulin binding Proteins 0.000 description 1
- 208000035269 cancer or benign tumor Diseases 0.000 description 1
- 238000005251 capillar electrophoresis Methods 0.000 description 1
- 150000004657 carbamic acid derivatives Chemical class 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 239000002041 carbon nanotube Substances 0.000 description 1
- 229910021393 carbon nanotube Inorganic materials 0.000 description 1
- 125000005392 carboxamide group Chemical group NC(=O)* 0.000 description 1
- 150000007942 carboxylates Chemical group 0.000 description 1
- 208000002458 carcinoid tumor Diseases 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 150000001767 cationic compounds Chemical class 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 230000011748 cell maturation Effects 0.000 description 1
- 230000005859 cell recognition Effects 0.000 description 1
- 239000002458 cell surface marker Substances 0.000 description 1
- 210000004520 cell wall skeleton Anatomy 0.000 description 1
- 230000002490 cerebral effect Effects 0.000 description 1
- 201000002891 ceruminous adenocarcinoma Diseases 0.000 description 1
- 208000024188 ceruminous carcinoma Diseases 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 239000007958 cherry flavor Substances 0.000 description 1
- 102000021178 chitin binding proteins Human genes 0.000 description 1
- 108091011157 chitin binding proteins Proteins 0.000 description 1
- 229940045110 chitosan Drugs 0.000 description 1
- VDQQXEISLMTGAB-UHFFFAOYSA-N chloramine T Chemical compound [Na+].CC1=CC=C(S(=O)(=O)[N-]Cl)C=C1 VDQQXEISLMTGAB-UHFFFAOYSA-N 0.000 description 1
- 229940106681 chloroacetic acid Drugs 0.000 description 1
- 208000006990 cholangiocarcinoma Diseases 0.000 description 1
- 235000012000 cholesterol Nutrition 0.000 description 1
- 201000005217 chondroblastoma Diseases 0.000 description 1
- 201000010240 chromophobe renal cell carcinoma Diseases 0.000 description 1
- 235000013985 cinnamic acid Nutrition 0.000 description 1
- 229930016911 cinnamic acid Natural products 0.000 description 1
- 235000015165 citric acid Nutrition 0.000 description 1
- 229960002173 citrulline Drugs 0.000 description 1
- 208000029664 classic familial adenomatous polyposis Diseases 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 238000007621 cluster analysis Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 208000011588 combined hepatocellular carcinoma and cholangiocarcinoma Diseases 0.000 description 1
- 230000000536 complexating effect Effects 0.000 description 1
- 239000002131 composite material Substances 0.000 description 1
- 238000009833 condensation Methods 0.000 description 1
- 230000005494 condensation Effects 0.000 description 1
- 230000009352 congenital transmission Effects 0.000 description 1
- 210000000795 conjunctiva Anatomy 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 238000011254 conventional chemotherapy Methods 0.000 description 1
- 229910001431 copper ion Inorganic materials 0.000 description 1
- 210000004087 cornea Anatomy 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- CNVQLPPZGABUCM-LIGYZCPXSA-N ctx toxin Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CC(N)=O)C(=O)N[C@H](C(=O)N[C@@H](CO)C(=O)N[C@H]1CSSC[C@H]2C(=O)N[C@H](C(N[C@@H](CC(N)=O)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@H]3CSSC[C@@H](C(N[C@@H](CC=4C5=CC=CC=C5NC=4)C(=O)N[C@@H](CO)C(=O)N[C@H](C(=O)N[C@@H](CSSC[C@H](NC(=O)[C@H](CCCNC(N)=N)NC3=O)C(=O)N[C@@H](CC=3C=CC(O)=CC=3)C(=O)N[C@@H](CO)C(O)=O)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC=3NC=NC=3)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N2)C(C)C)=O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H]([C@@H](C)O)NC1=O)=O)CCSC)C(C)C)[C@@H](C)O)NC(=O)[C@H]1NC(=O)CC1)C1=CC=CC=C1 CNVQLPPZGABUCM-LIGYZCPXSA-N 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 208000035250 cutaneous malignant susceptibility to 1 melanoma Diseases 0.000 description 1
- XLJMAIOERFSOGZ-UHFFFAOYSA-M cyanate Chemical compound [O-]C#N XLJMAIOERFSOGZ-UHFFFAOYSA-M 0.000 description 1
- 108010079940 cyanogenic beta-glucosidase Proteins 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- OILAIQUEIWYQPH-UHFFFAOYSA-N cyclohexane-1,2-dione Chemical compound O=C1CCCCC1=O OILAIQUEIWYQPH-UHFFFAOYSA-N 0.000 description 1
- 208000002445 cystadenocarcinoma Diseases 0.000 description 1
- LEVWYRKDKASIDU-IMJSIDKUSA-N cystine group Chemical group C([C@@H](C(=O)O)N)SSC[C@@H](C(=O)O)N LEVWYRKDKASIDU-IMJSIDKUSA-N 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 239000003145 cytotoxic factor Substances 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 125000001295 dansyl group Chemical group [H]C1=C([H])C(N(C([H])([H])[H])C([H])([H])[H])=C2C([H])=C([H])C([H])=C(C2=C1[H])S(*)(=O)=O 0.000 description 1
- 230000006240 deamidation Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 239000003405 delayed action preparation Substances 0.000 description 1
- YSMODUONRAFBET-UHFFFAOYSA-N delta-DL-hydroxylysine Natural products NCC(O)CCC(N)C(O)=O YSMODUONRAFBET-UHFFFAOYSA-N 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 108010017271 denileukin diftitox Proteins 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 239000003599 detergent Substances 0.000 description 1
- 238000006642 detritylation reaction Methods 0.000 description 1
- 125000004386 diacrylate group Chemical group 0.000 description 1
- GUJOJGAPFQRJSV-UHFFFAOYSA-N dialuminum;dioxosilane;oxygen(2-);hydrate Chemical compound O.[O-2].[O-2].[O-2].[Al+3].[Al+3].O=[Si]=O.O=[Si]=O.O=[Si]=O.O=[Si]=O GUJOJGAPFQRJSV-UHFFFAOYSA-N 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 150000004985 diamines Chemical class 0.000 description 1
- 229930191339 dianthin Natural products 0.000 description 1
- 229910000390 dicalcium phosphate Inorganic materials 0.000 description 1
- 235000019700 dicalcium phosphate Nutrition 0.000 description 1
- 229940038472 dicalcium phosphate Drugs 0.000 description 1
- 125000004989 dicarbonyl group Chemical group 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- FFYPMLJYZAEMQB-UHFFFAOYSA-N diethyl pyrocarbonate Chemical compound CCOC(=O)OC(=O)OCC FFYPMLJYZAEMQB-UHFFFAOYSA-N 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 206010013023 diphtheria Diseases 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 150000002019 disulfides Chemical class 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
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 201000009409 embryonal rhabdomyosarcoma Diseases 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 230000002121 endocytic effect Effects 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 108010028531 enomycin Proteins 0.000 description 1
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 description 1
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 description 1
- 201000010877 epithelioid cell melanoma Diseases 0.000 description 1
- YSMODUONRAFBET-UHNVWZDZSA-N erythro-5-hydroxy-L-lysine Chemical compound NC[C@H](O)CC[C@H](N)C(O)=O YSMODUONRAFBET-UHNVWZDZSA-N 0.000 description 1
- 201000005655 esophageal candidiasis Diseases 0.000 description 1
- CCIVGXIOQKPBKL-UHFFFAOYSA-M ethanesulfonate Chemical compound CCS([O-])(=O)=O CCIVGXIOQKPBKL-UHFFFAOYSA-M 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 238000001704 evaporation Methods 0.000 description 1
- 230000008020 evaporation Effects 0.000 description 1
- 230000017188 evasion or tolerance of host immune response Effects 0.000 description 1
- 210000001808 exosome Anatomy 0.000 description 1
- 238000010265 fast atom bombardment Methods 0.000 description 1
- 239000003925 fat Substances 0.000 description 1
- 108091022862 fatty acid binding Proteins 0.000 description 1
- 201000001169 fibrillary astrocytoma Diseases 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 230000022244 formylation Effects 0.000 description 1
- 238000006170 formylation reaction Methods 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 239000001530 fumaric acid Substances 0.000 description 1
- 235000011087 fumaric acid Nutrition 0.000 description 1
- 238000002825 functional assay Methods 0.000 description 1
- 125000000524 functional group Chemical group 0.000 description 1
- 230000002538 fungal effect Effects 0.000 description 1
- 125000003838 furazanyl group Chemical group 0.000 description 1
- 125000002541 furyl group Chemical group 0.000 description 1
- IECPWNUMDGFDKC-MZJAQBGESA-N fusidic acid Chemical class O[C@@H]([C@@H]12)C[C@H]3\C(=C(/CCC=C(C)C)C(O)=O)[C@@H](OC(C)=O)C[C@]3(C)[C@@]2(C)CC[C@@H]2[C@]1(C)CC[C@@H](O)[C@H]2C IECPWNUMDGFDKC-MZJAQBGESA-N 0.000 description 1
- BTCSSZJGUNDROE-UHFFFAOYSA-N gamma-aminobutyric acid Chemical compound NCCCC(O)=O BTCSSZJGUNDROE-UHFFFAOYSA-N 0.000 description 1
- 210000004475 gamma-delta t lymphocyte Anatomy 0.000 description 1
- 208000015419 gastrin-producing neuroendocrine tumor Diseases 0.000 description 1
- 201000000052 gastrinoma Diseases 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000007903 gelatin capsule Substances 0.000 description 1
- 238000001476 gene delivery Methods 0.000 description 1
- 102000034356 gene-regulatory proteins Human genes 0.000 description 1
- 108091006104 gene-regulatory proteins Proteins 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 230000001295 genetical effect Effects 0.000 description 1
- 208000005017 glioblastoma Diseases 0.000 description 1
- 201000002264 glomangiosarcoma Diseases 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- YMAWOPBAYDPSLA-UHFFFAOYSA-N glycylglycine Chemical compound [NH3+]CC(=O)NCC([O-])=O YMAWOPBAYDPSLA-UHFFFAOYSA-N 0.000 description 1
- 210000002288 golgi apparatus Anatomy 0.000 description 1
- 201000007574 granular cell carcinoma Diseases 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 229910052736 halogen Inorganic materials 0.000 description 1
- 150000002367 halogens Chemical class 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000035876 healing Effects 0.000 description 1
- 230000002440 hepatic effect Effects 0.000 description 1
- 208000006359 hepatoblastoma Diseases 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- 206010019847 hepatosplenomegaly Diseases 0.000 description 1
- 125000001072 heteroaryl group Chemical group 0.000 description 1
- 208000029824 high grade glioma Diseases 0.000 description 1
- 125000000487 histidyl group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C([H])=N1 0.000 description 1
- 230000001744 histochemical effect Effects 0.000 description 1
- 235000001050 hortel pimenta Nutrition 0.000 description 1
- BHEPBYXIRTUNPN-UHFFFAOYSA-N hydridophosphorus(.) (triplet) Chemical group [PH] BHEPBYXIRTUNPN-UHFFFAOYSA-N 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 1
- 229960002591 hydroxyproline Drugs 0.000 description 1
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 1
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 description 1
- 230000000148 hypercalcaemia Effects 0.000 description 1
- 208000030915 hypercalcemia disease Diseases 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 125000002632 imidazolidinyl group Chemical group 0.000 description 1
- 125000002636 imidazolinyl group Chemical group 0.000 description 1
- 125000002883 imidazolyl group Chemical group 0.000 description 1
- 125000001841 imino group Chemical group [H]N=* 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
- 210000002861 immature t-cell Anatomy 0.000 description 1
- 210000001822 immobilized cell Anatomy 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 239000012642 immune effector Substances 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 229940121354 immunomodulator Drugs 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 239000003022 immunostimulating agent Substances 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 230000002757 inflammatory effect Effects 0.000 description 1
- 206010022000 influenza Diseases 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 238000009413 insulation Methods 0.000 description 1
- 102000028416 insulin-like growth factor binding Human genes 0.000 description 1
- 108091022911 insulin-like growth factor binding Proteins 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 230000004073 interleukin-2 production Effects 0.000 description 1
- 108010074108 interleukin-21 Proteins 0.000 description 1
- 229940028885 interleukin-4 Drugs 0.000 description 1
- 238000001361 intraarterial administration Methods 0.000 description 1
- 230000010189 intracellular transport Effects 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 238000007914 intraventricular administration Methods 0.000 description 1
- 229940029339 inulin Drugs 0.000 description 1
- JYJIGFIDKWBXDU-MNNPPOADSA-N inulin Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)OC[C@]1(OC[C@]2(OC[C@]3(OC[C@]4(OC[C@]5(OC[C@]6(OC[C@]7(OC[C@]8(OC[C@]9(OC[C@]%10(OC[C@]%11(OC[C@]%12(OC[C@]%13(OC[C@]%14(OC[C@]%15(OC[C@]%16(OC[C@]%17(OC[C@]%18(OC[C@]%19(OC[C@]%20(OC[C@]%21(OC[C@]%22(OC[C@]%23(OC[C@]%24(OC[C@]%25(OC[C@]%26(OC[C@]%27(OC[C@]%28(OC[C@]%29(OC[C@]%30(OC[C@]%31(OC[C@]%32(OC[C@]%33(OC[C@]%34(OC[C@]%35(OC[C@]%36(O[C@@H]%37[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O%37)O)[C@H]([C@H](O)[C@@H](CO)O%36)O)[C@H]([C@H](O)[C@@H](CO)O%35)O)[C@H]([C@H](O)[C@@H](CO)O%34)O)[C@H]([C@H](O)[C@@H](CO)O%33)O)[C@H]([C@H](O)[C@@H](CO)O%32)O)[C@H]([C@H](O)[C@@H](CO)O%31)O)[C@H]([C@H](O)[C@@H](CO)O%30)O)[C@H]([C@H](O)[C@@H](CO)O%29)O)[C@H]([C@H](O)[C@@H](CO)O%28)O)[C@H]([C@H](O)[C@@H](CO)O%27)O)[C@H]([C@H](O)[C@@H](CO)O%26)O)[C@H]([C@H](O)[C@@H](CO)O%25)O)[C@H]([C@H](O)[C@@H](CO)O%24)O)[C@H]([C@H](O)[C@@H](CO)O%23)O)[C@H]([C@H](O)[C@@H](CO)O%22)O)[C@H]([C@H](O)[C@@H](CO)O%21)O)[C@H]([C@H](O)[C@@H](CO)O%20)O)[C@H]([C@H](O)[C@@H](CO)O%19)O)[C@H]([C@H](O)[C@@H](CO)O%18)O)[C@H]([C@H](O)[C@@H](CO)O%17)O)[C@H]([C@H](O)[C@@H](CO)O%16)O)[C@H]([C@H](O)[C@@H](CO)O%15)O)[C@H]([C@H](O)[C@@H](CO)O%14)O)[C@H]([C@H](O)[C@@H](CO)O%13)O)[C@H]([C@H](O)[C@@H](CO)O%12)O)[C@H]([C@H](O)[C@@H](CO)O%11)O)[C@H]([C@H](O)[C@@H](CO)O%10)O)[C@H]([C@H](O)[C@@H](CO)O9)O)[C@H]([C@H](O)[C@@H](CO)O8)O)[C@H]([C@H](O)[C@@H](CO)O7)O)[C@H]([C@H](O)[C@@H](CO)O6)O)[C@H]([C@H](O)[C@@H](CO)O5)O)[C@H]([C@H](O)[C@@H](CO)O4)O)[C@H]([C@H](O)[C@@H](CO)O3)O)[C@H]([C@H](O)[C@@H](CO)O2)O)[C@@H](O)[C@H](O)[C@@H](CO)O1 JYJIGFIDKWBXDU-MNNPPOADSA-N 0.000 description 1
- 108010028930 invariant chain Proteins 0.000 description 1
- 206010073096 invasive lobular breast carcinoma Diseases 0.000 description 1
- 239000011630 iodine Substances 0.000 description 1
- 229910052740 iodine Inorganic materials 0.000 description 1
- JDNTWHVOXJZDSN-UHFFFAOYSA-N iodoacetic acid Chemical compound OC(=O)CI JDNTWHVOXJZDSN-UHFFFAOYSA-N 0.000 description 1
- 238000004255 ion exchange chromatography Methods 0.000 description 1
- 239000003456 ion exchange resin Substances 0.000 description 1
- 229920003303 ion-exchange polymer Polymers 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- JXDYKVIHCLTXOP-UHFFFAOYSA-N isatin Chemical compound C1=CC=C2C(=O)C(=O)NC2=C1 JXDYKVIHCLTXOP-UHFFFAOYSA-N 0.000 description 1
- 208000028867 ischemia Diseases 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 125000001786 isothiazolyl group Chemical group 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 125000000842 isoxazolyl group Chemical group 0.000 description 1
- 230000000366 juvenile effect Effects 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 208000022013 kidney Wilms tumor Diseases 0.000 description 1
- 210000003292 kidney cell Anatomy 0.000 description 1
- 235000020061 kirsch Nutrition 0.000 description 1
- 239000004310 lactic acid Substances 0.000 description 1
- 235000014655 lactic acid Nutrition 0.000 description 1
- 229960000448 lactic acid Drugs 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 229950008325 levothyroxine Drugs 0.000 description 1
- XMGQYMWWDOXHJM-UHFFFAOYSA-N limonene Chemical compound CC(=C)C1CCC(C)=CC1 XMGQYMWWDOXHJM-UHFFFAOYSA-N 0.000 description 1
- 206010024627 liposarcoma Diseases 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 201000000014 lung giant cell carcinoma Diseases 0.000 description 1
- 210000002751 lymph Anatomy 0.000 description 1
- 208000012804 lymphangiosarcoma Diseases 0.000 description 1
- 208000018555 lymphatic system disease Diseases 0.000 description 1
- 230000000527 lymphocytic effect Effects 0.000 description 1
- 201000010953 lymphoepithelioma-like carcinoma Diseases 0.000 description 1
- 239000012931 lyophilized formulation Substances 0.000 description 1
- ZLNQQNXFFQJAID-UHFFFAOYSA-L magnesium carbonate Chemical compound [Mg+2].[O-]C([O-])=O ZLNQQNXFFQJAID-UHFFFAOYSA-L 0.000 description 1
- 239000001095 magnesium carbonate Substances 0.000 description 1
- 229910000021 magnesium carbonate Inorganic materials 0.000 description 1
- 238000012423 maintenance Methods 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
- 208000018013 malignant glomus tumor Diseases 0.000 description 1
- 201000004102 malignant granular cell myoblastoma Diseases 0.000 description 1
- 206010061526 malignant mesenchymoma Diseases 0.000 description 1
- 201000009020 malignant peripheral nerve sheath tumor Diseases 0.000 description 1
- 201000002338 malignant struma ovarii Diseases 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- 208000027202 mammary Paget disease Diseases 0.000 description 1
- 229960002510 mandelic acid Drugs 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 108010082117 matrigel Proteins 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 238000000816 matrix-assisted laser desorption--ionisation Methods 0.000 description 1
- 230000035800 maturation Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 208000023356 medullary thyroid gland carcinoma Diseases 0.000 description 1
- 210000004779 membrane envelope Anatomy 0.000 description 1
- 206010027191 meningioma Diseases 0.000 description 1
- 230000015689 metaplastic ossification Effects 0.000 description 1
- 208000021039 metastatic melanoma Diseases 0.000 description 1
- TWXDDNPPQUTEOV-FVGYRXGTSA-N methamphetamine hydrochloride Chemical compound Cl.CN[C@@H](C)CC1=CC=CC=C1 TWXDDNPPQUTEOV-FVGYRXGTSA-N 0.000 description 1
- 229940098779 methanesulfonic acid Drugs 0.000 description 1
- UZKWTJUDCOPSNM-UHFFFAOYSA-N methoxybenzene Substances CCCCOC=C UZKWTJUDCOPSNM-UHFFFAOYSA-N 0.000 description 1
- ZCQGVFNHUATAJY-UHFFFAOYSA-N methyl 2-[methyl(prop-2-enoyl)amino]acetate Chemical compound COC(=O)CN(C)C(=O)C=C ZCQGVFNHUATAJY-UHFFFAOYSA-N 0.000 description 1
- WBYWAXJHAXSJNI-UHFFFAOYSA-N methyl p-hydroxycinnamate Natural products OC(=O)C=CC1=CC=CC=C1 WBYWAXJHAXSJNI-UHFFFAOYSA-N 0.000 description 1
- OSWPMRLSEDHDFF-UHFFFAOYSA-N methyl salicylate Chemical compound COC(=O)C1=CC=CC=C1O OSWPMRLSEDHDFF-UHFFFAOYSA-N 0.000 description 1
- 125000000250 methylamino group Chemical group [H]N(*)C([H])([H])[H] 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 239000010445 mica Substances 0.000 description 1
- 229910052618 mica group Inorganic materials 0.000 description 1
- 238000002493 microarray Methods 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 210000004688 microtubule Anatomy 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 235000010755 mineral Nutrition 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 201000010225 mixed cell type cancer Diseases 0.000 description 1
- 208000029638 mixed neoplasm Diseases 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 108010010621 modeccin Proteins 0.000 description 1
- 239000002991 molded plastic Substances 0.000 description 1
- 238000001823 molecular biology technique Methods 0.000 description 1
- 239000002062 molecular scaffold Substances 0.000 description 1
- 229910052901 montmorillonite Inorganic materials 0.000 description 1
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 1
- 125000002757 morpholinyl group Chemical group 0.000 description 1
- 201000010879 mucinous adenocarcinoma Diseases 0.000 description 1
- 208000010492 mucinous cystadenocarcinoma Diseases 0.000 description 1
- 230000007498 myristoylation Effects 0.000 description 1
- 208000001611 myxosarcoma Diseases 0.000 description 1
- CMWYAOXYQATXSI-UHFFFAOYSA-N n,n-dimethylformamide;piperidine Chemical compound CN(C)C=O.C1CCNCC1 CMWYAOXYQATXSI-UHFFFAOYSA-N 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- OHDXDNUPVVYWOV-UHFFFAOYSA-N n-methyl-1-(2-naphthalen-1-ylsulfanylphenyl)methanamine Chemical compound CNCC1=CC=CC=C1SC1=CC=CC2=CC=CC=C12 OHDXDNUPVVYWOV-UHFFFAOYSA-N 0.000 description 1
- 239000005543 nano-size silicon particle Substances 0.000 description 1
- 229940037525 nasal preparations Drugs 0.000 description 1
- 239000007922 nasal spray Substances 0.000 description 1
- 239000006218 nasal suppository Substances 0.000 description 1
- 208000014761 nasopharyngeal type undifferentiated carcinoma Diseases 0.000 description 1
- 201000008026 nephroblastoma Diseases 0.000 description 1
- 208000007538 neurilemmoma Diseases 0.000 description 1
- 208000027831 neuroepithelial neoplasm Diseases 0.000 description 1
- 208000029974 neurofibrosarcoma Diseases 0.000 description 1
- 230000001272 neurogenic effect Effects 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- FEMOMIGRRWSMCU-UHFFFAOYSA-N ninhydrin Chemical compound C1=CC=C2C(=O)C(O)(O)C(=O)C2=C1 FEMOMIGRRWSMCU-UHFFFAOYSA-N 0.000 description 1
- VXAPDXVBDZRZKP-UHFFFAOYSA-N nitric acid phosphoric acid Chemical compound O[N+]([O-])=O.OP(O)(O)=O VXAPDXVBDZRZKP-UHFFFAOYSA-N 0.000 description 1
- 108020001162 nitroreductase Proteins 0.000 description 1
- 239000002687 nonaqueous vehicle Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000002414 normal-phase solid-phase extraction Methods 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 238000007899 nucleic acid hybridization Methods 0.000 description 1
- 239000002853 nucleic acid probe Substances 0.000 description 1
- 230000000269 nucleophilic effect Effects 0.000 description 1
- 125000003835 nucleoside group Chemical group 0.000 description 1
- 208000027825 odontogenic neoplasm Diseases 0.000 description 1
- 231100000590 oncogenic Toxicity 0.000 description 1
- 230000002246 oncogenic effect Effects 0.000 description 1
- 244000309459 oncolytic virus Species 0.000 description 1
- 229940100027 ontak Drugs 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 239000008203 oral pharmaceutical composition Substances 0.000 description 1
- 239000007968 orange flavor Substances 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 230000008520 organization Effects 0.000 description 1
- 229960003104 ornithine Drugs 0.000 description 1
- 201000008968 osteosarcoma Diseases 0.000 description 1
- 230000002611 ovarian Effects 0.000 description 1
- 208000012221 ovarian Sertoli-Leydig cell tumor Diseases 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 235000006408 oxalic acid Nutrition 0.000 description 1
- 125000002971 oxazolyl group Chemical group 0.000 description 1
- 108010090358 p300-CBP Transcription Factors Proteins 0.000 description 1
- 102000013034 p300-CBP Transcription Factors Human genes 0.000 description 1
- 229910052763 palladium Inorganic materials 0.000 description 1
- 230000026792 palmitoylation Effects 0.000 description 1
- FJKROLUGYXJWQN-UHFFFAOYSA-N papa-hydroxy-benzoic acid Natural products OC(=O)C1=CC=C(O)C=C1 FJKROLUGYXJWQN-UHFFFAOYSA-N 0.000 description 1
- 208000004019 papillary adenocarcinoma Diseases 0.000 description 1
- 201000010198 papillary carcinoma Diseases 0.000 description 1
- 201000010210 papillary cystadenocarcinoma Diseases 0.000 description 1
- 208000024641 papillary serous cystadenocarcinoma Diseases 0.000 description 1
- 201000001494 papillary transitional carcinoma Diseases 0.000 description 1
- 208000031101 papillary transitional cell carcinoma Diseases 0.000 description 1
- 244000052769 pathogen Species 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 229960001639 penicillamine Drugs 0.000 description 1
- LQRJAEQXMSMEDP-XCHBZYMASA-N peptide a Chemical compound N([C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](C)C(=O)NCCCC[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)C(\NC(=O)[C@@H](CCCCN)NC(=O)CNC(C)=O)=C/C=1C=CC=CC=1)C(N)=O)C(=O)C(\NC(=O)[C@@H](CCCCN)NC(=O)CNC(C)=O)=C\C1=CC=CC=C1 LQRJAEQXMSMEDP-XCHBZYMASA-N 0.000 description 1
- 239000000816 peptidomimetic Substances 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 108010076042 phenomycin Proteins 0.000 description 1
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 1
- 208000028591 pheochromocytoma Diseases 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 102000051624 phosphatidylethanolamine binding protein Human genes 0.000 description 1
- 108700021017 phosphatidylethanolamine binding protein Proteins 0.000 description 1
- 150000004713 phosphodiesters Chemical class 0.000 description 1
- 150000003904 phospholipids Chemical class 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 150000003016 phosphoric acids Chemical class 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
- 208000024724 pineal body neoplasm Diseases 0.000 description 1
- 201000004123 pineal gland cancer Diseases 0.000 description 1
- 125000004193 piperazinyl group Chemical group 0.000 description 1
- 125000000587 piperidin-1-yl group Chemical group [H]C1([H])N(*)C([H])([H])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 125000005936 piperidyl group Chemical group 0.000 description 1
- 208000021857 pituitary gland basophilic carcinoma Diseases 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Substances [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 1
- 230000004983 pleiotropic effect Effects 0.000 description 1
- 201000000317 pneumocystosis Diseases 0.000 description 1
- 108010089520 pol Gene Products Proteins 0.000 description 1
- 229920001993 poloxamer 188 Polymers 0.000 description 1
- 229940044519 poloxamer 188 Drugs 0.000 description 1
- 108700002563 poly ICLC Proteins 0.000 description 1
- 229940115270 poly iclc Drugs 0.000 description 1
- 229920000962 poly(amidoamine) Polymers 0.000 description 1
- 229920001606 poly(lactic acid-co-glycolic acid) Polymers 0.000 description 1
- 229920000333 poly(propyleneimine) Polymers 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 229920002647 polyamide Polymers 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 230000001884 polyglutamylation Effects 0.000 description 1
- 208000005987 polymyositis Diseases 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 235000010482 polyoxyethylene sorbitan monooleate Nutrition 0.000 description 1
- 230000006267 polysialylation Effects 0.000 description 1
- 229920000053 polysorbate 80 Polymers 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- GKKCIDNWFBPDBW-UHFFFAOYSA-M potassium cyanate Chemical compound [K]OC#N GKKCIDNWFBPDBW-UHFFFAOYSA-M 0.000 description 1
- 229940116317 potato starch Drugs 0.000 description 1
- 229940124606 potential therapeutic agent Drugs 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000002062 proliferating effect Effects 0.000 description 1
- 150000003147 proline derivatives Chemical class 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 125000001500 prolyl group Chemical group [H]N1C([H])(C(=O)[*])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 235000019260 propionic acid Nutrition 0.000 description 1
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical class CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 1
- 229940048914 protamine Drugs 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 108020001580 protein domains Proteins 0.000 description 1
- 230000004850 protein–protein interaction Effects 0.000 description 1
- 201000008520 protoplasmic astrocytoma Diseases 0.000 description 1
- 235000019423 pullulan Nutrition 0.000 description 1
- 230000002685 pulmonary effect Effects 0.000 description 1
- 125000004309 pyranyl group Chemical group O1C(C=CC=C1)* 0.000 description 1
- 125000003373 pyrazinyl group Chemical group 0.000 description 1
- 125000003072 pyrazolidinyl group Chemical group 0.000 description 1
- 125000002755 pyrazolinyl group Chemical group 0.000 description 1
- 125000003226 pyrazolyl group Chemical group 0.000 description 1
- 125000002098 pyridazinyl group Chemical group 0.000 description 1
- 125000004076 pyridyl group Chemical group 0.000 description 1
- 125000000714 pyrimidinyl group Chemical group 0.000 description 1
- 125000000719 pyrrolidinyl group Chemical group 0.000 description 1
- 125000001422 pyrrolinyl group Chemical group 0.000 description 1
- 125000000168 pyrrolyl group Chemical group 0.000 description 1
- 229940107700 pyruvic acid Drugs 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
- 239000002516 radical scavenger Substances 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 102000016914 ras Proteins Human genes 0.000 description 1
- 238000003259 recombinant expression Methods 0.000 description 1
- 238000001953 recrystallisation Methods 0.000 description 1
- 108010054624 red fluorescent protein Proteins 0.000 description 1
- 238000005932 reductive alkylation reaction Methods 0.000 description 1
- 238000005057 refrigeration Methods 0.000 description 1
- 230000027425 release of sequestered calcium ion into cytosol Effects 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 102000029752 retinol binding Human genes 0.000 description 1
- 108091000053 retinol binding Proteins 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 125000000548 ribosyl group Chemical group C1([C@H](O)[C@H](O)[C@H](O1)CO)* 0.000 description 1
- 229940081974 saccharin Drugs 0.000 description 1
- 235000019204 saccharin Nutrition 0.000 description 1
- 239000000901 saccharin and its Na,K and Ca salt Substances 0.000 description 1
- 229960004889 salicylic acid Drugs 0.000 description 1
- 201000007416 salivary gland adenoid cystic carcinoma Diseases 0.000 description 1
- 208000014212 sarcomatoid carcinoma Diseases 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 206010039667 schwannoma Diseases 0.000 description 1
- 238000003345 scintillation counting Methods 0.000 description 1
- 239000002795 scorpion venom Substances 0.000 description 1
- 201000008407 sebaceous adenocarcinoma Diseases 0.000 description 1
- 150000003335 secondary amines Chemical class 0.000 description 1
- 229940055619 selenocysteine Drugs 0.000 description 1
- ZKZBPNGNEQAJSX-UHFFFAOYSA-N selenocysteine Natural products [SeH]CC(N)C(O)=O ZKZBPNGNEQAJSX-UHFFFAOYSA-N 0.000 description 1
- 235000016491 selenocysteine Nutrition 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 210000000717 sertoli cell Anatomy 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 230000005582 sexual transmission Effects 0.000 description 1
- 239000004208 shellac Substances 0.000 description 1
- 229940113147 shellac Drugs 0.000 description 1
- ZLGIYFNHBLSMPS-ATJNOEHPSA-N shellac Chemical compound OCCCCCC(O)C(O)CCCCCCCC(O)=O.C1C23[C@H](C(O)=O)CCC2[C@](C)(CO)[C@@H]1C(C(O)=O)=C[C@@H]3O ZLGIYFNHBLSMPS-ATJNOEHPSA-N 0.000 description 1
- 235000013874 shellac Nutrition 0.000 description 1
- 201000008123 signet ring cell adenocarcinoma Diseases 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 239000004332 silver Substances 0.000 description 1
- 108091069025 single-strand RNA Proteins 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 229940074386 skatole Drugs 0.000 description 1
- 201000002078 skin pilomatrix carcinoma Diseases 0.000 description 1
- 208000000649 small cell carcinoma Diseases 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 1
- 229940079832 sodium starch glycolate Drugs 0.000 description 1
- 239000008109 sodium starch glycolate Substances 0.000 description 1
- 229920003109 sodium starch glycolate Polymers 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 239000006104 solid solution Substances 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 235000010356 sorbitol Nutrition 0.000 description 1
- 206010041823 squamous cell carcinoma Diseases 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 229940032147 starch Drugs 0.000 description 1
- 230000001954 sterilising effect Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 208000028210 stromal sarcoma Diseases 0.000 description 1
- 238000000547 structure data Methods 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 229940124530 sulfonamide Drugs 0.000 description 1
- 150000003456 sulfonamides Chemical class 0.000 description 1
- BDHFUVZGWQCTTF-UHFFFAOYSA-N sulfonic acid Chemical compound OS(=O)=O BDHFUVZGWQCTTF-UHFFFAOYSA-N 0.000 description 1
- 208000030457 superficial spreading melanoma Diseases 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 206010042863 synovial sarcoma Diseases 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 239000011975 tartaric acid Substances 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 229940034880 tencon Drugs 0.000 description 1
- 125000001981 tert-butyldimethylsilyl group Chemical group [H]C([H])([H])[Si]([H])(C([H])([H])[H])[*]C(C([H])([H])[H])(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 238000010998 test method Methods 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 229960000814 tetanus toxoid Drugs 0.000 description 1
- 108010013645 tetranectin Proteins 0.000 description 1
- 150000003536 tetrazoles Chemical class 0.000 description 1
- 208000001644 thecoma Diseases 0.000 description 1
- 229940021747 therapeutic vaccine Drugs 0.000 description 1
- 125000001113 thiadiazolyl group Chemical group 0.000 description 1
- 125000000335 thiazolyl group Chemical group 0.000 description 1
- 125000001544 thienyl group Chemical group 0.000 description 1
- 238000004809 thin layer chromatography Methods 0.000 description 1
- 125000003396 thiol group Chemical group [H]S* 0.000 description 1
- 125000005505 thiomorpholino group Chemical group 0.000 description 1
- 125000004568 thiomorpholinyl group Chemical group 0.000 description 1
- 210000001541 thymus gland Anatomy 0.000 description 1
- 208000030901 thyroid gland follicular carcinoma Diseases 0.000 description 1
- 208000015191 thyroid gland papillary and follicular carcinoma Diseases 0.000 description 1
- 239000003970 toll like receptor agonist Substances 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 231100000331 toxic Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 208000029335 trabecular adenocarcinoma Diseases 0.000 description 1
- 230000005026 transcription initiation Effects 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 238000003151 transfection method Methods 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000014723 transformation of host cell by virus Effects 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 206010044412 transitional cell carcinoma Diseases 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 102000035160 transmembrane proteins Human genes 0.000 description 1
- 108091005703 transmembrane proteins Proteins 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- 125000001425 triazolyl group Chemical group 0.000 description 1
- 229940035722 triiodothyronine Drugs 0.000 description 1
- 239000013638 trimer Substances 0.000 description 1
- 238000001665 trituration Methods 0.000 description 1
- 239000002753 trypsin inhibitor Substances 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 229960004441 tyrosine Drugs 0.000 description 1
- 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 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 238000010200 validation analysis Methods 0.000 description 1
- 230000007486 viral budding Effects 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- 230000009447 viral pathogenesis Effects 0.000 description 1
- 230000007919 viral pathogenicity Effects 0.000 description 1
- 230000008957 viral persistence Effects 0.000 description 1
- 239000000277 virosome Substances 0.000 description 1
- 230000029302 virus maturation Effects 0.000 description 1
- 102100032078 von Hippel-Lindau-like protein Human genes 0.000 description 1
- 235000012431 wafers Nutrition 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 230000003313 weakening effect Effects 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 239000009637 wintergreen oil Substances 0.000 description 1
- 238000002424 x-ray crystallography Methods 0.000 description 1
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
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70539—MHC-molecules, e.g. HLA-molecules
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
-
- 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
- C07K14/44—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from protozoa
- C07K14/445—Plasmodium
-
- 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
-
- 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
-
- 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
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/08—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
- C07K16/10—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
- C07K16/1036—Retroviridae, e.g. leukemia viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2833—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
-
- 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
- C12N7/00—Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
-
- 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
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/569—Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
- G01N33/56983—Viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/53—DNA (RNA) vaccination
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/545—Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/57—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
- A61K2039/572—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/58—Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
- A61K2039/585—Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/14011—Deltaretrovirus, e.g. bovine leukeamia virus
- C12N2740/14034—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- HTLV-1 human T-lymphotropic virus type-1
- BACKGROUND Human T-lymphotropic virus type-1 (HTLV-1) is a complex retrovirus which belongs to the Deltaretrovirus genus, infecting an estimated 5-10 million individuals worldwide.
- HTLV-1 has been identified as the most pathogenic and is the primary etiological agent of a malignant lymphoproliferation called adult T-cell leukemia/lymphoma (ATL), as well as an array of neurological pathologies known as HTLV-1-myelopathy/tropical spastic paraparesis (HAM/TSP).
- ATL adult T-cell leukemia/lymphoma
- HAM/TSP HTLV-1-myelopathy/tropical spastic paraparesis
- HTLV-1 is endemic in areas of Southeastern Japan, sub-Saharan Africa, South America, and the Caribbean, with foci in the Middle-East and Australo- Melanosia.
- HTLV-1 is predominately found in CD4 + T lymphocytes and can be spread by, e.g., sexual transmission, contaminated blood products, and mother-to-child transmission (parental/vertical). Further, HTLV-1 is a latent virus, and as such is unable to Attorney Docket No.250298.000682 be cleared from the host’s immune system, where 4-5% of HTLV-1 carriers develop ATL in their life-time and another 0.25-4% develop HAM/TSP. Despite scientific advances toward understanding the pathogenic progression of HTLV-1 infection, the prognosis of ATL, as well as the quality of life for those suffering from HAM/TSP, remains poor. There is no vaccine to prevent HTLV-1 infection or HTLV-1-associated disease.
- identifying HTLV-1-associated epitopes that can be amenable to targeting such as by therapeutic vaccines, immunotherapy, or other therapeutics that target these HTLV-1- associated antigens would be useful to combat HTLV-1-mediated infections and diseases.
- an isolated peptide comprising an amino acid sequence that is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof, wherein the isolated peptide is 5-20 amino acids in length.
- the isolated peptide comprises an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146.
- the isolated peptide consists essentially of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146.
- the isolated peptide consists of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146.
- an isolated peptide comprising two or more amino acid sequences selected from any one of SEQ ID NO: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof.
- the isolated peptide comprises one or more reverse peptide bonds, one or more non-peptide bonds, one or more D-isomers of amino acids, one or more chemical modifications, or any combination thereof.
- the isolated peptide is produced by expression in a heterologous host cell.
- the isolated peptide is produced synthetically.
- the isolated peptide, or pharmaceutically acceptable salt thereof, or fragment or derivative thereof induces a Human T-Lymphotropic Virus Type-1 (HTLV-1)-specific immune response in a subject when presented in a complex with a major histocompatibility complex (MHC) molecule on the surface of an antigen presenting cell (APC).
- MHC major histocompatibility complex
- APC antigen presenting cell
- a fusion protein comprising one or more isolated peptides described herein fused to one or more heterologous molecules.
- the one or more heterologous molecules enhance a peptide- specific immune response in a subject.
- the one or more heterologous molecules mediate peptide delivery to a specific site within a subject.
- the one or more heterologous molecules are a MHC molecule, or a fragment or derivative thereof.
- a conjugate comprising one or more isolated peptides described herein conjugated to one or more heterologous molecules.
- the one or more heterologous molecules enhance a peptide- specific immune response in a subject.
- the one or more heterologous molecules mediate peptide delivery to a specific site within a subject.
- the one or more heterologous molecules are an MHC molecule, or a fragment or derivative thereof.
- the one or more peptides are conjugated to a particle.
- an oligomeric complex comprising two or more isolated peptides described herein.
- a non-covalent complex comprising an isolated peptide described herein and an MHC molecule, or a fragment or derivative thereof.
- the MHC molecule, or the fragment thereof is a class I MHC molecule.
- Attorney Docket No.250298.000682 [0027]
- the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule.
- HLA human leukocyte antigen
- the MHC molecule, or the fragment thereof is a class II MHC molecule.
- the class II MHC molecule is a class II HLA molecule.
- a fusion protein comprising an isolated peptide described herein and an MHC molecule, or a fragment or derivative thereof.
- the MHC molecule, or the fragment thereof is a class I MHC molecule.
- the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule.
- HLA human leukocyte antigen
- the MHC molecule, or the fragment thereof is a class II MHC molecule.
- the class II MHC molecule is a class II HLA molecule.
- a conjugate comprising an isolated peptide described herein and a MHC molecule, or a fragment or derivative thereof.
- the MHC molecule, or the fragment thereof is a class I MHC molecule.
- the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule.
- HLA human leukocyte antigen
- the MHC molecule, or the fragment thereof is a class II MHC molecule.
- the class II MHC molecule is a class II HLA molecule.
- a pharmaceutical composition comprising (i) one or more isolated peptides described herein, one or more fusion proteins described herein, one or more conjugates described herein, one or more oligomeric complexes described herein, or one or more non-covalent complexes described herein, or any combination thereof; and (ii) a pharmaceutically acceptable carrier or excipient.
- the pharmaceutical composition can further comprise an adjuvant.
- Attorney Docket No.250298.000682 [0042]
- an isolated molecule that binds an isolated peptide described herein, a fusion protein described herein, a conjugate described herein, an oligomeric complex described herein, or a non-covalent complex described herein.
- the molecule is an antibody or an antigen-binding fragment thereof.
- the antibody is a bispecific antibody.
- the molecule is an alternative scaffold.
- the molecule is a chimeric antigen receptor (CAR).
- the molecule is a T cell receptor (TCR).
- TCR T cell receptor
- an isolated cell comprising a CAR described herein.
- the isolated cell is an immune cell.
- the immune cell is a T cell, an NK cell, or a macrophage.
- an isolated cell comprising a TCR described herein.
- the isolated cell is an immune cell.
- the immune cell is a T cell, an NK cell, or a macrophage.
- a pharmaceutical composition comprising (i) an isolated molecule described herein, or an isolated cell described herein; and (ii) a pharmaceutically acceptable carrier or excipient.
- an isolated polynucleotide comprising a nucleotide sequence encoding one or more isolated peptides described herein or a fusion protein described herein.
- the nucleotide sequence can be operably linked to a promoter.
- the isolated polynucleotide comprises DNA.
- the isolated polynucleotide comprises RNA.
- the RNA is mRNA.
- the RNA is self-replicating RNA.
- a vector comprising an isolated polynucleotide described herein. Attorney Docket No.250298.000682 [0062]
- the vector is an expression vector.
- the vector is a viral vector.
- a host cell comprising an isolated polynucleotide described herein or a vector described herein. [0065] In some embodiments, the host cell is a prokaryotic cell.
- the host cell is a eukaryotic cell.
- the host cell is an APC.
- a pharmaceutical composition comprising (i) an isolated polynucleotide described herein, or a vector described herein; and (ii) a pharmaceutically acceptable carrier or excipient.
- the pharmaceutically acceptable carrier is a lipid nanoparticle carrier.
- a method of inducing an immune response against HTLV-1 infection in a subject in need thereof comprising administering to the subject a therapeutically effective amount of: a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) a oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein.
- a method of inducing an immune response against HTLV-1 infection in a subject in need thereof comprising administering to the subject a therapeutically effective amount of one or more isolated peptides described herein.
- a method of inducing an immune response against a HTLV-1infection in a subject in need thereof comprising administering to the Attorney Docket No.250298.000682 subject an activated T cell that is produced by contacting a T cell with an APC that presents an isolated peptide described herein in complex with an MHC molecule.
- a method of treating a HTLV-1-induced disease or disorder in a subject in need thereof comprising administering to the subject an effective amount of: a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) an oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein.
- a method of preventing or reducing the likelihood of a HTLV-1-induced disease or disorder in a subject in need thereof comprising administering to the subject an effective amount of: a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) an oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein.
- a method of treating a HTLV-1-induced disease or disorder in a subject in need thereof comprising administering to the subject an effective amount of one or more isolated peptides described herein.
- Attorney Docket No.250298.000682 [0076]
- a method of preventing or reducing the likelihood of a HTLV-1-induced disease or disorder in a subject in need thereof the method comprising administering to the subject an effective amount of one or more isolated peptides described herein.
- the HTLV-1-induced disease or disorder is adult T-cell leukemia/lymphoma (ATL) or HTLV-1-myelopathy/tropical spastic paraparesis (HAM/TSP).
- ATL adult T-cell leukemia/lymphoma
- HAM/TSP HTLV-1-myelopathy/tropical spastic paraparesis
- kits comprising: (i) a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) an oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein; and (ii) packaging and/or instructions for use for the same.
- Figures 1A-1E illustrate affinity purification mass spectrometry for HTLV- transformed T-lymphoblast cell lines.
- An example of an anti-HLA-I W6/32 affinity purification workflow and subsequent peptide identifications by mass spectrometry is illustrated in Figure 1A.
- a description of peptide lengths and counts detected by mass spectrometry analysis for cell lines C8166 ( Figure 1B), MT-2 ( Figure 1C), MT-4 ( Figure 1D), and C5/MJ (Figure 1E) is shown in Figures 1B-1E, respectively.
- Figure 2 shows that eleven (11) HTLV peptides were detected from the C8166 cell line with good reproducibility, six (6) of which were predicted HLA binders to known Attorney Docket No.250298.000682 C8166 HLA alleles by NetMHCpan 4.0 (see, e.g., Table 3).
- Figure discloses SEQ ID NOs: 85, 63, 74, 83, 78, 81, 51, 24, 29, 70 and 31, respectively, in order of columns.
- Figure 3 shows that thirty-two (32) HTLV peptides were detected from the MT-2 cell line, twenty-one (21) of which were found in all three replicates, and a majority of which were predicted binders to known MT-2 alleles by NetMHCpan 4.0 (see, e.g., Table 4).
- Figure discloses SEQ ID NOs: 55, 46, 86, 80, 21 and 5, respectively, in order of appearance and SEQ ID NOs: 84, 89, 88, 87, 72, 11, 9, 52, 67, 20, 44, 51, 57, 4, 45, 17, 60, 62, 54, 35, 77, 49, 76, 71, 82, and 73, respectively, in order of columns.
- Figure 5 shows that HLA-I immunopeptidomics from C5/MJ cells with interferon (IFN) and without IFN (no IFN) treatment yielded nine (9) overlapping HTLV peptides, all of which were predicted binders to C5/MJ HLA alleles by NetMHCpan 4.0 (see, e.g., Table 6).
- Figures 6A-6E show that sixteen (16) and twenty-one (21) HTLV peptides were detected from no IFN- and IFN-treated ED-41214(-) primary cells by mass spectrometry, with good reproducibility among biological replicates (see, e.g., Figure 6B and Figure 6D, respectively). Fifteen (15) HTLV peptides were shared between no IFN- and IFN-treated ED-41214(-) cells ( Figure 6E). Only a few HTLV peptides were predicted binders to the common alleles by NetMHCpan 4.0 (see, e.g., Table 7).
- Figure 6A and Figure 6C Descriptions of peptide lengths and counts detected by mass spectrometry analysis for no IFN- and IFN-treated ED- 41214(-) cells are shown in Figure 6A and Figure 6C, respectively.
- Figure discloses SEQ ID NOs: 32, 7, 3, 25, 30, 52, 33, 61, 39, 19, 6, 28, 40, 23, 10, 43, 48, 32, 3, 7, 30, 34, 13, 52, 12, 33, 61, 39, 6, 19, 28, 40, 23, 10, 43, 16, and 22, respectively, order of appearance.
- Figures 7A-7D illustrate peptide distributions were obtained for all HTLV primary samples tested ((see, e.g., Figure 6A for ED41214(-), no IFN 1; ED40515(-), no IFN 1 Attorney Docket No.250298.000682 (Figure 7A); ED40515(+), no IFN 1 ( Figure 7B); ATL43T(+), no IFN 1 ( Figure 7C); and ATL43Tb(-) IFN 1 ( Figure 7E)); however, HTLV peptides were detected only for ED- 41214(-) (see, e.g., Figure 6A) (combined search against human UniProt + HTLV sequence).
- Figures 8A-8D show examples of peptide distributions from HLA-I peptidome analysis of ATL patient samples by Thermo Orbitrap ( Figures 8A-8B) and Bruker timsTOF SCP ( Figures 8C-8D) mass spectrometers.
- Figures 9A-9B show an example of a workflow used to reconstruct patient-specific HTLV genomes from RNA sequencing. RNA sequencing and RNA contig reconstruction steps are illustrated in Figure 9A.
- HTLV genome reconstruction including BLAST analysis against the virus reference genome, viral genome reconstruction, and coding sequence extraction steps, are illustrated in Figure 9B.
- Figure 10 shows RNA sequencing read coverage of patient-specific reconstructed genomes.
- the present disclosure provides, among other things, isolated peptides derived from human T-lymphotropic virus type-1 (HTLV-1), and fragments or derivatives thereof.
- Various peptide-based molecules including complexes e.g., peptide-MHC (pMHC) complexes
- fusion proteins e.g., fusion proteins, and conjugates comprising the peptides are also provided.
- Binding moieties e.g., antibodies, alternative scaffolds, T-cell receptors (TCRs) or chimeric antigen receptors (CARs) that bind to the peptides or peptide-based molecules are also provided.
- the compositions of the present disclosure can be used to induce an immune response against HTLV infection (e.g., HTLV-1, HTLV-2, HTLV-3, and/or HTLV-4 infection) and/or for treating, preventing, and/or reducing the likelihood of an HTLV-induced disease or disorder (e.g., an HTLV-1-, HTLV-2-, HTLV- 3-, and/or HTLV-4-induced disease or disorder).
- HTLV infection e.g., HTLV-1, HTLV-2, HTLV-3, and/or HTLV-4 infection
- an HTLV-induced disease or disorder e.g., an HTLV-1-, HTLV-2-, HTLV- 3-, and/or HTLV
- the present disclosure also provides methods for identifying an immunogenic virus-derived peptide.
- Lymphocytes such as T cells, play important roles in adaptive anti-infection, antitumor, autoimmune, and transplant rejection responses.
- a T cell mediated Attorney Docket No.250298.000682 immune response involves close contact, e.g., an immunological synapse, between a T cell and an antigen presenting cell (APC).
- APC antigen presenting cell
- TCR T-cell receptor
- MHC major histocompatibility complex
- CD28 CD28
- T cell receptors are heterodimeric structures composed of two types of chains (an ⁇ (alpha) and ⁇ (beta) chain, or a ⁇ (gamma) and ⁇ (delta) chain).
- the ⁇ chain is encoded by the nucleic acid sequence located within the ⁇ locus (on human or mouse chromosome 14), which also encompasses the entire ⁇ locus that encodes the ⁇ chain, and the ⁇ chain is encoded by the nucleic acid sequence located within the ⁇ locus (on mouse chromosome 6 or human chromosome 7).
- the majority of T cells have an ⁇ ⁇ TCR, while a minority of T cells bear a ⁇ ⁇ TCR.
- T cell receptor ⁇ and ⁇ polypeptides are linked to each other via a disulfide bond.
- Each of the two polypeptides that make up the TCR contains an extracellular domain comprising constant and variable regions, a transmembrane domain, and a cytoplasmic tail (the transmembrane domain and the cytoplasmic tail also being a part of the constant region).
- the variable region of each TCR comprises a unique and characteristic structure, i.e., an idiotope or idiotype, that determines the specificity of the TCR.
- a TCR will bind to a pMHC complex only if the TCR comprises an idiotype that recognizes the peptide being presented in the context of MHC, e.g., the unique conformation of a particular pMHC complex.
- Immunotherapeutic approaches to treating disease work to regulate T cell activity in vivo, e.g., to enhance anti-infection and anti-tumor responses, or, for example, to downregulate autoimmune and transplant rejection responses.
- immunotherapies can target signaling by the TCR complex by binding CD3 and/or the pairing of costimulatory molecules.
- therapies that take advantage of the uniquely specific interaction between a TCR and pMHC complex may provide the ability to specifically modulate the activity of specific T cells in vivo, and provide treatments based on T cell modulation.
- the presentation of virus-derived peptides by MHC molecules on the surface of an infected cell and the recognition of these pMHC complexes by, and subsequent activation of, CD8+ cytotoxic T cells provides an important mechanism for immunity-based protection against viruses.
- Cells infected with HTLV-1 including those that developed into cancer cells, can express various HTLV-1-associated antigens.
- Peptides derived from these antigens may be displayed on the cell surface in complex with MHC molecules. Detection of an MHC-presented HTLV-1-derived peptide by a T cell bearing the corresponding TCR, leads to targeted killing of the infected cell. However, because of the selection processes which occur during T cell maturation in the thymus, there is often a scarcity of T cells in the circulating repertoire, which recognize HTLV-1-derived peptides with a sufficiently high level of affinity. As a result, infected cells often escape elimination by the immune system.
- HTLV-1-derived peptides presented on infected cells can allow for the development of immunotherapeutic reagents designed to specifically target and destroy HTLV-1-infected cells (e.g., HTLV-1- induced cancer cells).
- immunotherapeutic reagents designed to specifically target and destroy HTLV-1-infected cells (e.g., HTLV-1- induced cancer cells).
- Such reagents may be moieties that bind to the HTLV-1-derived peptide and/or pMHC complexes and the reagents, e.g., moieties that can function by inducing a T cell response.
- such reagents may be based on antibodies, TCRs, and/or CARs.
- the present disclosure is based on a proteogenomic approach that detects MHC-associated HTLV-1 peptides from HTLV-1 infected cells.
- the repertoire of HLA-I- HTLV-1 peptides that are expressed in HTLV-1-transformed cells and ATL primary cells characterized herein can provide an accurate representation of HTLV-1 epitopes in a population.
- HLA-restricted viral peptides as potential targets may be leveraged for delivering immunotherapeutics (such as antibodies (e.g., bispecific antibodies), engineered TCR- or CAR- based cellular therapies) to infected tissues.
- genomic variability between strains of a virus in combination with differences in patient HLA alleles can be a challenge Attorney Docket No.250298.000682 in developing therapeutics against these peptide targets.
- the present disclosure provides a proteogenomics approach for generating patient- specific databases that allow for the comprehensive identification of viral peptides, e.g., HTLV-1-derived peptides, based on the viral transcriptomes sequenced from individual patient samples.
- the HTLV-1-HLA-associated peptides disclosed herein may be used in the development of immunotherapeutics (such as antibodies (e.g., bispecific antibodies), engineered TCR- or CAR- based cellular therapies) for the treatment of HTLV-1-related diseases or disorders including T-cell leukemia/lymphoma (ATL), HTLV-1- myelopathy/tropical spastic paraparesis (HAM/TSP), and hypersensitivity reactions, e.g., arthritis, uveitis, and HTLV-1-associated infective dermatitis (IDH).
- immunotherapeutics such as antibodies (e.g., bispecific antibodies), engineered TCR- or CAR- based cellular therapies
- ATL T-cell leukemia/lymphoma
- HAM/TSP HTLV-1- myelopathy/tropical spastic paraparesis
- IDH HTLV-1-associated infective dermatitis
- the proteogenomic discovery platform described herein provides
- Such a range can be within an order of magnitude, preferably within 50%, more preferably within 20%, still more preferably within 10%, and even more preferably within 5% of a given value or range.
- the allowable variation encompassed by the term “about” or “approximately” depends on the particular system under study, and can be readily appreciated by one of ordinary skill in the art.
- the term “antigen” encompasses any agent (e.g., protein, peptide, polysaccharide, glycoprotein, glycolipid, nucleotide, portions thereof, or combinations thereof) that, when introduced into an immunocompetent host (directly or upon expression as in, e.g., DNA or RNA vaccines) is recognized by the immune system of the host and is Attorney Docket No.250298.000682 capable of eliciting an immune response by the host.
- the T-cell receptor (TCR) recognizes a peptide presented in the context of a major histocompatibility complex (MHC) as part of an immunological synapse.
- MHC major histocompatibility complex
- the peptide-MHC (pMHC) complex is recognized by TCR, with the peptide (antigenic determinant) and the TCR idiotype providing the specificity of the interaction.
- the term “antigen” encompasses peptides presented in the context of MHCs, e.g., pMHC complexes.
- the peptide displayed on MHC may also be referred to as an “epitope” or an “antigenic determinant”.
- peptide encompass not only those presented naturally by antigen- presenting cells (APCs), but may be any desired peptide so long as it is recognized by an immune cell, e.g., when presented appropriately to the cells of an immune system.
- a single antigen such as an antigenic polypeptide
- a single antigen may have more than one epitope.
- Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of structural epitopes and are defined as those residues that directly contribute to the affinity of the interaction between an MHC molecule and the antigen.
- Epitopes may also be conformational, that is, composed of non-linear amino acids.
- epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
- MHC major histocompatibility complex
- HLA human leukocyte antigen
- MHC class I molecule comprising MHC class I ⁇ (heavy) chain and ⁇ 2 microglobulin
- MHC class II molecule comprising MHC class II ⁇ chain and MHC class II ⁇ chain
- individual chains of MHC molecules e.g., MHC class I ⁇ (heavy) chain, MHC class II ⁇ chain, and MHC class II ⁇ chain
- individual subunits of such chains of MHC molecules e.g., ⁇ 1, ⁇ 2, and/or ⁇ 3 subunits of MHC class I ⁇ chain, ⁇ 1- ⁇ 2 subunits of MHC class II ⁇ chain, ⁇ 1- ⁇ 2 subunits Attorney Docket No.250298.000682 of MHC class II ⁇ chain
- portions e.g.,
- An MHC class I molecule comprises a peptide binding groove formed by the ⁇ 1 and ⁇ 2 domains of the heavy a chain that can stow a peptide of around 8-10 amino acids.
- both classes of MHC bind a core of about 9 amino acids (e.g., 5 to 17 amino acids) within peptides
- the open-ended nature of MHC class II peptide binding groove (the ⁇ 1 domain of a class II MHC ⁇ polypeptide in association with the ⁇ 1 domain of a class II MHC ⁇ polypeptide) allows for a wider range of peptide lengths.
- Peptides binding MHC class II usually vary between 13 and 17 amino acids in length, though shorter or longer lengths are not uncommon.
- the peptide-MHC complex described herein may be a peptide-MHC complex from a non-human animal.
- the peptide-MHC complex described herein may include an peptide-HLA complex, i.e., a peptide-MHC complex from a human. Conventional identifications of particular MHC variants are used herein.
- HLA-A11 refers to a human leucocyte antigen from the A gene group (hence a class I type MHC) gene position (known as a gene locus) number 11; gene HLA-DR11, refers to a human leucocyte antigen coded by a gene from the DR region (hence a class II type MHC) locus number 11.
- MHC-peptide complex examples include (i) an MHC molecule, e.g., a human and/or non- human animal MHC molecule, or portion thereof (e.g., the peptide-binding groove thereof, and e.g., the extracellular portion thereof), and (ii) an antigenic peptide (e.g., a HTLV-1- derived peptide), where the MHC molecule and the antigenic peptide are complexed in such a manner that the pMHC complex can specifically bind a T-cell receptor.
- an MHC molecule e.g., a human and/or non- human animal MHC molecule, or portion thereof (e.g., the peptide-binding groove thereof, and e.g., the extracellular portion thereof)
- an antigenic peptide e.g., a HTLV-1- derived peptide
- HLA-peptide complex encompasses cell surface expressed pMHC complexes and soluble pMHC complexes.
- HLA-peptide complex refers to an MHC-peptide complex wherein the MHC molecule is a Human Leukocyte Antigen (HLA) molecule.
- T cell or “T lymphocyte” is used herein in its broadest sense to refer to all types of immune cells expressing CD3, including, but not limited to, T-helper cells (CD4+ cells), cytotoxic T-cells (CD8+ cells), tumor infiltrating cytotoxic T cells (TIL; CD8+ T cell), CD4+CD8+ T cells, T-regulatory cells (Treg), and NK-T cells.
- T cells can include thymocytes, na ⁇ ve T cells, memory T cells, immature T cells, mature T cells, resting T cells, or activated T cells.
- T cells may also include “gamma-delta T cells ( ⁇ T cells),” which refer to a specialized population that to a small subset of T cells possessing a distinct TCR on their surface, and unlike the majority of T cells in which the TCR is composed of two glycoprotein chains designated ⁇ - and ⁇ -TCR chains, the TCR in ⁇ T cells is made up of a ⁇ -chain and a ⁇ -chain.
- ⁇ T cells gamma-delta T cells
- the term “antigen presenting cell” or “APC” refers to any cell that presents on the surface of the cell an antigen in association with a major histocompatibility complex molecule, either MHC class I or MHC class II molecule, or both.
- the terms include monoclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked Fvs (sdFv), intrabodies, minibodies, diabodies and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antigen-specific TCR), and epitope-binding fragments of any of the above.
- antibody and “antibodies” also refer to covalent diabodies such as those disclosed in U.S. Pat. Appl. Pub.
- a peptide binding moiety e.g., an antibody, an alternative scaffold, a CAR, or a TCR
- a peptide-based molecule such as a complex (e.g., a pMHC complex), fusion protein, or conjugate comprising the described peptide
- the peptide binding moiety forms a stable intermolecular non-covalent bonds with the HTLV-1- derived peptide or peptide-based molecule (such as a complex (e.g., a pMHC complex), fusion protein, or conjugate comprising the described peptide).
- polynucleotide and nucleic acid include polymeric forms of nucleotides of any length, including ribonucleotides (RNA), deoxyribonucleotides (DNA), or analogs or modified versions thereof. They include single-, double-, and multi-stranded DNA or RNA, genomic DNA, complementary DNA (cDNA), DNA-RNA hybrids, and polymers comprising purine bases, pyrimidine bases, or other natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases.
- RNA ribonucleotides
- DNA deoxyribonucleotides
- analogs or modified versions thereof include single-, double-, and multi-stranded DNA or RNA, genomic DNA, complementary DNA (cDNA), DNA-RNA hybrids, and polymers comprising purine bases, pyrimidine bases, or other natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases.
- operably linked refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
- a control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
- “Operably linked” sequences include both expression control sequences that are contiguous with a gene of interest and expression control sequences that act in trans or at a distance to control a gene of interest (or sequence of interest).
- expression control sequence includes polynucleotide sequences, which are necessary to affect the expression and processing of coding sequences to which they are ligated.
- “Expression control sequences” include: appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance polypeptide stability; and when desired, sequences that enhance polypeptide secretion.
- the nature of such control sequences differs depending upon the host organism. For example, in prokaryotes, such control sequences generally include promoter, ribosomal binding site and transcription termination sequence, while in eukaryotes typically such control sequences include promoters and transcription termination sequence.
- control sequences is intended to include components whose presence is essential for expression and processing and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
- Attorney Docket No.250298.000682 [00114]
- isolated refers to a homogenous population of molecules (such as polynucleotides or polypeptides) which have been substantially separated and/or purified away from other components of the system the molecules are produced in, such as a recombinant cell, as well as a protein that has been subjected to at least one purification or isolation step.
- isolated refers to a molecule that is substantially free of other cellular material and/or chemicals and encompasses molecules that are isolated to a higher purity, such as to 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% purity.
- derivative refers to a peptide, polypeptide, or polynucleotide, or a variant or analog thereof, comprising one or more mutations and/or chemical modifications as compared to a reference peptide, polypeptide or polynucleotide.
- the terms “treat” or “treatment” of a state, disorder, disease, or condition include: (1) preventing, delaying, or reducing the incidence and/or likelihood of the appearance of at least one clinical or sub-clinical symptom of the state, disorder, disease, or condition developing in a subject that may be afflicted with or predisposed to the state, disorder, disease, or condition, but does not yet experience or display clinical or subclinical symptoms of the state, disorder, disease, or condition; or (2) inhibiting the state, disorder, disease, or condition, i.e., arresting, reducing or delaying the development of the disease or a relapse thereof or at least one clinical or sub-clinical symptom thereof; or (3) relieving the state, disorder, disease, or condition, i.e., causing regression of the state,
- the benefit to a subject to be treated is either statistically significant or at least perceptible to the patient or to the physician.
- An “individual” or “subject” or “animal” refers to humans, veterinary animals (e.g., cats, dogs, cows, horses, sheep, pigs, etc.) and experimental animal models of diseases (e.g., mice, rats). In a preferred embodiment, the subject is a human.
- the term “effective” applied to dose or amount refers to that quantity of a compound or pharmaceutical composition that is sufficient to result in a desired activity Attorney Docket No.250298.000682 upon administration to a subject in need thereof.
- the effective amount of the combination may or may not include amounts of each ingredient that would have been effective if administered individually.
- the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition being treated, the particular drug or drugs employed, the mode of administration, and the like.
- pharmaceutically acceptable refers to molecular entities and other ingredients of such compositions that are physiologically tolerable and do not typically produce untoward reactions when administered to a mammal (e.g., a human).
- the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in mammals, and more particularly in humans.
- the term “administration” and the like refers to and includes the administration of a composition to a subject or system (e.g., to a cell, organ, tissue, organism, or relevant component or set of components thereof). The skilled artisan will appreciate that route of administration may vary depending, for example, on the subject or system to which the composition is being administered, the nature of the composition, the purpose of the administration, etc.
- administration to an animal subject may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal and/or vitreal.
- administration may involve intermittent dosing.
- administration may involve continuous dosing (e.g., perfusion) for at least a selected period of time.
- conventional molecular biology, microbiology, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual, Second Edition. Attorney Docket No.250298.000682 Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press, 1989 (herein “Sambrook et al., 1989”); DNA Cloning: A Practical Approach, Volumes I and II (D.N. Glover ed.
- the present disclosure provides isolated peptides comprising an amino acid sequence derived from human T-lymphotropic virus type-1 (HTLV-1).
- the HTLV genome follows a canonical structure of replication competent retroviruses comprising GAG, POL, and ENV domains flanked by two long terminal repeat (LTR) domains on either end of the provirus.
- LTR long terminal repeat
- HTLV-1 has a monopartite, linear, dimeric single strand RNA (+) genome of 8.5 kb, comprising a 5’-cap and a 3’ poly- A tail.
- the two LTRs are about 600 nucleotide residues in length and are located at the 5’ and 3’ ends of the genome.
- the LTRs comprise U3, R, and U5 regions, which together can Attorney Docket No.250298.000682 function as a transcription unit.
- the 5’ end of the genome further comprises a primer binding site and the 3’ end further comprises a polypurin tract.
- the integrated virus utilizes promoter elements in the 5’ LTR to trigger and drive transcription, resulting in an unspliced full-length mRNA that can serve, e.g., as genomic RNA for packaging into virions.
- HTLV-1 While similar to other retroviruses, the genomic RNA of HTLV-1 encodes the structural and enzymatic proteins Gag (an internal group specific antigen), Pol (a polymerase, reverse transcriptase and integrase enzymes required for viral replication and maturation), and Env (envelope glycoprotein), HTLV-1 differs from other retroviruses due to a pX region located toward the 3’ end between the ENV and 3’ LTR.
- the pX region encodes several alternatively spliced regulatory genes including, without limitation, Tax and HTLV-I basic leucine zipper factor (HBZ), both of which can be implicated in viral pathogenesis.
- Tax is a 40 kDa protein which mainly localizes to the nucleus but can also be within the cytoplasm of infected cells. Tax interacts with a variety of host proteins and plays a key role in transactivating proviral transcription from the 5’ LTR. Tax can functionally inactivate p53 and target pRB for degradation, allowing for survival of infected cells. Tax can also dysregulate several signaling pathways such as but not limited to activator protein 1 (AP-1), nuclear factor kappa B (NF- ⁇ B), serum responsive factor (SRF), and cyclic AMP response element-binding protein (CREB) pathways.
- AP-1 activator protein 1
- NF- ⁇ B nuclear factor kappa B
- SRF serum responsive factor
- CREB cyclic AMP response element-binding protein
- HBZ is a nuclear protein but can also be localized to the cytoplasm. HBZ comprises three domains: an activation domain, a central domain, and a basic leucine zipper domain. HBZ antagonizes several Tax-mediated functions and participates in viral persistence and immune evasion.
- the activation domain of HBZ comprises two LXXLL - like motifs (where L is leucine and X is any amino acid) which bind can bind to a kinase- inducible domain interacting (KIX) domain of p300-CBP transcription coactivators.
- HTLV-1 After infection of a host, HTLV-1 infects cells via its glycoprotein gp62. HTLV-1 can then achieve latency by integrating into the host genome and increasing proviral load by proliferation of infected cells.
- the HTLV-1 life cycle initiates when the HTLV-1 virion attaches to a host cell such as by way of a surface receptor(s), e.g., GLUT1/ HSPG/NRP-1, via the HTLV- 1 envelope surface and/or transmembrane domains of the Env protein.
- a host cell such as by way of a surface receptor(s), e.g., GLUT1/ HSPG/NRP-1
- the contents of the virions can enter the cytoplasm of the host cell and the viral genomic RNA (gRNA) can undergo reverse transcription to convert gRNA into double stranded DNA (dsDNA) using the virally encoded reverse transcriptase (RNA-dependent/DNA-dependent polymerase).
- the host cell machinery can then drive transcription of the proviral DNA using host cell RNA polymerase II to produce viral mRNA.
- the unspliced full-length viral genomic RNA ( ⁇ 9 kb) can be used to produce progeny virions and to produce, e.g., Gag protein, Pol protein, and Pro protein (a protease enzyme required for maturation of structural and enzymatic proteins).
- the singly spliced subgenomic mRNA ( ⁇ 4.3 kb) can be used to produce Env protein, and the doubly-spliced RNA ( ⁇ 2.1 kb) can be used to translate the regulatory proteins Tax (open reading frame (ORF) IV) and Rex (ORF III).
- Alternative splicing can also lead to the production of a number of accessory proteins from pX ORF I and II.
- Immature virions can be assembled at the plasma membrane and are considered mature following viral budding.
- Amino acid sequences derived from HTLV-1 disclosed herein may include naturally occurring proteogenic amino acids as well as non-proteogenic amino acids and non-naturally occurring amino acids such as amino acid analogs.
- the amino acids that may be used in the practice of the present disclosure may include, for example, without limitation, naturally occurring proteogenic (L)-amino acids, their optical (D)-isomers, chemically modified amino acids, including, e.g., amino acid analogs such as, e.g., selenocysteine (Sec), penicillamine (3-mercapto-D-valine), pyroglutamic acid (5- oxoproline), etc., naturally occurring non-proteogenic amino acids such as norleucine, and chemically synthesized amino acids that have properties known in the art to be characteristic of an amino acid, and amino acid equivalents.
- proteins e.g., selenocysteine (Sec), penicillamine (3-mercapto-D-valine), pyroglutamic acid (5- oxoproline), etc.
- non-proteogenic amino acids such as norleucine
- chemically synthesized amino acids that have properties known in the art to be characteristic of an
- an isolated peptide of the present disclosure comprises an amino acid sequence that is at least about 60%, at least about 65%, at least Attorney Docket No.250298.000682 about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% identical to the amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof.
- an isolated peptide of the present disclosure comprises an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143- 146.
- an isolated peptide of the present disclosure consists essentially of an amino acid sequence of any of SEQ ID NOs: 1-89 and 143-146. In some embodiments, the isolated peptide consists of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. In some embodiments, the isolated peptide comprises two or more sequences selected from any one of SEQ ID NOs: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof. [00130] A list of non-limiting examples of HTLV-1-derived peptides are provided in Table 1 below. Table 1.
- a peptide of the disclosure may be synthetically produced or produced by hydrolysis. Synthetically produced peptides can include randomly generated peptides, specifically designed peptides, and peptides where at least some of the amino acid positions are conserved among several peptides and the remaining positions are random. Alternatively, a peptide of the present disclosure may be produced by expression in a heterologous host cell.
- peptides that are produced by hydrolysis undergo hydrolysis prior to binding of the antigen to an MHC molecule.
- Class I MHC typically present peptides Attorney Docket No.250298.000682 derived from proteins actively synthesized in the cytoplasm of the cell.
- class II MHC typically present peptides derived either from exogenous proteins that enter a cell’s endocytic pathway or from proteins synthesized in the endoplasmic reticulum (ER). Intracellular trafficking permits a peptide to become associated with an MHC molecule.
- the binding of a peptide to an MHC peptide binding groove can control the spatial arrangement of MHC and/or peptide amino acid residues recognized by a TCR. Such spatial control is due in part to hydrogen bonds formed between a peptide and an MHC molecule. Based on the knowledge on how peptides bind to various MHC molecules, the major MHC anchor amino acids and the surface exposed amino acids that are varied among different peptides can be determined.
- the length of an MHC-binding peptide is from about 5 to about 40 amino acid residues, more preferably from about 6 to about 30 amino acid residues, and even more preferably from about 8 to about 20 amino acid residues, and even more preferably between about 9 and 11 amino acid residues, including any size peptide between 5 and 40 amino acids in length, in whole integer increments (i.e., 5, 6, 7, 8, 9...40). While naturally MHC class II-bound peptides vary from about 9-40 amino acids, in nearly all cases the peptide can be truncated to an about 9-11 amino acid core without loss of MHC binding activity or T cell recognition.
- the isolated peptides of the disclosure may be about 8-12 amino acids in length.
- a peptide disclosed herein may be 8 amino acids, 9 amino acids, 10 amino acids, 11 amino acids, or 12 amino acids in length.
- the peptides of the disclosure may comprise one or more reverse peptide bonds, one or more non-peptide bonds, one or more chemical modifications, one or more D-isomers of amino acids, or any combination thereof.
- the peptide may be modified to comprise one or more reverse peptide bonds or non-peptide bonds. Such modification may improve stability and/or binding of the peptide to MHC molecules to elicit a stronger immune response.
- a reverse peptide bond amino acid residues are not joined by peptide (—CO—NH—) linkages but the peptide bond is reversed.
- Such retro-inverso peptidomimetics may be made using methods known in the art, for example such as those described in Meziere et al., 1997 (Meziere C., et al. J Immunol 1997). This approach involves making Attorney Docket No.250298.000682 pseudopeptides containing changes involving the backbone, and not the orientation of side chains. Such pseudopeptides may be useful, for example, for desired MHC binding and/or T helper cell responses.
- Retro-inverse peptides which contain NH—CO bonds instead of CO—NH peptide bonds, are much more resistant to proteolysis. Additional non-peptide bond that may be used are, for example, —CH2—NH, —CH2S—, —CH2CH2—, — CH ⁇ CH—, —COCH2—, —CH(OH)CH2—, and —CH2SO—.
- the amino acid residues comprising the peptides of the disclosure may be chemically modified.
- Non-limiting examples of chemical modifications include, for example, phosphorylation, acetylation, deamidation acylation, amidination, pyridoxylation of lysine, reductive alkylation, trinitrobenzylation of amino groups with 2,4,6- trinitrobenzene sulphonic acid (TNBS), amide modification of carboxyl groups and sulphydryl modification by performic acid oxidation of cysteine to cysteic acid, formation of mercurial derivatives, formation of mixed disulfides with other thiol compounds, reaction with maleimide, carboxymethylation with iodoacetic acid or iodoacetamide and carbamoylation with cyanate at alkaline pH.
- TNBS 2,4,6- trinitrobenzene sulphonic acid
- modification of, for example, arginyl residues in proteins may be based on the reaction of vicinal dicarbonyl compounds such as phenylglyoxal, 2,3- butanedione, and 1,2-cyclohexanedione to form an adduct.
- Another example is the reaction of methylglyoxal with arginine residues.
- Cysteine can be modified without concomitant modification of other nucleophilic sites such as lysine and histidine.
- Selective reduction of disulfide bonds in proteins can also be performed. Disulfide bonds can be formed and oxidized during the heat treatment of biopharmaceuticals.
- Woodward’s Reagent K may be used to modify specific glutamic acid residues.
- N-(3-(dimethylamino)propyl)-N′- ethylcarbodiimide can be used to form intra-molecular crosslinks between a lysine residue and a glutamic acid residue.
- diethylpyrocarbonate and 4-hydroxy-2-nonenal can be used to modify histidyl residues in proteins.
- the reaction of lysine residues and other ⁇ -amino groups is, for example, useful in binding of peptides to surfaces or the cross- linking of proteins/peptides.
- the preferred amino acids in position 2 for HLA- A*02 are Leu, lie, Val, or Met and at the C-terminus are Val or Leu.
- Multiple positions may be important for stable peptide binding to HLA-A*02, including positions 2, 3, 5-7, and 9.
- the anchor residues at position 2 and 9 may be of prime importance for peptide binding to HLA-A2.
- other peptide side chains, e.g., at position 3 may contribute to the stability of the interaction.
- the optimal length for peptide binding can be longer than 9 residues.
- Non-limiting examples Attorney Docket No.250298.000682 of linkers include a part of human CD8 ⁇ chain, extracellular domain of CD28, an Ig hinge from IgG, IgM, IgA, IgD, or IgE, FcyRllla receptor, or a functional fragment thereof.
- co-stimulatory domains may be derived from one or more co-stimulatory molecules, such as, but not limited to, 4-1BB, CD2, CD7, CD27, CD28, CD30, CD40, CD54 (ICAM), CD83, CD134 (OX40), CD150 (SLAMF1), CD152 (CTLA4), CD223 (LAG3), CD270 (HVEM), CD278 (ICOS), DAP10, LAT, NKD2C SLP76, TRIM, BTLA, GITR, CD226, HVEM, and ZAP70.
- co-stimulatory molecules such as, but not limited to, 4-1BB, CD2, CD7, CD27, CD28, CD30, CD40, CD54 (ICAM), CD83, CD134 (OX40), CD150 (SLAMF1), CD152 (CTLA4), CD223 (LAG3), CD270 (HVEM), CD278 (ICOS), DAP10, LAT, NKD2C SLP76, TRIM,
- Non-limiting examples of classes of additional genes include (a) a second targeting moiety, such as antibodies, including fragments thereof and bispecific antibodies (e.g., bispecific T cell engagers (BiTEs)), (b) secretable cytokines (e.g., GM-CSF, IL-7, IL-12, IL-15, IL-18), (c) membrane bound cytokines (e.g., IL-15), (d) chimeric cytokine receptors (e.g., IL-2/IL-7, IL-4/IL-7), (e) constitutive active cytokine receptors (e.g., C7R), (f) dominant negative receptors (DNR; e.g., TGFRII DNR), (g) ligands of co-stimulatory molecules (e.g., CD80, 4-1BBL), (h) nuclear factor of activated T cells (NFATs) (e.g., NFATc1, NFATc2, NFATc3, NFATc
- compositions of the disclosure may comprise multiple peptides, e.g., 2 to 50, 2 to 40, 2 to 30, 5 to 25, 5 to 20, or 10 to 15 peptides as described herein (e.g., SEQ ID NO: 1-89 and 143-146).
- the pharmaceutical composition may be adapted for administration by any appropriate route such as, e.g., parenteral (including subcutaneous, intramuscular, or intravenous), enteral (including oral or rectal), inhalation, or intranasal routes.
- parenteral including subcutaneous, intramuscular, or intravenous
- enteral including oral or rectal
- inhalation or intranasal routes.
- compositions based on the peptides, peptide-based molecules such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties disclosed herein can be formulated in any conventional manner using one or more physiologically acceptable carriers and/or excipients.
- complexes e.g., peptide-MHC (pMHC) complexes
- fusion proteins e.g., fusion proteins, or conjugates comprising the peptide(s)
- nucleic acid molecules, vectors, cells, or binding moieties disclosed herein can be formulated in any conventional manner using one or more physiologically acceptable carriers and/or excipients.
- the lyophilization formulation may comprise the peptides disclosed herein, mannitol and poloxamer 188.
- the pharmaceutical composition may comprise a lyophilization formulation comprising a reconstituted-liquid composition.
- pharmaceutical compositions of the present disclosure may provide a formulation with an enhanced solubility and/or moistening of the lyophilizate over previously known compositions.
- enhanced solubility and/or moistening of the lyophilizate may be achieved using an appropriate composition of excipients.
- compositions of the present disclosure comprising peptides of SEQ ID NO: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof, may be developed to show a Attorney Docket No.250298.000682 desired shelf stability at (e.g., at ⁇ 20° C, +5° C, or +25° C) and can be easily resolubilized such that the lyophilizate can be completely dissolved through the use of a buffer or other excipients from seconds up to two or more minutes, with or without the use of an of ultrasonic homogenizer.
- the pharmaceutical compositions can further be formulated as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain other agents including suspending, stabilizing and/or dispersing agents.
- the pharmaceutical compositions can also be formulated as a depot preparation. These long acting formulations can be administered by implantation (e.g. subcutaneously or intramuscularly) or by intramuscular injection.
- the compounds may be formulated with suitable polymeric or hydrophobic materials (e.g. as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
- penetrants are generally known in the art, and include, for example, for transmucosal administration, bile salts, and fusidic acid derivatives.
- detergents may be used to facilitate permeation.
- Transmucosal administration can occur using nasal sprays or suppositories.
- the vector particles described herein can be formulated into ointments, salves, gels, or creams as generally known in the art.
- a wash solution can also be used locally to treat an injury or inflammation in order to accelerate healing.
- Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
- Sterile injectable solutions can be prepared by incorporating the active compounds or constructs in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
- solutions can be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
- the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but slow release capsules or microparticles and microspheres and the like can also be employed.
- aqueous solutions for parenteral administration in an aqueous solution
- the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
- aqueous solutions are especially suitable for intravenous, intratumorally, intramuscular, subcutaneous and intraperitoneal administration.
- sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
- one dosage could Attorney Docket No.250298.000682 be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion.
- nasal solutions can be aqueous solutions designed to be administered to the nasal passages in drops or sprays. Nasal solutions can be prepared so that they are similar in many respects to nasal secretions. Thus, the aqueous nasal solutions usually are isotonic and slightly buffered to maintain a pH of 5.5 to 7.5.
- oral pharmaceutical compositions will include an inert diluent or assimilable edible carrier, or they may be enclosed in hard or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
- the active compounds may be incorporated Attorney Docket No.250298.000682 with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
- the tablets, troches, pills, capsules and the like may also contain the following: a binder, as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring.
- a binder as gum tragacanth, acacia, cornstarch, or gelatin
- excipients such as dicalcium phosphate
- a disintegrating agent such as corn starch, potato starch, alginic acid and the like
- a lubricant such as magnesium stearate
- a sweetening agent such as sucrose, lactose or saccharin may be added or a flavor
- kits for use with methods and compositions can also include a suitable container, for example, vials, tubes, mini- or microfuge tubes, test tube, flask, bottle, syringe or other container. Where an additional component or agent is provided, the kit can contain one or more additional containers into which this agent or component may be placed.
- Compositions may include administration to a subject intravenously, intratumorally, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostaticaly, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, intramuscularly, intrathecally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, orally, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, via a catheter, via a lavage, in a cream, or in a lipid composition.
- the vaccine composition may take the form of an APC displaying the peptide of the disclosure in complex with MHC.
- the APC is an immune cell, more preferably a dendritic cell or a B cell.
- the peptide may be pulsed onto the surface of the cell (Thurner, J Exp Med. 1999; 190(11):1669-78), or nucleic acid encoding for the peptide of the disclosure may be introduced into dendritic cells or B cells (e.g., by electroporation. Van Tendeloo, Blood.2001; 98(1):49-56).
- compositions of the disclosure may be administered directly into the patient, into the affected organ or systemically i.d., i.m., s.c., i.p. and i.v., or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation of immune cells derived from the patient, which are then re-administered to the patient.
- the nucleic acid is administered to cells in vitro, it may be useful for the cells to be transfected so as to co-express immune-stimulating cytokines, such as interleukin-2.
- the peptide or peptide-based molecule may be substantially pure, or combined with an immune- stimulating adjuvant or used in combination with immune-stimulatory cytokines, or be administered with a suitable delivery system, e.g., liposomes, viral particles, VLPs.
- a suitable delivery system e.g., liposomes, viral particles, VLPs.
- the Attorney Docket No.250298.000682 peptide or peptide-based molecule may also be conjugated to a suitable carrier such as keyhole limpet haemocyanin (KLH) or mannan (see, e.g., WO 95/18145 and Longenecker et al., 1993).
- KLH keyhole limpet haemocyanin
- mannan see, e.g., WO 95/18145 and Longenecker et al., 1993.
- the peptide-containing compositions described herein further comprise an accessory molecule which can modulate a survival or an activity of TCR-expressing cells.
- an accessory molecule which can modulate a survival or an activity of TCR-expressing cells.
- useful accessory molecules include, e.g., an anti- CD28 antibody, an anti-CD80 (B7.1) antibody, an anti-CD86 (B7.2) antibody, an anti-anti- CD3 antibody, an anti-CD2 antibody, an anti-CD4 antibody, an anti-CD8 antibody, an anti- CD47 antibody, and functional derivatives, mutants and fragments thereof.
- Accessory molecules used in the peptide-containing compositions described herein include molecules that provide a signal which, in addition to the primary signal provided by, for instance, binding of a TCR/CD3 complex with a pMHC complex, mediates a T cell response, including, but not limited to, proliferation, activation, differentiation, and the like.
- the accessory molecule can be, for example, an inhibitory or stimulatory antibody, a peptide ligand, a costimulatory peptide, a cytokine, etc.
- Non-limiting examples of accessory molecules that can be used in the peptide-containing compositions described herein include, e.g., CD7, B7.1 (CD80), B7.2 (CD86), PD-L1 , PD-L2, 4-1BBL, OX40L, Fas ligand (FasL), inducible co stimulatory ligand (ICOS-L), intercellular adhesion molecule (ICAM), CD30L, CD40, CD70, CD83, HLA-G, MICA, MICB, FIVEM, lymphotoxin ⁇ receptor, 3/TR6, ILT3, ILT4, HVEM, an agonist or antibody that binds Toll ligand receptor and a ligand that specifically binds to B7-H3 as well as antibodies that specifically bind to CD27, CD28, B7.1 (CD80), B7.2 (CD86), 4-1BB, OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD
- accessory molecules include, e.g., TNF/TNF family members (e.g., OX40L, ICOSL, FASL, LTA, LTB TRAIL, CD153, TNFSF9, RANKL, TWEAK, TNFSF13, TNFSF13b, TNFSF14, TNFSF15, TNFSF18, CD40LG, CD70); members of the Immunoglobulin superfamily (e.g., VISTA, PD1, PD- L1 , PD-L2, B71 , B72, CTLA4, CD28, TIM3, CD4, CD8, CD19, T cell receptor chains, Attorney Docket No.250298.000682 ICOS, ICOS ligand, HHLA2, butyrophilms, BTLA, B7-H3, B7-H4, CD3, CD79a, CD79b, IgSF CAMS (including CD2, CD58, CD48, CD150, CD229, CD244, ICAM-1),
- TNF/TNF family members
- the peptide-containing compositions described herein further comprise a cytotoxic agent.
- the cytotoxic agent is a toxin or a radioactive isotope (e.g., a radioconjugate) or a suicide gene.
- toxins which can be used in the peptide-containing compositions described herein include, e.g., enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments, mutants or derivatives thereof.
- Enzymatically active toxins and fragments thereof that can be used include, for example, diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, ⁇ -sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
- diphtheria A chain nonbinding active fragments of diphtheria toxin
- exotoxin A chain from Pseudomonas aeruginosa
- ricin A chain abrin A chain
- Non-limiting examples of suicide genes include, e.g., thymidine kinase, cytosine deaminase, purine nucleoside phosphorylase, nitroreductase, ⁇ -galactosidase, hepatic cytochrome P450-2B1, linamarase, horseradish peroxidase, and carboxypeptidase.
- Methods for introducing polypeptide or polynucleotides of the present disclosure into a cell or subject can include, for example, vector delivery, particle-mediated delivery, exosome-mediated delivery, lipid-nanoparticle-mediated delivery, cell- penetrating-peptide-mediated delivery, or implantable-device-mediated delivery.
- a nucleic acid or protein can be introduced into a cell or subject in a carrier such as a poly(lactic acid) (PLA) microsphere, a poly(D,L-lactic-coglycolic-acid) (PLGA) microsphere, a liposome, a micelle, an inverse micelle, a lipid cochleate, or a lipid microtubule.
- a carrier such as a poly(lactic acid) (PLA) microsphere, a poly(D,L-lactic-coglycolic-acid) (PLGA) microsphere, a liposome, a micelle, an inverse micelle, a lipid cochleate, or a lipid microtubule.
- Exemplary nanoparticles include, but are not limited to, polymeric nanoparticles, inorganic nanoparticles, liposomes, lipid nanoparticles (LNP), an immune stimulating complex (ISCOM), a virus-like particle (VLP), or a self-assembling protein.
- the nanoparticles may be calcium phosphate nanoparticles, silicon nanoparticles or gold nanoparticles.
- the polymeric nanoparticles may comprise one or more synthetic polymers, such as poly(d,l-lactide-co- glycolide) (PLG), poly(d,l-lactic-coglycolic acid) (PLGA), poly(g-glutamic acid) (g- PGA), poly(ethylene glycol) (PEG), or polystyrene or one or more natural polymers such as a polysaccharide, for example pullulan, alginate, inulin, and chitosan.
- PEG poly(ethylene glycol)
- polystyrene or one or more natural polymers such as a polysaccharide, for example pullulan, alginate, inulin, and chitosan.
- the use of a polymeric nanoparticles may be advantageous due to the properties of the polymers that may be include in the nanoparticle.
- the natural and synthetic polymers recited above may have good biocompatibility and biodegradability, a non-toxic nature and/or the ability to be manipulated into desired shapes and sizes.
- the polymeric nanoparticle may also form hydrogel nanoparticles, hydrophilic three-dimensional polymer networks with favorable properties including flexible mesh size, large surface area for multivalent conjugation, high water content, and high loading capacity for antigens.
- Polymers such as Poly(L-lactic acid) (PLA), PLGA, PEG, and polysaccharides are suitable for forming hydrogel nanoparticles.
- Inorganic nanoparticles typically have a rigid structure and comprise a shell in which an antigen is encapsulated or a core to which the antigen may be covalently attached.
- the core may comprise one or more atoms such as gold (Au), silver (Ag), copper (Cu) atoms, Au/Ag, Au/Cu, Au/Ag/Cu, Au/Pt, Au/Pd or Au/Ag/Cu/Pd or calcium phosphate (CaP).
- Other molecules suitable for complexing with the polypeptide or polynucleotides of the disclosure include cationic molecules, such as, polyamidoamine , dendritic polylysine, polyethylene irinine or polypropylene imine, polylysine, chitosan, DNA-gelatin coarcervates, DEAE dextran, dendrimers, or polyethylenimine (PEI).
- Nanoparticles that may be used for conjugation with antibodies of the present disclosure include but not are limited to PEGylated liposomes, poly(d,l-lactide-co-glycolide)/montmorillonite nanoparticles (PLGA/MMT NPs), Attorney Docket No.250298.000682 poly(lactide-co-glycolide) (PLGA) nanoparticles, poly-(malic acid)-based nanoparticles, chitosan-shelled nanoparticles, carbon nanotubes, and other inorganic nanoparticles (such as nanoparticles made of magnesium–aluminium layered double hydroxides with disuccinimidyl carbonate (DSC), and TiO 2 nanoparticles).
- PEGylated liposomes poly(d,l-lactide-co-glycolide)/montmorillonite nanoparticles (PLGA/MMT NPs)
- PLGA/MMT NPs PLGA/M
- Nanoparticles can be developed and conjugated to an antibody contained in a pharmaceutical composition for targeting virus-infected cells.
- Treatment Methods [00342]
- Compositions of the present disclosure including the peptides, peptide- based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties of the disclosure, may be used in the prophylaxis and/or treatment of a viral infection (e.g., HTLV-1 infection) and/or diseases or disorders caused by the viral infection (e.g., HTLV-1 infection).
- a viral infection e.g., HTLV-1 infection
- diseases or disorders caused by the viral infection e.g., HTLV-1 infection
- a method for modulating an activity, proliferation or survival of a cell comprising a TCR comprising contacting the cell with a composition (e.g., peptide, complex (e.g., pMHC complex), fusion protein, or conjugate) of the disclosure.
- a composition e.g., peptide, complex (e.g., pMHC complex), fusion protein, or conjugate
- the cell is a lymphocyte such as, e.g., a T-cell (e.g., a CD4+ T-cell or a CD8+ T-cell).
- the target T cell is a CD4+ T cell such as, e.g., a helper T cell (e.g., a Th1, Th2, or Th17 cell) or a CD4+/CD25+/FOXP3+ regulatory T (Treg) cell.
- the target T cell is a CD8+ T cell such as, e.g., a cytotoxic T cell.
- the target T cell is a memory T cell, which can be a CD4+ T cell or a CD8+ T cell, where memory T cells are generally CD45RO+.
- the target T cell is an NK-T cell.
- the contacting is ex vivo.
- the contacting is in vivo in a subject (e.g., human).
- the cell is a mammalian cell (e.g., a human cell).
- Attorney Docket No.250298.000682 [00347]
- the peptide is presented by a class I MHC polypeptide.
- the peptide is presented by class II MHC polypeptides.
- T cell The interaction of a T cell with the peptides described herein can result in, e.g., activation, induction of anergy, or death of a T cell that occurs when the TCR of the T cell is bound by a TCR-binding molecule (e.g., pMHC complex).
- Activation of a T cell refers to induction of signal transduction pathways in the T cell resulting in production of cellular products (e.g., interleukin-2) by that T cell.
- cellular products e.g., interleukin-2
- Activation and anergy can be measured by, for example, measuring the amount of IL-2 produced by a T cell after a pMHC complex has bound to the TCR.
- Anergic cells will have decreased IL-2 production when compared with stimulated T cells.
- Another method for measuring the diminished activity of anergic T cells includes measuring intracellular and/or extracellular calcium mobilization by a T cell upon engagement of its TCR’s. “T cell death” refers to the permanent cessation of substantially all functions of the T cell.
- a method of inducing an immune response against HTLV infection comprising administering to the subject a therapeutically effective amount of a composition (e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, cells, or binding moieties) of the present disclosure.
- a composition e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, cells, or binding moieties
- generating an immune response comprises an increase in target antigen-specific cytotoxic T lymphocytes (CTL) activity of about 1.5- fold to 20-fold, or more fold in a subject administered a composition of the disclosure as compared to a control. In certain embodiments, generating an immune response comprises an increase in target-specific CTL activity of about 1.5-fold to 20-fold, or more fold in a subject administered the composition of the disclosure as compared to a control.
- CTL cytotoxic T lymphocytes
- generating an immune response that comprises an increase in target antigen- specific cell-mediated immunity activity as measured by ELISpot assays measuring cytokine secretion, such as interferon-gamma (IFN- ⁇ ), interleukin-2 (IL-2), tumor necrosis Attorney Docket No.250298.000682 factor-alpha (TNF- ⁇ ), or other cytokines, of about 1.5-fold to 20-fold, or more fold as compared to a control.
- generating an immune response comprises an increase in target- specific antibody production of between 1.5-fold and 5-fold in a subject administered the composition of the disclosure as compared to an appropriate control.
- generating an immune response comprises an increase in target- specific antibody production of about 1.5-fold to 20-fold, or more fold in a subject administered the composition of the disclosure as compared to a control.
- T cell activation may be determined, e.g., by measuring changes in the level of expression of cytokines and/or T cell activation markers, and/or the induction of antigen- specific proliferating cells. Techniques known to those of skill in the art, including, but not limited to, immunoprecipitation followed by western blot analysis, ELISAs, flow cytometry, northern blot analysis, and RT-PCR can be used to measure the expression cytokines and T cell activation markers.
- Cytokine release may be measured by measuring secretion of cytokines including but not limited to Interleukin-2 (IL-2), Interleukin-4 (IL- 4), Interleukin-6 (IL-6), Interleukin-12 (IL-12), Interleukin-16 (IL-16), PDGF, TGF- ⁇ , TGF- ⁇ , TNF- ⁇ , TNF- ⁇ , GCSF, GM-CSF, MCSF, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , TFN- ⁇ , IGF-I, and IGF-II.
- IL-2 Interleukin-2
- IL-4 Interleukin-4
- IL-6 Interleukin-6
- IL-12 Interleukin-12
- IL-16 Interleukin-16
- T cell modulation may also be evaluated by measuring, e.g., proliferation by, e.g., 3 H-thymidine incorporation, trypan blue cell counts, and fluorescence activated cell sorting (FACS).
- FACS fluorescence activated cell sorting
- the anti-tumor responses of T cells may be determined in xenograft tumor models. Tumors may be established using any human cancer cell line expressing the relevant tumor associated antigen. To establish xenograft tumor models, about 5 ⁇ 10 6 viable cells, may be injected, e.g., subcutaneously into nude athymic mice using for example Matrigel (Becton Dickinson).
- the endpoint of the xenograft tumor models can be determined based on the size of the tumors, weight of animals, survival time and histochemical and histopathological examination of the cancer, using methods known to one skilled in the art.
- a method of treating an HTLV-1 infection in a subject in need thereof comprising administering to the subject Attorney Docket No.250298.000682 an effective amount of a composition (e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, cells, binding moieties) of the present disclosure.
- a composition e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, cells, binding moieties
- a method of preventing or reducing the likelihood of an HTLV-1-induced disease or disorder in a subject in need thereof comprising administering to the subject an effective amount of a composition (e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, a cells, binding moieties) of the present disclosure.
- a composition e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, a cells, binding moieties
- the HTLV-1-induced disease or disorder can be, e.g., adult T-cell leukemia/lymphoma (ATL) or HTLV-1-myelopathy/tropical spastic paraparesis (HAM/TSP).
- the HTLV-1-induced disease or disorder can be, e.g., a hypersensitivity reaction such as, without limitation, arthritis, uveitis, or HTLV-1- associated Infective Dermatitis (IDH).
- a hypersensitivity reaction such as, without limitation, arthritis, uveitis, or HTLV-1- associated Infective Dermatitis (IDH).
- IDH HTLV-1-associated Infective Dermatitis
- ATL The most benign but detectable form of ATL is an asymptomatic pre-leukemic phase which can be diagnosed, e.g., incidentally upon examination of a peripheral blood smear which reveals abnormal lymphocytes with characteristics of lobulated nuclei Smoldering
- ATL is the most benign symptomatic form of ATL and can be characterized by a normal peripheral blood leukocyte count, a few circulating leukemic cells, and cutaneous lesions but not visceral lesions.
- the development of chronic ATL is associated with visceral involvement and is evidenced by hepatosplenomegaly, lymphadenopathy, and peripheral blood leukocytosis.
- Patients with acute ATL can have elevated levels of lactate dehydrogenase and bilirubin, peripheral blood leukocytosis, hypercalcemia, and cutaneous and visceral involvement.
- ATL can be accompanied by severe immunosuppression and many ATL patients can be highly susceptible to various infections such, but not limited to cryptococcal meningitis, pneumocystis carinii pneumonia, disseminated cytomegalovirus infection, and candida esophagitis.
- Acute ATL is typically unresponsive to conventional chemotherapy and the median life expectancy of patients with acute ATL is approximately 11 months.
- HTLV-1 is thought to cause approximately 80 percent of the cases of HAM/TSP such as by weakening the immune system.
- HAM/TSP A majority of patients with Attorney Docket No.250298.000682 HAM/TSP have few or no symptoms over the lifespan. In addition to neurological symptoms of weakness and muscle spasms and/or stiffness, in rare instances patients with HAM/TSP may also have, e.g., arthritis (inflammation of one or more joints); infectious dermatitis (inflammation of the skin); keratoconjunctivitis sicca (persistent dryness of the cornea and/or conjunctiva); polymyositis (an inflammatory muscle disease); pulmonary lymphocytic alveolitis (inflammation of the lung); and/or uveitis (inflammation of the uveal tract of the eye).
- arthritis inflammation of one or more joints
- infectious dermatitis infectious dermatitis
- keratoconjunctivitis sicca persistent dryness of the cornea and/or conjunctiva
- polymyositis an inflammatory muscle disease
- the cancer may specifically be of the following histological type, though it is not limited to these: neoplasm, malignant; carcinoma; carcinoma, undifferentiated; giant and spindle cell carcinoma; small cell carcinoma; papillary carcinoma; squamous cell carcinoma; lymphoepithelial carcinoma; basal cell carcinoma; pilomatrix carcinoma; transitional cell carcinoma; papillary transitional cell carcinoma; adenocarcinoma; gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma; adenoid cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma, familial polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchiolo-alveolar adenocarcinoma; papillary adenocarcinom
- compositions and methods can be combined with other therapeutic agents suitable for the same or similar diseases.
- two or more embodiments described herein may be also co-administered to generate additive or synergistic effects.
- the embodiment described herein and the second therapeutic agent may be simultaneously or sequentially (in any order). Suitable therapeutically effective dosages for each agent may be lowered due to the additive action or synergy.
- the compositions and methods disclosed herein are useful to enhance the efficacy of vaccines directed to HTLV-1 infection or HTLV-1- induced diseases.
- compositions and methods described herein can be administered to a subject either simultaneously with or before (e.g., 1-30 days before) a reagent (including but not limited to small molecules, antibodies, or cellular reagents) that acts to elicit an immune response (e.g., to treat HTLV-1, HTLV-2, HTLV-3, and/or HTLV- 4 infection or cancer) is administered to the subject.
- a reagent including but not limited to small molecules, antibodies, or cellular reagents
- an immune response e.g., to treat HTLV-1, HTLV-2, HTLV-3, and/or HTLV- 4 infection or cancer
- the compositions and methods described herein can be also administered in combination with an anti-tumor antibody or an antibody directed at a pathogenic antigen (e.g., HTLV-1) or allergen.
- compositions and methods described herein can be combined with other immunomodulatory treatments such as, e.g., therapeutic vaccines (including but not limited to GVAX, DC-based vaccines, etc.), checkpoint inhibitors (including but not limited to agents that block CTLA4, PD1, LAG3, TIM3, etc.) or activators (including but not limited to agents that enhance 41BB, OX40, etc.).
- therapeutic vaccines including but not limited to GVAX, DC-based vaccines, etc.
- checkpoint inhibitors including but not limited to agents that block CTLA4, PD1, LAG3, TIM3, etc.
- activators including but not limited to agents that enhance 41BB, OX40, etc.
- the inhibitory treatments described herein can be also combined with other treatments that possess the ability to modulate NKT function or stability, including but not limited to CD1d, CD1d-fusion proteins, CD1d dimers or larger polymers of CD1d either unloaded or loaded with antigens, CD1d- chimeric antigen receptors (CD1d-CAR), or any other of the five known CD1 isomers existing in humans (CD1a, CD1b, CD1c, CD1e), in any of the aforementioned forms or formulations, alone or in combination with each other or other agents.
- Therapeutic methods described herein can be combined with additional immunotherapies and therapies.
- NKT cells described herein when used for treating cancer, can be used in combination with cancer therapies, such as, e.g., surgery, radiotherapy, chemotherapy or combinations thereof, depending on type of the tumor, patient condition, other health issues, and a variety of factors.
- cancer therapies such as, e.g., surgery, radiotherapy, chemotherapy or combinations thereof, depending on type of the tumor, patient condition, other health issues, and a variety of factors.
- other therapeutic agents useful for combination cancer therapy with the inhibitors described herein include anti-angiogenic agents.
- anti-angiogenic agents have been identified Attorney Docket No.250298.000682 and are known in the art, including, e.g., TNP-470, platelet factor 4, thrombospondin-1, tissue inhibitors of metalloproteases (TIMP1 and TIMP2), prolactin (16-Kd fragment), angiostatin (38-Kd fragment of plasminogen), endostatin, bFGF soluble receptor, transforming growth factor ⁇ , interferon- ⁇ , soluble KDR and FLT-1 receptors, placental proliferin-related protein, as well as those listed by Carmeliet and Jain (2000).
- the inhibitors described herein can be used in combination with a VEGF antagonist or a VEGF receptor antagonist such as anti-VEGF antibodies, VEGF variants, soluble VEGF receptor fragments, aptamers capable of blocking VEGF or VEGFR, neutralizing anti-VEGFR antibodies, inhibitors of VEGFR tyrosine kinases and any combinations thereof (e.g., anti-hVEGF antibody A4.6.1, bevacizumab or ranibizumab).
- a VEGF antagonist or a VEGF receptor antagonist such as anti-VEGF antibodies, VEGF variants, soluble VEGF receptor fragments, aptamers capable of blocking VEGF or VEGFR, neutralizing anti-VEGFR antibodies, inhibitors of VEGFR tyrosine kinases and any combinations thereof (e.g., anti-hVEGF antibody A4.6.1, bevacizumab or ranibizumab).
- Non-limiting examples of chemotherapeutic compounds which can be used in combination treatments include, for example, aminoglutethimide, amsacrine, anastrozole, asparaginase, bcg, bicalutamide, bleomycin, buserelin, busulfan, campothecin, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, estradiol, estramnustine, etoposide, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone, flutamide
- chemotherapeutic compounds may be categorized by their mechanism of action into, for example, following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine)); Attorney Docket No.250298.000682 antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide), DNA damaging agents (actino
- compositions and methods described herein can encompass co-administering compositions and methods described herein with an anti-viral drug.
- useful anti-viral drugs include, adefovir and entecavir, telbivudine, immune system modulators such as interferon- ⁇ , - ⁇ or - ⁇ , didanosine, lamivudine, zanamavir, lopanivir, nelfinavir, efavirenz, indinavir, valacyclovir, zidovudine, amantadine, rimantidine, ribavirin, ganciclovir, foscarnet, and acyclovir or any salts or variants thereof.
- Kits [00369] The present disclosure further comprises a kit which may comprise any of various compositions of the present disclosure, including the peptides, peptide-based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties of the disclosure.
- peptide-based molecules such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)
- the present disclosure may include a kit comprising, for example: (a) a container that contains a pharmaceutical composition disclosed herein, for example, a pharmaceutical composition in solution or in lyophilized form; (b) optionally, a second container containing a diluent or reconstituting solution for the lyophilized formulation; and/or (c) optionally, instructions for (i) use of the solution or (ii) reconstitution and/or use of the lyophilized formulation.
- a kit comprising, for example: (a) a container that contains a pharmaceutical composition disclosed herein, for example, a pharmaceutical composition in solution or in lyophilized form; (b) optionally, a second container containing a diluent or reconstituting solution for the lyophilized formulation; and/or (c) optionally, instructions for (i) use of the solution or (ii) reconstitution and/or use of the lyophilized formulation.
- the kit may further comprise, for example, without limitation, one or more of (i) a buffer, (ii) a diluent, (iii) a filter, (iv) a needle, and/or (v) a syringe.
- the container may be a bottle, a vial, a syringe or test tube.
- the container may be a multi-use container.
- the pharmaceutical composition may be lyophilized.
- Kits of the present disclosure may comprise a lyophilized formulation of the present disclosure in a suitable container and instructions for its reconstitution and/or use.
- Suitable containers include, for example, bottles, vials (e.g. dual chamber vials), syringes (such as dual chamber syringes) and test tubes.
- the container may be formed from a variety of materials such as glass or plastic.
- the kit and/or container may contain instructions on or associated with the container that indicate directions for reconstitution of the lyophilized formulation and/or use of the kit.
- the label may indicate that the lyophilized formulation is to be reconstituted to an appropriate peptide concentration.
- the label may indicate that the formulation is useful or intended for any route of administration disclosed herein, e.g., parenteral administration routes disclosed herein.
- the container holding the formulation may be a multi-use vial, which may allow for repeat administrations (e.g., from 2-6 administrations) of the reconstituted formulation.
- the kit may further comprise a second container comprising a suitable diluent (e.g., sodium bicarbonate solution).
- a suitable diluent e.g., sodium bicarbonate solution.
- the kit may further include other materials desirable from a commercial and/or user standpoint, including, for example, without limitation, other buffers, diluents, filters, needles, syringes, and/or package inserts which may comprise, e.g., instructions for use.
- Kits of the present disclosure may have a single container that contains the formulation of the pharmaceutical compositions according to the present disclosure with or without other components (e.g., other compounds or pharmaceutical compositions of these other compounds) or may have a distinct container for each component.
- kits of the disclosure may include a formulation of the disclosure packaged for use in combination with the coadministration of a second compound (such as adjuvants (e.g., GM-CSF, a chemotherapeutic agent, a natural product, Attorney Docket No.250298.000682 a hormone or antagonist, an anti-angiogenesis agent or inhibitor, an apoptosis-inducing agent or a chelator) or a pharmaceutical composition thereof.
- adjuvants e.g., GM-CSF, a chemotherapeutic agent, a natural product, Attorney Docket No.250298.000682 a hormone or antagonist, an anti-angiogenesis agent or inhibitor, an apoptosis-inducing agent or a chelator
- the components of the kit may be pre-complexed or each component may be in a separate distinct container prior to administration to a patient.
- the components of the kit may be provided in one or more liquid solutions.
- a liquid solutions described herein may be an aqueous solution, for example, a sterile aqueous solution.
- the components of the kit may also be provided as solids, which may be converted into liquids such as by addition of suitable solvents, which may be provided in another distinct container.
- the container of a therapeutic kit may be a vial, test tube, flask, bottle, syringe, or any other means of enclosing a solid or liquid.
- the kit may contain a second vial or other container, which may allow for separate dosing.
- the kit may also contain another container for a pharmaceutically acceptable liquid.
- a kit may contain an apparatus (e.g., one or more needles, syringes, eye droppers, pipettes, etc.), which may allow for administration of the agents of the disclosure that are components of the present kit.
- Method and System for Identifying an Immunogenic Virus-Derived Peptide [00378] The present disclosure further comprises a method and system for identifying an immunogenic virus-derived peptide. Some or all the steps of the method can be executed by a computational device.
- the present disclosure may include a method for identifying an immunogenic virus-derived peptide that includes the following steps: (a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus, wherein the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences and a plurality of infected-subject endogenous RNA contig sequences; (b) identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences; (c) assembling a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; (d) identifying a protein sequence based Attorney Docket No.250298.000682 on the viral RNA sequence; and (e)
- a system configured to perform functions associated with the foregoing method can include a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device.
- the non-transitory computer-readable medium may include instructions thereon, that when executed by the processor(s), cause the computational device to: (a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences and a plurality of infected-subject endogenous RNA contig sequences, and wherein the infected subject is infected with a virus; (b) identify the plurality of virus- derived RNA contig sequences from within the plurality of RNA contig sequences; (c) assemble a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; (d) identify a protein sequence based on the
- the plurality of RNA contig sequences may be derived from one infected subject.
- the infected subject may be a human.
- the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject.
- the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject.
- identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences can further include: comparing at least a portion of contig sequences of the plurality of RNA contig sequences to a reference viral sequence; and identifying the plurality of virus-derived RNA contig sequences such that each contig sequence of the plurality of virus-derived RNA contig sequences comprises at least a portion that corresponds to the reference viral sequence.
- Attorney Docket No.250298.000682 [00386]
- each contig sequence of the plurality of virus- derived RNA contig may be distinct from the plurality of infected-subject endogenous RNA contig sequences.
- each contig sequence of the plurality of virus- derived RNA contig sequences may lack infected-subject endogenous RNA contig sequences.
- the reference viral sequence may include a reference genome.
- the reference genome may include a HTLV-1 virus genome.
- assembling the viral RNA sequence based on the plurality of virus-derived RNA contig sequences may include: overlapping common sequence portions at ends of at least a portion of the plurality of virus-derived RNA contig sequences such that the at least a portion of the plurality of virus-derived RNA contig sequences overlap linearly to assemble the viral RNA sequence.
- identifying a protein sequence based on the viral RNA sequence such that the protein sequence includes a translation of the viral RNA sequence may include: identifying the protein sequence without requiring a comparison to a database of viral proteins.
- identifying the protein sequence based on the viral RNA sequence such that the protein sequence includes a translation of the viral RNA sequence may include: identifying a plurality of protein sequences each based on the viral RNA sequence such that each of the plurality of protein sequences respectively include a translation of the viral RNA sequence, and identifying the protein sequence as a frequently occurring protein sequence within the plurality of protein sequences.
- the protein sequence may be identified based on the viral RNA sequence associated with a single infected subject.
- identifying the immunogenic virus-derived peptide based at least in part on the protein sequence may include: identifying a MHC molecule associated with the single infected subject; identifying one or more peptides based at least in part on the protein sequence such that the one or more peptides each form Attorney Docket No.250298.000682 a respective MHC-peptide complex with the MHC molecule; and identifying the immunogenic virus-derived peptide based on the one or more peptides.
- Method and System for Identifying an Integration Site of a Viral Gene within a Subject Gene [00395] The present disclosure further comprises a method and system for identifying an integration site of a viral gene within a subject gene.
- the present disclosure may include a method for identifying an integration site of a viral gene within a subject gene that includes the following steps: (a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus- derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and a plurality of hybrid RNA contig sequences comprising viral and infected- subject endogenous portions; (b) identifying the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; (c) comparing, for at least a portion of the plurality
- a system configured to perform functions associated with the foregoing method can include a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device.
- the non-transitory computer-readable medium may include instructions thereon, that when executed by the processor(s), cause the computational device to: (a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and plurality Attorney Docket No.250298.000682 of hybrid RNA contig sequences comprising viral and infected-subject endogenous portions; (b) identify the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; (c) compare, for at least a portion of the plurality of hybrid RNA contig
- the plurality of RNA contig sequences may be derived from one infected subject.
- the infected subject may be a human.
- the plurality of virus-derived RNA contig sequences may be derived from the virus infecting the infected subject.
- the virus may include a HTLV-1 virus.
- the plurality of infected-subject endogenous RNA contig sequences may be derived from RNA endogenous to the infected subject.
- the subject reference genome can include the human genome.
- These computer program instructions may also be stored in a computer- readable memory that can direct a computer or other programmable data processing Attorney Docket No.250298.000682 apparatus to function in a particular manner, such that the instructions stored in the computer-readable memory produce an article of manufacture including instruction means that implement one or more functions associated with method steps.
- Non-transitory computer-readable media may include, but is not limited to, random access memory (RAM), read-only memory (ROM), electronically erasable programmable ROM (EEPROM), flash memory or other memory technology, compact disc ROM (CD-ROM), digital versatile disks (DVD) or other optical storage, magnetic cassettes, magnetic tape, magnetic disk storage or other magnetic storage devices, or any other tangible, physical medium which can be used to store computer readable information.
- RAM random access memory
- ROM read-only memory
- EEPROM electronically erasable programmable ROM
- CD-ROM compact disc ROM
- DVD digital versatile disks
- Embodiments or implementations of the disclosed technology may provide for a computer program product, including a computer-usable medium having a computer- readable program code or program instructions embodied therein, said computer-readable program code adapted to be executed to implement one or more functions related to example methods presented herein.
- the computer program instructions may be loaded onto a computer or other programmable data processing apparatus to cause a series of operational elements or steps to be performed on the computer or other programmable apparatus to produce a computer-implemented process such that the instructions that execute on the computer or other programmable apparatus provide elements or steps for implementing the functions related to example methods presented herein.
- illustrations and descriptions of methods support combinations of means for performing the specified functions, combinations of elements or steps for performing the specified functions, and program instruction means for performing the specified functions. It will also be understood that at least some method steps, and combinations of method steps, can be implemented by special-purpose, hardware-based computer systems that perform the specified functions, elements or steps, or combinations of special-purpose hardware and computer instructions.
- Certain implementations of the disclosed technology can be utilized with customer devices that may include mobile computing devices.
- customer devices that may include mobile computing devices.
- mobile computing devices there are several categories of mobile devices, generally known as portable computing devices that can run on batteries but are not usually classified as laptops.
- mobile devices can include, but are not limited to portable computers, tablet PCs, internet tablets, PDAs, ultra-mobile PCs (UMPCs), wearable devices, and smart phones.
- UMPCs ultra-mobile PCs
- Certain implementations of the disclosed technology can be utilized with medical equipment, medical devices, and/or associated peripherals.
- the present disclosure provides a method for identifying an immunogenic virus-derived peptide, the method comprising: a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus, wherein the plurality of RNA contig sequences comprises a plurality of virus-derived RNA contig sequences and a plurality of infected-subject endogenous RNA contig sequences; b) identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences; c) assembling a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; d) identifying a protein sequence based on the viral RNA sequence; and e) identifying the immunogenic virus-derived peptide based at least in part on the identified protein sequence.
- the plurality of RNA contig sequences are derived from one infected subject. [00413] In some embodiments, the infected subject is a human. [00414] In some embodiments, the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject. [00415] In some embodiments, the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject.
- the identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences comprises: comparing at least a portion of contig sequences of the plurality of RNA contig sequences to a reference viral sequence; and Attorney Docket No.250298.000682 identifying the plurality of virus-derived RNA contig sequences such that each contig sequence of the plurality of virus-derived RNA contig sequences comprises at least a portion that corresponds to the reference viral sequence.
- each contig sequence of the plurality of virus- derived RNA contig is distinct from the plurality of infected-subject endogenous RNA contig sequences.
- each contig sequence of the plurality of virus- derived RNA contig sequences lacks infected-subject endogenous RNA contig sequences.
- the reference viral sequence comprises a reference genome.
- the reference genome comprises a HTLV-1 virus genome.
- the assembling the viral RNA sequence based on the plurality of virus-derived RNA contig sequences comprises: overlapping common sequence portions at ends of at least a portion of the plurality of virus-derived RNA contig sequences such that the at least a portion of the plurality of virus-derived RNA contig sequences overlap linearly to assemble the viral RNA sequence.
- the identifying a protein sequence based on the viral RNA sequence such that the identified protein sequence includes a translation of the viral RNA sequence comprises: identifying the protein sequence without requiring a comparison to a database of viral proteins. [00423] In some embodiments, the identifying the protein sequence based on the viral RNA sequence such that the identified protein sequence includes a translation of the viral RNA sequence further comprises: identifying a plurality of protein sequences each based on the viral RNA sequence such that each of the plurality of protein sequences respectively include a translation of the viral RNA sequence, and identifying the protein sequence as a frequently occurring protein sequence within the plurality of protein sequences.
- the protein sequence identified based on the viral RNA sequence is associated with a single infected subject.
- the identifying the immunogenic virus-derived peptide based at least in part on the protein sequence comprises: identifying an MHC molecule associated with the single infected subject; identifying one or more peptides based at least in part on the protein sequence such that the one or more peptides each form a respective MHC-peptide complex with the MHC molecule; and identifying the immunogenic virus-derived peptide based on the one or more peptides.
- the present disclosure provides a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device, the non-transitory computer-readable medium including instructions thereon, that when executed by the processor(s), cause the computational device to: a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences and a plurality of infected- subject endogenous RNA contig sequences, and wherein the infected subject is infected with a virus; b) identify the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences; c) assemble a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; d) identify a protein sequence based on the viral RNA sequence; e) identify an immunogenic virus
- the plurality of RNA contig sequences are derived from only one infected subject.
- the infected subject comprises a human. Attorney Docket No.250298.000682
- the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject.
- the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject.
- the instructions which cause the computational device to identify the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences further comprise instructions, that when executed by the processor(s), cause the computational device to: compare at least a portion of contig sequences of the plurality of RNA contig sequences to a reference viral sequence; and identify the plurality of virus-derived RNA contig sequences such that each contig sequence of the plurality of virus-derived RNA contig sequences comprises at least a portion that corresponds to the reference viral sequence.
- each contig sequence of the plurality of virus- derived RNA contig is distinct from the plurality of infected-subject endogenous RNA contig sequences.
- each contig sequence of the plurality of virus- derived RNA contig sequences lacks portions of infected-subject endogenous RNA contig sequences.
- the reference viral sequences comprises a reference genome.
- the reference genome comprises a HTLV-1 genome.
- the instructions which cause the computational device to assemble the viral RNA sequence based on the plurality of virus-derived RNA contig sequences further comprise instructions, that when executed by the processor(s), cause the computational device to: overlap common sequence portions at ends of at least a portion of the plurality of virus-derived RNA contig sequences such that the at least a portion of the plurality of virus- derived RNA contig sequences overlap linearly to assemble the viral RNA sequence.
- the instructions which cause the computational device to identify a protein sequence based on the viral RNA sequence such that the protein Attorney Docket No.250298.000682 sequence includes a translation of the viral RNA sequence further comprise instructions, that when executed by the processor, cause the computational device to: identify the protein sequence without requiring a comparison to a database of viral proteins.
- the protein sequence identified based on the viral RNA sequence is a novel protein.
- the protein sequence identified based on the viral RNA sequence is associated with a single infected subject.
- the instructions which cause the computational device to identify a protein sequence based on the viral RNA sequence such that the protein sequence includes a translation of the viral RNA sequence further comprise instructions, that when executed by the processor(s), cause the computational device to: identify a plurality of protein sequences each based on the viral RNA sequence such that each of the plurality of protein sequences respectively include a translation of the viral RNA sequence; and identify the protein sequence as a frequently occurring protein sequence within the plurality of protein sequences.
- the instructions which cause the computational device to identify the immunogenic virus-derived peptide based at least in part on the protein sequence further comprise instructions, that when executed by the processor(s), cause the computational device to: identify a major histocompatibility complex (MHC) molecule associated with the single infected subject; identify one or more peptides based at least in part on the protein sequence such that the one or more peptides are each capable of forming a respective MHC-peptide complex with the MHC molecule; and identify the immunogenic virus-derived peptide based on the one or more peptides.
- MHC major histocompatibility complex
- the instructions when executed by the processor(s), further cause the computational device to: store the protein sequence to a database such that the protein sequence is associated with the infected subject within the database.
- Attorney Docket No.250298.000682 [00443]
- the present disclosure provides a method for identifying an integration site of a viral gene within a subject gene, the method comprising: a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and a plurality of hybrid RNA contig sequences comprising viral and infected-subject endogenous portions; b) identifying the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; c) comparing, for at least a portion of the
- the plurality of RNA contig sequences are derived from one infected subject.
- the infected subject is a human.
- the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject.
- the virus is a HTLV-1.
- the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject.
- the subject reference genome comprises the human genome.
- the present disclosure provides a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device, the non-transitory computer-readable medium including instructions thereon, that when executed by the processor(s), cause the computational device to: a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences Attorney Docket No.250298.000682 comprise a plurality of virus-derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and plurality of hybrid RNA contig sequences comprising viral and infected-subject endogenous portions; b) identify the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; c) compare, for at least a portion of the plurality of hybrid RNA contig sequences, infected-subject endogenous portions
- the plurality of RNA contig sequences are derived from only one infected subject.
- the infected subject comprises a human.
- the plurality of virus-derived RNA contig sequences are derived from a virus infecting the infected subject.
- the virus comprises a HTLV-1.
- the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject.
- the subject reference genome comprises the human genome.
- HTLV-1 Human T-lymphotropic Virus, type 1
- ATL Adult T-cell Leukemia/Lymphoma
- HAM HTLV-1 associated myelopathy
- IDH HTLV-1-associated infective dermatitis
- HTLV-1 is the most well-studied of the four HTLV genotypes (HTLV-1, HTLV-2, HTLV-3 and HTLV-4) the immunopeptidome of HTLV-1-infected T-cells and cell lines have not yet been characterized.
- HLA-I immunopeptidomics were performed on four model HTLV- transformed cell lines: MT-4, C8166, MT-2 and C5/MJ; primary cells and ATL patient samples.
- Example 1 Immunopeptidomics for HTLV-transformed cells.
- HLA-I complexes were purified from the 4 HTLV-transformed cell lines MT-4, C8166, MT-2 and C5/MJ, using anti-HLA-I W6/32 affinity purification ( Figure 1A).
- the mass spectrometry raw files were searched against a combined FASTA database containing both human (Uniprot) and HTLV-1 (Uniprot) protein sequences. More than 13,000 peptides were detected from the MT-4 cell line ( Figure 1D) and between 8,000 to 10,000 peptides were detected from the C8166, MT-2, and C5/MJ cell lines ( Figures 1B-1C and Figure 1E), with nonamers being the most abundant peptide length. [00460] Furthermore, a number of unique HTLV peptides were also identified. For example, 19 HTLV-1 peptides were detected from MT-4 cells, and 16 of the peptides overlapped between the three biological replicates ( Figure 4 and Table 5).
- panNetMHC 4.0 algorithm was used to predict the binding affinity of the identified HTLV-1 peptides with the HLA alleles of the respective cell lines.
- Most of the HTLV-1 peptides identified from the four cell lines were strong (panNetMHC 4.0 Rank below 0.5) or weak (panNetMHC 4.0 Rank between 0.5 – 2.0) predicted binders to the HLA of described cell lines (Tables 3-6). Table 3.
- HTLV-1 Peptides Detected from MT-2 Cells HLA- HLA- HLA- HLA- HLA- HLA- Attorney Docket No.250298.000682 Peptide Le Protein nM Ran nM Ran nM Ran nM Ran nM Ran nM Ran nM Ran nM Ran nM Ran nk 72 0 .26 .23 2 6 3 .87 2 .71 .02 .34 9 2 0 0 0
- HTLV-1 Peptides Detected from MT-4 Cells HLA- HLA- HLA- HLA- HLA- A02:01 A11:01 A24:02 B07:02 B15:01 n 1 .25 .75 .34 .14 5 .04 .49 .91 .50 .72 .00 .59 .45
- Attorney Docket No.250298.000682 LPAPHLTLP 9
- ENV_HTL1 3401 51.133821 60.744133 52.693154 2.47 3207 56.66 (SEQ ID NO: 53) M 6 4 8 7 55 .17 .80 n Table 6.
- Example 3 Validation of HTLV peptides with heavy analogues in cell lines and calculation of copy number.
- a few HTLV peptides are also selected for validation using the synthetic heavy analogues. Briefly, synthetic peptides containing heavy Leucine ( 13 C(6) 15 N(1)) (Thermo) are mixed with the HLA-eluted peptides from HTLV-transformed cell lines and are analyzed by LC-MS/MS. The co-elution and similarity in fragmentation profile of endogenous and heavy peptides are used to confirm the endogenous peptide identity.
- Example 4 Identification of HTLV-specific peptides from HLA-I immunopeptidomics of ATL patient samples.
- ATL adult T-cell leukemia patient samples procured.
- Peripheral blood mononuclear cells (PBMCs) were suspended in lysis buffer containing 1% NP-40 and incubated at 4°C for 1 hour.
- HLA class I complexes were then purified using a pan class I anti-HLA W6/32 antibody covalently conjugated to sepharose beads, followed by subsequent washing of the beads and elution of the HLA complexes in acidic solution (pH 2.7).
- a C18-based solid-phase extraction was utilized to separate the HLA proteins from the associated peptides, which were then further analyzed by liquid chromatography-mass spectrometry (LC-MS).
- LC-MS liquid chromatography-mass spectrometry
- HLA genotyping of ATL patient samples highlighted the diversity of HLA-I in the patient population with a high proportion of patients with HLA-A02 (12 patient samples), HLA-A11 (6 patient samples) and HLA-A24 (9 patient samples) alleles (Table 8). Table 8.
- the two unique HTLV peptides detected at the highest frequencies in the ATL patient samples were QSSSFIFHK (SEQ ID NO: 143) and EYTNIPISLL (SEQ ID NO: 144), as determined using analysis from the Bruker timsTOF SCP mass spectrometer (Table 10). Both peptides originated from the HTLV Tax protein and were detected in 2/6 HLA-A11 (33%) and 3/6 HLA-A24 (50%) of patient samples respectively.
- the HTLV peptides identified from the individual patient samples were matched with their respective HLA alleles using the panNetMHC 4.0 binding prediction algorithm to further enhance the confidence in peptide identifications.
- RNAseq-based approach was employed to generate a database of patient-specific HTLV genome sequences directly reconstituted from transcribed virus RNA sequences amplified from patient samples (Figure 9).
- Total human RNA reads were first converted into large contigs using a de novo sequencing approach, and contigs with Attorney Docket No.250298.000682 loose homology to the HTLV reference genome (NCBI Accession No. NC_001436.1) were marked as HTLV-specific sequences.
- Fully-mapped contigs to the HTLV reference genome were re-arranged in a reference-free fashion to assemble patient-specific HTLV genomic sequences. Using this approach, seven patient-specific HTLV genome sequences were reconstructed.
- Genome reconstruction was partial, ranging from 13% to 42% of the HTLV reference genome.
- the patient-specific viral sequence was then used to extract and translate all HTLV coding sequences.
- Non-canonical protein sequences with start codons embedded within canonical protein sequences were also identified in each sequence.
- the RNA sequence read coverage of the patient-specific reconstructed genomes is depicted in Figure 10. [00468]
- a summary of the RNAseq data of the present Example, including patient- specific reconstructed genome length and percentage (%) of the HTLV reference genome, is provided in Table 11. Table 11.
- RNA-seq reads were aligned to the HTLV reference genome (NCBI Accession No. NC_001436.1) using minimap2 (v2.17) (see, e.g., Li 2018). Alignments were then sorted by coordinates and quality controlled by samtools flagstats (v1.9) (see, e.g., Li et al., 2009) and bedtools genomeCoverageBed (v2.17.0) (see, e.g., Quinlan et al., 2010) inspection.
- RNAseq Data Paired-end Illumina RNA reads from each sample were de novo assembled into large contigs using megahit (see, e.g., Li et al., 2015) (options: --min-count 3 --k-min 27 --k-max 127 --prune-level 2) and mapped using BLAST to the HTLV reference genome (NCBI Accession No. NC_001436.1) to select HTLV-specific sequences.
- Contigs without BLAST matches were discarded, as well as BLAST results with E values greater than 0.001, percentage identity below 79%, or alignment length of less than 50 nucleotides. Overlapping contigs were merged using custom scripts and final sequences that covered the entire length or partial length of the reference genome sequence with the highest identity were selected.
- RNA or DNA is sequenced from a cancer patient’s tumor cell(s) and/or healthy tissue(s)/cell(s) to identify sequences which may contain HTLV-1-associated insertions in genes expressed in the tumor cell.
- Tumor material is assessed for HTLV-1-associated insertions by sequencing such as by RNA sequencing. Sequencing data are used to investigate HTLV- 1-associated insertions expressed in genes.
- HTLV-1 epitopes are determined by sequencing the genome and/or exome of tumor tissue and/or healthy tissue from a cancer patient using next generation sequencing (NGS) approaches.
- NGS next generation sequencing
- NGS applies to, without limitation, genome sequencing, genome resequencing, epigenome characterization, DNA-protein interactions (ChIP-sequencing), and transcriptome profiling (RNA-Seq). Similar to DNA-based assays using, e.g., NGS or massively parallel sequencing (MPS) approaches, RNA from tumors is analyzed by conversion to cDNA and generation of a library suitable for sequencing. [00476] Assays are employed to identify HTLV-1 epitopes in biological samples.
- Suitable assays used to identify HTLV-1 epitopes in biological samples include, but are not limited to, proteomics, NGS, solution hybridization, array hybridization nucleic amplification, polymerase chain reaction (PCR), RT-PCR, quantitative PCR, branched Attorney Docket No.250298.000682 DNA (bDNA) assay, rolling circle amplification (RCA), in situ hybridization, Northern hybridization, hybridization protection assay (HPA), single molecule hybridization detection, Invader assay, and/or Oligo Ligation Assay (OLA), hybridization, and array analysis.
- the sequencing data derived from determining the presence of HTLV-1 epitopes in a cancer patient is analyzed to predict personal HTLV-1 peptides that can bind to HLA molecules of the individual.
- the data are analyzed using a computer.
- the sequence data are analyzed, in particular, for the presence of HTLV-1 antigens.
- HTLV-1 antigens are assessed by their affinity to MHC molecules.
- Neural network-based learning approaches with validated binding and non- binding peptides are used in prediction algorithms for the major HLA-A and -B alleles. Algorithms are used for predicting missense mutations that create strong binding peptides to a cancer patient’s cognate MHC molecules.
- a peptide binding tool can be one of the following: Antibody Epitope Prediction, ANTIGENIC, BepiPred, CTLPred, DiscoTope, EPIPREDICT, Epitope Cluster Analysis, Epitope conserveancy Analysis, EUiPro, HLA Peptide Binding Predictions, HLABinding, MAPPP, MHCBench, MHC-I processing predictions, Mosaic Vaccine Tool Suite, NetChop, NetCTL, NetMHC, NetMHCII, NetMHCpan, nHLAPred- I, OptiTope, PAProC, POPI, PREDEP, Prediction of Antigenic Determinants, ProPred, ProPred-1, RankPep, SMM, SVMHC, T
- IEDB Immune Epitope Database and Analysis Resource
- ANN artificial neural networks
- NetMHC Longegaard C, et al. Nucleic Acids Res. 2008;36 (Web Server issue):W509-W512.
- Programs such as SMMPMBEC (Kim Y, et al.
- BMC Bio informatics.2009; 10:394) and/or SMM are used. These programs use position-weight matrices to describe statistical preferences from peptide-MHC I binding data. This approach suppresses noise caused, for example, by a limited number of data points present in the training set and/or experimental error.
- SNPs single nucleotide polymorphisms
- SNPs contain a range of molecular variation: (1) SNPs, (2) multinucleotide polymorphisms (MNPs), (3) short deletion and insertion polymorphisms (indels/DIPs), (4) micro satellite markers or short tandem repeats (STRs), (5) heterozygous sequences, and (6) named variants.
- MNPs multinucleotide polymorphisms
- Indels/DIPs short deletion and insertion polymorphisms
- STRs micro satellite markers or short tandem repeats
- heterozygous sequences eomic-based methods for identifying tumor specific HTLV-1 antigens such as direct protein sequencing are used. Protein sequencing of enzymatic digests using multidimensional MS techniques including tandem mass spectrometry (MS/MS) is used to identify HTLV-1 antigens.
- MS/MS tandem mass spectrometry
- High-throughput methods for de novo sequencing of unknown proteins is used, for example, to analyze the proteome of a cancer patient's tumor to identify expressed HTLV-1 antigens.
- meta-shotgun protein sequencing is used to identify expressed HTLV-1 antigens.
- Tumor specific HTLV-1 antigens are identified using MHC multimers to identify HTLV-1 antigen-specific T-cell responses.
- high-throughput analysis of HTLV-1 antigen- specific T-cell responses in cancer patient samples may be performed using MHC tetramer-based screening approaches.
- Such tetramer-based screening approaches are used for the identification of tumor specific HTLV-1 antigens, or as a secondary screening protocol to assess HTLV-1 antigens to which a patient may have already been exposed, which may support the selection of candidate HTLV-1 antigens.
- filters are applied to eliminate (1) epitopes with lower binding affinity than the corresponding wild-type sequences and/or (2) epitopes predicted to be poorly processed by the immunoproteasome.
- Candidate mutated peptides are synthesized and screened to identify T cell HTLV-1 antigens.
- Pulsing antigen presenting cells (APCs) with relatively long synthetic peptides that encompass minimal T cell epitopes is used to identify HTLV-1 epitopes.
- Nonsynonymous mutated epitopes are identified in tumors by evaluating the response of CD4+ tumor infiltrating lymphocytes (TILs) to autologous B cells that are pulsed with Attorney Docket No.250298.000682 peptides encompassing individual mutations. Use of this approach results in the identification of mutated cell epitopes.
- a peptide screening assay is performed based on the combination of two peptide libraries: (1) overlapping long-peptides (2) peptides according to MHC-binding prediction. Screening leads to identification of mutated HTLV- 1-reactive T cells. [00486]
- a tandem minigene screening approach is used to identify HTLV-1 epitopes.
- a tandem minigene construct comprised, for example, without limitation 6 to 24 minigenes that encoded polypeptides comprising a mutated amino acid residue flanked on the N- and/or C-terminus by, e.g., 12 amino acids. Tandem minigene constructs are synthesized and used to transfect autologous APCs and/or cell lines co-expressing autologous HLA molecules. Using this approach, HTLV-1 epitopes are identified in cancer patients, e.g., patients with adult T-cell leukemia/lymphoma (ATL) or HTLV-1- myelopathy/tropical spastic paraparesis (HAM/TSP).
- ATL adult T-cell leukemia/lymphoma
- HAM/TSP HTLV-1- myelopathy/tropical spastic paraparesis
- HTLV-1 epitopes are identified using an approach combining whole- exome/transcriptome sequencing analysis, MHC binding prediction, and mass spectrometric technique to detect peptides eluted from HLA molecules. Predicted high- binding peptides are confirmed by mass spectrometry. Example 7. Determination of MHC binding capacity. [00488] Candidate peptides according to the present disclosure are tested for their MHC binding capacity (affinity). The individual peptide-MHC (pMHC) complexes are produced by UV-ligand exchange. A UV-sensitive peptide is cleaved upon UV-irradiation, and exchanged with the peptide of interest.
- Peptide candidates that effectively bind and stabilize the peptide-receptive MHC molecules prevent dissociation of the MHC complexes.
- an ELISA is performed based on the detection of the light chain ( ⁇ 2m) of stabilized MHC complexes. Briefly, 96-well plates are coated with streptavidin, washed, and blocked. Refolded HLA-A monomers serve as standards, covering a pre-determined concentration range. Peptide-MHC monomers of the UV-exchange reaction are diluted in blocking buffer. Samples are incubated, washed, incubated with HRP conjugated anti- ⁇ 2m, washed again and detected with a chromogenic substrate solution that is stopped per the manufacturer’s protocol.
- HLA- heavy chain additionally comprises a C-terminal biotinylation tag which replaces, for example, the transmembrane and/or cytoplasmic domains.
- E. coli cells are lysed and inclusion bodies processed to approximately 80% purity.
- Inclusion bodies of ⁇ 2m and heavy chain are denatured separately in denaturation buffer. Refolding buffer is prepared. Synthetic peptides are dissolved to a final concentration and added to the refold buffer. Then ⁇ 2m followed by heavy chain are added. Refolding is performed to completion. [00492] The refold mixture is then dialyzed.
- the protein solution is subsequently filtered through a filter and loaded onto an exchange column (pre-equilibrated). Protein is eluted such as by way of a linear salt gradient using additional purifier. HLA-peptide complex is eluted, and peak fractions are collected. A cocktail of protease inhibitors is added and the fractions are chilled on ice.
- Biotin-tagged pHLA molecules are buffer exchanged into a buffer using a fast desalting column equilibrated in the same buffer. Upon elution, the protein-containing fractions are chilled on ice and protease inhibitor cocktail is added. Biotinylation reagents are then added. The mixture is then allowed to incubate.
- the biotinylated pHLA molecules are further purified, for example, by gel filtration chromatography using purifier with a column pre-equilibrated with filtered PBS.
- the biotinylated pHLA mixture is concentrated to a final volume, loaded onto the column and developed.
- Biotinylated pHLA molecules elute, e.g., as a single peak. Fractions containing protein are pooled, chilled on ice, and protease inhibitor cocktail is added. Protein concentration is determined and aliquots of biotinylated pHLA molecules are stored frozen.
- Such peptide-MHC (pMHC) complexes are used in soluble form or immobilized through their C-terminal biotin moiety on to a solid support, to be used for the detection of T cells and T cell receptors which bind the peptide-MHC complex.
- pMHC peptide-MHC
- Such complexes are used in panning phage libraries, performing ELISA assays and/or preparing sensor chips for measurements of affinity and binding kinetics.
- Example 9 Identification of T cell receptors (TCRs) that bind to pMHC complexes.
- Antigen binding T cell receptors are obtained using peptides disclosed herein to pan a TCR phage library.
- the library is constructed using ⁇ - and ⁇ - chain sequences obtained from a natural repertoire. The random combination of these ⁇ - and ⁇ - chain sequences occurs during library creation, thereby producing a non-natural repertoire of ⁇ / ⁇ chain combinations.
- TCRs obtained from the library are assessed by enzyme-linked immunoassay (ELISA) to confirm specific antigen recognition. ELISA plates are coated with streptavidin and incubated with the biotinylated peptide-HLA complex. TCR-bearing phage clones are added to each well and detection is carried out using an HRP antibody conjugate. Bound antibody is detected using a peroxidase Substrate System.
- TCRs can bind a peptide-HLA complex of the disclosure by surface plasmon resonance (SPR) using isolated TCRs.
- SPR surface plasmon resonance
- ⁇ - and ⁇ - chain sequences are expressed in E. coli as soluble TCRs.
- Binding of the soluble TCRs to the complexes is analyzed by surface plasmon resonance.
- Biotinylated peptide-HLA monomers are prepared and immobilized on a streptavidin-coupled sensor chip.
- TCRs that specifically recognize peptide-HLA complexes of the disclosure are obtained from the library. Data generated according to the above-described experiments confirm that antigen specific TCRs can be isolated. Attorney Docket No.250298.000682 Example 10. Characterization of binding to MHC and stability of pMHC complex. [00500] T2 cell-based peptide binding assay.
- T2 cells which do not express the transporter associated with antigen processing (TAP), and as such do not assemble stable MHC class I on the cell surface, are pulsed with different concentrations of peptides (controls or HTLV-1 peptide of interest [POI]), washed, detected with fluorescently-tagged antibody recognizing MHC class I (e.g., A2 allele), and run through a FACS Scan analyzer.
- the difference between the MFI (mean fluorescence intensity) corresponding to a given concentration of POI and the negative control (non-MHC binder) is a function of the number of stabilized pMHC complexes displayed on the cell surface.
- RA relative affinity
- RA the peptide concentration responsible for 50% of the signal corresponding to saturation
- Monomer-coated plates are first stripped, releasing the placeholder peptide and leaving only the MHC heavy chain bound to the plate.
- Test peptides are then introduced under optimal folding conditions, along with the anti-MHC-FITC mAb. Plates were incubated.
- the anti-MHC-FITC mAb binds preferentially to a refolded MHC complex.
- the fluorescence intensity resultant from each peptide is related to the peptide's capacity to complex with MHC molecule.
- Each Attorney Docket No.250298.000682 peptide's binding is evaluated relative to a positive control peptide, and the results are expressed, for example, as percent (%) binding.
- Affinity assay For the affinity assay, after the initial stripping of the placeholder peptide, increasing concentrations of POI are added to a series of wells and incubated under the conditions described previously. Plates are read on the fluorometer. Dose response curves are generated. The amount of peptide required to achieve 50% of the maximum is recorded as ED50 value.
- Off-Rate assay Plates are washed after incubation under conditions to remove excess peptide. The plates are then incubated on allele-specific monomer plates. The plates are measured at multiple time points (e.g., 0, 0.5, 1, 1.5, 2, 4, 6 and 8 hrs) for relative fluorescence intensity.
- iScore calculation an iScore is a multi-parameter calculation provided within the iTopia software. Its value is calculated based on the binding, affinity, and stability data. Example 11. Determination of responses against tumor cells. [00506] A suitable number of groups of mice are immunized with a plasmid expressing HTLV-1 peptides disclosed herein by direct inoculation.
- mice are inoculated into lymph nodes, e.g., inguinal lymph nodes, with plasmids at an appropriate concentration at day 0, and at subsequent days over the time course of the experiment, e.g., at days 3, 14, and 17. In certain cases, this is followed by one or more additional peptide boost(s) on following days, e.g., days 28 and 31, using a negative control peptide and POI.
- Splenocytes are stimulated ex vivo with POI and tested against Chromium-51 ( 51 Cr)--labeled tumor cells at various E:T ratios.
- Example 12 In vivo assessment of enhanced immunity against HTLV-1 peptides.
- mice are immunized with a plasmid expressing HTLV-1 peptides disclosed herein by direct inoculation.
- lymph nodes e.g., inguinal lymph nodes
- plasmids at an appropriate concentration at day 0, and at subsequent days over the time course of the experiment, e.g., at days 3, 14, and 17.
- this is followed by Attorney Docket No.250298.000682 one or more additional peptide boost(s) on following days, e.g., days 28 and 31, using a negative control peptide and POI.
- splenocytes were isolated from littermate control mice and incubated with one or more appropriate concentrations of POI for a pre-determined period of time. These cells were then stained with CFSEhi fluorescence and intravenously co-injected into immunized mice with an equal number of control splenocytes stained with CFSElo fluorescence. After a pre- determined period of time, the specific elimination of target cells was measured by removing spleen and PBMCs from challenged animals and measuring CFSE fluorescence by flow cytometry.
- POIs are used in in vitro for immunization of blood to generate cytotoxic T lymphocytes (CTLs).
- CTLs cytotoxic T lymphocytes
- PBMCs from normal donors are purified from buffy coats by centrifugation in standard sterile medium designed for isolating lymphocytes. Cultures are carried out using autologous plasma (AP).
- AP autologous plasma
- DCs autologous dendritic cells
- APCs antigen presenting cells
- DCs are generated and CTLs are induced with DCs and peptides from PBMCs.
- Monocyte-enriched cell fractions are cultured to induce maturation.
- Specific numbers of CD8+-enriched T lymphocytes and peptide-pulsed DCs are co-cultured. Cultures are restimulated on various days with autologous irradiated peptide-pulsed DCs. Immunogenicity is assayed using in vitro cytotoxicity and cytokine production assays.
- Example 14 CD8+ T cell responses against peptides. [00511] Based on the predicted or experimentally verified HLA-binding peptides, whether T cells can be generated to recognize the tumor-specific peptides is determined. Peptides with appropriate binding scores are synthesized.
- T cells are stimulated with peptide-pulsed (individual peptide or peptide pool) autologous APCs such as dendritic cells and/or CD40L-expanded autologous B cells on Attorney Docket No.250298.000682 a predetermined schedule, for example, in the presence of IL-2 and IL-7.
- peptide-pulsed APCs such as dendritic cells and/or CD40L-expanded autologous B cells on Attorney Docket No.250298.000682 a predetermined schedule, for example, in the presence of IL-2 and IL-7.
- the expanded CD8+ cells are tested on ELISpot for evidence of reactivity against the peptide based on IFN ⁇ secretion.
- Example 15 Cytokine production assay.
- cytokine e.g., IL-2 and IFN ⁇
- T cells are harvested after contact with the peptide-pulsed APC, centrifuged, and both cell pellets and supernatants are collected. Cytokine production is measured by ELISA from the supernatant.
- cytokine e.g., IL-2 and IFN ⁇
- MHC I molecules e.g., HLA-A2, HLA-A24, HLA-A11
- POI HTLV-1 viral peptide of interest
- OVA OVA 257-264 peptide
- a cohort of transgenic mice that express human HLA molecule(s) is initially injected with the MHC I molecule/peptide or MHC I molecule/OVA complexes and the level of effector and memory CD8+ T cells is monitored at different time points post- injection (from spleen and/or blood). Mock injected mice will serve as a negative control group in this experiment.
- a second cohort of mice are then injected with the MHC I molecule/POI complex and subsequently challenged using AAV-HTLV-1, at post- injection time points that will be determined according to the measurements of effector and memory CD8 + T cells levels from the first experiment.
- Control groups are constituted with (i) mice not injected with the MHC I molecule/POI complex but challenged with AAV-HTLV-1, (ii) mice injected with MHC I molecule/OVA complexes, and (iii) mice injected with MHC I molecule/POI complex but not challenged with AAV-HTLV-1.
- the viral burden is analyzed at different time points post-injection to determine MHC I molecule/POI limitation of viral infection from CD8+ T cells activation.
- Attorney Docket No.250298.000682 Example 17. In vivo activation of antigen-specific T cells by the viral peptide as a method to decrease viral production.
- HTLV-1 viral peptide of interest in the context of MHC I molecules (e.g., HLA-A2, HLA-A24, HLA-A11) to enhance a T cell response against HTLV-1 virus following AAV-HTLV-1 challenge is tested in transgenic mice that express human HLA molecule(s). More particularly, the in vivo activation of peptide-specific CD8+ T cells after (i) POI immunization, (ii) acute challenge with AAV-HTLV-1, and/or (iii) chronic challenge with AAV-HTLV-1, is compared.
- POI HTLV-1 viral peptide of interest
- MHC class I molecule(s) displaying the HTLV-1 viral POI or (ii) a control MHC class I molecule displaying the OVA 257-262 peptide (OVA) are generated.
- Transgenic mice that express human HLA molecule(s) are first challenged with AAV-HTLV-1.
- the MHC class I molecule/peptide or MHC class I molecule/OVA complexes produced for this example are then injected 1 day and 7 days after infection with the acute challenge, and 1 day, 7 days, and 14 days after the chronic challenge.
- the viral burden is analyzed at different time points post-peptide injection by blood collection and post-mortem organs collection.
- the levels of effector and memory CD8+ T cells are measured and compared to the control conditions to determine the capacity of MHC class I molecule/peptide to decrease viral production in response to an AAV-HTLV-1 challenge.
- Example 18 Enhancement of a peptide-specific CD8+ T cell immunity in a peptide- presenting tumor model.
- the ability of the HTLV-1 peptide of interest in the context of MHC I molecules to enhance a CD8+ T cell response against peptide-presenting tumors is tested in a general purpose strain mice, e.g., C57B1/6 mice, grafted with a peptide-expressing tumor cell line.
- the peptide-expressing tumor cell line is first grown in vitro, then injected (e.g., subcutaneously) into the mice. After tumor cell injection, when tumors are of palpable size, the HTLV-1 peptide, for example, in a specific groove of the MHC I or control peptide (e.g., ovalbumin [OVA] peptide) in an alternate groove of the MHC I molecule, are injected intratumorally. Tumor cell volume is measured. Tumor, blood, and/or spleen is collected, and homogenized in single cell suspensions, when applicable. Attorney Docket No.250298.000682 The level of CD8+ T cells in the homogenized samples is determined at all suitable time points.
- a specific groove of the MHC I or control peptide e.g., ovalbumin [OVA] peptide
- Splenocytes and antigen presenting cell primary cultures Spleens of adult general purpose strain mice, e.g., C57Bl/6 mice, are excised and placed in cold buffer. Tissues are then homogenized to break apart the spleens. Dissociated cells are centrifuged. After centrifugation, the cell pellet is resuspended in a suitable volume of lysis buffer designed to remove red blood cells, e.g., ACK (Ammonium-Chloride-Potassium) lysis buffer, and incubated in the buffer. After incubation the cell suspension is added to buffer, and centrifuged.
- ACK Ammonium-Chloride-Potassium
- APC global antigen-presenting cells
- Peptide-pulsed APC are harvested and washed prior to addition to any of various suitable T cell lines (e.g., J.RT3-T3.5 derived cells), after infection with the viral particles.
- the T cells are co-cultured with the pulsed APC and subsequently used in proliferation, luciferase and/or cytokines production assays.
- T cell activation post transduction Alternatively, to co-culture with peptides-pulsed APCs, suitable T cell lines (e.g., J.RT3-T3.5-derived cell lines) are activated after transduction with either phytohemagglutinin (PHA,), phorbol 12-myristate 13-acetate (PMA), or a combination of PHA and PMA.
- PHA phytohemagglutinin
- PMA phorbol 12-myristate 13-acetate
- Another alternative method to activate T cells after transduction is to use soluble or immobilized antibodies such as, but not limited to anti-CD3 and/or anti-CD28 monoclonal antibodies. Soluble antibodies are added at an appropriate concentration. In some experiments, plates pre-coated with anti-CD3 antibodies, for example, are used in combination with soluble anti-CD28 antibody. Attorney Docket No.250298.000682 [00526] Another alternative method to activate T cell lines is activating beads coupled with anti-CD3/anti-CD28 antibodies. After transduction, infected cells are counted, and anti-CD3/anti-CD28 beads are added to the culture medium at a given ratio. [00527] Cells staining and FACS analysis.
- Fluorescence-activated cell sorting is performed after the transduction. Transduced cells are counted and seeded into a cell culture plate. Cells are spun, washed, and then spun again any number of times. Cells are incubated with a dye for determining viability of cells, e.g., Live/Dead TM Fixable Near- IR stain, washed, and incubated with Fc block in buffer, e.g., FACS stain buffer. After being washed any number of times again with FACS stain buffer, cells are subsequently incubated with antibodies such as, but not limited to, fluorescent-labeled antibodies targeting TCR and control.
- FACS Fluorescence-activated cell sorting
- cytokines production assay For transcription factor activity analysis and cytokines production assay, cells are harvested after contact with the peptide-pulsed APC, centrifuged, and cell pellets and supernatants are collected. [00530] Cell pellets are processed for RNA extraction for transcriptomics analysis via qPCR. Cytokine production is measured by ELISA from the supernatant. Cell pellets are processed for luciferase detection assay to measure programmed cell death, e.g., AP1 activity. [00531] Splenocytes isolation and CD8+T cell culture. Spleens of adult general purpose strain mice, e.g., C57Bl/6 mice, are excised and placed in cold buffer.
- Tissues are then homogenized to break apart the spleens.
- Dissociated cells are centrifuged. After centrifugation, the cell pellet is resuspended in a suitable volume of lysis buffer designed to remove red blood cells, e.g., ACK (Ammonium-Chloride-Potassium) lysis buffer, and incubated in the buffer. After incubation, the cell suspension is added to buffer, and centrifuged. The cell pellet is then resuspended in buffer and the cell solution is filtered. Attorney Docket No.250298.000682 The filtered cell suspension is centrifuged again and the subsequent pellet is resuspended in a suitable volume of buffer.
- ACK Ammonium-Chloride-Potassium
- CD8+ T cells are then isolated such as, for example, by using a CD8a+ T cell Isolation Kit.
- Peptide immunization of mice For immunization, a suitable amount of HTLV-1 peptide or control peptide is diluted in buffer and emulsified at a given ratio with an adjuvant such as, but not limited to, complete Freund’s adjuvant (CFA) or incomplete Freund’s adjuvant (IFA) using, for example, a double syringes system, to reach a final desired volume of peptide/adjuvant emulsion.
- CFA complete Freund’s adjuvant
- IFA incomplete Freund’s adjuvant
- a measured volumed (e.g., 200 ⁇ l) of the peptide/adjuvant emulsion is then injected by a given route of administration, for example, subcutaneously, into mice (e.g., C57Bl/6 mice) under appropriate anesthesia in appropriate location(s).
- a given route of administration for example, subcutaneously
- mice e.g., C57Bl/6 mice
- Quantification of cytotoxicity activity by flow cytometry Quantification of cytotoxicity activity by flow cytometry.
- Preparation of target cells For preparation of target cells (e.g., autologous B cells), the target cells are isolated from splenocytes of OT-1 or P14 mice by using a Mouse B cell Kit (or the like, according to the manufacturer's instruction), and are subsequently stimulated for a predetermined duration in the presence of a set concentration of IFN- ⁇ .
- Targets cells are then counted, divided into tubes, and washed in buffer. A proportion of the target cells are stained with a high concentration (e.g., 0.2 ⁇ M) of CFSE (CFSEHigh) and a separate proportion with a low concentration (e.g, 0.02 ⁇ M) of CFSE (CFSELow) in buffer. After the incubation, cells are pelleted and resuspended in an appropriate media to quench the labeling reaction. Target cells stained with the low concentration of CFSE are pulsed by adding the HTLV-1 peptides or control OVA peptides at a suitable final concentration under appropriate culture conditions.
- CFSEHigh a high concentration of e.g., 0.2 ⁇ M of CFSE
- CFSELow a separate proportion with a low concentration of CFSE
- Effector cells Total CD8+ T cells containing the effector cells are enriched from splenocytes of P14 and OT-1 mice using a Negative Selection Human CD8 T cell isolation Kit. CD8+ T cells are counted, resuspended in complete T cell medium, and serially diluted volume:volume in a given volume of complete T cell medium. From each dilution, a given volume of cells are seeded in duplicate to multi-well cell culture plate.
- a therapeutic DNA or RNA vaccine comprising polynucleotides or vectors encoding polynucleotides to be used is prepared by GMP manufacturing of the plasmid vaccine according to regulatory authorities' guidelines.
- the vaccine is appropriately formulated, for example, by dissolving in a saline solution, at a suitable concentration.
- the vaccine may be administered either intradermal or intramuscular with or without following electroporation or alternatively with a jet injector. * * * [00538]
- the claimed subject matter is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the claimed subject matter in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims. [00539] All patents, applications, publications, test methods, literature, and other materials cited herein are hereby incorporated by reference in their entirety as if physically present in this specification.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Immunology (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Virology (AREA)
- Biophysics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Engineering & Computer Science (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Microbiology (AREA)
- Biomedical Technology (AREA)
- Cell Biology (AREA)
- Hematology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Tropical Medicine & Parasitology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Oncology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Urology & Nephrology (AREA)
- Wood Science & Technology (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Communicable Diseases (AREA)
- Food Science & Technology (AREA)
Abstract
The present disclosure provides isolated peptides derived from human T-lymphotropic virus type-1 (HTLV-1), peptide-based molecules (e.g., peptide-MHC (pMHC) complexes), polynucleotides and vectors encoding the peptides or peptide-based molecules, pharmaceutical compositions (e.g., vaccine compositions), and their use for treatment, prevention, or reduction of the likelihood of HTLV-1 infection and/or HTLV-1 -induced diseases. The present disclosure also provides binding moieties that bind to the peptides or peptide-based molecules disclosed herein, and their use for treatment, prevention, or reduction of the likelihood of HTLV-1 infection and/or HTLV-1 -induced diseases. The present disclosure further provides methods and systems for identifying immunogenic virus-derived peptides.
Description
Attorney Docket No.250298.000682 VIRAL PEPTIDES AND USES THEREOF CROSS-REFERENCE TO RELATED APPLICATION [0001] This patent application claims the benefit of U.S. Provisional Application No. 63/583,834, filed September 19, 2023, the disclosure of which is incorporated by reference herein in its entirety for all purposes. SEQUENCE LISTING [0002] The instant application contains a Sequence Listing which has been submitted electronically in XML file format and is hereby incorporated by reference in its entirety. Said XML copy, created on August 29, 2024, is named 250298_000682_SL.xml and is 126,068 bytes in size. TECHNICAL FIELD [0003] The present disclosure relates to methods and compositions that involve isolated peptides derived from human T-lymphotropic virus type-1 (HTLV-1), and the use of such methods and compositions for treating, preventing, or reducing the likelihood of HTLV-1 infection and/or HTLV-1-induced diseases. BACKGROUND [0004] Human T-lymphotropic virus type-1 (HTLV-1) is a complex retrovirus which belongs to the Deltaretrovirus genus, infecting an estimated 5-10 million individuals worldwide. Of the four known types of HTLV (HTLV-1, HTLV-2 HTLV-3, and HTLV- 4), HTLV-1 has been identified as the most pathogenic and is the primary etiological agent of a malignant lymphoproliferation called adult T-cell leukemia/lymphoma (ATL), as well as an array of neurological pathologies known as HTLV-1-myelopathy/tropical spastic paraparesis (HAM/TSP). HTLV-1 is endemic in areas of Southwestern Japan, sub-Saharan Africa, South America, and the Caribbean, with foci in the Middle-East and Australo- Melanosia. HTLV-1 is predominately found in CD4+ T lymphocytes and can be spread by, e.g., sexual transmission, contaminated blood products, and mother-to-child transmission (parental/vertical). Further, HTLV-1 is a latent virus, and as such is unable to
Attorney Docket No.250298.000682 be cleared from the host’s immune system, where 4-5% of HTLV-1 carriers develop ATL in their life-time and another 0.25-4% develop HAM/TSP. Despite scientific advances toward understanding the pathogenic progression of HTLV-1 infection, the prognosis of ATL, as well as the quality of life for those suffering from HAM/TSP, remains poor. There is no vaccine to prevent HTLV-1 infection or HTLV-1-associated disease. Accordingly, identifying HTLV-1-associated epitopes that can be amenable to targeting such as by therapeutic vaccines, immunotherapy, or other therapeutics that target these HTLV-1- associated antigens would be useful to combat HTLV-1-mediated infections and diseases. SUMMARY [0005] As specified in the Background section above, there is a need in the art for development of methods to identify HTLV-1 epitopes that could be helpful for patients infected with HTLV-1. The present application addresses these and other needs. [0006] In one aspect, provided herein is an isolated peptide comprising an amino acid sequence that is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof, wherein the isolated peptide is 5-20 amino acids in length. [0007] In some embodiments, the isolated peptide comprises an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. [0008] In some embodiments, the isolated peptide consists essentially of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. [0009] In some embodiments, the isolated peptide consists of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. [0010] In another aspect, provided herein is an isolated peptide comprising two or more amino acid sequences selected from any one of SEQ ID NO: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof. [0011] In some embodiments, the isolated peptide comprises one or more reverse peptide bonds, one or more non-peptide bonds, one or more D-isomers of amino acids, one or more chemical modifications, or any combination thereof. [0012] In some embodiments, the isolated peptide is produced by expression in a heterologous host cell.
Attorney Docket No.250298.000682 [0013] In some embodiments, the isolated peptide is produced synthetically. [0014] In some embodiments, the isolated peptide, or pharmaceutically acceptable salt thereof, or fragment or derivative thereof induces a Human T-Lymphotropic Virus Type-1 (HTLV-1)-specific immune response in a subject when presented in a complex with a major histocompatibility complex (MHC) molecule on the surface of an antigen presenting cell (APC). [0015] In another aspect, provided herein is a fusion protein comprising one or more isolated peptides described herein fused to one or more heterologous molecules. [0016] In some embodiments, the one or more heterologous molecules enhance a peptide- specific immune response in a subject. [0017] In some embodiments, the one or more heterologous molecules mediate peptide delivery to a specific site within a subject. [0018] In some embodiments, the one or more heterologous molecules are a MHC molecule, or a fragment or derivative thereof. [0019] In another aspect, provided herein is a conjugate comprising one or more isolated peptides described herein conjugated to one or more heterologous molecules. [0020] In some embodiments, the one or more heterologous molecules enhance a peptide- specific immune response in a subject. [0021] In some embodiments, the one or more heterologous molecules mediate peptide delivery to a specific site within a subject. [0022] In some embodiments, the one or more heterologous molecules are an MHC molecule, or a fragment or derivative thereof. [0023] In some embodiments, the one or more peptides are conjugated to a particle. [0024] In another aspect, provided herein is an oligomeric complex comprising two or more isolated peptides described herein. [0025] In another aspect, provided herein is a non-covalent complex comprising an isolated peptide described herein and an MHC molecule, or a fragment or derivative thereof. [0026] In some embodiments, the MHC molecule, or the fragment thereof, is a class I MHC molecule.
Attorney Docket No.250298.000682 [0027] In some embodiments, the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule. [0028] In some embodiments, the MHC molecule, or the fragment thereof, is a class II MHC molecule. [0029] In some embodiments, the class II MHC molecule is a class II HLA molecule. [0030] In another aspect, provided herein is a fusion protein comprising an isolated peptide described herein and an MHC molecule, or a fragment or derivative thereof. [0031] In some embodiments, the MHC molecule, or the fragment thereof, is a class I MHC molecule. [0032] In some embodiments, the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule. [0033] In some embodiments, the MHC molecule, or the fragment thereof, is a class II MHC molecule. [0034] In some embodiments, the class II MHC molecule is a class II HLA molecule. [0035] In another aspect, provided herein is a conjugate comprising an isolated peptide described herein and a MHC molecule, or a fragment or derivative thereof. [0036] In some embodiments, the MHC molecule, or the fragment thereof, is a class I MHC molecule. [0037] In some embodiments, the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule. [0038] In some embodiments, the MHC molecule, or the fragment thereof, is a class II MHC molecule. [0039] In some embodiments, the class II MHC molecule is a class II HLA molecule. [0040] In another aspect, provided herein is a pharmaceutical composition comprising (i) one or more isolated peptides described herein, one or more fusion proteins described herein, one or more conjugates described herein, one or more oligomeric complexes described herein, or one or more non-covalent complexes described herein, or any combination thereof; and (ii) a pharmaceutically acceptable carrier or excipient. [0041] In some embodiments, the pharmaceutical composition can further comprise an adjuvant.
Attorney Docket No.250298.000682 [0042] In another aspect, provided herein is an isolated molecule that binds an isolated peptide described herein, a fusion protein described herein, a conjugate described herein, an oligomeric complex described herein, or a non-covalent complex described herein. [0043] In some embodiments, the molecule is an antibody or an antigen-binding fragment thereof. [0044] In some embodiments, the antibody is a bispecific antibody. [0045] In some embodiments, the molecule is an alternative scaffold. [0046] In some embodiments, the molecule is a chimeric antigen receptor (CAR). [0047] In some embodiments, the molecule is a T cell receptor (TCR). [0048] In another aspect, provided herein is an isolated cell comprising a CAR described herein. [0049] In some embodiments, the isolated cell is an immune cell. [0050] In some embodiments, the immune cell is a T cell, an NK cell, or a macrophage. [0051] In another aspect, provided herein is an isolated cell comprising a TCR described herein. [0052] In some embodiments, the isolated cell is an immune cell. [0053] In some embodiments, the immune cell is a T cell, an NK cell, or a macrophage. [0054] In another aspect, provided herein is a pharmaceutical composition comprising (i) an isolated molecule described herein, or an isolated cell described herein; and (ii) a pharmaceutically acceptable carrier or excipient. [0055] In another aspect, provided herein is an isolated polynucleotide comprising a nucleotide sequence encoding one or more isolated peptides described herein or a fusion protein described herein. [0056] In some embodiments, the nucleotide sequence can be operably linked to a promoter. [0057] In some embodiments, the isolated polynucleotide comprises DNA. [0058] In some embodiments, the isolated polynucleotide comprises RNA. [0059] In some embodiments, the RNA is mRNA. [0060] In some embodiments, the RNA is self-replicating RNA. [0061] In another aspect, provided herein is a vector comprising an isolated polynucleotide described herein.
Attorney Docket No.250298.000682 [0062] In some embodiments, the vector is an expression vector. [0063] In some embodiments, the vector is a viral vector. [0064] In another aspect, provided herein is a host cell comprising an isolated polynucleotide described herein or a vector described herein. [0065] In some embodiments, the host cell is a prokaryotic cell. [0066] In some embodiments, the host cell is a eukaryotic cell. [0067] In some embodiments, the host cell is an APC. [0068] In another aspect, provided herein is a pharmaceutical composition comprising (i) an isolated polynucleotide described herein, or a vector described herein; and (ii) a pharmaceutically acceptable carrier or excipient. [0069] In some embodiments, the pharmaceutically acceptable carrier is a lipid nanoparticle carrier. [0070] In another aspect, provided herein is a method of inducing an immune response against HTLV-1 infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of: a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) a oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein. [0071] In another aspect, provided herein is a method of inducing an immune response against HTLV-1 infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of one or more isolated peptides described herein. [0072] In another aspect, provided herein is a method of inducing an immune response against a HTLV-1infection in a subject in need thereof, comprising administering to the
Attorney Docket No.250298.000682 subject an activated T cell that is produced by contacting a T cell with an APC that presents an isolated peptide described herein in complex with an MHC molecule. [0073] In another aspect, provided herein is a method of treating a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of: a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) an oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein. [0074] In another aspect, provided herein is a method of preventing or reducing the likelihood of a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of: a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) an oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein. [0075] In another aspect, provided herein is a method of treating a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of one or more isolated peptides described herein.
Attorney Docket No.250298.000682 [0076] In another aspect, provided herein is a method of preventing or reducing the likelihood of a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of one or more isolated peptides described herein. [0077] In some embodiments, the HTLV-1-induced disease or disorder is adult T-cell leukemia/lymphoma (ATL) or HTLV-1-myelopathy/tropical spastic paraparesis (HAM/TSP). [0078] In another aspect, provided herein is a kit comprising: (i) a) one or more isolated peptides described herein; b) a fusion protein described herein; c) a conjugate described herein; d) an oligomeric complex described herein; e) a non-covalent complex described herein; f) a pharmaceutical composition described herein; g) a molecule described herein; h) an isolated cell described herein; i) an isolated polynucleotide described herein; or j) a vector described herein; and (ii) packaging and/or instructions for use for the same. [0079] These and other aspects of the present invention will be apparent to those of ordinary skill in the art in the following description, claims and drawings. BRIEF DESCRIPTION OF THE DRAWINGS [0080] Figures 1A-1E illustrate affinity purification mass spectrometry for HTLV- transformed T-lymphoblast cell lines. An example of an anti-HLA-I W6/32 affinity purification workflow and subsequent peptide identifications by mass spectrometry is illustrated in Figure 1A. A description of peptide lengths and counts detected by mass spectrometry analysis for cell lines C8166 (Figure 1B), MT-2 (Figure 1C), MT-4 (Figure 1D), and C5/MJ (Figure 1E) is shown in Figures 1B-1E, respectively. [0081] Figure 2 shows that eleven (11) HTLV peptides were detected from the C8166 cell line with good reproducibility, six (6) of which were predicted HLA binders to known
Attorney Docket No.250298.000682 C8166 HLA alleles by NetMHCpan 4.0 (see, e.g., Table 3). Figure discloses SEQ ID NOs: 85, 63, 74, 83, 78, 81, 51, 24, 29, 70 and 31, respectively, in order of columns. [0082] Figure 3 shows that thirty-two (32) HTLV peptides were detected from the MT-2 cell line, twenty-one (21) of which were found in all three replicates, and a majority of which were predicted binders to known MT-2 alleles by NetMHCpan 4.0 (see, e.g., Table 4). Figure discloses SEQ ID NOs: 55, 46, 86, 80, 21 and 5, respectively, in order of appearance and SEQ ID NOs: 84, 89, 88, 87, 72, 11, 9, 52, 67, 20, 44, 51, 57, 4, 45, 17, 60, 62, 54, 35, 77, 49, 76, 71, 82, and 73, respectively, in order of columns. [0083] Figure 4 shows that nineteen (19) HTLV peptides were detected from the MT-4 cell line, with sixteen (16) peptides overlapping between replicates. NetMHCpan 4.0 predicted a few of these peptides to be binders to common HLA alleles, e.g., HLA-A02, HLA-A11, and HLA-A24, in the patient population (see, e.g., Table 5). Figure discloses SEQ ID NOs: 2, 1, 65, 66, 70, 68, 18, 56, 41, 64, 59, 47, 27, 31, 26, 42, 75, 15, and 53, respectively, in order of columns. [0084] Figure 5 shows that HLA-I immunopeptidomics from C5/MJ cells with interferon (IFN) and without IFN (no IFN) treatment yielded nine (9) overlapping HTLV peptides, all of which were predicted binders to C5/MJ HLA alleles by NetMHCpan 4.0 (see, e.g., Table 6). Figure discloses SEQ ID NOs: 79, 50, 20, 58, 64, 36, 69, 9, 38, 8, 14, 89, and 37, respectively, in order of columns. [0085] Figures 6A-6E show that sixteen (16) and twenty-one (21) HTLV peptides were detected from no IFN- and IFN-treated ED-41214(-) primary cells by mass spectrometry, with good reproducibility among biological replicates (see, e.g., Figure 6B and Figure 6D, respectively). Fifteen (15) HTLV peptides were shared between no IFN- and IFN-treated ED-41214(-) cells (Figure 6E). Only a few HTLV peptides were predicted binders to the common alleles by NetMHCpan 4.0 (see, e.g., Table 7). Descriptions of peptide lengths and counts detected by mass spectrometry analysis for no IFN- and IFN-treated ED- 41214(-) cells are shown in Figure 6A and Figure 6C, respectively. Figure discloses SEQ ID NOs: 32, 7, 3, 25, 30, 52, 33, 61, 39, 19, 6, 28, 40, 23, 10, 43, 48, 32, 3, 7, 30, 34, 13, 52, 12, 33, 61, 39, 6, 19, 28, 40, 23, 10, 43, 16, and 22, respectively, order of appearance. [0086] Figures 7A-7D illustrate peptide distributions were obtained for all HTLV primary samples tested ((see, e.g., Figure 6A for ED41214(-), no IFN 1; ED40515(-), no IFN 1
Attorney Docket No.250298.000682 (Figure 7A); ED40515(+), no IFN 1 (Figure 7B); ATL43T(+), no IFN 1 (Figure 7C); and ATL43Tb(-) IFN 1 (Figure 7E)); however, HTLV peptides were detected only for ED- 41214(-) (see, e.g., Figure 6A) (combined search against human UniProt + HTLV sequence). [0087] Figures 8A-8D show examples of peptide distributions from HLA-I peptidome analysis of ATL patient samples by Thermo Orbitrap (Figures 8A-8B) and Bruker timsTOF SCP (Figures 8C-8D) mass spectrometers. [0088] Figures 9A-9B show an example of a workflow used to reconstruct patient-specific HTLV genomes from RNA sequencing. RNA sequencing and RNA contig reconstruction steps are illustrated in Figure 9A. HTLV genome reconstruction, including BLAST analysis against the virus reference genome, viral genome reconstruction, and coding sequence extraction steps, are illustrated in Figure 9B. [0089] Figure 10 shows RNA sequencing read coverage of patient-specific reconstructed genomes. DETAILED DESCRIPTION [0090] The present disclosure provides, among other things, isolated peptides derived from human T-lymphotropic virus type-1 (HTLV-1), and fragments or derivatives thereof. Various peptide-based molecules including complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, and conjugates comprising the peptides are also provided. Further provided herein are polynucleotides and vectors encoding the peptides or peptide- based molecules described herein. Binding moieties (e.g., antibodies, alternative scaffolds, T-cell receptors (TCRs) or chimeric antigen receptors (CARs)) that bind to the peptides or peptide-based molecules are also provided. The compositions of the present disclosure can be used to induce an immune response against HTLV infection (e.g., HTLV-1, HTLV-2, HTLV-3, and/or HTLV-4 infection) and/or for treating, preventing, and/or reducing the likelihood of an HTLV-induced disease or disorder (e.g., an HTLV-1-, HTLV-2-, HTLV- 3-, and/or HTLV-4-induced disease or disorder). In one aspect, the present disclosure also provides methods for identifying an immunogenic virus-derived peptide. [0091] Lymphocytes, such as T cells, play important roles in adaptive anti-infection, antitumor, autoimmune, and transplant rejection responses. Generally, a T cell mediated
Attorney Docket No.250298.000682 immune response involves close contact, e.g., an immunological synapse, between a T cell and an antigen presenting cell (APC). The pairing of several molecules is involved in the formation of the immunological synapse, including, but not limited to: (a) a T-cell receptor (TCR) on a T cell, which specifically binds to a peptide presented in the peptide binding groove of a major histocompatibility complex (MHC) molecule on an APC; and (b) CD28 (on the T cell), which pairs with a B7 molecule on the APC. A TCR, together with CD3 molecules, form a TCR complex, and upon pairing of the TCR to the peptide-MHC (pMHC) complex, a signal is sent through CD3. Signaling through both the TCR complex and CD28 on the T cell results in activation of the T cell. [0092] T cell receptors are heterodimeric structures composed of two types of chains (an ^ (alpha) and ^ (beta) chain, or a ^ (gamma) and ^ (delta) chain). The ^ chain is encoded by the nucleic acid sequence located within the ^ locus (on human or mouse chromosome 14), which also encompasses the entire ^ locus that encodes the δ chain, and the ^ chain is encoded by the nucleic acid sequence located within the ^ locus (on mouse chromosome 6 or human chromosome 7). The majority of T cells have an ^ ^ TCR, while a minority of T cells bear a ^ ^ TCR. T cell receptor ^ and ^ polypeptides (and similarly ^ and ^ polypeptides) are linked to each other via a disulfide bond. Each of the two polypeptides that make up the TCR contains an extracellular domain comprising constant and variable regions, a transmembrane domain, and a cytoplasmic tail (the transmembrane domain and the cytoplasmic tail also being a part of the constant region). [0093] The variable region of each TCR comprises a unique and characteristic structure, i.e., an idiotope or idiotype, that determines the specificity of the TCR. Generally, a TCR will bind to a pMHC complex only if the TCR comprises an idiotype that recognizes the peptide being presented in the context of MHC, e.g., the unique conformation of a particular pMHC complex. [0094] Immunotherapeutic approaches to treating disease work to regulate T cell activity in vivo, e.g., to enhance anti-infection and anti-tumor responses, or, for example, to downregulate autoimmune and transplant rejection responses. However, such methods can lack specificity since immunotherapies can target signaling by the TCR complex by binding CD3 and/or the pairing of costimulatory molecules. Such approaches can result in undesirable side effects, e.g., a hyperactive immune response or generalized immune
Attorney Docket No.250298.000682 suppression. Accordingly, therapies that take advantage of the uniquely specific interaction between a TCR and pMHC complex may provide the ability to specifically modulate the activity of specific T cells in vivo, and provide treatments based on T cell modulation. [0095] The presentation of virus-derived peptides by MHC molecules on the surface of an infected cell and the recognition of these pMHC complexes by, and subsequent activation of, CD8+ cytotoxic T cells provides an important mechanism for immunity-based protection against viruses. Cells infected with HTLV-1, including those that developed into cancer cells, can express various HTLV-1-associated antigens. Peptides derived from these antigens may be displayed on the cell surface in complex with MHC molecules. Detection of an MHC-presented HTLV-1-derived peptide by a T cell bearing the corresponding TCR, leads to targeted killing of the infected cell. However, because of the selection processes which occur during T cell maturation in the thymus, there is often a scarcity of T cells in the circulating repertoire, which recognize HTLV-1-derived peptides with a sufficiently high level of affinity. As a result, infected cells often escape elimination by the immune system. [0096] The identification of HTLV-1-derived peptides presented on infected cells (e.g., HTLV-1-induced cancer cells) can allow for the development of immunotherapeutic reagents designed to specifically target and destroy HTLV-1-infected cells (e.g., HTLV-1- induced cancer cells). Such reagents may be moieties that bind to the HTLV-1-derived peptide and/or pMHC complexes and the reagents, e.g., moieties that can function by inducing a T cell response. For example, such reagents may be based on antibodies, TCRs, and/or CARs. [0097] The present disclosure, in part, is based on a proteogenomic approach that detects MHC-associated HTLV-1 peptides from HTLV-1 infected cells. The repertoire of HLA-I- HTLV-1 peptides that are expressed in HTLV-1-transformed cells and ATL primary cells characterized herein can provide an accurate representation of HTLV-1 epitopes in a population. [0098] HLA-restricted viral peptides as potential targets may be leveraged for delivering immunotherapeutics (such as antibodies (e.g., bispecific antibodies), engineered TCR- or CAR- based cellular therapies) to infected tissues. However, genomic variability between strains of a virus in combination with differences in patient HLA alleles can be a challenge
Attorney Docket No.250298.000682 in developing therapeutics against these peptide targets. To address this challenge, in one aspect the present disclosure provides a proteogenomics approach for generating patient- specific databases that allow for the comprehensive identification of viral peptides, e.g., HTLV-1-derived peptides, based on the viral transcriptomes sequenced from individual patient samples. The HTLV-1-HLA-associated peptides disclosed herein may be used in the development of immunotherapeutics (such as antibodies (e.g., bispecific antibodies), engineered TCR- or CAR- based cellular therapies) for the treatment of HTLV-1-related diseases or disorders including T-cell leukemia/lymphoma (ATL), HTLV-1- myelopathy/tropical spastic paraparesis (HAM/TSP), and hypersensitivity reactions, e.g., arthritis, uveitis, and HTLV-1-associated infective dermatitis (IDH). The proteogenomic discovery platform described herein provides a method for identifying viral-derived peptides as targets for anti-viral related immunotherapy. Definitions [0099] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. [00100] Singular forms “a”, “an”, and “the” include plural references unless the context clearly dictates otherwise. Thus, for example, a reference to “a method” includes one or more methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure. [00101] The term “about” or “approximately” includes being within a statistically meaningful range of a value. Such a range can be within an order of magnitude, preferably within 50%, more preferably within 20%, still more preferably within 10%, and even more preferably within 5% of a given value or range. The allowable variation encompassed by the term “about” or “approximately” depends on the particular system under study, and can be readily appreciated by one of ordinary skill in the art. [00102] The term “antigen” encompasses any agent (e.g., protein, peptide, polysaccharide, glycoprotein, glycolipid, nucleotide, portions thereof, or combinations thereof) that, when introduced into an immunocompetent host (directly or upon expression as in, e.g., DNA or RNA vaccines) is recognized by the immune system of the host and is
Attorney Docket No.250298.000682 capable of eliciting an immune response by the host. The T-cell receptor (TCR) recognizes a peptide presented in the context of a major histocompatibility complex (MHC) as part of an immunological synapse. The peptide-MHC (pMHC) complex is recognized by TCR, with the peptide (antigenic determinant) and the TCR idiotype providing the specificity of the interaction. Accordingly, the term “antigen” encompasses peptides presented in the context of MHCs, e.g., pMHC complexes. The peptide displayed on MHC may also be referred to as an “epitope” or an “antigenic determinant”. The terms “peptide,” “antigenic determinant,” “epitopes,” etc., encompass not only those presented naturally by antigen- presenting cells (APCs), but may be any desired peptide so long as it is recognized by an immune cell, e.g., when presented appropriately to the cells of an immune system. For example, a peptide having an artificially prepared amino acid sequence may also be used as the epitope. [00103] A single antigen (such as an antigenic polypeptide) may have more than one epitope. Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of structural epitopes and are defined as those residues that directly contribute to the affinity of the interaction between an MHC molecule and the antigen. Epitopes may also be conformational, that is, composed of non-linear amino acids. In certain embodiments, epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents. [00104] The terms “major histocompatibility complex,” and “MHC” encompass the terms “human leukocyte antigen” or “HLA” (the latter two of which are generally reserved for human MHC molecules), naturally occurring MHC molecules (e.g., MHC class I molecule comprising MHC class I α (heavy) chain and β2 microglobulin; MHC class II molecule comprising MHC class II α chain and MHC class II β chain), individual chains of MHC molecules (e.g., MHC class I α (heavy) chain, MHC class II α chain, and MHC class II β chain), individual subunits of such chains of MHC molecules (e.g., α1, α2, and/or α3 subunits of MHC class I α chain, α1-α2 subunits of MHC class II α chain, β1-β2 subunits
Attorney Docket No.250298.000682 of MHC class II β chain) as well as portions (e.g., the peptide-binding portions, e.g., the peptide-binding grooves), mutants and various derivatives thereof (including fusions proteins), wherein such portion, mutants and derivatives retain the ability to display an antigenic peptide for recognition by a TCR, e.g., an antigen-specific TCR. An MHC class I molecule comprises a peptide binding groove formed by the α1 and α2 domains of the heavy a chain that can stow a peptide of around 8-10 amino acids. Despite the fact that both classes of MHC bind a core of about 9 amino acids (e.g., 5 to 17 amino acids) within peptides, the open-ended nature of MHC class II peptide binding groove (the α1 domain of a class II MHC α polypeptide in association with the β1 domain of a class II MHC β polypeptide) allows for a wider range of peptide lengths. Peptides binding MHC class II usually vary between 13 and 17 amino acids in length, though shorter or longer lengths are not uncommon. As a result, peptides may shift within the MHC class II peptide binding groove, changing which 9-mer sits directly within the groove at any given time. In some embodiments, the peptide-MHC complex described herein may be a peptide-MHC complex from a non-human animal. In other embodiments, the peptide-MHC complex described herein may include an peptide-HLA complex, i.e., a peptide-MHC complex from a human. Conventional identifications of particular MHC variants are used herein. For example, HLA-A11 refers to a human leucocyte antigen from the A gene group (hence a class I type MHC) gene position (known as a gene locus) number 11; gene HLA-DR11, refers to a human leucocyte antigen coded by a gene from the DR region (hence a class II type MHC) locus number 11. [00105] “MHC-peptide complex,” “peptide-MHC complex,” “pMHC complex,” “peptide-in-groove,” and the like include (i) an MHC molecule, e.g., a human and/or non- human animal MHC molecule, or portion thereof (e.g., the peptide-binding groove thereof, and e.g., the extracellular portion thereof), and (ii) an antigenic peptide (e.g., a HTLV-1- derived peptide), where the MHC molecule and the antigenic peptide are complexed in such a manner that the pMHC complex can specifically bind a T-cell receptor. A pMHC complex encompasses cell surface expressed pMHC complexes and soluble pMHC complexes.
Attorney Docket No.250298.000682 [00106] “HLA-peptide complex,” “peptide-HLA complex,” “pHLA complex,” and the like refer to an MHC-peptide complex wherein the MHC molecule is a Human Leukocyte Antigen (HLA) molecule. [00107] The term “T cell” or “T lymphocyte” is used herein in its broadest sense to refer to all types of immune cells expressing CD3, including, but not limited to, T-helper cells (CD4+ cells), cytotoxic T-cells (CD8+ cells), tumor infiltrating cytotoxic T cells (TIL; CD8+ T cell), CD4+CD8+ T cells, T-regulatory cells (Treg), and NK-T cells. T cells can include thymocytes, naïve T cells, memory T cells, immature T cells, mature T cells, resting T cells, or activated T cells. T cells may also include “gamma-delta T cells (γδ T cells),” which refer to a specialized population that to a small subset of T cells possessing a distinct TCR on their surface, and unlike the majority of T cells in which the TCR is composed of two glycoprotein chains designated α- and β-TCR chains, the TCR in γδ T cells is made up of a γ-chain and a δ-chain. [00108] The term “antigen presenting cell” or “APC” refers to any cell that presents on the surface of the cell an antigen in association with a major histocompatibility complex molecule, either MHC class I or MHC class II molecule, or both. [00109] Terms “antibody,” “antibodies,” “immunoglobulin”, and the like refer to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen, whether natural or partly or wholly synthetically produced. The terms include monoclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab′) fragments, disulfide-linked Fvs (sdFv), intrabodies, minibodies, diabodies and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antigen-specific TCR), and epitope-binding fragments of any of the above. The terms “antibody” and “antibodies” also refer to covalent diabodies such as those disclosed in U.S. Pat. Appl. Pub. 2007/0004909, incorporated herein by reference in its entirety, and Ig-DARTS such as those disclosed in U.S. Pat. Appl. Pub. 2009/0060910, incorporated herein by reference in its entirety. Antibodies useful in the present disclosure include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an antigen binding site.
Attorney Docket No.250298.000682 Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass. [00110] The term “specifically binds,” “binds in a specific manner,” “antigen- specific” or the like, indicates that the molecules involved in the specific binding are able to form a complex with each other that is relatively stable under physiological conditions, and are unable to form stable complexes non-specifically with other molecules outside the specified binding pair. Accordingly, a peptide binding moiety (e.g., an antibody, an alternative scaffold, a CAR, or a TCR) that binds in a specific manner to an HTLV-1- derived peptide, or a peptide-based molecule (such as a complex (e.g., a pMHC complex), fusion protein, or conjugate comprising the described peptide) indicates that the peptide binding moiety forms a stable intermolecular non-covalent bonds with the HTLV-1- derived peptide or peptide-based molecule (such as a complex (e.g., a pMHC complex), fusion protein, or conjugate comprising the described peptide). Specific binding can be characterized by an equilibrium dissociation constant (KD) in the low micromolar to picomolar range (i.e., a smaller KD denotes a tighter binding). High specificity may be in the low nanomolar range, with very high specificity being in the picomolar range. For example, a peptide binding moiety may exhibit binding to an HTLV-1-derived peptide or peptide-based molecule (such as a complex (e.g., a pMHC complex), fusion protein, or conjugate comprising the described peptide) with a KD of about 3000 nM or less, about 2000 nM or less, about 1000 nM or less, about 500 nM or less, about 300 nM or less, about 200 nM or less, about 100 nM or less, about 50 nM or less, about 1 nM or less, or about 0.5 nM or less. Methods for determining whether two molecules specifically bind to one another are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like. [00111] The terms “protein” and “polypeptide”, used interchangeably herein, encompass all kinds of naturally occurring and synthetic proteins, including protein fragments of all lengths, fusion proteins and modified proteins, including without limitation, glycoproteins, as well as all other types of modified proteins (e.g., proteins resulting from phosphorylation, acetylation, myristoylation, palmitoylation, glycosylation, oxidation, formylation, amidation, polyglutamylation, ADP-ribosylation, PEGylation,
Attorney Docket No.250298.000682 biotinylation, etc.). Small polypeptides of less than 100 amino acids, preferably less than 50 amino acids, may be referred to as “peptides”. [00112] The terms “polynucleotide” and “nucleic acid”, used interchangeably herein, include polymeric forms of nucleotides of any length, including ribonucleotides (RNA), deoxyribonucleotides (DNA), or analogs or modified versions thereof. They include single-, double-, and multi-stranded DNA or RNA, genomic DNA, complementary DNA (cDNA), DNA-RNA hybrids, and polymers comprising purine bases, pyrimidine bases, or other natural, chemically modified, biochemically modified, non-natural, or derivatized nucleotide bases. [00113] The term “operably linked” or the like refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. For example, a control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences. “Operably linked” sequences include both expression control sequences that are contiguous with a gene of interest and expression control sequences that act in trans or at a distance to control a gene of interest (or sequence of interest). The term “expression control sequence” includes polynucleotide sequences, which are necessary to affect the expression and processing of coding sequences to which they are ligated. “Expression control sequences” include: appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signals such as splicing and polyadenylation signals; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance polypeptide stability; and when desired, sequences that enhance polypeptide secretion. The nature of such control sequences differs depending upon the host organism. For example, in prokaryotes, such control sequences generally include promoter, ribosomal binding site and transcription termination sequence, while in eukaryotes typically such control sequences include promoters and transcription termination sequence. The term “control sequences” is intended to include components whose presence is essential for expression and processing and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
Attorney Docket No.250298.000682 [00114] The term “isolated” refers to a homogenous population of molecules (such as polynucleotides or polypeptides) which have been substantially separated and/or purified away from other components of the system the molecules are produced in, such as a recombinant cell, as well as a protein that has been subjected to at least one purification or isolation step. “Isolated” refers to a molecule that is substantially free of other cellular material and/or chemicals and encompasses molecules that are isolated to a higher purity, such as to 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% purity. [00115] The term “derivative” as used herein refers to a peptide, polypeptide, or polynucleotide, or a variant or analog thereof, comprising one or more mutations and/or chemical modifications as compared to a reference peptide, polypeptide or polynucleotide. Mutations and/or chemical modifications are further detailed below and can include, for example, insertions, substitutions, deletions, transversions, and/or inversions at one or more locations in the amino acid or nucleotide sequence. [00116] The terms “treat” or “treatment” of a state, disorder, disease, or condition include: (1) preventing, delaying, or reducing the incidence and/or likelihood of the appearance of at least one clinical or sub-clinical symptom of the state, disorder, disease, or condition developing in a subject that may be afflicted with or predisposed to the state, disorder, disease, or condition, but does not yet experience or display clinical or subclinical symptoms of the state, disorder, disease, or condition; or (2) inhibiting the state, disorder, disease, or condition, i.e., arresting, reducing or delaying the development of the disease or a relapse thereof or at least one clinical or sub-clinical symptom thereof; or (3) relieving the state, disorder, disease, or condition, i.e., causing regression of the state, disorder, disease, or condition or at least one of the clinical or sub-clinical symptoms of the state, disorder, disease, or condition. The benefit to a subject to be treated is either statistically significant or at least perceptible to the patient or to the physician. [00117] An “individual” or “subject” or “animal” refers to humans, veterinary animals (e.g., cats, dogs, cows, horses, sheep, pigs, etc.) and experimental animal models of diseases (e.g., mice, rats). In a preferred embodiment, the subject is a human. [00118] The term “effective” applied to dose or amount refers to that quantity of a compound or pharmaceutical composition that is sufficient to result in a desired activity
Attorney Docket No.250298.000682 upon administration to a subject in need thereof. Note that when a combination of active ingredients is administered, the effective amount of the combination may or may not include amounts of each ingredient that would have been effective if administered individually. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition being treated, the particular drug or drugs employed, the mode of administration, and the like. [00119] The phrase “pharmaceutically acceptable”, as used in connection with compositions described herein, refers to molecular entities and other ingredients of such compositions that are physiologically tolerable and do not typically produce untoward reactions when administered to a mammal (e.g., a human). Preferably, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in mammals, and more particularly in humans. [00120] The term “administration” and the like refers to and includes the administration of a composition to a subject or system (e.g., to a cell, organ, tissue, organism, or relevant component or set of components thereof). The skilled artisan will appreciate that route of administration may vary depending, for example, on the subject or system to which the composition is being administered, the nature of the composition, the purpose of the administration, etc. For example, in certain embodiments, administration to an animal subject (e.g., to a human or a rodent) may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal and/or vitreal. In some embodiments, administration may involve intermittent dosing. In some embodiments, administration may involve continuous dosing (e.g., perfusion) for at least a selected period of time. [00121] In accordance with the disclosure herein, there may be employed conventional molecular biology, microbiology, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Sambrook, Fritsch & Maniatis, Molecular Cloning: A Laboratory Manual, Second Edition.
Attorney Docket No.250298.000682 Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press, 1989 (herein “Sambrook et al., 1989”); DNA Cloning: A Practical Approach, Volumes I and II (D.N. Glover ed. 1985); Oligonucleotide Synthesis (M.J. Gait ed.1984); Nucleic Acid Hybridization [B.D. Hames & S.J. Higgins eds. (1985)]; Transcription And Translation [B.D. Hames & S.J. Higgins, eds. (1984)]; Animal Cell Culture [R.I. Freshney, ed. (1986)]; Immobilized Cells And Enzymes [IRL Press, (1986)]; B. Perbal, A Practical Guide To Molecular Cloning (1984); Ausubel, F.M. et al. (eds.). Current Protocols in Molecular Biology. John Wiley & Sons, Inc., 1994. These techniques include site directed mutagenesis as described in Kunkel, Proc. Natl. Acad. Sci. USA 82: 488- 492 (1985), U. S. Patent No. 5,071, 743, Fukuoka et al., Biochem. Biophys. Res. Commun. 263: 357-360 (1999); Kim and Maas, BioTech.28: 196-198 (2000); Parikh and Guengerich, BioTech.24: 428-431 (1998); Ray and Nickoloff, BioTech. 13: 342-346 (1992); Wang et al., BioTech.19: 556-559 (1995); Wang and Malcolm, BioTech. 26: 680-682 (1999); Xu and Gong, BioTech.26: 639-641 (1999), U.S. Patents Nos. 5,789, 166 and 5,932, 419, Hogrefe, Strategies l4. 3: 74-75 (2001), U. S. Patents Nos. 5,702,931, 5,780,270, and 6,242,222, Angag and Schutz, Biotech.30: 486-488 (2001), Wang and Wilkinson, Biotech.29: 976-978 (2000), Kang et al., Biotech.20: 44-46 (1996), Ogel and McPherson, Protein Engineer.5: 467-468 (1992), Kirsch and Joly, Nucl. Acids. Res.26: 1848-1850 (1998), Rhem and Hancock, J. Bacteriol. 178: 3346-3349 (1996), Boles and Miogsa, Curr. Genet.28: 197-198 (1995), Barrenttino et al., Nuc. Acids. Res.22: 541-542 (1993), Tessier and Thomas, Meths. Molec. Biol.57: 229-237, and Pons et al., Meth. Molec. Biol.67: 209-218. Peptides Disclosed Herein [00122] In one aspect, the present disclosure provides isolated peptides comprising an amino acid sequence derived from human T-lymphotropic virus type-1 (HTLV-1). [00123] The HTLV genome follows a canonical structure of replication competent retroviruses comprising GAG, POL, and ENV domains flanked by two long terminal repeat (LTR) domains on either end of the provirus. In particular, HTLV-1 has a monopartite, linear, dimeric single strand RNA (+) genome of 8.5 kb, comprising a 5’-cap and a 3’ poly- A tail. The two LTRs are about 600 nucleotide residues in length and are located at the 5’ and 3’ ends of the genome. The LTRs comprise U3, R, and U5 regions, which together can
Attorney Docket No.250298.000682 function as a transcription unit. The 5’ end of the genome further comprises a primer binding site and the 3’ end further comprises a polypurin tract. The integrated virus utilizes promoter elements in the 5’ LTR to trigger and drive transcription, resulting in an unspliced full-length mRNA that can serve, e.g., as genomic RNA for packaging into virions. While similar to other retroviruses, the genomic RNA of HTLV-1 encodes the structural and enzymatic proteins Gag (an internal group specific antigen), Pol (a polymerase, reverse transcriptase and integrase enzymes required for viral replication and maturation), and Env (envelope glycoprotein), HTLV-1 differs from other retroviruses due to a pX region located toward the 3’ end between the ENV and 3’ LTR. The pX region encodes several alternatively spliced regulatory genes including, without limitation, Tax and HTLV-I basic leucine zipper factor (HBZ), both of which can be implicated in viral pathogenesis. [00124] The viral gene product Tax is a 40 kDa protein which mainly localizes to the nucleus but can also be within the cytoplasm of infected cells. Tax interacts with a variety of host proteins and plays a key role in transactivating proviral transcription from the 5’ LTR. Tax can functionally inactivate p53 and target pRB for degradation, allowing for survival of infected cells. Tax can also dysregulate several signaling pathways such as but not limited to activator protein 1 (AP-1), nuclear factor kappa B (NF-κB), serum responsive factor (SRF), and cyclic AMP response element-binding protein (CREB) pathways. The pleiotropic functions of Tax can contribute to viral pathogenicity and transformation of HTLV-1 infected cells. [00125] HBZ is a nuclear protein but can also be localized to the cytoplasm. HBZ comprises three domains: an activation domain, a central domain, and a basic leucine zipper domain. HBZ antagonizes several Tax-mediated functions and participates in viral persistence and immune evasion. The activation domain of HBZ comprises two LXXLL - like motifs (where L is leucine and X is any amino acid) which bind can bind to a kinase- inducible domain interacting (KIX) domain of p300-CBP transcription coactivators. The LXXLL-like motifs and KIX domain can be a requisite for HBZ activation of TGF-β (Transforming growth factor beta)/Smad signaling. [00126] After infection of a host, HTLV-1 infects cells via its glycoprotein gp62. HTLV-1 can then achieve latency by integrating into the host genome and increasing proviral load by proliferation of infected cells.
Attorney Docket No.250298.000682 [00127] The HTLV-1 life cycle initiates when the HTLV-1 virion attaches to a host cell such as by way of a surface receptor(s), e.g., GLUT1/ HSPG/NRP-1, via the HTLV- 1 envelope surface and/or transmembrane domains of the Env protein. After the outer membrane of the viral particle fuses with the host cell membrane, the contents of the virions can enter the cytoplasm of the host cell and the viral genomic RNA (gRNA) can undergo reverse transcription to convert gRNA into double stranded DNA (dsDNA) using the virally encoded reverse transcriptase (RNA-dependent/DNA-dependent polymerase). Viral dsDNA traffics to the nucleus and stochastically integrates into the host genome using a viral encoded integrase at which point the viral genome is now a provirus. The host cell machinery can then drive transcription of the proviral DNA using host cell RNA polymerase II to produce viral mRNA. The unspliced full-length viral genomic RNA (~9 kb) can be used to produce progeny virions and to produce, e.g., Gag protein, Pol protein, and Pro protein (a protease enzyme required for maturation of structural and enzymatic proteins). The singly spliced subgenomic mRNA (~4.3 kb) can be used to produce Env protein, and the doubly-spliced RNA (~ 2.1 kb) can be used to translate the regulatory proteins Tax (open reading frame (ORF) IV) and Rex (ORF III). Alternative splicing can also lead to the production of a number of accessory proteins from pX ORF I and II. Immature virions can be assembled at the plasma membrane and are considered mature following viral budding. [00128] Amino acid sequences derived from HTLV-1 disclosed herein may include naturally occurring proteogenic amino acids as well as non-proteogenic amino acids and non-naturally occurring amino acids such as amino acid analogs. In some embodiments, the amino acids that may be used in the practice of the present disclosure may include, for example, without limitation, naturally occurring proteogenic (L)-amino acids, their optical (D)-isomers, chemically modified amino acids, including, e.g., amino acid analogs such as, e.g., selenocysteine (Sec), penicillamine (3-mercapto-D-valine), pyroglutamic acid (5- oxoproline), etc., naturally occurring non-proteogenic amino acids such as norleucine, and chemically synthesized amino acids that have properties known in the art to be characteristic of an amino acid, and amino acid equivalents. [00129] In some embodiments, an isolated peptide of the present disclosure comprises an amino acid sequence that is at least about 60%, at least about 65%, at least
Attorney Docket No.250298.000682 about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% identical to the amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof. In some embodiments, an isolated peptide of the present disclosure comprises an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143- 146. In some embodiments, an isolated peptide of the present disclosure consists essentially of an amino acid sequence of any of SEQ ID NOs: 1-89 and 143-146. In some embodiments, the isolated peptide consists of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. In some embodiments, the isolated peptide comprises two or more sequences selected from any one of SEQ ID NOs: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof. [00130] A list of non-limiting examples of HTLV-1-derived peptides are provided in Table 1 below. Table 1. Examples of HTLV-1-derived Peptides SEQ ID NO: Peptide Sequence 1 ATMASLISH
Attorney Docket No.250298.000682 KTRWQLHHSPR LENRVLTGW
Attorney Docket No.250298.000682 64 SPCHNSLIL 65 VVCMYLYQL G
[00131] A peptide of the disclosure may be synthetically produced or produced by hydrolysis. Synthetically produced peptides can include randomly generated peptides, specifically designed peptides, and peptides where at least some of the amino acid positions are conserved among several peptides and the remaining positions are random. Alternatively, a peptide of the present disclosure may be produced by expression in a heterologous host cell. [00132] In nature, peptides that are produced by hydrolysis undergo hydrolysis prior to binding of the antigen to an MHC molecule. Class I MHC typically present peptides
Attorney Docket No.250298.000682 derived from proteins actively synthesized in the cytoplasm of the cell. In contrast, class II MHC typically present peptides derived either from exogenous proteins that enter a cell’s endocytic pathway or from proteins synthesized in the endoplasmic reticulum (ER). Intracellular trafficking permits a peptide to become associated with an MHC molecule. [00133] The binding of a peptide to an MHC peptide binding groove can control the spatial arrangement of MHC and/or peptide amino acid residues recognized by a TCR. Such spatial control is due in part to hydrogen bonds formed between a peptide and an MHC molecule. Based on the knowledge on how peptides bind to various MHC molecules, the major MHC anchor amino acids and the surface exposed amino acids that are varied among different peptides can be determined. [00134] Preferably, the length of an MHC-binding peptide is from about 5 to about 40 amino acid residues, more preferably from about 6 to about 30 amino acid residues, and even more preferably from about 8 to about 20 amino acid residues, and even more preferably between about 9 and 11 amino acid residues, including any size peptide between 5 and 40 amino acids in length, in whole integer increments (i.e., 5, 6, 7, 8, 9...40). While naturally MHC class II-bound peptides vary from about 9-40 amino acids, in nearly all cases the peptide can be truncated to an about 9-11 amino acid core without loss of MHC binding activity or T cell recognition. [00135] In some embodiments, the isolated peptides of the disclosure may be about 8-12 amino acids in length. For example, a peptide disclosed herein may be 8 amino acids, 9 amino acids, 10 amino acids, 11 amino acids, or 12 amino acids in length. [00136] The peptides of the disclosure may comprise one or more reverse peptide bonds, one or more non-peptide bonds, one or more chemical modifications, one or more D-isomers of amino acids, or any combination thereof. [00137] In some embodiments, the peptide may be modified to comprise one or more reverse peptide bonds or non-peptide bonds. Such modification may improve stability and/or binding of the peptide to MHC molecules to elicit a stronger immune response. In a reverse peptide bond, amino acid residues are not joined by peptide (—CO—NH—) linkages but the peptide bond is reversed. Such retro-inverso peptidomimetics may be made using methods known in the art, for example such as those described in Meziere et al., 1997 (Meziere C., et al. J Immunol 1997). This approach involves making
Attorney Docket No.250298.000682 pseudopeptides containing changes involving the backbone, and not the orientation of side chains. Such pseudopeptides may be useful, for example, for desired MHC binding and/or T helper cell responses. Retro-inverse peptides, which contain NH—CO bonds instead of CO—NH peptide bonds, are much more resistant to proteolysis. Additional non-peptide bond that may be used are, for example, —CH2—NH, —CH2S—, —CH2CH2—, — CH═CH—, —COCH2—, —CH(OH)CH2—, and —CH2SO—. [00138] The amino acid residues comprising the peptides of the disclosure may be chemically modified. Non-limiting examples of chemical modifications include, for example, phosphorylation, acetylation, deamidation acylation, amidination, pyridoxylation of lysine, reductive alkylation, trinitrobenzylation of amino groups with 2,4,6- trinitrobenzene sulphonic acid (TNBS), amide modification of carboxyl groups and sulphydryl modification by performic acid oxidation of cysteine to cysteic acid, formation of mercurial derivatives, formation of mixed disulfides with other thiol compounds, reaction with maleimide, carboxymethylation with iodoacetic acid or iodoacetamide and carbamoylation with cyanate at alkaline pH. Chemical modifications may not correspond to those that may be present in vivo. [00139] For example, modification of, for example, arginyl residues in proteins may be based on the reaction of vicinal dicarbonyl compounds such as phenylglyoxal, 2,3- butanedione, and 1,2-cyclohexanedione to form an adduct. Another example is the reaction of methylglyoxal with arginine residues. Cysteine can be modified without concomitant modification of other nucleophilic sites such as lysine and histidine. Selective reduction of disulfide bonds in proteins can also be performed. Disulfide bonds can be formed and oxidized during the heat treatment of biopharmaceuticals. Woodward’s Reagent K may be used to modify specific glutamic acid residues. N-(3-(dimethylamino)propyl)-N′- ethylcarbodiimide can be used to form intra-molecular crosslinks between a lysine residue and a glutamic acid residue. For example, diethylpyrocarbonate and 4-hydroxy-2-nonenal can be used to modify histidyl residues in proteins. The reaction of lysine residues and other α-amino groups is, for example, useful in binding of peptides to surfaces or the cross- linking of proteins/peptides. Lysine is the site of attachment of poly(ethylene)glycol and the major site of modification in the glycosylation of proteins. Methionine residues in proteins can be modified with e.g. iodoacetamide, bromoethylamine, and chloramine T.
Attorney Docket No.250298.000682 Tetranitromethane and N-acetylimidazole can be used for the modification of tyrosyl residues. Cross-linking via the formation of dityrosine can be accomplished with hydrogen peroxide/copper ions. N-bromosuccinimide, 2-hydroxy-5-nitrobenzyl bromide or 3- bromo-3-methyl-2-(2-nitrophenylmercapto)-3H-indole (BPNS-skatole) have been used in recent studies for the modification of tryptophan. Successful modification of therapeutic proteins and peptides with PEG can lead to an extension of circulatory half-life while cross- linking of proteins/ peptides with glutaraldehyde, polyethylene glycol diacrylate and formaldehyde can be used for the preparation of hydrogels. Chemical modification of allergens for immunotherapy can be achieved by carbamylation with potassium cyanate. [00140] Peptides of the present disclosure may also be synthesized with additional chemical groups present at their N- and/or C-termini, to enhance the stability, bioavailability, and/or affinity of the peptides. [00141] N-terminal modifications can include methylation (e.g., —NHCH3 or — N(CH3)2), acetylation (e.g., with acetic acid or a halogenated derivative thereof such as ^- chloroacetic acid, ^-bromoacetic acid, or ^-iodoacetic acid), adding a benzyloxycarbonyl (Cbz) group, or blocking the amino terminus with any blocking group containing a carboxylate functionality defined by RCOO— or sulfonyl functionality defined by R— SO2—, where R is selected from alkyl, aryl, heteroaryl, alkyl aryl, and the like, and similar groups. One can also incorporate a desamino acid at the N-terminus (so that there is no N- terminal amino group) to decrease susceptibility to proteases or to restrict the conformation of the peptide. Additionally, hydrophobic groups such as carbobenzoxyl, dansyl, or t- butyloxycarbonyl groups may be added to the N-terminus. Likewise, an acetyl group or a 9-fluorenylmethoxy-carbonyl group may be placed at the N-terminus. [00142] C-terminal modifications can include replacing the free acid with a carboxamide group or forming a cyclic lactam at the carboxy terminus to introduce structural constraints. One can also cyclize the peptides of the disclosure, or incorporate a desamino or descarboxy residue at the termini of the peptide, so that there is no terminal amino or carboxyl group, to decrease susceptibility to proteases or to restrict the conformation of the peptide. C-terminal functional groups of the compounds of the present disclosure include amide, amide lower alkyl, amide di(lower alkyl), lower alkoxy, hydroxy, and carboxy, and the lower ester derivatives thereof, and the pharmaceutically acceptable
Attorney Docket No.250298.000682 salts thereof. Additionally, the hydrophobic group, t-butyloxycarbonyl, or an amido group may be added to the C-terminus. [00143] Further examples of non-natural modifications include incorporation of non-encoded α-amino acids, photoreactive cross-linking amino acids, N-methylated amino acids, and β-amino acids, backbone reduction, retroinversion by using D-amino acids, and C-terminal amidation and PEGylation. [00144] Peptides described herein may comprise one or more (e.g., 1, 2, 3, or 4) amino acid substitutions and/or insertions and/or deletions. Amino acid substitution means that an amino acid residue is substituted for a replacement amino acid residue at the same position. Inserted amino acid residues may be inserted at any position and may be inserted such that some or all of the inserted amino acid residues are immediately adjacent one another or may be inserted such that none of the inserted amino acid residues is immediately adjacent another inserted amino acid residue. One or more (e.g., 1, 2, 3 or 4) amino acids may be substituted and/or inserted and/or deleted from the sequence of any one of SEQ ID NOs: 1-89 and 143-146. Each substitution and/or insertion and/or deletion can take place at any position of any one of SEQ ID NOs: 1-89 and 143-146. [00145] In some embodiments, the peptides of the disclosure may comprise additional amino acids (e.g., 1, 2, 3 or 4) at the C-terminal end and/or at the N-terminal end of the sequence of any one SEQ ID NOs: 1-89 and 143-146. A peptide of the disclosure may comprise the amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146 except for one or more (e.g., 1, 2, 3, or 4) amino acid substitutions, insertions or deletions. [00146] Inserted amino acids and substituted amino acids may be naturally occurring amino acids or may be non-naturally occurring amino acids and, for example, may contain a non-natural side chain, and/or be linked together via non-native peptide bonds. Such altered peptide ligands are discussed further in Douat-Casassus et al., J. Med. Chem, 2007; 50(7):1598-609 and Hoppes et al., J. Immunol 2014; 193(10):4803-13 and references therein. If more than one amino acid residue is substituted and/or inserted, the replacement/inserted amino acid residues may be the same as each other or different from one another. Each replacement amino acid may have a different side chain to the amino acid being replaced.
Attorney Docket No.250298.000682 [00147] D-amino acids may be substituted for the L-amino acids in the antigenic peptides of the disclosure. In addition, non-standard amino acids (i.e., other than the common naturally occurring proteinogenic amino acids such as β-γ-δ-amino acids, as well as many derivatives of L-α-amino acids) may also be used for substitutions or additions to produce peptides of the present disclosure. [00148] Amino acid substitutions may be conservative, by which it is meant the substituted amino acid has similar chemical properties to the original amino acid. For example, the following groups of amino acids share similar chemical properties such as size, charge, and polarity: Group 1 - Ala, Ser, Thr, Pro, Gly; Group 2 - Asp, Asn, Glu, Gln; Group 3 - His, Arg, Lys; Group 4 - Met, Leu, Ile, Val, Cys; Group 5 - Phe, Thy, Trp. [00149] Substantial changes in function (e.g., affinity for MHC molecules and/or TCRs) can be made by selecting substitutions that are less conservative than those described above, in other words, selecting residues that differ more significantly in their effect on maintaining the structure of the peptide backbone in the area of the substitution (e.g., as a sheet or helical conformation), the bulk of the side chain, or the charge or hydrophobicity of the peptide at the positions involved for MHC or TCR binding. The substitutions which in general are expected to produce the greatest changes in peptide properties will be those in which (a) a hydrophilic residue, e.g. Ser, is substituted for (or by) a hydrophobic residue, e.g. Leu, Ile, Phe, Val or Ala; (b) a residue having an electropositive side chain, e.g., Lys, Arg, or His, is substituted for (or by) an electronegative residue, e.g. Glu or Asp; or (c) a residue having a bulky side chain, e.g. Phe, is substituted for (or by) a residue not having a side chain, e.g., Gly. [00150] One can also replace the naturally occurring side chains of the 20 genetically encoded amino acids (or the stereoisomeric D-amino acids) with other side chains, for instance with groups such as alkyl, lower alkyl, cyclic 4-, 5-, 6-, to 7-membered alkyl, amide, amide lower alkyl, amide di(lower alkyl), lower alkoxy, hydroxy, carboxy and the lower ester derivatives thereof, and with 4-, 5-, 6-, to 7-membered heterocyclic. For example, proline analogues in which the ring size of the proline residue is changed from 5 members to 4, 6, or 7 members can be employed. Cyclic groups can be saturated or unsaturated, and if unsaturated, can be aromatic or non-aromatic. Heterocyclic groups preferably contain one or more nitrogen, oxygen, and/or sulfur heteroatoms. Examples of
Attorney Docket No.250298.000682 such groups include the furazanyl, furyl, imidazolidinyl, imidazolyl, imidazolinyl, isothiazolyl, isoxazolyl, morpholinyl (e.g. morpholino), oxazolyl, piperazinyl (e.g., 1- piperazinyl), piperidyl (e.g., 1-piperidyl, piperidino), pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolidinyl (e.g., 1- pyrrolidinyl), pyrrolinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, thiomorpholinyl (e.g., thiomorpholino), and triazolyl. These heterocyclic groups can be substituted or unsubstituted. Where a group is substituted, the substituent can be alkyl, alkoxy, halogen, oxygen, or substituted or unsubstituted phenyl. [00151] Other examples of amino acid replacements include stereoisomers (e.g., D- amino acids) and unnatural amino acids such as, for example, L-ornithine, L-homocysteine, L-homoserine, L-citrulline, 3-sulfino-L-alanine, N-(L-arginino)succinate, 3,4-dihydroxy- L-phenylalanine, 3-iodo-L-tyrosine, 3,5-diiodo-L-tyrosine, triiodothyronine, L-thyroxine, L-selenocysteine, N-(L-arginino)taurine, 4-aminobutylate, (R,S)-3-amino-2- methylpropanoate, a,a-disubstituted amino acids, N-alkyl amino acids, lactic acid, β- alanine, 3-pyridylalanine, 4-hydroxyproline, O-phosphoserine, N-methylglycine, N- acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, nor-leucine, and other similar amino acids and imino acids. [00152] The amino acid residues that do not substantially contribute to interactions with the T-cell receptor can be modified by replacement with other amino acid whose incorporation does not substantially affect T-cell reactivity and does not eliminate binding to the relevant MHC. [00153] The peptides may also comprise isosteres of two or more residues. An “isostere” as used here refers to a sequence of two or more residues that can be substituted for a second sequence because the steric conformation of the first sequence fits a binding site specific for the second sequence. The term specifically includes peptide backbone modifications well known to those skilled in the art. Such modifications include modifications of the amide nitrogen, the α-carbon, amide carbonyl, complete replacement of the amide bond, extensions, deletions or backbone crosslinks. [00154] Combinations of several substitutions/additions/deletions at more than one position can be developed and tested to determine if the combination results in an additive
Attorney Docket No.250298.000682 or synergistic effects on the immunogenicity of the peptide. In some embodiments, no more than 4 positions within the peptide are simultaneously altered. [00155] Preferably, peptides of the disclosure bind to an MHC molecule in the peptide binding groove of the MHC molecule. Generally, the amino acid modifications described above will not impair the ability of the peptide to bind to the MHC molecule. In some embodiments, the amino acid modifications improve the ability of the peptide to bind to the MHC molecule. For example, mutations may be made at positions which anchor the peptide to the MHC molecule. Such anchor positions and the preferred residues at these locations for peptides which bind, in particular, HLA-A*02 may comprise, e.g., amino acids residues at position 2, and/or at the C-terminus of the peptide, which may be considered primary anchor positions. Preferred anchor residues may be different for each HLA type. As a non-limiting example, the preferred amino acids in position 2 for HLA- A*02 are Leu, lie, Val, or Met and at the C-terminus are Val or Leu. Multiple positions may be important for stable peptide binding to HLA-A*02, including positions 2, 3, 5-7, and 9. The anchor residues at position 2 and 9 may be of prime importance for peptide binding to HLA-A2. However, other peptide side chains, e.g., at position 3, may contribute to the stability of the interaction. In certain cases, the optimal length for peptide binding can be longer than 9 residues. [00156] The immunologic properties of peptides can be described as a function of binding to MHC molecules (Kon and Koff) and TCR (affinity of interaction between TCR and MHC-peptide complexes). Modifications of primary MHC anchor residues exhibit a significant degree of predictability about overall impact on binding to MHC molecules. Modifications of secondary MHC anchor residues can impact the affinity of interaction of the MHC-peptide complex to TCR as well as with the Kon and Koff relative to peptide-MHC interaction. [00157] When the HTLV-1 peptide is a mutant peptide, T cell lines against a natural (non-mutated) epitope are generated, and an immunization strategy potent enough to generate a useful response in transgenic mice carrying human MHC (such as the A2 allele) is used. HTLV-1 peptides are interrogated ex vivo in the presence of competent APCs and the functional impact of T cells specific for natural (non-mutated) epitopes is measured. The evaluation is done at various concentrations of HTLV-1 peptide because the expected
Attorney Docket No.250298.000682 effect is biphasic in the instance of cross reactive peptides (activating at limited concentrations and inhibiting at higher concentrations due to antigen-induced cell death [AICD]). Measurement of the following three parameters can be used to describe characteristics of the disclosed HTLV-1 peptides: [00158] 1. Minimal required concentration of HTLV-1 metrics to induce effects indicative of T cell activation (e.g., cytokine [e.g., IFN-γ] production); 2. Maximal (peak value) effect (e.g., cytokine [e.g., IFN-γ] production) at any HTLV-1 peptide concentration; and 3. HTLV-1 peptide concentration at peak value of activating effect (e.g., cytokine [e.g., IFN-γ] concentration). [00159] By way of a non-limiting example, HTLV-1 peptides that result in reduced values associated with parameters number 1 and number 3, but increased number 2, can be useful. Use of natural epitope and/or unrelated non-cross-reactive peptide as references is valuable for identifying classes of peptides of possible value. Peptides possessing properties quantitatively comparable to or even moderately attenuated from those of natural epitopes are still considered useful since, while they retain cross-reactivity, they may exhibit immunologic properties that are distinct from those of the natural peptide, e.g., reduced capacity to break tolerance or reestablish responsiveness in vivo or lower propensity to induce AICD. [00160] In addition to practicality and rapidity, additional advantages of this screening approach include, but are not limited to, use of more relevant polyclonal T cell lines instead of potentially biased T cell clones as a read out, and the composite value, integrating parameters such as Kon, Koff, and TCR affinity that can translate into cross- reactivity and functional avidity of peptide-MHC complexes relative to TCR. These parameters can be predictive of the in vivo immunologic properties and thus can define useful panels of peptides eligible for further evaluation, optimization, and practical applications. Peptides that bind to MHC and retain cross-reactivity against TCR specific for the nominal wild-type peptide are predicted to elicit a measurable effect in this assay. [00161] A peptide of the disclosure, or a pharmaceutically acceptable salt thereof, or fragment or derivative thereof, may be used to induce an immune response. If this is the case, it is important that the immune response is specific to the intended target (e.g., HTLV-
Attorney Docket No.250298.000682 1, HTLV-2, HTLV-3, and/or HTLV-4) to avoid the risk of unwanted side effects that may be associated with an “off target” immune response. Therefore, it is preferred that the amino acid sequence of a peptide of the disclosure does not match the amino acid sequence of a peptide from any other endogenous protein(s), particularly that of another human protein. Also, the amino acid modifications described herein should not impair the ability of the peptide inducing an antigen-specific immune response when presented in a complex with an MHC molecule on the surface of an antigen presenting cell (APC). [00162] The peptides may be also modified to improve half-life and/or bioavailability, for example, by PEGylation, glycosylation, polysialylation, HESylation, recombinant PEG mimetics, Fc fusion, albumin fusion, nanoparticle attachment, nanoparticulate encapsulation, cholesterol fusion, iron fusion, or acylation. [00163] The peptides of the disclosure can also serve as structural models for non- peptidic compounds with similar biological activity. A variety of techniques can be used to construct compounds with the same or similar desired biological activity as the lead peptide compound, but with more favorable activity than the lead with respect to solubility, stability, and susceptibility to hydrolysis and proteolysis. These techniques include replacing the peptide backbone with a backbone composed of amidates, phosphonates, carbamates, sulfonamides, secondary amines, and N-methylamino acids. [00164] Multiple peptides described herein may be operably linked together. Accordingly, in one aspect, the present disclosure provides an isolated peptide or polypeptide comprising two or more amino acid sequences selected from SEQ ID NO: 1- 89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof. For example, such a multi-epitope peptide or polypeptide may comprise include 2 to 50, 2 to 40, 2 to 30, 5 to 25, 5 to 20, or 10 to 15 single-epitope peptides as described herein (e.g., SEQ ID NO: 1-89 and 143-146). The single-epitope peptides (e.g., SEQ ID NO: 1-89 and 143-146) may be arranged in any order, and may be identical or different. [00165] In some embodiments, the present disclosure provides an isolated peptide or polypeptide comprising 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92,
Attorney Docket No.250298.000682 or 93 amino acid sequences selected from SEQ ID NO: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof. [00166] The single-epitope peptides may be linked via a linker. The linker may comprise relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions. The linker can be selected from, e.g., Table 2, or other neutral spacers of nonpolar amino acids or neutral polar amino acids. It will be understood that the optionally present linker need not be comprised of the same residues and thus may be a hetero- or homo-oligomer. When present, the linker will usually be at least one or two residues, more usually three to six residues. [00167] Peptides of the disclosure can be synthesized by e.g., solid phase synthesis. As such, the peptides may be immobilized, for example to a solid support such as a bead. Peptides of the disclosure may be synthesized by the Fmoc-polyamide mode of solid-phase peptide synthesis. Temporary N-amino group protection is afforded by the 9- fluorenylmethyloxycarbonyl (Fmoc) group. Repetitive cleavage of this highly base-labile protecting group is done using 20% piperidine in N, N-dimethylformamide. Side-chain functionalities may be protected as their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), butyloxycarbonyl derivative (in the case of lysine and histidine), trityl derivative (in the case of cysteine) and 4-methoxy-2,3,6-trimethylbenzenesulphonyl derivative (in the case of arginine). Where glutamine or asparagine are C-terminal residues, use is made of the 4,4′- dimethoxybenzhydryl group for protection of the side chain amido functionalities. The solid-phase support is based on a polydimethyl-acrylamide polymer constituted from the three monomers dimethylacrylamide (backbone-monomer), bisacryloylethylene diamine (cross linker) and acryloylsarcosine methyl ester (functionalizing agent). The peptide-to- resin cleavable linked agent used is the acid-labile 4-hydroxymethyl-phenoxyacetic acid derivative. All amino acid derivatives are added as their preformed symmetrical anhydride derivatives except for asparagine and glutamine, which are added using a reversed N, N- dicyclohexyl-carbodiimide/1-hydroxybenzotriazole mediated coupling procedure. All coupling and deprotection reactions are monitored using ninhydrin, trinitrobenzene sulphonic acid or isotin test procedures. Upon completion of synthesis, peptides are cleaved
Attorney Docket No.250298.000682 from the resin support with concomitant removal of side-chain protecting groups by treatment with 95% trifluoroacetic acid containing a 50% scavenger mix. Scavengers commonly used include ethanedithiol, phenol, anisole and water, the exact choice depending on the constituent amino acids of the peptide being synthesized. Also, a combination of solid phase and solution phase methodologies for the synthesis of peptides is possible. [00168] Trifluoroacetic acid is removed by evaporation in vacuo, with subsequent trituration with diethyl ether affording the crude peptide. Any scavengers present are removed by a simple extraction procedure which on lyophilization of the aqueous phase affords the crude peptide free of scavengers. [00169] Purification may be performed by techniques such as re-crystallization, ion- exchange chromatography, size exclusion chromatography, hydrophobic interaction chromatography and reverse-phase high performance liquid chromatography using e.g. acetonitrile/water gradient separation, or a combination thereof. [00170] Peptides may be analyzed using thin layer chromatography, electrophoresis, in particular capillary electrophoresis, solid phase extraction (CSPE), reverse-phase high performance liquid chromatography, amino-acid analysis after acid hydrolysis and by fast atom bombardment (FAB) mass spectrometric analysis, as well as MALDI and ESI-Q- TOF mass spectrometric analysis. [00171] Alternatively, the peptide may be produced by recombinant expression in a heterologous host cell. Such methods typically involve the use of a vector comprising a nucleic acid sequence encoding the peptide to be expressed, to express the polypeptide in vivo; for example, in bacteria, yeast, insect or mammalian cells. [00172] In further embodiments, in vitro cell-free systems may be used. The peptides may be isolated and/or may be provided in substantially pure form. For example, they may be provided in a form which is substantially free of other peptides or proteins. Peptide-MHC (pMHC) Complexes Disclosed Herein [00173] In another aspect, the disclosure provides a complex of a peptide of the disclosure and an MHC molecule. Preferably, the peptide is bound to the peptide binding groove of the MHC molecule. In some embodiments, the peptide and the MHC molecule
Attorney Docket No.250298.000682 form a non-covalent complex. In other embodiments, the peptide and the MHC molecule may be covalently linked, for example, via a linker. [00174] MHC molecules are generally classified into two categories: class I and class II MHC molecules. An MHC class I molecule is an integral membrane protein comprising a glycoprotein heavy chain, also referred to herein as the α chain, which has three extracellular domains (i.e., α1, α2 and α3) and two intracellular domains (i.e., a transmembrane domain (TM) and a cytoplasmic domain (CYT)). The heavy chain is noncovalently associated with a soluble subunit called β2 microglobulin (β2m or β2M). An MHC class II molecule or MHC class II protein is a heterodimeric integral membrane protein comprising one α chain and one β chain in noncovalent association. The α chain has two extracellular domains (α1 and α2), and two intracellular domains (a TM domain and a CYT domain). The β chain contains two extracellular domains (β1 and β2), and two intracellular domains (a TM domain and CYT domain). [00175] The domain organization of class I and class II MHC molecules forms the antigenic determinant binding site, e.g., the peptide-binding portion or peptide binding groove, of the MHC molecule. A peptide binding groove refers to a portion of an MHC molecule that forms a cavity in which a peptide, e.g., antigenic determinant, can bind. The conformation of a peptide binding groove is capable of being altered upon binding of a peptide to enable proper alignment of amino acid residues important for TCR binding to the peptide-MHC (pMHC) complex. [00176] In some embodiments, MHC molecules include fragments of MHC chains that are sufficient to form a peptide binding groove. For example, a peptide binding groove of a class I protein can comprise portions of the α1 and α2 domains of the heavy chain capable of forming two β-pleated sheets and two α helices. Inclusion of a portion of the β2 microglobulin chain stabilizes the MHC class I molecule. While for most versions of MHC class II molecules, interaction of the α and β chains can occur in the absence of a peptide, the two-chain molecule of MHC class II is unstable until the binding groove is filled with a peptide. A peptide binding groove of a class II protein can comprise portions of the α1 and β1 domains capable of forming two β-pleated sheets and two α helices. A first portion of the α1 domain forms a first β-pleated sheet and a second portion of the α1 domain forms a first a helix. A first portion of the β1 domain forms a second β-pleated sheet and a second
Attorney Docket No.250298.000682 portion of the β1 domain forms a second a helix. The X-ray crystallographic structure of class II protein with a peptide engaged in the binding groove of the protein shows that one or both ends of the engaged peptide can project beyond the MHC protein. Thus, the ends of the α1 and β1 α helices of class II form an open cavity such that the ends of the peptide bound to the binding groove are not buried in the cavity. Moreover, the X-ray crystallographic structure of class II proteins shows that the N-terminal end of the MHC β chain apparently projects from the side of the MHC protein in an unstructured manner since the first 4 amino acid residues of the β chain could not be assigned by X-ray crystallography. [00177] The peptides of the present disclosure can bind to an MHC molecule in a manner such that the pMHC complex can bind to a TCR, preferably in a specific manner. In certain embodiments, binding of the pMHC complex to the TCR may induce a T cell response. [00178] Whether or not a given peptide will form a complex with an MHC molecule can be determined by assessing whether the MHC can be refolded in the presence of the peptide using the process described in, for example, PCT Application WO2018/083505, which is incorporated herein in its entirety for all purposes. If the peptide does not form a complex with MHC then MHC will not refold. Refolding can be confirmed using an antibody that recognizes MHC in a folded state only. Alternatively, the ability of a peptide to stabilize MHC on the surface of transporter associated with antigen processing (TAP)- deficient cell lines such as T2 cells, which lack the capacity for TAP-mediated translocation of cytosolic peptides into the endoplasmic reticulum (ER) for peptide loading onto MHC class I molecules, or other biophysical methods to determine interaction parameters can be determined. [00179] The peptides according to the present disclosure may be provided as an MHC groove-binding peptide. In some embodiments, the MHC groove-binding peptide can be designed such that the peptide may vary in some or all the positions involved in MHC binding. For example, the MHCBN is a comprehensive database of MHC binding and non-binding peptides compiled from published literature and existing databases. The latest version of the database has 25,860 entries including 20,717 MHC binders and 4,022 MHC non-binders for more than 450 MHCs. The database has sequence and structure data
Attorney Docket No.250298.000682 of (a) source proteins of peptides and (b) MHCs. MHCBN has a number of web tools that include: (i) mapping of peptide on query sequence; (ii) search on any field; (iii) creation of data sets; and (iv) online data submission. [00180] In some cases, a peptide binding tool for prediction of binding to MHC-I or MHC-II can be, for example, Antibody Epitope Prediction, ANTIGENIC, BepiPred, CTLPred, DiscoTope, EPIPREDICT, Epitope Cluster Analysis, Epitope Conservancy Analysis, EUiPro, HLA Peptide Binding Predictions, HLABinding, MAPPP, MHCBench, MHC-I processing predictions, Mosaic Vaccine Tool Suite, NetChop, NetCTL, NetMHC, NetMHCII, NetMHCpan, nHLAPred-I, OptiTope, PAProC, POPI, PREDEP, Prediction of Antigenic Determinants, ProPred, ProPred-1, RankPep, SMM, SVMHC, TAPPred, VaxiJen, or combinations thereof. Additional example programs are used such as BIMAS, SYFPEITHI, or Rankpep. [00181] In one specific embodiment, a library of altered peptides is produced by genetically engineering the library using polymerase chain reaction (PCR) or any other suitable technique to construct a DNA fragment encoding the peptide. With PCR techniques, by using oligonucleotides that are randomly mutated within particular triplet codons, the resultant fragment pool encodes all possible combination of codons at these positions. Preferably, certain of the amino acid positions are maintained constant, which are the conserved amino acids that are required for binding to the MHC peptide binding groove, and which do not contact the T cell receptor (TCR). [00182] In some embodiments, when a library of altered peptides is produced by genetically engineering the library using polymerase chain reaction (PCR) or any other suitable technique to construct a DNA fragment encoding the peptide, the target TCR is a TCR for which it is desired to identify the peptide epitope recognized by the receptor. In some embodiments, the target TCR is from a patient with an HTLV-1 infection and/or an HTLV-1-induced disease or disorder). In some embodiments, the TCR includes an α-chain and a β-chain. [00183] MHC molecules used in pMHC complexes described herein include naturally occurring full-length MHC molecules as well as individual chains of MHC molecules (e.g., MHC class I α (heavy) chain, β2-microglobulin, MHC class II α chain, and MHC class II β chain), individual subunits of such chains of MHCs (e.g., α1, α2 and/or
Attorney Docket No.250298.000682 α3 subunits of MHC class I α chain, α1 and/or α2 subunits of MHC class II α chain, β1 and/or β2 subunits of MHC class II β chain) as well as fragments, mutants, and various derivatives thereof (including fusion proteins, e.g., fusions with viral envelope proteins or fusogens), wherein such fragments, mutants, and derivatives retain the ability to display an antigenic determinant for recognition by an antigen-specific TCR. In one specific embodiment, the MHC comprises a transmembrane domain embedded in the lipid envelope of a liposome, a recombinant viral particle, or a virus-like particle (VLP). [00184] Naturally-occurring MHC molecules are encoded by a cluster of genes on human chromosome 6 or mouse chromosome 17. MHCs are also referred to as H-2 in mice and Human Leucocyte Antigen (HLA) in humans. MHC class I molecules specifically bind CD8 molecules expressed on cytotoxic T lymphocytes (CD8+ T cells), whereas MHC class II molecules specifically bind CD4 molecules expressed on helper T lymphocytes (CD4+ T cells). MHCs include, but are not limited to, HLA specificities such as A (e.g. A1-A74), B (e.g., B 1-B77), C (e.g., C1-C11), D (e.g., D1-D26), E, G, DR (e.g., DR1- DR8), DQ (e.g., DQ1-DQ9) and DP (e.g. DP1-DP6). More preferably, HLA specificities include A1, A2, A3, A11, A23, A24, A28, A30, A33, B7, B8, B35, B44, B53, B60, B62, DR1, DR2, DR3, DR4, DR7, DR8, and DR-11. [00185] In some embodiments, the MHC molecule in a pMHC complex of the present disclosure is a human leukocyte antigen (HLA) molecule. The MHC molecule may be a human HLA molecule selected from the group consisting of HLA-A, HLA-B, HLA- C, HLA-E, HLA-F, and HLA-G. In some embodiments, the MHC class I or MHC II polypeptides may be derived from any functional human HLA-A, B, C, DR, or DQ molecules. Non-limiting examples of HLA-A alleles comprise, without limitation, A*0101, A*0201, A*0202, A*0301, A*1101, A*2301, A*2402, A*2501, A*2601, A*2901 , A*2902, A*3101, A*3201, A*3301, A*3401, A*3601, A*4301, A*6601, A*6801, A*6901, A*7401, and A*8001. Non-limiting examples of HLA-B alleles comprise, without limitation, B*0702. B*0801, B*1301, B*1401, B*1402, B*1501, B*1801, B*1802, B*2701, B*2702, B*3501, B*3502, B*3701, B*3801, B*3901, B*4001, B*4101, B*4201, B*4402, B*4501, B*4601, B*4701, B*4801, B*4901, B*5001, B*5101, B*5201, B*5301, B*5401, B*5501, B*5502, B*5601, B*5701, B*5801, B*5901, B*6701, B*7301, B*1517, B*8101, B*8201, and B*8301. Non-limiting examples of HLA-C alleles
Attorney Docket No.250298.000682 comprise, without limitation, Cw*0101, Cw*0202, Cw*0303, Cw*0401, Cw*0501, Cw*0602, Cw*0701, Cw*0702, Cw*0802, Cw*1203, Cw*1401, Cw*1502, Cw*1601, Cw*1701, and. Cw*1801. Non-limiting examples of HLA-DR alleles comprise, without limitation, DRB1*0101, DRB1*0103, DRB1*1501, DRB1*1502, DRB1*1601, DRB1*1602, DRB1*0301, DRB1*0401, DRB1*0404, DRB1*1101, DRB1*1201, DRB1*1301 , DRB1*1302, DRB1*1401, DRB1*1402, DRB1*0701, DRB1*0801, DRB1*0802, DRB1*0803, DRB1*0901, and DRB1*1001. [00186] In some embodiments, the MHC class I molecule may be selected from HLA-A*02, HLA-A*01, HLA-A*03, HLA-A*11, HLA-A*23, HLA-A*24, HLA-B*07, HLA-B*08, HLA-B*40, HLA-B*44, HLA-B*15, HLA-C*04, HLA*C*03 HLA-C*07. There are also allelic variants of the above HLA types, all of which are encompassed by the present disclosure. In some embodiments, the MHC molecule may be HLA-A*02 or HLA-A*11. [00187] The MHC molecules used herein may also be from any other mammalian or avian species, for example, non-human primates, rodents (e.g., mice), rabbits, equines, bovines, canines, felines, pigs, etc. [00188] Naturally occurring MHC class I molecules bind peptides derived from proteolytically degraded proteins, especially endogenously synthesized proteins, by a cell. Small peptides obtained accordingly are transported into the endoplasmic reticulum where they associate with nascent MHC class I molecules before being routed through the Golgi apparatus and displayed on the cell surface for recognition by cytotoxic T lymphocytes. [00189] Naturally occurring MHC class I molecules consist of an α (heavy) chain associated with β2-microglobulin. The heavy chain consists of subunits α1-α3. The β2- microglobulin protein and α3 subunit of the heavy chain are associated. In certain embodiments, β2-microglobulin and α3 subunit are covalently bound. In certain embodiments, β2-microglobulin and α3 subunit are non-covalently bound. The α1 and α2 subunits of the heavy chain fold to form a groove for a peptide, e.g., antigenic determinant, to be displayed and recognized by TCR. [00190] Class I molecules can bind peptides of about 8-10 amino acids in length. All humans have between three and six different class I molecules, which can each bind many different types of peptides.
Attorney Docket No.250298.000682 [00191] In some embodiments, the MHC contained in the pMHC complexes of the disclosure comprises (i) a class I MHC polypeptide or a fragment, mutant or derivative thereof, and, optionally, (ii) a β2 microglobulin polypeptide or a fragment, mutant or derivative thereof. In one specific embodiment, the class I MHC polypeptide is linked to the β2 microglobulin polypeptide by a peptide linker. [00192] In one specific embodiment, the class I MHC polypeptide is a human class I MHC polypeptide selected from the group consisting of HLA-A, HLA-B, HLA-C, HLA- E, HLA-F, and HLA-G. In another specific embodiment, the class I MHC polypeptide is a murine class I MHC polypeptide selected from the group consisting of H-2K, H-2D, H- 2L, H2-IA, H2-IB, H2-IJ, H2-IE, and H2-IC. [00193] In some embodiments, the peptide disclosed herein forms a complex with one or more MHC class I α heavy chains. In some embodiments, the MHC class I α heavy chain is fully human. In some embodiments, the MHC class I α heavy chain is humanized. Humanized MHC class I α heavy chains are described, e.g., in U.S. Pat. Pub. Nos. 2013/0111617, 2013/0185819 and 2014/0245467. In some embodiments, the MHC class I α heavy chain comprises a human extracellular domain (human α1, α2, and/or α3 domains) and a cytoplasmic domain of another species. In some embodiments, the class I α heavy chain polypeptide is HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, HLA-G, HLA- K, or HLA-L. In some embodiments, the HLA-A sequence can be an HLA-A*0201 sequence. In various aspects, the peptide-MHC can include all the domains of an MHC class I heavy chain. [00194] In some embodiments, the MHC molecule comprises a β2-microglobulin. In some embodiments, the β2-microglobulin is fully human. In some embodiments, the β2-microglobulin is humanized. [00195] In some embodiments, the MHC class I molecule comprises a mutation in a β2-microglobulin (β2m or Β2M) polypeptide and in the Heavy Chain sequence to effect a disulfide bond between the Β2M and the Heavy Chain. In some cases, the Heavy Chain is an HLA and wherein the disulfide bond links one of the following pairs of residues: Β2M residue 12, HLA residue 236; Β2M residue 12, HLA residue 237; Β2M residue 8, HLA residue 234; Β2M residue 10, HLA residue 235; Β2M residue 24, HLA residue 236; Β2M residue 28, HLA residue 232; Β2M residue 98, HLA residue 192; Β2M residue 99, HLA
Attorney Docket No.250298.000682 residue 234; Β2M residue 3, HLA residue 120; Β2M residue 31, HLA residue 96; Β2M residue 53, HLA residue 35; Β2M residue 60, HLA residue 96; Β2M residue 60, HLA residue 122; Β2M residue 63, HLA residue 27; Β2M residue Arg3, HLA residue Gly120; Β2M residue His31, HLA residue Gln96; Β2M residue Asp53, HLA residue Arg35; Β2M residue Trp60, HLA residue Gln96; Β2M residue Trp60, HLA residue Asp122; Β2M residue Tyr63, HLA residue Tyr27; Β2M residue Lys6, HLA residue Glu232; Β2M residue Gln8, HLA residue Arg234; Β2M residue Tyr10, HLA residue Pro235; Β2M residue Ser11, HLA residue Gln242; Β2M residue Asn24, HLA residue Ala236; Β2M residue Ser28, HLA residue Glu232; Β2M residue Asp98, HLA residue His192; and Β2M residue Met99, HLA residue Arg234, first linker position Gly2, Heavy Chain (HLA) position Tyr84; Light Chain (Β2M) position Arg12, HLA Ala236; and/or Β2M residue Arg12, HLA residue Gly237. [00196] In some embodiments, the antigenic determinant amino acid sequence can be that of a peptide described herein, which can be presented by an MHC class I molecule. In certain embodiments, the sequence can comprise from about 8 to about 15 contiguous amino acids. In certain embodiments, the sequence can comprise from about 8 to about 12 contiguous amino acids. [00197] In some embodiments, at least one chain of the MHC and the peptide are comprised within a fusion protein. In one specific embodiment, the MHC and the peptide are separated by a linker sequence. For example, the single chain molecule can comprise, from amino to carboxy terminal, an antigenic determinant, a β2-microglobulin sequence, and a class I α (heavy) chain sequence. Alternatively, the single chain molecule can comprise, from amino to carboxy terminal, an antigenic determinant, a class I α (heavy) chain sequence, and a β2-microglobulin sequence. The single-chain molecule can further comprise a signal peptide sequence at the amino terminal. In certain embodiments, there can be a linker sequence between the peptide sequence and the β2-microglobulin sequence. In certain embodiments, there can be a linker sequence between the β2-microglobulin sequence and the class I α (heavy) chain sequence. A single-chain molecule can further comprise a signal peptide sequence at the amino terminal, as well as first linker sequence extending between the peptide sequence and the β2-microglobulin sequence, and/or a second linker sequence extending between the β2-microglobulin sequence and the class I
Attorney Docket No.250298.000682 heavy chain sequence. In certain embodiments, the β2-microglobulin and the class I α (heavy) chain sequences can be human, murine, or porcine. [00198] In some embodiments, a single-chain molecule can comprise a first flexible linker between the peptide ligand segment and the β2-microglobulin segment. For example, linkers can extend from and connect the carboxy terminal of the peptide ligand segment to the amino terminal of the β2-microglobulin segment. Preferably, the linkers are structured to allow the linked peptide ligand to fold into the binding groove resulting in a functional MHC-antigen peptide. In some embodiments, this linker can comprise at least about 10 amino acids, up to about 15 amino acids. In some embodiments, a single- chain molecule can comprise a second flexible linker inserted between the β2- microglobulin and heavy chain segments. For example, linkers can extend from and connect the carboxy terminal of the β2-microglobulin segment to the amino terminal of the heavy chain segment. In certain embodiments, the β2-microglobulin and the heavy chain can fold into the binding groove resulting in a molecule which can function in promoting T cell expansion. [00199] Suitable linkers used in the MHCs can be of any of a number of suitable lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and can be 1, 2, 3, 4, 5, 6, or 7 amino acids. Non-limiting examples of linkers include, e.g., glycine polymers (G)n, glycine-serine polymers (including, for example, (GS)n, (GSGGS)n (SEQ ID NO: 92) and (GGGS)n (SEQ ID NO: 93), where n is an integer of at least one), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers. Glycine and glycine-serine polymers can be used; both Gly and Ser are relatively unstructured, and therefore can serve as a neutral tether between components. Glycine polymers can be used; glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains). Exemplary linkers can comprise amino acid sequences including, but not limited to, those listed in Table 2. In some embodiments, a linker peptide includes a cysteine residue that can form a disulfide bond with a cysteine residue present in a second polypeptide.
Attorney Docket No.250298.000682 Table 2. Examples of Linker Sequences Linker amino SEQ Linker codon- SEQ Linker codon- SEQ acid sequence ID optimized nucleotide ID optimized nucleotide ID :
Attorney Docket No.250298.000682 GGGGSGGGGS 109 GGAGGTGGAGGT 125 GGGGGTGGAGGAT 141 GGGGSGGGGS AGTGGCGGTGGT CAGGAGGCGGTGG
de covalently attached to an MHC class I α (heavy) chain via a disulfide bridge (i.e., a disulfide bond between two cystines). In certain embodiments, the disulfide bond comprises a first cysteine, comprising a linker extending from the carboxy terminal of an antigen peptide, and a second cysteine comprising an MHC class I heavy chain (e.g., an MHC class I α (heavy) chain which has a non-covalent binding site for the antigen peptide). In certain embodiments, the second cysteine can be a mutation (addition or substitution) in the MHC class I α (heavy) chain. In certain embodiments, the single-chain molecule can comprise one contiguous polypeptide chain as well as a disulfide bridge. In certain embodiments, the single-chain molecule can comprise two contiguous polypeptide chains which are attached via the disulfide bridge as the only covalent linkage. In some embodiments, the linking sequences can comprise at least one amino acid in addition to the Cys residues, including one or more Gly residues, one or more Ala residues, and/or one or more Ser residues. [00201] In certain embodiments, the disulfide bridge can link an antigen peptide described herein in the class I groove of the pMHC complex if the pMHC complex comprises a first cysteine in a Gly-Ser linker extending between the C-terminus of the peptide and the β2-microglobulin, and a second cysteine in a proximal heavy chain position. [00202] Attaching the peptide to the MHC class I or MHC class II molecule via a flexible linker has the can help ensure that the peptide will occupy and stay associated with the MHC molecule during biosynthesis, transport, and display. However, there may be situations in which this linker can interfere with peptide binding to the MHC molecule or with TCR recognition of the complex. As an alternate approach, in some embodiments, the MHC molecule and the peptide are expressed separately. [00203] In some embodiments, the β2-microglobulin sequence can comprise a full- length β2-microglobulin sequence. In certain embodiments, the β2-microglobulin
Attorney Docket No.250298.000682 sequence lacks the leader peptide sequence. As such, in some configurations, the β2- microglobulin sequence can comprise about 99 amino acids, and can be a mouse ^2- microglobulin sequence (e.g., GenBank Accession No. X01838). In some other configurations, the β2-microglobulin sequence can comprise about 99 amino acids, and can be a human ^2-microglobulin sequence (e.g., GenBank Accession No. AF072097.1). [00204] In some embodiments, the pMHC complex can contain MHC sequences as disclosed in U.S. Patent Nos.4,478,823; 6,011,146; 8,518,697; 8,895,020; 8,992,937; WO 96/04314; Mottez et al. J. Exp. Med. 181: 493-502, 1995; Madden et al. Cell 70: 1035- 1048, 1992; Matsumura et al., Science 257: 927-934, 1992; Mage et al., Proc. Natl. Acad. Sci. USA 89: 10658-10662, 1992; Toshitani et al, Proc. Nat’l Acad. Sci.93: 236-240, 1996; Chung et al, J. Immunol.163:3699-3708, 1999; Uger and Barber, J. Immunol.160: 1598- 1605, 1998; Uger et al., J. Immunol.162, pp.6024-6028, 1999; White et al., J. Immunol. 162: 2671-2676, 1999; Yu et al., J. Immunol. 168:3145-3149, 2002; Truscott et al., J. Immunol. 178: 6280–6289, 2007, all of which are incorporated by reference in their entireties. [00205] In some embodiments, the MHC comprises a class II MHC polypeptide or a fragment, mutant or derivative thereof. In one specific embodiment, the MHC comprises α and β polypeptides of a class II MHC complex or a fragment, mutant or derivative thereof. In one specific embodiment, the α and β polypeptides are linked by a peptide linker. In one specific embodiment, the MHC comprises α and β polypeptides of a human class II MHC complex selected from the group consisting of HLA-DP, HLA-DR, HLA- DQ, HLA-DM and HLA-DO. In another specific embodiment, the MHC comprises α and β polypeptides of a murine H-2A or H-2E class II MHC complex. [00206] Naturally occurring MHC class II molecules can contain two polypeptide chains, α and β. The chains may come from the DP, DQ, or DR gene groups. There are about 40 known different human MHC class II molecules. All have the same basic structure but can vary subtly in their molecular structure. MHC class II molecules can bind peptides of 13-18 amino acids in length. [00207] In some embodiments, the MHC class II α chain is fully human. In some embodiments, the MHC class II α chain is humanized. Humanized MHC class II α chains are described, e.g., in U.S. Pat. Nos. 8,847,005, 9,043,996, and 10,154,658, which are
Attorney Docket No.250298.000682 incorporated herein by reference in their entireties. In some embodiments, the humanized MHC class II α chain polypeptide comprises a human extracellular domain and a cytoplasmic domain of another species. In some embodiments, the class II α chain is HLA- DMA, HLA-DOA, HLA-DPA, HLA-DQA or HLA-DRA. In some embodiments, the class II α chain polypeptide is humanized HLA-DMA, HLA-DOA, HLA-DPA, HLA-DQA and/or HLA-DRA. [00208] In some embodiments, the peptide of the present disclosure forms a complex with one or more MHC class II β chains. In some embodiments, the MHC class II β chain is fully human. In some embodiments, the MHC class II β chain polypeptide is humanized. Humanized MHC class II β chain polypeptides are described, e.g., in U.S. Pat. Nos.8,847,005, 9,043,996, and 10,154,658, which are incorporated herein by reference in their entireties. In some embodiments, the humanized MHC class II β chain comprises a human extracellular domain and a cytoplasmic domain of another species. In some embodiments, the class II β chain is HLA-DMB, HLA-DOB, HLA-DPB, HLA-DQB or HLA-DRB. In some embodiments, the class II β chain is humanized HLA-DMB, HLA- DOB, HLA-DPB, HLA-DQB and/or HLA-DRB. [00209] The pMHC complexes of the disclosure may be isolated and/or in a substantially pure form. For example, the complex may be provided in a form which is substantially free of other peptides or proteins. MHC molecules as disclosed herein can include recombinant MHC molecules, non-naturally occurring MHC molecules, and functionally equivalent fragments of MHC, including derivatives or variants thereof, provided that peptide binding is retained. For example, MHC molecules may be fused to a therapeutic moiety, attached to a solid support, in soluble form, attached to a tag, biotinylated and/or in multimeric form. A peptide disclosed herein may be covalently attached to the MHC. [00210] Methods to produce soluble recombinant MHC molecules with which peptides disclosed herein can form a complex include, but are not limited to, expression and purification from E. coli cells or insect cells. Alternatively, MHC molecules may be produced synthetically, or using cell free systems. [00211] The peptides disclosed herein may be presented on the surface of a cell in complex with MHC. Thus, the present disclosure also provides a cell presenting on its
Attorney Docket No.250298.000682 surface a pMHC complex disclosed herein. Such a cell may be a mammalian cell, preferably a cell of the immune system, and a specialized antigen-presenting cell (APC) such as a dendritic cell or a B cell. Other preferred cells include T2 cells. Cells presenting the peptide or pMHC complex of the disclosure may be isolated, preferably in the form of a homogenous population, or provided in a substantially pure form. Such cells may not naturally present the complex of the disclosure, or alternatively said cells may present the complex at a level higher than they would in nature. Such cells may be obtained by pulsing said cells with one or more peptides (e.g., 2 to 50, 2 to 40, 2 to 30, 5 to 25, 5 to 20, or 10 to 15 peptides) of the disclosure, or genetically modifying the cells (via DNA or RNA transfer) to express one or more peptides (e.g., 2 to 50, 2 to 40, 2 to 30, 5 to 25, 5 to 20, or 10 to 15 peptides) of the disclosure. Pulsing involves incubating the cells with the peptide for several hours using peptide concentrations typically ranging from 10−5 to 10−12 M. Such cells may additionally be transduced with HLA molecules, such as HLA-A*02 to further induce presentation of the peptide(s). Cells may be produced recombinantly. Cells presenting peptides of the disclosure may be used to isolate T cells and TCRs which are activated by, or bind to, the cells. Fusion Proteins, Conjugates and Oligomeric Complexes Disclosed Herein [00212] Peptides or pMHC complexes disclosed herein may be fused or conjugated to one or more heterologous molecules. Peptides or pMHC complexes of the disclosed herein may also be in multimeric form. Accordingly, the present disclosure also provides fusion proteins, conjugates, and oligomeric complexes comprising a peptide or a pMHC complex of the disclosure. [00213] In some embodiments, peptides are fused or conjugated to one or more heterologous molecules which includes an MHC molecule (or fragments thereof). [00214] Heterologous molecules suitable for genetical fusion and/or chemical conjugation with the peptides or the pMHC complexes of the disclosure include, but are not limited to, peptides, polypeptides, small molecules, polymers, nucleic acids, lipids, sugars, etc. The heterologous molecule(s) may be fused at the N- and/or C-terminus of the peptide and/or another polypeptide chain in the pMHC complex.
Attorney Docket No.250298.000682 [00215] Heterologous peptides and polypeptides include, but are not limited to, an epitope (e.g., FLAG) or a tag sequence (e.g., His6 (SEQ ID NO: 147), and the like) to allow for the detection and/or isolation of a fusion protein; a transmembrane receptor protein or a portion thereof, such as an extracellular domain or a transmembrane and intracellular domain; a ligand or a portion thereof which binds to a transmembrane receptor protein; an enzyme or portion thereof which is catalytically active; a polypeptide or peptide which promotes oligomerization, such as a leucine zipper domain; a polypeptide or peptide which increases stability, such as an immunoglobulin constant region (e.g., an Fc domain); a half- life-extending sequence comprising a combination of two or more (e.g., 2, 5, 10, 15, 20, 25, etc.) naturally occurring or non-naturally occurring charged and/or uncharged amino acids (e.g., Ser, Gly, Glu or Asp) designed to form a predominantly hydrophilic or predominantly hydrophobic fusion partner for a fusion protein; a functional or non- functional antibody (e.g., an antibody that is specific for dendritic cells), or a heavy or light chain thereof; and a polypeptide which has an activity, such as a therapeutic activity, different from fusion proteins of the present disclosure. In some embodiments, the one or more heterologous molecules enhances a peptide-specific immune response in a subject. In some embodiments, the one or more heterologous molecules mediates peptide delivery to a specific site within a subject. [00216] In some embodiments, fusion proteins of the disclosure may comprise one or more affinity tags, e.g., to allow for affinity purification or coupling to another molecule. Examples of affinity tags include, but are not limited to, a His6 tag (SEQ ID NO: 147), an Avi-tag, a biotin, a hemagglutinin (HA) tag, a FLAG tag, a Myc tag, a GST tag, a MBP tag, a chitin binding protein tag, a calmodulin tag, a V5 tag, a streptavidin binding tag, a green fluorescent protein (GFP), YFP, RFP, CFP, mCherry, tdTomato, SUMO tag, and Ubiquitin tag. [00217] In some embodiments, fusion proteins of the disclosure may comprise one or more epitopes that is not present in the antigen. One such example is the use of fusion peptides where a promiscuous T helper epitope is covalently linked (e.g., via a polypeptide linker or spacer) to the peptide sequence. Non-limiting examples of promiscuous T helper epitopes include the PADRE peptide, tetanus toxoid peptide (830-843), or influenza haemagglutinin, HA(307-319).
Attorney Docket No.250298.000682 [00218] Peptides or pMHC complexes of the disclosure may be conjugated to additional moieties such as carrier molecules or adjuvants for use as vaccines. Examples of adjuvants used in vaccines include microbes, such as the bacterium Bacillus Calmette- Guérin (BCG), and/or substances produced by bacteria, such as Detox B (an oil droplet emulsion of monophosphoryl lipid A and mycobacterial cell wall skeleton). KLH (keyhole limpet hemocyanin), bovine serum albumin (BSA), the E2 core protein of the pyruvate dehydrogenase complex are examples of suitable carrier proteins used in vaccine compositions. Additional examples of carrier proteins suitable for use in the compositions of the present disclosure include, but are not limited to, ovalbumin (OVA), blue carrier protein (BCP), thyroglobulin (THY), a soybean trypsin inhibitor (STI), and multiple attachment peptide (MAP), albumin, serum albumin, c-reactive protein, conalbumin, lactalbumin, ion carrier protein, acyl carrier protein, signal transduction adapter protein, androgen binding protein, calcium binding protein, calmodulin binding protein, ceruloplasmin, cholesterol Ester transfer protein, f box protein, fatty acid binding protein, follistatin, follistatin related protein, GTP binding protein, insulin-like growth factor binding protein, iron binding protein, latent TGFbeta binding protein, light-harvesting protein complex, lymph Sphere antigen, membrane transport protein, neurophysin, periplasmic binding protein, phosphate binding protein, phosphatidylethanolamine binding protein, phospholipid transport protein, retinol binding protein, RNA binding protein, s- phase kinase related protein, sex hormone binding globulin, Thyroxine binding protein, transcobalamin, transcortin, transferrin binding protein, and/or vitamin D binding protein. [00219] As a further example, a peptide or pMHC complex of the present disclosure may be fused to, for example, the 80 N-terminal amino acids of the HLA-DR antigen- associated invariant chain (p33 or Ii) as derived from the NCBI, GenBank Accession- number X00497). The Ii fragment may facilitate an efficient introduction of the peptide or pMHC complex into the cells. [00220] Peptides or pMHC complexes of the present disclosure may also be attached, covalently (e.g., via a linker) or non-covalently, to a moiety capable of eliciting a therapeutic effect, such as antibodies, or cytokines, such as interleukin 2, interferon-α, and granulocyte-macrophage colony-stimulating factor. Alternatively or additionally, the peptides or pMHC complexes may be encapsulated into liposomes.
Attorney Docket No.250298.000682 [00221] Other suitable heterologous molecules include, but are not limited to, fluorescent, or luminescent labels, radiolabels, nucleic acid probes, and contrast reagents, antibodies, or enzymes that produce a detectable product. Methods for detecting heterologous molecules may include flow cytometry, microscopy, electrophoresis, or scintillation counting. [00222] In some embodiments, peptides or pMHC complexes of the disclosure may be conjugated with fluorocarbon to increase cellular immunogenicity. Where the peptide or another polypeptide chain of the pMHC complex is linked to a fluorocarbon, the terminus of the peptide or polypeptide chain, such as the terminus that is not conjugated to the fluorocarbon, or other attachment, can be altered, for example to promote solubility of the fluorocarbon-peptide/polypeptide construct via the formation of micelles. To facilitate large-scale synthesis of the construct, the N- or C-terminal amino acid residues of the peptide or another polypeptide chain of the pMHC complex can be modified. When the desired peptide or another polypeptide chain of the pMHC complex is particularly sensitive to cleavage by peptidases, the normal peptide bond can be replaced by a non-cleavable peptide mimetic. Such bonds and methods of synthesis are well known in the art. [00223] Peptides or pMHC complexes of the disclosure may be provided in soluble form, or may be immobilized by attachment to a suitable solid support. Examples of solid supports include, but are not limited to, a bead, a membrane, sepharose, a magnetic bead, a plate, a tube, a column. pMHC complexes may be attached to an ELISA plate, a magnetic bead, or a surface plasmon resonance biosensor chip. Methods of attaching peptides or pMHC complexes to a solid support are known to the skilled person, and include, for example, using an affinity binding pair, e.g. biotin and streptavidin, or antibodies and antigens. In some embodiments, peptides or pMHC complexes are labeled with biotin and attached to streptavidin-coated surfaces. [00224] Peptides or pMHC complexes of the disclosure may be in multimeric form, for example, dimeric, or tetrameric, or pentameric, or octameric, or greater. Accordingly, in some aspects, the present disclosure provides oligomeric complexes comprising the peptides or pMHC complexes of the present disclosure. As used herein, the terms “oligomer”, “oligomeric”, “oligomerize” and “oligomerization” or the like encompass a dimer, trimer, tetramer, pentamer, hexamer, heptamer, octamer, or higher species of
Attorney Docket No.250298.000682 polymerized monomers that comprise the peptide or pMHC complex. Having multiple copies of the peptides or pMHC complexes in a large complex may enhance their biological activity, e.g., immunogenic activity. [00225] For example, the peptides of the disclosure may be oligomerized using the biotin/streptavidin system. Biotinylated analogs of peptide monomers may be synthesized by standard techniques. For example, the peptide may be C-terminally biotinylated. These biotinylated peptide monomers are then oligomerized by incubation with streptavidin [e.g., at a 4:1 molar ratio at room temperature in phosphate buffered saline (PBS) or HEPES- buffered RPMI medium for 1 hour]. In a variation of this embodiment, biotinylated peptide monomers may be oligomerized by incubation with anti-biotin antibodies [e.g., goat anti- biotin IgG]. [00226] In general, oligomeric pMHC complexes may be produced using pMHC tagged with a biotin residue and complexed through fluorescently labeled streptavidin. A biotinylation site may be introduced to the pMHC complex to which biotin can be added, for example, using the BirA enzyme. Alternatively, oligomeric pMHC complexes may be formed by using immunoglobulin as a molecular scaffold. In this system, the extracellular domains of MHC molecules are fused with the constant region of an immunoglobulin heavy chain separated by a short amino acid linker. Oligomeric pMHC complexes have also been produced using carrier molecules such as dextran. Oligomeric pMHC complexes can be useful for improving the detection of binding moieties, such as T cell receptors, which bind said complex, because of avidity effects. [00227] In other embodiments, the peptides or pMHC complexes of the disclosure can be oligomerized by covalent attachment to at least one linker. The linker moiety can be a peptide linker, such as those described herein (e.g., in Table 2). In some embodiments, polyethylene glycol (PEG) may serve as the linker that oligomerizes the peptide monomers. For example, a single PEG moiety may be simultaneously attached to the N-termini of both peptide chains of a peptide dimer. [00228] Alternatively, oligomeric peptide or pMHC complexes may also contain one or more intramolecular disulfide bonds between cysteine residues of the peptide or pMHC monomers. Preferably, the two monomers contain at least one intramolecular disulfide bond. Most preferably, both monomers contain an intramolecular disulfide bond,
Attorney Docket No.250298.000682 such that each monomer contains a cyclic group. Such disulfide bonds may be formed by oxidation of the cysteine residues of the peptide core sequence. In one embodiment the control of cysteine bond formation is exercised by choosing an oxidizing agent of the type and concentration effective to optimize formation of the desired isomer. For example, oxidation of a peptide dimer to form two intramolecular disulfide bonds (one on each peptide chain) is preferentially achieved (over formation of intermolecular disulfide bonds) when the oxidizing agent is DMSO. The formation of cysteine bonds can be controlled by the selective use of thiol-protecting groups during peptide synthesis. [00229] In some embodiments, peptides or pMHC complexes described herein may be fused or conjugated to a dimerization moiety. The dimerization moiety may contain, for example, an immunoglobulin domain, such as from an IgG antibody (e.g., human IgG), which connects two monomers generating a homodimer or heterodimer molecule. As a non-limiting example, the dimerization motif in the proteins according to the present disclosure may be constructed to include a hinge region and an immunoglobulin domain (e.g. Cy3 domain), e.g., carboxyterminal C domain (CH3 domain), or a sequence that is substantially identical to the C domain. The hinge region may be Ig derived and contributes to the dimerization through the formation of an interchain covalent bond(s), e.g. disulfide bridge(s). In addition, such homodimer or heterodimer molecules may further comprise one or more targeting moieties that bind to target molecules present on, for example, antigen-presenting cells (APCs) such as dendritic cells or B cells. In such instances, the hinge region may function as a flexible spacer between the domains allowing the two targeting units to bind simultaneously to two target molecules on the APC expressed with variable distances. The immunoglobulin domains contribute to dimerization through non- covalent interactions, e.g. hydrophobic interactions. In a preferred embodiment the CH3 domain is derived from IgG. These dimerization moieties may be exchanged with other multimerization moieties from e.g., other Ig isotypes/subclasses. Preferably the dimerization motif is derived from native human proteins, such as human IgG. Examples of such homodimer protein construct are described in US 10,590,195, which is incorporated herein by reference in its entirety.
Attorney Docket No.250298.000682 Nucleic Acids and Vectors [00230] In another aspect, the disclosure provides an isolated polynucleotide comprising a nucleic acid sequence encoding one or more peptide(s) and/or peptide-based molecules (such as complexes (e.g., pMHC complexes), fusion proteins, or conjugates comprising the described peptides) of the disclosure. The polynucleotide may be, for example, DNA, cDNA, PNA, RNA or combinations thereof, either single- and/or double- stranded, or native or stabilized forms of polynucleotides, such as, for example, polynucleotides with a phosphorothioate backbone and it may or may not contain introns so long as it codes for the peptide. [00231] In some embodiments, the polynucleotide described herein encodes a peptide comprising an amino acid sequence that is at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99% identical to the amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146, or a fragment or derivative thereof. In some embodiments, the polynucleotide described herein encodes a peptide comprising an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146, or a fragment or derivative thereof. [00232] In some embodiments, the polynucleotide described herein encodes more than one peptide selected from any one of SEQ ID NOs: 1-89 and 143-146, or a fragment or derivative thereof . For example, the polynucleotide described herein may encodes 2 to 50, 2 to 40, 2 to 30, 5 to 25, 5 to 20, or 10 to 15 peptides as described herein (e.g., SEQ ID NO: 1-89 and 143-146), or a fragment or derivative thereof. The peptides may be arranged in any order, and may be identical or different. [00233] In some embodiment, the polynucleotide described herein encodes 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, or 93 amino acid sequences selected from SEQ ID NO: 1-89 and 143-146, or a fragment or derivative thereof. [00234] In some embodiment, the polynucleotide described herein is a DNA molecule.
Attorney Docket No.250298.000682 [00235] Methods to deliver DNA to a subject include, for example, direct delivery, as naked DNA. Delivery may also be achieved by nanoparticles; gene gun, microneedle array and in situ electroporation. The nucleic acids can also be administered using ballistic delivery. Particles comprised solely of DNA can be administered. Alternatively, DNA can be adhered to particles, such as gold particles. [00236] In some embodiments, the polynucleotide described herein is an RNA molecule. For example, the RNA molecule may be mRNA or a self-replicating RNA. [00237] A polynucleotide encoding RNA disclosed herein can be used to make a vaccine. RNA cannot integrate into the genome and has therefore little oncogenic potential; thus, RNA can be useful for making a vaccine. Also, RNA only needs to enter the cytoplasm, contrary to DNA which needs to enter the nucleus. An RNA molecule disclosed herein may be chemically modified and/or incorporate modified nucleosides to overcome susceptibility to degradation. RNA vaccines may comprise mRNA and/or self-replicating RNA (also known as RNA replicons). Delivery techniques for RNA vaccines may also encompass, for example, condensation with protamine and encapsulation into liposomes or nanoparticles.. [00238] The nucleic acids (either DNA or RNA) can also be delivered complexed to cationic compounds, such as cationic lipids. Lipid-mediated gene delivery methods are described, for example, in WO 91/06309; WO 93/24640; WO 96/18372; U.S. Pat No. 5,279,833, which are incorporated herein by reference in their entireties. [00239] A nucleic acid molecule described herein may be generated synthetically. One method is the phosphoramidite method. Without wishing to be bound by theory, in this chemistry, a phosphoramidite (a nucleoside with side protecting groups that preserve the integrity of the sugar, the phosphodiester linkage, and the base during chain extension steps) is coupled through its reactive 3' phosphorous group to the 5' hydroxyl group of a nucleoside immobilized on a solid support column. The steps of oligonucleotide synthesis can include the following: (1) Detritylation, in which the dimethoxytrityl (DMT or trityl) group on the 5' hydroxyl of the support nucleoside is removed by treatment with trichloroacetic acid (TCA). (2) In the coupling step, a phosphoramidite, made reactive by tetrazole (a weak acid), is chemically coupled to the last base added to the column support material. (3) In the capping step, any free 5' hydroxyl groups of unreacted column
Attorney Docket No.250298.000682 nucleotides are acetylated by treatment with acetic anhydride and N-methylimidazole. (4) In the oxidation step, the unstable internucleotide phosphate linkage between the previously coupled base and the most recently added base is oxidized by treatment with iodine and water to a more stable phosphotriester linkage. Following coupling of all bases in the oligonucleotide's sequence, the completed nucleic acid chain may be cleaved from the column by treatment with ammonium hydroxide, and the base protecting groups are removed by heating in the ammonium hydroxide solution. [00240] By way of a non-limiting example, a synthesis cycle may comprise growth of the nucleotide chain from an initial protected nucleoside derivatized via its terminal 3′ hydroxyl to a solid support. Reagents and solvents can be pumped through the support to induce the consecutive removal and addition of sugar protecting groups in order to isolate the reactivity of a specific chemical moiety on the monomer and effect its stepwise addition to the growing oligonucleotide chain. Assembly of the protected oligonucleotide chain can be carried out in chemical steps, for example, without limitation, deblocking, activation/coupling, oxidation, and capping. Cleavage and deprotection then reveal the single-stranded nucleic acid. [00241] Nucleic acid synthesis methods disclosed herein can comprise, for example, oligonucleotide synthesis, column-based oligonucleotide synthesis, microarray-based oligonucleotide synthesis, gene synthesis from oligonucleotides, gene synthesis from array-derived oligonucleotide pools, and any of various error correction and sequence validation steps, or any combination thereof. [00242] RNA chemical synthesis may be similar to that used for DNA. In some embodiments, RNA chemical synthesis methods may comprise an additional protecting group at the 2′ hydroxyl of ribose. The 2′ hydroxyl of ribose position may be protected with tert-butyldimethyl silyl groups, which can be stable throughout the synthesis, and can be removed at the final deprotection step by addition of a basic fluoride ion such as tetrabutylammonium fluoride (TBAF). The remaining positions on both the sugar and the bases can be protected in the same fashion as for DNA. By adjusting several parameters in the DNA synthesis protocol such as, but not limited to the coupling times, monomer delivery rate, frequency of washing steps, and types of capping reagents, stepwise coupling efficiencies of up to 99% can be obtained.
Attorney Docket No.250298.000682 [00243] Viral nucleic acid synthesis may be catalyzed by both viral and host enzymes, the relative contribution of which can be determined by the type of virus and the specific molecule. Viruses with RNA genomes, except for the retroviruses, synthesize mRNA and replicate their genomes using virus-encoded RNA-dependent RNA polymerases. In contrast, retroviruses synthesize a double-stranded complementary DNA (cDNA) copy of their single-stranded RNA genome using a virion-encoded RNA- dependent DNA polymerase (reverse transcriptase). In subsequent steps, the retroviral cDNA may be integrated into the host chromosome and transcribed by host-encoded DNA- dependent RNA polymerase II (pol II) to yield viral messages and genomic RNA. DNA viruses, except for poxviruses, also use host-encoded pol II to transcribe their messages. Poxviruses, because they replicate in the cytoplasm and do not have access to pol II, assemble a novel transcriptase composed of multiple poxvirus-specific (and possibly one or more host-derived) subunits. Most DNA virus families (e.g. Poxviridae, Iridoviridae, Herpesviridae, Adenoviridae) synthesize a virus-encoded DNA-dependent, DNA polymerase. However, two families (i.e., Parvoviridae and Papovaviridae) utilize host DNA polymerase, and the Hepadnaviridae replicate viral DNA through an RNA intermediate using a virus-encoded reverse transcriptase. [00244] Due to the degeneracy of the genetic code, nucleic acid molecules of different nucleotide sequence can encode the same amino acid sequence. For expression in various hosts, the polynucleotides may be codon-optimized. [00245] In a further aspect, the disclosure provides a vector comprising a nucleic acid sequence according to the third aspect of the disclosure. The vector may include, in addition to a nucleic acid sequence encoding only a peptide of the disclosure, one or more additional nucleic acid sequences encoding one or more additional peptides. Such additional peptides may, once expressed, be fused to the N-terminus or the C-terminus of the peptide of the disclosure. Examples of such additional peptides are detailed in the sections above. In one embodiment, the vector includes a nucleic acid sequence encoding a peptide or protein tag such as, for example, a biotinylation site, a FLAG-tag, a MYC-tag, an HA-tag, a GST-tag, a Strep-tag or a poly-histidine tag. [00246] The vector utilized in the context of the present disclosure desirably comprises sequences appropriate for introduction into cells. For instance, the vector may
Attorney Docket No.250298.000682 be an expression vector, a vector in which the coding sequence of the polypeptide is under the control of its own cis-acting regulatory elements, a vector designed to facilitate gene integration or gene replacement in host cells, and the like. [00247] In the context of the present disclosure, the term “vector” encompasses a DNA molecule, such as a plasmid, bacteriophage, phagemid, virus or other vehicle, which contains one or more heterologous or recombinant nucleotide sequences (e.g., an above- described nucleic acid molecule of the disclosure, under the control of a functional promoter and, possibly, also an enhancer) and is capable of functioning as a vector in the sense understood by those of ordinary skill in the art. [00248] The following vectors are provided by way of example: bacteriophages such as lambda (X) bacteriophage, EMBL bacteriophage; bacterial vectors such as pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a; pTrc99A, pKK223-3, pKK233-3, pDR540, and pRIT5; eukaryotic vectors such as pWLneo, pSV2cat, pOG44, PXR1, pSG, pSVK3, pBPV, pMSG and pSVL; and transposons such as Sleeping Beauty transposon and PiggyBac transposon. [00249] In some embodiments, the vector is a viral vector. Viral vectors can be derived from naturally occurring virus genomes, which typically are modified to be replication incompetent, e.g. non-replicating. Non-replicating viruses require the provision of proteins in trans for replication. Typically, those proteins are stably or transiently expressed in a viral producer cell line, thereby allowing replication of the virus. The viral vectors are, thus, typically infectious and non-replicating. Viral vectors may be adenovirus vectors, adeno-associated virus (AAV) vectors (e.g., AAV type 5 and type 2), alphavirus vectors (e.g., Venezuelan equine encephalitis virus (VEE), Sindbis virus (SIN), Semliki forest virus (SFV), and VEE-SIN chimeras), herpes virus vectors (e.g., vectors derived from cytomegaloviruses, like rhesus cytomegalovirus (RhCMV)), arena virus vectors (e.g. lymphocytic choriomeningitis virus (LCMV) vectors), measles virus vectors, pox virus vectors (e.g., vaccinia virus, modified vaccinia virus Ankara (MVA), NYVAC (derived from the Copenhagen strain of vaccinia), and avipox vectors (canarypox (ALVAC) and fowlpox (FPV) vectors), vesicular stomatitis virus (VSV) vectors, retrovirus vectors, lentivirus vectors, simian virus 40 (SV40), bovine papilloma viruses, Epstein-Barr viruses, Moloney murine leukemia viruses, Harvey murine sarcoma viruses, murine mammary
Attorney Docket No.250298.000682 tumor viruses, Rous sarcoma viruses, poxvirus viral like particles, baculoviral vectors and bacterial spores. [00250] As further examples, adenovirus vectors may be derived from human adenovirus (Ad) but also from adenoviruses that infect other species, such as bovine adenovirus (e.g. bovine adenovirus 3, BAdV3), a canine adenovirus (e.g. CAdV2), a porcine adenovirus (e.g. PAdV3 or 5), or great apes, such as Chimpanzee (Pan), Gorilla (Gorilla), Orangutan (Pongo), Bonobo (Pan paniscus) and common chimpanzee (Pan troglodytes). Poxvirus (Poxviridae) vectors may be derived from smallpox virus (variola), vaccinia virus, cowpox virus or monkeypox virus. Exemplary vaccinia viruses are the Copenhagen vaccinia virus (W), New York Attenuated Vaccinia Virus (NYVAC), ALVAC, TROVAC and Modified Vaccinia Ankara (MVA). [00251] Many expression systems are known in the art, including bacteria (for example E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae), filamentous fungi (for example Aspergillus spec.), plant cells, animal cells (e.g., mammalian cells), and insect cells. [00252] In yet another aspect, the disclosure provides a host cell comprising the vector of the disclosure. The host cell can be either prokaryotic or eukaryotic. Bacterial cells may be preferred prokaryotic host cells in some circumstances and typically are a strain of E. coli such as, for example, the E. coli strains DH5 and RR1. Non-limiting examples of eukaryotic host cells include yeast, insect, and mammalian cells (e.g., from a mouse, rat, monkey, or human cell lines). Non-limiting examples of yeast host cells include, e.g., YPH499, YPH500, and YPH501. Non-limiting examples of mammalian host cells include Chinese hamster ovary (CHO) cells, NIH Swiss mouse embryo cells NIH/3T3, monkey kidney-derived COS-1 cells, and 293 cells which are human embryonic kidney cells. Examples of insect cells include Sf9 cells, which can be transfected with baculovirus expression vectors. [00253] Transformation of appropriate cell hosts with a DNA construct of the present disclosure is accomplished by well-known methods that typically depend on the type of vector used. Successfully transformed cells, i.e. cells that contain a DNA construct of the present disclosure, can be identified by, for example, PCR. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
Attorney Docket No.250298.000682 [00254] It will be appreciated that certain host cells of the disclosure are useful in the preparation of the peptides or peptide-based molecules of the disclosure, for example bacterial, yeast, and insect cells. However, other host cells may be useful in certain therapeutic methods. For example, antigen-presenting cells (APCs), such as dendritic cells or B cells, may be used to express the peptides of the disclosure such that the peptides may be loaded into appropriate MHC molecules. [00255] A further aspect of the disclosure provides a method of producing peptides or peptide-based molecules of the disclosure, the method comprising culturing a host cell and isolating the peptide or peptide-based molecule from the host cell or its culture medium. Peptide and pMHC Binding Moieties [00256] Peptides, pMHC complex, or other peptide-based molecules (such as a complex, fusion protein, or conjugate comprising a peptide disclosed herein) of the present disclosure can be used to identify and/or isolate binding moieties that bind specifically to a peptide, pMHC complex, or other peptide-based molecule of the disclosure. Such binding moieties may be used as immunotherapeutic reagents and may include, e.g., antibodies (or antigen-binding fragments thereof), alternative scaffolds, TCRs, and CARs. [00257] In one aspect, the disclosure provides a peptide binding moiety that binds a peptide of the disclosure. Preferably the peptide binding moiety binds a peptide when the peptide is in complex with MHC. In the latter instance, the peptide binding moiety may bind partially to the MHC, provided that the peptide binding moiety also binds to the peptide. The peptide binding moiety may bind only the peptide, and that binding may be specific. The peptide binding moiety may bind only the pMHC complex and that binding may be specific. [00258] The disclosure also provides a method of identifying a peptide binding moiety that binds a pMHC complex of the disclosure, the method comprising contacting a candidate peptide binding moiety with the pMHC complex and determining whether the candidate peptide binding moiety binds the complex. Methods to determine binding to pMHC complexes include, for example, surface plasmon resonance, or any other biosensor technique, ELISA, flow cytometry, chromatography, microscopy. Alternatively, or in
Attorney Docket No.250298.000682 addition, binding may be determined by functional assays in which a biological response is detected upon binding, for example, cytokine release or cell apoptosis. [00259] The candidate peptide binding moiety may be a peptide binding moiety of the type already described, such as an antibody or a TCR. [00260] For example, antibodies and TCRs may be obtained from display libraries in which the pMHC complex of the disclosure is used to pan the library. TCRs can be displayed on the surface of phage particles and yeast particles, for example, and such libraries have been used for the isolation of high affinity variants of TCR derived from T cell clones. TCR phage libraries can be used to isolate TCRs with novel antigen specificity. Such libraries can be constructed with α- and β- chain sequences corresponding to those found in a natural repertoire. However, the random combination of these α- and β- chain sequences, which occurs during library creation, can produce a repertoire of TCRs that may not be naturally occurring. [00261] In some embodiments, the pMHC complex of the disclosure may be used to screen a library of diverse TCRs displayed on the surface of phage particles. The TCRs displayed by said library may not correspond to those contained in a natural repertoire, for example, they may contain α- and β- chain pairing that would not be present in vivo, and/or the TCRs may contain non-natural mutations and/or the TCRs may be in soluble form. Screening may involve panning the phage library with pMHC complexes of the disclosure and subsequently isolating bound phage particles. For this purpose, pMHC complexes may be attached to a solid support, such as a magnet bead, or column matrix and phage bound pMHC complexes isolated, with a magnet, or by chromatography, respectively. The panning steps may be repeated several times. Isolated phage may be further expanded in E. coli cells. Isolated phage particles may be tested for specific binding to pMHC complexes of the disclosure. Binding can be detected using techniques including, but not limited to, ELISA, or SPR for example using a BiaCore instrument. The DNA sequence of the T cell receptor displayed by pMHC binding phage can be further identified by PCR methods. [00262] Alternatively, antigen binding T cells and TCRs can be isolated from fresh blood obtained from patients or healthy donors. Such a method involves stimulating T cells using autologous dendritic cells (DCs), followed by autologous B cells, and then pulsed
Attorney Docket No.250298.000682 with a peptide disclosed herein. Several rounds of stimulation may be carried out, for example three or four rounds. Activated T cells may then be tested for specificity by measuring cytokine release in the presence of T2 cells pulsed with the peptide of the disclosure (for example using an IFNγ ELISpot assay). Activated cells may then be sorted by fluorescence-activated cell sorting (FACS) using labelled antibodies to detect intracellular cytokine production (e.g. IFNγ), or expression of a cell surface marker (such as CD137). Sorted cells may be expanded and further validated, for example, by ELISpot assay and/or cytotoxicity against target cells and/or staining by peptide-MHC tetramer. The TCR chains from validated clones may then be amplified by rapid amplification of cDNA ends (RACE) and sequenced. [00263] A peptide binding moiety disclosed herein can include, for example, without limitation, an antibody, a TCR, or a CAR. [00264] In some embodiments, the peptide binding moiety of the disclosure may be an antibody or antigen-binding fragment thereof. Antibodies or antigen-binding fragments thereof encompass derivatives, functional equivalents, and homologues of antibodies, humanized antibodies, including any polypeptide comprising an immunoglobulin binding domain, whether natural or wholly or partially synthetic and any polypeptide or protein having a binding domain which is, or is homologous to, an antibody binding domain. Chimeric molecules comprising an immunoglobulin binding domain, or equivalent, fused to another polypeptide are therefore included. A humanized antibody may be a modified antibody having the variable regions of a non-human, e.g. murine, antibody, and the constant region of a human antibody. Examples of antibodies are the immunoglobulin isotypes (e.g., IgG, IgE, IgM, IgD and IgA) and their isotypic subclasses; or fragments that comprise an antigen binding domain such as Fab, scFv, Fv, dAb, Fd; and diabodies. Antibodies may be polyclonal or monoclonal. A monoclonal antibody may be referred to herein as “mAb”. [00265] In some embodiments, the antibody is a multispecific antibody. In some embodiments, the antibody is a bispecific antibody. The bispecific antibody may comprise a second targeting moiety that targets to the desired cell or tissue or to another desired antigen associated with the same or similar disease or disorder.
Attorney Docket No.250298.000682 [00266] It is possible to take an antibody, for example a monoclonal antibody, and use recombinant DNA technology to produce other antibodies or chimeric molecules which retain the specificity of the original antibody. Such techniques may involve introducing DNA encoding the immunoglobulin variable region, or the complementary determining regions (CDRs), of an antibody to the constant regions, or constant regions plus framework regions, of a different immunoglobulin. A hybridoma (or other cell that produces antibodies) may be subject to genetic mutation or other changes, which may or may not alter the binding specificity of antibodies produced. [00267] It has been shown that fragments of a whole antibody can perform the function of binding antigens. Examples of binding fragments are (i) the Fab fragment consisting of VL, VH, CL and CH1 domains; (ii) the Fd fragment consisting of the VH and CH1 domains; (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment which consists of a VH domain; (v) isolated CDR regions; (vi) F(ab′)2 fragments, a bivalent fragment comprising two linked Fab fragments; (vii) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site; (viii) bispecific single chain Fv dimers, (ix) “diabodies”, multivalent or multispecific fragments constructed by gene fusion, and (x) VHH or VNAR antibodies, also known as single-domain antibodies or nanobodies (Nb), which may be derived from heavy-chain antibodies from e.g., dromedaries, camels, llamas, alpacas, or sharks. [00268] Diabodies are multimers of polypeptides, each polypeptide comprising a first domain comprising a binding region of an immunoglobulin light chain and a second domain comprising a binding region of an immunoglobulin heavy chain, the two domains being linked (e.g. by a peptide linker) but unable to associate with each other to form an antigen binding site: antigen binding sites are formed by the association of the first domain of one polypeptide within the multimer with the second domain of another polypeptide within the multimer (WO94/13804). Where bispecific antibodies are to be used, these may be conventional bispecific antibodies, which can be manufactured in a variety of ways, e.g. prepared chemically or from hybrid hybridomas, or may be any of the bispecific antibody fragments mentioned above. It may be preferable to use scFv dimers or diabodies rather than whole antibodies. Diabodies and scFv can be constructed without an Fc region, using
Attorney Docket No.250298.000682 only variable domains, potentially reducing the effects of anti-idiotypic reaction. Other forms of bispecific antibodies include the single chain “Janusins”. Bispecific diabodies, as opposed to bispecific whole antibodies, may also be useful because they can be readily constructed and expressed in E. coli. Diabodies (and many other polypeptides such as antibody fragments) of appropriate binding specificities can be readily selected using phage display from libraries. If one arm of the diabody is to be kept constant, for instance, with a specificity directed against an antigen of interest, then a library can be made where the other arm is varied, and an antibody of appropriate specificity selected. An “antigen binding domain” is the part of an antibody which comprises the area which specifically binds to and is complementary to part or all of an antigen. Where an antigen is large, an antibody may only bind to a particular part of the antigen, which part is termed an epitope. An antigen binding domain may be provided by one or more antibody variable domains. An antigen binding domain may comprise an antibody light chain variable region (VL) and an antibody heavy chain variable region (VH). [00269] In some embodiments, the peptide binding moiety may be an antibody-like molecule that has been designed to specifically bind a peptide or peptide-MHC complex of the disclosure. In some embodiments the peptide binding moiety may comprise a TCR- mimic antibody. In some embodiments, such TCR-mimic antibodies can comprise high- affinity soluble antibody molecules endowed with a TCR-like specificity towards tumor or viral epitopes that can target tumor and/or virus-infected cells and mediate their specific killing. [00270] Also encompassed within the present disclosure are binding moieties based on engineered protein scaffolds or “alternative scaffolds”. Alternative scaffolds are derived from stable, soluble, natural protein structures which have been modified to provide a binding site for a target molecule of interest. Examples of alternative scaffolds include, but are not limited to, affibodies, which are based on the Z-domain of staphylococcal protein A that provides a binding interface on two of its α-helices; anticalins, derived from lipocalins, that incorporate binding sites for small ligands at the open end of a β-barrel fold; monobodies, designed to incorporate the fibronectin type III domain (Fn3) of fibronectin or tenascin as a protein scaffold or synthetic FN3 domains (e.g., tencon); nanobodies, and DARPins. Additional alternative scaffolds include Adnectin™, iMab, EETI-II/AGRP,
Attorney Docket No.250298.000682 Kunitz domain, thioredoxin peptide aptamer, Affilin, Tetranectin, Fynomer, and Avimer. Alternative scaffolds are typically targeted to bind the same antigenic proteins as antibodies, and are potential therapeutic agents. They may act as inhibitors or antagonists, or as delivery vehicles to target molecules, such as toxins, to a specific tissue in vivo. Short peptides may also be used to bind a target protein. Phylomers are natural structured peptides derived from bacterial genomes. Such peptides represent a diverse array of protein structural folds and can be used to inhibit/disrupt protein-protein interactions in vivo. [00271] Alternative scaffolds are typically single chain polypeptidic frameworks that contain a highly structured core associated with variable domains of high conformational tolerance allowing insertions, deletions, or other substitutions within the variable domains. Libraries introducing diversity to one or more variable domains, and in some cases to the structured core, may be generated using known protocols and the resulting libraries may be screened for binding to the peptide and/or the pMHC complex of the disclosure, and the identified binders may be further characterized for their specificity using known methods. Alternative scaffolds may be derived from Protein A, in particular, the Z-domain thereof (affibodies), ImmE7 (immunity proteins), BPTI/APPI (Kunitz domains), CTLA-4, charybdotoxin (Scorpion toxin), Min-23 (knottins), lipocalins (anticalins), Ras-binding protein AF-6 (PDZ-domains), neokarzinostatin, a fibronectin domain, an ankyrin consensus repeat domain, or thioredoxin. [00272] In some embodiments, the antibodies or alternative scaffolds described herein can be immobilized on viral vectors. Such modified recombinant viral vectors can be useful for the targeted introduction of genetic materials encoded by the viral vectors into cells and/or tissues. Various means can be used to mobilize the antibodies or alternative scaffolds to the viral vectors, for example, by using an affinity binding pair, such as c- Myc/anti-Myc antibody, streptavidin/biotin, or via spy-tag/spy-catcher system. Exemplary vectors that may be modified with the antibodies or alternative scaffolds described herein include, but are not limited to, adeno-associated virus (AAV) vectors (e.g., AAV1, AAV2, AAV6, AAV9, or AAV9.PHP), retroviral vectors, lentiviral vectors, and targeted oncolytic viruses (e.g., herpes simplex virus (HSV)).
Attorney Docket No.250298.000682 [00273] In some embodiments, the peptide binding moiety may be a TCR. TCRs are described using the International Immunogenetics (IMGT) TCR nomenclature, and the IMGT public database of TCR sequences. [00274] The TCRs of the present disclosure may be in any format. For example, the TCRs may be αβ heterodimers, or αα or ββ homodimers. [00275] α/β heterodimeric TCRs have an α-chain and a β-chain. Broadly, each chain comprises variable, joining and constant region, and the β-chain also usually contains a short diversity region between the variable and joining regions, but this diversity region is often considered as part of the joining region. Each variable region comprises three hypervariable CDRs (Complementarity Determining Regions) embedded in a framework sequence; CDR3 is believed to be the main mediator of antigen recognition. There are several types of α- chain variable (Vα) regions and several types of β-chain variable (Vβ) regions distinguished by their framework, CDR1 and CDR2 sequences, and by a partly defined CDR3 sequence. [00276] The TCRs of the disclosure may not correspond to TCRs as they exist in nature. For example, they may comprise α- and β- chain combinations that are not present in a natural repertoire. Alternatively or additionally, a TCR described herein may be soluble, and/or the α- and/or β- chain constant domain may be truncated relative to the native/naturally occurring TRAC/TRBC sequences such that, for example, the C-terminal transmembrane domain and intracellular regions are not present. Such truncation may result in removal of the cysteine residues from TRAC/TRBC that form the native interchain disulfide bond. [00277] In addition, the TRAC/TRBC domains may contain modifications. For example, the α-chain extracellular sequence may include a modification relative to the native/naturally occurring TRAC whereby amino acid T48 of TRAC, with reference to IMGT numbering, is replaced with C48. Likewise, the β-chain extracellular sequence may include a modification relative to the native/naturally occurring TRBC1 or TRBC2 whereby S57 of TRBC1 or TRBC2, with reference to IMGT numbering, is replaced with C57. These cysteine substitutions relative to the native α- and β- chain extracellular sequences enable the formation of a non-native interchain disulfide bond which stabilizes the refolded soluble TCR, i.e. the TCR formed by refolding extracellular α- and β- chains.
Attorney Docket No.250298.000682 This non-native disulfide bond facilitates the display of correctly folded TCRs on phage. In addition, the use of the stable disulfide linked soluble TCR enables more convenient assessment of binding affinity and binding half-life. Alternative positions for the formation of a non-native disulfide include, for example, Thr 45 of exon 1 of TRAC*01 and Ser 77 of exon 1 of TRBC1*01 or TRBC2*01; Tyr 10 of exon 1 of TRAC*01 and Ser 17 of exon 1 of TRBC1*01 or TRBC2*01; Thr 45 of exon 1 of TRAC*01 and Asp 59 of exon 1 of TRBC1*01 or TRBC2*01; and Ser 15 of exon 1 of TRAC*01 and Glu 15 of exon 1 of TRBC1*01 or TRBC2*01. TCRs with a non-native disulfide bond may be full length or may be truncated. [00278] TCRs of the disclosure may be in single chain format. Single chain TCRs include αβ TCR polypeptides of the type: Vα-L-Vβ, Vβ-L-Vα, Vα-Cα-L-Vβ, Vα-L-Vβ-Cβ or Vα-Cα-L-Vβ-Cβ, optionally in the reverse orientation, wherein Vα and Vβ are TCR α and β variable regions respectively, Cα and Cβ are TCR α and β constant regions respectively, and L is a linker sequence. Single chain TCRs may contain a non-native disulfide bond. The TCR may be in a soluble form (i.e. having no transmembrane or cytoplasmic domains) or may contain full length α- and β- chains. The TCR may be provided on the surface of a cell, such as a T cell. [00279] TCRs of the disclosure may be engineered to include mutations. Methods for producing mutated high affinity TCR variants such as phage display and site directed mutagenesis. Preferably, mutations to improve affinity are made within the variable regions of α- and/or β- chains. More preferably mutations to improve affinity are made within the CDRs. There may be between 1 and 15 mutations in the α- and or β- chain variable regions. [00280] TCRs of the disclosure may also be labeled with an imaging compound, for example a label that is suitable for diagnostic purposes. Such labelled high affinity TCRs are useful in a method for detecting a TCR ligand selected from CD1-antigen complexes, bacterial superantigens, and MHC-peptide/superantigen complexes, which method comprises contacting the TCR ligand with a high affinity TCR (or a multimeric high affinity TCR complex) which is specific for the TCR ligand; and detecting binding to the TCR ligand. In multimeric high affinity TCR complexes (formed, for example, using biotinylated heterodimers) fluorescent streptavidin can be used to provide a detectable label. A fluorescently-labelled multimer is suitable for use in FACS analysis, for example
Attorney Docket No.250298.000682 to detect antigen presenting cells carrying the peptide for which the high affinity TCR is specific. [00281] A TCR of the present disclosure (or multivalent complex thereof) may alternatively or additionally be associated with (e.g. covalently or otherwise linked to) a therapeutic agent which may be, for example, a toxic moiety for use in cell killing, or an immunostimulating agent such as an interleukin or a cytokine. A multivalent high affinity TCR complex of the present disclosure may have enhanced binding capability for a TCR ligand compared to a non-multimeric wild-type or high affinity T cell receptor heterodimer. Thus, the multivalent high affinity TCR complexes according to the disclosure are particularly useful for tracking or targeting cells presenting particular antigens in vitro or in vivo, and are also useful as intermediates for the production of further multivalent high affinity TCR complexes having such uses. The high affinity TCR or multivalent high affinity TCR complex may therefore be provided in a pharmaceutically acceptable formulation for use in vivo. [00282] High affinity TCRs of the disclosure may be used in the production of soluble bi-specific reagents. A preferred embodiment is a reagent which comprises a soluble TCR, fused via a linker to an anti-CD3 specific antibody fragment. [00283] In a further aspect, the disclosure provides nucleic acid encoding the TCR of the disclosure, a TCR expression vector comprising nucleic acid encoding a TCR of the disclosure, as well as a cell harboring such a vector. The TCR may be encoded either in a single open reading frame or two distinct open reading frames. Also included in the scope of the disclosure is a cell harboring a first expression vector which comprises nucleic acid encoding an α- chain of a TCR of the disclosure, and a second expression vector which comprises nucleic acid encoding a β-chain of a TCR of the disclosure. Alternatively, one vector may encode both an α- and a β- chain of a TCR of the disclosure. [00284] A further aspect of the present disclosure provides a cell displaying on its surface a TCR of the disclosure. The cell may be a T cell, or other immune cell. The T cell may be modified such that it does not correspond to a T cell as it exists in nature. For example, the cell may be transfected with a vector encoding a TCR of the disclosure such that the T cell expresses a further TCR in addition to the native TCR. Additionally or alternatively, the T cell may be modified such that it is not able to present the native TCR.
Attorney Docket No.250298.000682 There are a number of methods suitable for the transfection of T cells with DNA or RNA encoding the TCRs of the disclosure. As a non-limiting example, the transfection method may comprise a rapid RNA-based transfection system. T cells expressing the TCRs of the disclosure are suitable for use in adoptive therapy-based treatment of diseases such as cancers. There are a number of suitable methods by which adoptive therapy can be carried out. For example, adoptive cell therapy (ACT) may comprise use of autologous tumor- infiltrating lymphocytes, and may include a lymphodepletion preparative regimen prior to ACT. In some embodiments, viruses, e.g., retroviruses, that encode TCRs may be used for genetic modification of lymphocytes to convert normal lymphocytes into lymphocytes with anti-cancer activity. The adoptive transfer of lymphocytes with anti-cancer activity into patients requiring treatment of, e.g., metastatic melanoma, can mediate tumor regression. In some embodiments, ACT may comprise treatment of patients with cancers expressing viral or alloantigens, treatment of patients with cancers expressing viral antigens, and/or ACT using gene-modified lymphocytes. In some embodiments, ACT methods may include, for example, genetic modification of lymphocytes to introduce new recognition specificities using, e.g., αβTCR(s) and/or chimeric TCR(s); genetic modification of lymphocytes to alter function of T cells using, e.g., co-stimulatory molecules (e.g., CD28, 41BB), cytokines (e.g., IL2, IL15), homing molecules (e.g., CD62L, CCR7), and/or molecules capable of preventing apoptosis (BCL2); modification of host lymphodepletion using, e.g., selective depletion of CD4+ cells or T regulatory cells; blocking of inhibitory signals on reactive lymphocytes using, e.g., antibodies to CTLA4 and/or PD-1; administration of vaccines to stimulate transferred cells using, e.g., recombinant virus encoding antigen(s); administration of alternative cytokines to support cell growth using, e.g., IL15 and/or IL21; stimulation of APCs using, e.g., toll-like receptor agonists; generation of less differentiated lymphocytes using, e.g., alternate culture conditions and growth promoting cytokines in vitro; and, overcoming antigen escape variants using, e.g., natural killer cells. [00285] The TCRs of the disclosure intended for use in adoptive therapy are generally glycosylated when expressed by the transfected T cells. The glycosylation pattern of transfected TCRs may be modified by mutations of the transfected gene.
Attorney Docket No.250298.000682 [00286] In some embodiments, the peptide binding moiety may be a chimeric antigen receptor (CAR). CARs are genetically engineered receptors. CARs may be generated that bind the peptides or pMHC complexes of the present disclosure by incorporating an antigen binding domain that specifically binds the peptide or pMHC complex to the extracellular domain of the CAR. CARs may be introduced into and expressed by immune cells, such as T cells, NK cells, or macrophages. CARs can be programmed to both recognize a specific antigen and, when bound to that antigen, activate the immune cell to attack and destroy the cell presenting that antigen. When these antigens exist on tumor cells, an immune cell that expresses the CAR can target and kill the tumor cell. [00287] The general structure of a CAR typically comprises an extracellular domain that binds the antigen (e.g. the peptides or pMHC complexes of the present disclosure), a hinge, a transmembrane domain, and an intercellular domain comprising a signaling domain and optionally one or more co-stimulatory domains. [00288] Extracellular domains of the CAR may contain any polypeptide that specifically binds the desired antigen (e.g. the peptides or pMHC complexes of the present disclosure). For example, the extracellular domain may comprise an antibody fragment such as scFv or VHH. The CARs may also be engineered to bind two or more desired antigens that may be arranged in tandem and separated by linker sequences. For example, one or more domain antibodies, scFvs, llama VHH antibodies or other VH only antibody fragments may be organized in tandem via a linker to provide bispecificity or multispecificity to the CAR. [00289] A hinge domain may be present between the extracellular domain and the transmembrane domain of the CAR, e.g., to provide flexibility to allow effective binding of the extracellular domain to its intended target. The hinge domain may be a polypeptide of about 2 to 100 amino acids in length. The hinge may include or be composed of flexible residues such as Gly and Ser so that the adjacent protein domains are free to move relative to one another. Longer hinges may be used when it is desirable to ensure that two adjacent domains do not sterically interfere with one another. The hinge may be derived from a hinge region or portion of the hinge region of any immunoglobulin. Non-limiting examples
Attorney Docket No.250298.000682 of linkers include a part of human CD8α chain, extracellular domain of CD28, an Ig hinge from IgG, IgM, IgA, IgD, or IgE, FcyRllla receptor, or a functional fragment thereof. [00290] Transmembrane domains of the CAR may be derived transmembrane proteins, such as an alpha, beta or zeta chain of a T-cell receptor, CD28, CD3 epsilon, CD2, CD4, CD5, CD8, CD9, CD16, CD18, CD19, CD22, CD27, CD29, CD33, CD37, CD40, CD45, CD49a, CD64, CD80, CD84, CD86, CD96 (Tactile), CD100 (SEMA4D), CD103, CD134, CD154, CD160 (BY55), KIRDS2, OX40, LFA-1 (CD11a, CD18), CD11b, CD11c, CD11d, ICOS (CD278), 4-1 BB (CD137), 4-1 BBL, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRFI), IL2R beta, IL2R gamma, IL7Ra, ITGA1, VLA1, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, ITGAE, ITGAL, LFA-1, ITGAM, ITGAX, ITGB1, ITGB2, LFA-1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CEACAM1, CRT AM, Ly9 (CD229), PSGL1, SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKp30, NKp44, NKp46, NKG2D, and NKG2C, or functional fragment thereof. [00291] The intracellular signaling domain of a CAR participates in transducing the signal of effective CAR binding to a target antigen into the interior of the immune effector cell to elicit an effector cell function, e.g., activation, cytokine production, proliferation, and cytotoxic activity, including the release of cytotoxic factors to the CAR-bound target cell, or other cellular responses elicited following antigen binding to the extracellular CAR domain. Non-limiting examples of intracellular signaling domains of the CAR include those derived from CD3ζ, CD3 ^, CD3δ, CD3γ, CD5, CD22, CD39, CD79A, CD79B, CD66d, CD226, DAP10, DAP12, Fc epsilon receptor I gamma chain (FCER1G), or FcR β. [00292] Intracellular co-stimulatory domains of the CAR can provide a second signal required for efficient activation and function of T lymphocytes upon binding to antigen. Such co-stimulatory domains may be derived from one or more co-stimulatory molecules, such as, but not limited to, 4-1BB, CD2, CD7, CD27, CD28, CD30, CD40, CD54 (ICAM), CD83, CD134 (OX40), CD150 (SLAMF1), CD152 (CTLA4), CD223 (LAG3), CD270 (HVEM), CD278 (ICOS), DAP10, LAT, NKD2C SLP76, TRIM, BTLA, GITR, CD226, HVEM, and ZAP70.
Attorney Docket No.250298.000682 [00293] The CARs can be generated by standard molecular biology techniques. The extracellular domain that binds the desired antigen may be derived from the antibodies or their antigen binding fragments described herein. [00294] In another aspect, the disclosure further provides cells that comprise peptide a binding moiety (e.g., TCRs and CARs) of the present disclosure. In some embodiments, the host cell is an immune cell. In some embodiments, the immune cell is T cell, NK cell, or a macrophage. The host cell may be autologous or allogeneic with respective to the subject receiving the cell (as treatment). [00295] In some embodiments, TCRs of the present disclosure are provided as TCR- T cells. In some embodiments, CARs of the present disclosure are provided as CAR-T cells. Any methods known in the art for modifying T cells to express a TCR or CAR can be employed to generate the TCR-T or CAR-T cells of the present disclosure. [00296] The cells expressing a peptide binding moiety (e.g., TCRs and CARs) of the present disclosure may also contain one or more additional genes. The additional genes can be used to increase the effector function of the cells expressing the peptide binding moiety (e.g., TCRs and CARs). Non-limiting examples of classes of additional genes include (a) a second targeting moiety, such as antibodies, including fragments thereof and bispecific antibodies (e.g., bispecific T cell engagers (BiTEs)), (b) secretable cytokines (e.g., GM-CSF, IL-7, IL-12, IL-15, IL-18), (c) membrane bound cytokines (e.g., IL-15), (d) chimeric cytokine receptors (e.g., IL-2/IL-7, IL-4/IL-7), (e) constitutive active cytokine receptors (e.g., C7R), (f) dominant negative receptors (DNR; e.g., TGFRII DNR), (g) ligands of co-stimulatory molecules (e.g., CD80, 4-1BBL), (h) nuclear factor of activated T cells (NFATs) (e.g., NFATc1, NFATc2, NFATc3, NFATc4, and NFAT5), or (j) suicide genes (e.g., CD20, truncated EGFR or HER2, inducible caspase 9 molecules). In some embodiments, the cells expressing a peptide binding moiety (e.g., TCRs and CARs) of the present disclosure may express a second targeting moiety that targets to a specific tissue and/or cell type or to a known cancer antigen. Pharmaceutical Compositions, Dosage Forms, and Administration [00297] In a further aspect, the disclosure provides a pharmaceutical composition comprising a peptide, a peptide-based molecule (such as a complex (e.g., peptide-MHC
Attorney Docket No.250298.000682 (pMHC) complex), fusion protein, or conjugate comprising the peptide), a nucleic acid molecule, a vector, a cell, or a peptide binding moiety of the disclosure together with a pharmaceutically acceptable carrier and/or excipient. The pharmaceutical compositions of the disclosure may be in any suitable form (depending upon the desired method of administering to a patient). Suitable compositions and methods of administration are known to those skilled in the art, for example see, Johnson et al., Blood.2009; 114(3):535- 46. [00298] The pharmaceutical compositions may comprise the peptides or peptide- based molecules of the disclosure either in the free form or in the form of a pharmaceutically acceptable salt. The term “pharmaceutically acceptable salt” as used herein refers to a derivative of the disclosed peptides wherein the peptide is modified by making acid or base salts of the agent. For example, acid salts are prepared from the free base (typically wherein the neutral form of the drug has a neutral —NH2 group) involving reaction with a suitable acid. Suitable acids for preparing acid salts include both organic acids, e.g., acetic acid, benzoic acid, citric acid, propionic acid, glycolic acid, trifluoroacetic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, maleic acid, succinic acid, fumaric acid, tartaric acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like, as well as inorganic acids, e.g., hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid phosphoric acid and the like. Conversely, preparation of basic salts of acid moieties which may be present on a peptide are prepared using a pharmaceutically acceptable base such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, trimethylamine or the like. [00299] Compositions of the disclosure may comprise multiple peptides, e.g., 2 to 50, 2 to 40, 2 to 30, 5 to 25, 5 to 20, or 10 to 15 peptides as described herein (e.g., SEQ ID NO: 1-89 and 143-146). In some embodiment, the compositions of the disclosure may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, or 93 amino acid
Attorney Docket No.250298.000682 sequences selected from SEQ ID NO: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or fragment or derivative thereof. [00300] In some embodiments, the peptides or peptide-based molecules may be present in a solution at a concentration of about 1 μg/mL to 50 mg/mL, for example, about 0.1 mg/mL to 10 mg/mL, about 0.2 mg/mL to 5 mg/mL, about 0.5 mg/mL to 8 mg/mL, about 0.8 mg/mL to 12 mg/mL, about 1 mg/mL to 15 mg/mL, about 2 mg/mL to 20 mg/mL, or about 5 mg/mL to 25 mg/mL, or about 0.1 mg/mL, 0.2 mg/mL, 0.3 mg/mL, 0.4 mg/mL, 0.5 mg/mL, 0.6 mg/mL, 0.7 mg/mL, 0.8 mg/mL, 0.9 mg/mL, 1 mg/mL, 1.25 mg/mL, 1.5 mg/mL, 1.75 mg/mL, 2 mg/mL, 2.25 mg/mL, 2. 5 mg/mL, 2.75 mg/mL, 3 mg/mL, 3.25 mg/mL, 3.5 mg/mL, 3.75 mg/mL, 4 mg/mL, 5 mg/mL, 6 mg/mL, 7 mg/mL, 8 mg/mL, 9 mg/mL, 10 mg/mL, 11 mg/mL, 12 mg/mL, 13 mg/mL, 14 mg/mL, 15 mg/mL or 20 mg/mL. [00301] The pharmaceutical composition may be adapted for administration by any appropriate route such as, e.g., parenteral (including subcutaneous, intramuscular, or intravenous), enteral (including oral or rectal), inhalation, or intranasal routes. [00302] Such compositions may be prepared by any method known in the art of pharmacy, for example, by mixing the active ingredient with the carrier(s) or excipient(s) under sterile conditions. [00303] In addition, disclosed herein are pharmaceutical dosage forms comprising the peptides, peptide-based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties of the disclosure. [00304] Pharmaceutical compositions based on the peptides, peptide-based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties disclosed herein can be formulated in any conventional manner using one or more physiologically acceptable carriers and/or excipients. The peptides, peptide-based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties may be formulated for administration by, for example, injection, inhalation, or insulation (either through the mouth or the nose) or by oral, buccal, parenteral or rectal administration, or by administration directly to an organ or tissue.
Attorney Docket No.250298.000682 [00305] The pharmaceutical compositions can be formulated for a variety of modes of administration, including systemic, topical, or localized administration. Techniques and formulations can be found in, for example, Remington's Pharmaceutical Sciences, Meade Publishing Co., Easton, Pa. For systemic administration, injection is preferred, including intramuscular, intravenous, intraperitoneal, and subcutaneous. For the purposes of injection, the pharmaceutical compositions can be formulated in liquid solutions, preferably in physiologically compatible buffers, such as Hank’s solution or Ringer’s solution. In addition, the pharmaceutical compositions may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms of the pharmaceutical composition are also suitable. [00306] In some embodiments, the pharmaceutical compositions of the present disclosure may be lyophilized. As a non-limiting example, the obtained lyophilizate can be reconstituted into a hydrous composition by adding a hydrous solvent. In some embodiments, the hydrous composition may be able to be directly administered parenterally to a patient. Therefore, a further embodiment of the present disclosure is a hydrous pharmaceutical composition, obtainable via reconstitution of the lyophilizate with a hydrous solvent. [00307] In some embodiments, the pharmaceutical composition disclosed herein may comprise a lyophilized formulation. As a non-limiting example, the lyophilization formulation may comprise peptides of the disclosure, mannitol, and/or TWEEN 80®. As another non-limiting example, the lyophilization formulation may comprise the peptides disclosed herein, mannitol and poloxamer 188. In some embodiments, the pharmaceutical composition may comprise a lyophilization formulation comprising a reconstituted-liquid composition. [00308] In some embodiments, pharmaceutical compositions of the present disclosure may provide a formulation with an enhanced solubility and/or moistening of the lyophilizate over previously known compositions. As a non-limiting example, enhanced solubility and/or moistening of the lyophilizate may be achieved using an appropriate composition of excipients. In this way, pharmaceutical compositions of the present disclosure comprising peptides of SEQ ID NO: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof, may be developed to show a
Attorney Docket No.250298.000682 desired shelf stability at (e.g., at −20° C, +5° C, or +25° C) and can be easily resolubilized such that the lyophilizate can be completely dissolved through the use of a buffer or other excipients from seconds up to two or more minutes, with or without the use of an of ultrasonic homogenizer. Furthermore, the composition can be easily provided to a patient in need of treatment via any appropriate delivery route disclosed herein, e.g., parenteral (including subcutaneous, intramuscular, or intravenous), enteral (including oral or rectal), inhalation, or intranasal routes. As a non-limiting example, the pH-value of the resulting solution may be between pH 2.7 and pH 9. [00309] For oral administration, the pharmaceutical compositions may take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g. pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g. lactose, microcrystalline cellulose or calcium hydrogen phosphate); lubricants (e.g. magnesium stearate, talc or silica); disintegrants (e.g. potato starch or sodium starch glycolate); or wetting agents (e.g. sodium lauryl sulfate). The tablets can also be coated by methods well known in the art. Liquid preparations for oral administration may take the form of, for example, solutions, syrups or suspensions, or they may be presented as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g. sorbitol syrup, cellulose derivatives or hydrogenated edible fats); emulsifying agents (e.g. lecithin or acacia); non-aqueous vehicles (e.g. ationd oil, oily esters, ethyl alcohol or fractionated vegetable oils); and preservatives (e.g. methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations can also contain buffer salts, flavoring, coloring and sweetening agents as appropriate. [00310] The pharmaceutical compositions can be formulated for parenteral administration by injection, e.g. by bolus injection or continuous infusion. Formulations for injection can be presented in a unit dosage form, e.g. in ampoules or in multi-dose containers, with an optionally added preservative. The pharmaceutical compositions can further be formulated as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain other agents including suspending, stabilizing and/or dispersing agents.
Attorney Docket No.250298.000682 [00311] Additionally, the pharmaceutical compositions can also be formulated as a depot preparation. These long acting formulations can be administered by implantation (e.g. subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (e.g. as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. Other suitable delivery systems include microspheres, which offer the possibility of local noninvasive delivery of drugs over an extended period of time. This technology can include microspheres having a precapillary size, which can be injected via a coronary catheter into any selected part of an organ without causing inflammation or ischemia. The administered therapeutic is men slowly released from the microspheres and absorbed by the surrounding cells present in the selected tissue. [00312] Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, bile salts, and fusidic acid derivatives. In addition, detergents may be used to facilitate permeation. Transmucosal administration can occur using nasal sprays or suppositories. For topical administration, the vector particles described herein can be formulated into ointments, salves, gels, or creams as generally known in the art. A wash solution can also be used locally to treat an injury or inflammation in order to accelerate healing. [00313] Pharmaceutical forms suitable for injectable use can include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid. It must be stable under the conditions of manufacture and certain storage parameters (e.g. refrigeration and freezing) and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. [00314] If formulations disclosed herein are used as a therapeutic to boost an immune response in a subject, a therapeutic agent can be formulated into a composition in a neutral or salt form. Pharmaceutically acceptable salts, include the acid addition salts
Attorney Docket No.250298.000682 (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. [00315] A carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents known in the art. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. [00316] Sterile injectable solutions can be prepared by incorporating the active compounds or constructs in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. [00317] Upon formulation, solutions can be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but slow release capsules or microparticles and microspheres and the like can also be employed. [00318] For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intratumorally, intramuscular, subcutaneous and intraperitoneal administration. In this context, sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage could
Attorney Docket No.250298.000682 be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. [00319] The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. For example, a subject may be administered peptides, peptide-based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties described herein on a daily or weekly basis for a time period or on a monthly, bi-yearly or yearly basis depending on need or exposure to a pathogenic organism (e.g., HTLV-1) or to a condition in the subject (e.g. cancer). [00320] In addition to the compounds formulated for parenteral administration, such as intravenous, intratumorally, intradermal or intramuscular injection, other pharmaceutically acceptable forms include, e.g., tablets or other solids for oral administration; liposomal formulations; time release capsules; biodegradable and any other form currently used. [00321] One may also use intranasal or inhalable solutions or sprays, aerosols or inhalants. Nasal solutions can be aqueous solutions designed to be administered to the nasal passages in drops or sprays. Nasal solutions can be prepared so that they are similar in many respects to nasal secretions. Thus, the aqueous nasal solutions usually are isotonic and slightly buffered to maintain a pH of 5.5 to 7.5. In addition, antimicrobial preservatives, similar to those used in ophthalmic preparations, and appropriate drug stabilizers, if required, may be included in the formulation. Various commercial nasal preparations are known and can include, for example, antibiotics and antihistamines and are used for asthma prophylaxis. [00322] Oral formulations can include excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders. In certain defined embodiments, oral pharmaceutical compositions will include an inert diluent or assimilable edible carrier, or they may be enclosed in hard or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compounds may be incorporated
Attorney Docket No.250298.000682 with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. [00323] The tablets, troches, pills, capsules and the like may also contain the following: a binder, as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar, or both. A syrup of elixir may contain the active compounds sucrose as a sweetening agent methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor. [00324] Further embodiments disclosed herein can concern kits for use with methods and compositions. Kits can also include a suitable container, for example, vials, tubes, mini- or microfuge tubes, test tube, flask, bottle, syringe or other container. Where an additional component or agent is provided, the kit can contain one or more additional containers into which this agent or component may be placed. Kits herein will also typically include a means for containing the peptides, peptide-based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained. Optionally, one or more additional active agents may be needed for compositions described. [00325] Dose ranges and frequency of administration can vary depending on the nature of the composition and the medical condition as well as parameters of a specific patient and the route of administration used. A dose can also depend on the subject in which it is being administered. For example, a lower dose may be required if the subject is juvenile, and a higher dose may be required if the subject is an adult human subject. In certain embodiments, a more accurate dose can depend on the weight of the subject. A
Attorney Docket No.250298.000682 suitable, non-limiting example of a dosage of a pharmaceutical composition containing the same disclosed herein may vary depending upon the age and the size of a subject to be administered, target disease, the purpose of the treatment, conditions, route of administration, and the like. Non-limiting examples of suitable dosages include, e.g., 0.01 to about 20 mg/kg body weight, more preferably about 0.02 to about 7, about 0.03 to about 5, or about 0.05 to about 3 mg/kg body weight. Depending on the severity of the condition, the frequency and the duration of the treatment can be adjusted. In certain embodiments, the initial dose may be followed by administration of a second or a plurality of subsequent doses in an amount that can be approximately the same or less than that of the initial dose, wherein the subsequent doses are separated by at least 1 day to 3 days; at least one week, at least 2 weeks; at least 3 weeks; at least 4 weeks; at least 5 weeks; at least 6 weeks; at least 7 weeks; at least 8 weeks; at least 9 weeks; at least 10 weeks; at least 12 weeks; or at least 14 weeks. [00326] Compositions may include administration to a subject intravenously, intratumorally, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostaticaly, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, intramuscularly, intrathecally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, orally, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, via a catheter, via a lavage, in a cream, or in a lipid composition. [00327] Certain additional agents used in the combination therapies can be formulated and administered by any means known in the art. [00328] Compositions as disclosed herein can also include adjuvants such as aluminum salts and other mineral adjuvants, tensoactive agents, bacterial derivatives, vehicles and cytokines. Adjuvants can also have antagonizing immunomodulating properties. For example, adjuvants can stimulate Th1 or Th2 immunity. Compositions and methods as disclosed herein can also include adjuvant therapy. [00329] The peptides or peptides-based molecules of the disclosure may be provided in the form of a vaccine composition. The vaccine composition may be useful for the treatment, prevention, or reduction of the likelihood of HTLV-1 infection and/or HTLV-
Attorney Docket No.250298.000682 1-induced diseases or disorders. As will be appreciated, vaccines may take several forms (see, e.g., Schlom, J Natl Cancer Inst.2012; 104(8):599-613; Salgaller, Cancer Res.1996; 56(20):4749-57 and Marchand, Int J Cancer. 1999; 80(2):219-30). The vaccine composition may include additional peptides or peptides-based molecules such that the peptide or peptides-based molecule of the disclosure is one of a mixture of peptides or peptides-based molecules. Adjuvants may be added to the vaccine composition to augment the immune response. In particular for peptide-containing vaccines compositions of the disclosure, pharmaceutically acceptable adjuvants include, but are not limited to, aluminum salts, Amplivax, AS 15, Aquila’s QS21 stimulon, AsA404 (DMXAA), beta- glucan, BCG, CP-870,893, CpG7909, CyaA, dSLIM, GM-CSF, IC30, IC31, Imiquimod, ImuFact EV1P321, IS Patch, ISS, 1018 ISS, ISCOMATRIX, Juvlmmune, LipoVac, MF59, monophosphoryl lipid A, Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, OK-432, OM-174, OM-197-MP-EC, ONTAK, poly-ICLC, PepTel®, Pam3Cys, PLGA microparticles, resiquimod, SRL172, Virosomes and other Virus-like particles, YF-17D, VEGF trap, R848, and/or vadimezan. [00330] Alternatively, the vaccine composition may take the form of an APC displaying the peptide of the disclosure in complex with MHC. Preferably the APC is an immune cell, more preferably a dendritic cell or a B cell. The peptide may be pulsed onto the surface of the cell (Thurner, J Exp Med. 1999; 190(11):1669-78), or nucleic acid encoding for the peptide of the disclosure may be introduced into dendritic cells or B cells (e.g., by electroporation. Van Tendeloo, Blood.2001; 98(1):49-56). [00331] The pharmaceutical compositions of the disclosure may be administered directly into the patient, into the affected organ or systemically i.d., i.m., s.c., i.p. and i.v., or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation of immune cells derived from the patient, which are then re-administered to the patient. If the nucleic acid is administered to cells in vitro, it may be useful for the cells to be transfected so as to co-express immune-stimulating cytokines, such as interleukin-2. The peptide or peptide-based molecule may be substantially pure, or combined with an immune- stimulating adjuvant or used in combination with immune-stimulatory cytokines, or be administered with a suitable delivery system, e.g., liposomes, viral particles, VLPs. The
Attorney Docket No.250298.000682 peptide or peptide-based molecule may also be conjugated to a suitable carrier such as keyhole limpet haemocyanin (KLH) or mannan (see, e.g., WO 95/18145 and Longenecker et al., 1993). [00332] In some embodiments, the peptide-containing compositions described herein further comprise an accessory molecule which can modulate a survival or an activity of TCR-expressing cells. [00333] Non-limiting examples of useful accessory molecules include, e.g., an anti- CD28 antibody, an anti-CD80 (B7.1) antibody, an anti-CD86 (B7.2) antibody, an anti-anti- CD3 antibody, an anti-CD2 antibody, an anti-CD4 antibody, an anti-CD8 antibody, an anti- CD47 antibody, and functional derivatives, mutants and fragments thereof. [00334] Accessory molecules used in the peptide-containing compositions described herein include molecules that provide a signal which, in addition to the primary signal provided by, for instance, binding of a TCR/CD3 complex with a pMHC complex, mediates a T cell response, including, but not limited to, proliferation, activation, differentiation, and the like. [00335] The accessory molecule can be, for example, an inhibitory or stimulatory antibody, a peptide ligand, a costimulatory peptide, a cytokine, etc. Non-limiting examples of accessory molecules that can be used in the peptide-containing compositions described herein include, e.g., CD7, B7.1 (CD80), B7.2 (CD86), PD-L1 , PD-L2, 4-1BBL, OX40L, Fas ligand (FasL), inducible co stimulatory ligand (ICOS-L), intercellular adhesion molecule (ICAM), CD30L, CD40, CD70, CD83, HLA-G, MICA, MICB, FIVEM, lymphotoxin β receptor, 3/TR6, ILT3, ILT4, HVEM, an agonist or antibody that binds Toll ligand receptor and a ligand that specifically binds to B7-H3 as well as antibodies that specifically bind to CD27, CD28, B7.1 (CD80), B7.2 (CD86), 4-1BB, OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD3, CD7, LIGHT, NKG2C, B7-H3, and a ligand that specifically binds to CD83. [00336] Additional non-limiting examples of accessory molecules include, e.g., TNF/TNF family members (e.g., OX40L, ICOSL, FASL, LTA, LTB TRAIL, CD153, TNFSF9, RANKL, TWEAK, TNFSF13, TNFSF13b, TNFSF14, TNFSF15, TNFSF18, CD40LG, CD70); members of the Immunoglobulin superfamily (e.g., VISTA, PD1, PD- L1 , PD-L2, B71 , B72, CTLA4, CD28, TIM3, CD4, CD8, CD19, T cell receptor chains,
Attorney Docket No.250298.000682 ICOS, ICOS ligand, HHLA2, butyrophilms, BTLA, B7-H3, B7-H4, CD3, CD79a, CD79b, IgSF CAMS (including CD2, CD58, CD48, CD150, CD229, CD244, ICAM-1), Leukocyte immunoglobulin like receptors (LILR), killer cell immunoglobulin like receptors (KIR)), lectin superfamily members, selectins, cytokines/chemokine and cytokine/chemokine receptors, growth factors and growth factor receptors), adhesion molecules (integrins, fibronectins, cadherins), or ecto-domains of multi-span integral membrane proteins, or antibodies directed to any of these molecules. [00337] In some embodiments, the peptide-containing compositions described herein further comprise a cytotoxic agent. In one specific embodiment, the cytotoxic agent is a toxin or a radioactive isotope (e.g., a radioconjugate) or a suicide gene. Non-limiting examples of toxins which can be used in the peptide-containing compositions described herein include, e.g., enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments, mutants or derivatives thereof. Enzymatically active toxins and fragments thereof that can be used include, for example, diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, α-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. Non-limiting examples of suicide genes include, e.g., thymidine kinase, cytosine deaminase, purine nucleoside phosphorylase, nitroreductase, β-galactosidase, hepatic cytochrome P450-2B1, linamarase, horseradish peroxidase, and carboxypeptidase. [00338] Methods for introducing polypeptide or polynucleotides of the present disclosure into a cell or subject can include, for example, vector delivery, particle-mediated delivery, exosome-mediated delivery, lipid-nanoparticle-mediated delivery, cell- penetrating-peptide-mediated delivery, or implantable-device-mediated delivery. In some embodiments, a nucleic acid or protein can be introduced into a cell or subject in a carrier such as a poly(lactic acid) (PLA) microsphere, a poly(D,L-lactic-coglycolic-acid) (PLGA) microsphere, a liposome, a micelle, an inverse micelle, a lipid cochleate, or a lipid microtubule.
Attorney Docket No.250298.000682 [00339] The use of nanoparticles to deliver the polypeptide or polynucleotides compositions of the disclosure is contemplated herein. Exemplary nanoparticles include, but are not limited to, polymeric nanoparticles, inorganic nanoparticles, liposomes, lipid nanoparticles (LNP), an immune stimulating complex (ISCOM), a virus-like particle (VLP), or a self-assembling protein. The nanoparticles may be calcium phosphate nanoparticles, silicon nanoparticles or gold nanoparticles. For examples, the polymeric nanoparticles may comprise one or more synthetic polymers, such as poly(d,l-lactide-co- glycolide) (PLG), poly(d,l-lactic-coglycolic acid) (PLGA), poly(g-glutamic acid) (g- PGA), poly(ethylene glycol) (PEG), or polystyrene or one or more natural polymers such as a polysaccharide, for example pullulan, alginate, inulin, and chitosan. The use of a polymeric nanoparticles may be advantageous due to the properties of the polymers that may be include in the nanoparticle. For instance, the natural and synthetic polymers recited above may have good biocompatibility and biodegradability, a non-toxic nature and/or the ability to be manipulated into desired shapes and sizes. The polymeric nanoparticle may also form hydrogel nanoparticles, hydrophilic three-dimensional polymer networks with favorable properties including flexible mesh size, large surface area for multivalent conjugation, high water content, and high loading capacity for antigens. Polymers such as Poly(L-lactic acid) (PLA), PLGA, PEG, and polysaccharides are suitable for forming hydrogel nanoparticles. Inorganic nanoparticles typically have a rigid structure and comprise a shell in which an antigen is encapsulated or a core to which the antigen may be covalently attached. The core may comprise one or more atoms such as gold (Au), silver (Ag), copper (Cu) atoms, Au/Ag, Au/Cu, Au/Ag/Cu, Au/Pt, Au/Pd or Au/Ag/Cu/Pd or calcium phosphate (CaP). [00340] Other molecules suitable for complexing with the polypeptide or polynucleotides of the disclosure include cationic molecules, such as, polyamidoamine , dendritic polylysine, polyethylene irinine or polypropylene imine, polylysine, chitosan, DNA-gelatin coarcervates, DEAE dextran, dendrimers, or polyethylenimine (PEI). [00341] In some embodiments, antibodies of the present disclosure can be conjugated to nanoparticles. Nanoparticles that may be used for conjugation with antibodies of the present disclosure include but not are limited to PEGylated liposomes, poly(d,l-lactide-co-glycolide)/montmorillonite nanoparticles (PLGA/MMT NPs),
Attorney Docket No.250298.000682 poly(lactide-co-glycolide) (PLGA) nanoparticles, poly-(malic acid)-based nanoparticles, chitosan-shelled nanoparticles, carbon nanotubes, and other inorganic nanoparticles (such as nanoparticles made of magnesium–aluminium layered double hydroxides with disuccinimidyl carbonate (DSC), and TiO2 nanoparticles). Nanoparticles can be developed and conjugated to an antibody contained in a pharmaceutical composition for targeting virus-infected cells. Treatment Methods [00342] Compositions of the present disclosure, including the peptides, peptide- based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties of the disclosure, may be used in the prophylaxis and/or treatment of a viral infection (e.g., HTLV-1 infection) and/or diseases or disorders caused by the viral infection (e.g., HTLV-1 infection). [00343] In one aspect, disclosed herein is a method for modulating an activity, proliferation or survival of a cell comprising a TCR, comprising contacting the cell with a composition (e.g., peptide, complex (e.g., pMHC complex), fusion protein, or conjugate) of the disclosure. [00344] In some embodiments, the cell is a lymphocyte such as, e.g., a T-cell (e.g., a CD4+ T-cell or a CD8+ T-cell). In some embodiments, the target T cell is a CD4+ T cell such as, e.g., a helper T cell (e.g., a Th1, Th2, or Th17 cell) or a CD4+/CD25+/FOXP3+ regulatory T (Treg) cell. In some cases, the target T cell is a CD8+ T cell such as, e.g., a cytotoxic T cell. In some cases, the target T cell is a memory T cell, which can be a CD4+ T cell or a CD8+ T cell, where memory T cells are generally CD45RO+. In some cases, the target T cell is an NK-T cell. [00345] In some embodiments, the contacting is ex vivo. In some embodiments, the contacting is in vivo in a subject (e.g., human). [00346] In some embodiments, the cell is a mammalian cell (e.g., a human cell).
Attorney Docket No.250298.000682 [00347] In some embodiments, e.g., where the target T cell is a CD8+ T cell, the peptide is presented by a class I MHC polypeptide. In some embodiments, e.g., where the target T cell is a CD4+ T cell, the peptide is presented by class II MHC polypeptides. [00348] The interaction of a T cell with the peptides described herein can result in, e.g., activation, induction of anergy, or death of a T cell that occurs when the TCR of the T cell is bound by a TCR-binding molecule (e.g., pMHC complex). “Activation of a T cell” refers to induction of signal transduction pathways in the T cell resulting in production of cellular products (e.g., interleukin-2) by that T cell. “Anergy” refers to the diminished reactivity by a T cell to an antigen. Activation and anergy can be measured by, for example, measuring the amount of IL-2 produced by a T cell after a pMHC complex has bound to the TCR. Anergic cells will have decreased IL-2 production when compared with stimulated T cells. Another method for measuring the diminished activity of anergic T cells includes measuring intracellular and/or extracellular calcium mobilization by a T cell upon engagement of its TCR’s. “T cell death” refers to the permanent cessation of substantially all functions of the T cell. [00349] In another aspect, provided herein is a method of inducing an immune response against HTLV infection (e.g., a HTLV-1, HTLV-2, HTLV-3, and/or HTLV-4 infection) in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of a composition (e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, cells, or binding moieties) of the present disclosure. [00350] In certain embodiments, generating an immune response comprises an increase in target antigen-specific cytotoxic T lymphocytes (CTL) activity of about 1.5- fold to 20-fold, or more fold in a subject administered a composition of the disclosure as compared to a control. In certain embodiments, generating an immune response comprises an increase in target-specific CTL activity of about 1.5-fold to 20-fold, or more fold in a subject administered the composition of the disclosure as compared to a control. In a further embodiment, generating an immune response that comprises an increase in target antigen- specific cell-mediated immunity activity as measured by ELISpot assays measuring cytokine secretion, such as interferon-gamma (IFN-γ), interleukin-2 (IL-2), tumor necrosis
Attorney Docket No.250298.000682 factor-alpha (TNF-α), or other cytokines, of about 1.5-fold to 20-fold, or more fold as compared to a control. [00351] In a further embodiment, generating an immune response comprises an increase in target- specific antibody production of between 1.5-fold and 5-fold in a subject administered the composition of the disclosure as compared to an appropriate control. In another embodiment, generating an immune response comprises an increase in target- specific antibody production of about 1.5-fold to 20-fold, or more fold in a subject administered the composition of the disclosure as compared to a control. [00352] T cell activation may be determined, e.g., by measuring changes in the level of expression of cytokines and/or T cell activation markers, and/or the induction of antigen- specific proliferating cells. Techniques known to those of skill in the art, including, but not limited to, immunoprecipitation followed by western blot analysis, ELISAs, flow cytometry, northern blot analysis, and RT-PCR can be used to measure the expression cytokines and T cell activation markers. Cytokine release may be measured by measuring secretion of cytokines including but not limited to Interleukin-2 (IL-2), Interleukin-4 (IL- 4), Interleukin-6 (IL-6), Interleukin-12 (IL-12), Interleukin-16 (IL-16), PDGF, TGF-α, TGF-β, TNF-α, TNF-β, GCSF, GM-CSF, MCSF, IFN-α, IFN-β, IFN-γ, TFN-γ, IGF-I, and IGF-II. [00353] T cell modulation may also be evaluated by measuring, e.g., proliferation by, e.g., 3H-thymidine incorporation, trypan blue cell counts, and fluorescence activated cell sorting (FACS). [00354] The anti-tumor responses of T cells may be determined in xenograft tumor models. Tumors may be established using any human cancer cell line expressing the relevant tumor associated antigen. To establish xenograft tumor models, about 5×106 viable cells, may be injected, e.g., subcutaneously into nude athymic mice using for example Matrigel (Becton Dickinson). The endpoint of the xenograft tumor models can be determined based on the size of the tumors, weight of animals, survival time and histochemical and histopathological examination of the cancer, using methods known to one skilled in the art. [00355] In a related aspect, disclosed herein is a method of treating an HTLV-1 infection in a subject in need thereof, the method comprising administering to the subject
Attorney Docket No.250298.000682 an effective amount of a composition (e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, cells, binding moieties) of the present disclosure. [00356] In a related aspect, disclosed herein is a method of preventing or reducing the likelihood of an HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of a composition (e.g., one or more peptides, complexes (e.g., pMHC complex), fusion proteins, conjugates, nucleic acid molecules, vectors, a cells, binding moieties) of the present disclosure. [00357] The HTLV-1-induced disease or disorder can be, e.g., adult T-cell leukemia/lymphoma (ATL) or HTLV-1-myelopathy/tropical spastic paraparesis (HAM/TSP). In some embodiments, the HTLV-1-induced disease or disorder can be, e.g., a hypersensitivity reaction such as, without limitation, arthritis, uveitis, or HTLV-1- associated Infective Dermatitis (IDH). [00358] Non-limiting examples of ATL are acute ATL, lymphoma ATL, chronic ATL, and smoldering ATL. The most benign but detectable form of ATL is an asymptomatic pre-leukemic phase which can be diagnosed, e.g., incidentally upon examination of a peripheral blood smear which reveals abnormal lymphocytes with characteristics of lobulated nuclei Smoldering ATL is the most benign symptomatic form of ATL and can be characterized by a normal peripheral blood leukocyte count, a few circulating leukemic cells, and cutaneous lesions but not visceral lesions. The development of chronic ATL is associated with visceral involvement and is evidenced by hepatosplenomegaly, lymphadenopathy, and peripheral blood leukocytosis. Patients with acute ATL can have elevated levels of lactate dehydrogenase and bilirubin, peripheral blood leukocytosis, hypercalcemia, and cutaneous and visceral involvement. ATL can be accompanied by severe immunosuppression and many ATL patients can be highly susceptible to various infections such, but not limited to cryptococcal meningitis, pneumocystis carinii pneumonia, disseminated cytomegalovirus infection, and candida esophagitis. Acute ATL is typically unresponsive to conventional chemotherapy and the median life expectancy of patients with acute ATL is approximately 11 months. [00359] HTLV-1 is thought to cause approximately 80 percent of the cases of HAM/TSP such as by weakening the immune system. A majority of patients with
Attorney Docket No.250298.000682 HAM/TSP have few or no symptoms over the lifespan. In addition to neurological symptoms of weakness and muscle spasms and/or stiffness, in rare instances patients with HAM/TSP may also have, e.g., arthritis (inflammation of one or more joints); infectious dermatitis (inflammation of the skin); keratoconjunctivitis sicca (persistent dryness of the cornea and/or conjunctiva); polymyositis (an inflammatory muscle disease); pulmonary lymphocytic alveolitis (inflammation of the lung); and/or uveitis (inflammation of the uveal tract of the eye). [00360] When the disease being treated is a cancer, the cancer may specifically be of the following histological type, though it is not limited to these: neoplasm, malignant; carcinoma; carcinoma, undifferentiated; giant and spindle cell carcinoma; small cell carcinoma; papillary carcinoma; squamous cell carcinoma; lymphoepithelial carcinoma; basal cell carcinoma; pilomatrix carcinoma; transitional cell carcinoma; papillary transitional cell carcinoma; adenocarcinoma; gastrinoma, malignant; cholangiocarcinoma; hepatocellular carcinoma; combined hepatocellular carcinoma and cholangiocarcinoma; trabecular adenocarcinoma; adenoid cystic carcinoma; adenocarcinoma in adenomatous polyp; adenocarcinoma, familial polyposis coli; solid carcinoma; carcinoid tumor, malignant; branchiolo-alveolar adenocarcinoma; papillary adenocarcinoma; chromophobe carcinoma; acidophil carcinoma; oxyphilic adenocarcinoma; basophil carcinoma; clear cell adenocarcinoma; granular cell carcinoma; follicular adenocarcinoma; papillary and follicular adenocarcinoma; nonencapsulating sclerosing carcinoma; adrenal cortical carcinoma; endometroid carcinoma; skin appendage carcinoma; apocrine adenocarcinoma; sebaceous adenocarcinoma; ceruminous adenocarcinoma; mucoepidermoid carcinoma; cystadenocarcinoma; papillary cystadenocarcinoma; papillary serous cystadenocarcinoma; mucinous cystadenocarcinoma; mucinous adenocarcinoma; signet ring cell carcinoma; infiltrating duct carcinoma; medullary carcinoma; lobular carcinoma; inflammatory carcinoma; paget's disease, mammary; acinar cell carcinoma; adenosquamous carcinoma; adenocarcinoma w/squamous metaplasia; thymoma, malignant; ovarian stromal tumor, malignant; thecoma, malignant; granulosa cell tumor, malignant; and roblastoma, malignant; sertoli cell carcinoma; leydig cell tumor, malignant; lipid cell tumor, malignant; paraganglioma, malignant; extra-mammary paraganglioma, malignant; pheochromocytoma; glomangiosarcoma; malignant melanoma; amelanotic melanoma;
Attorney Docket No.250298.000682 superficial spreading melanoma; malig melanoma in giant pigmented nevus; epithelioid cell melanoma; blue nevus, malignant; sarcoma; fibrosarcoma; fibrous histiocytoma, malignant; myxosarcoma; liposarcoma; leiomyosarcoma; rhabdomyosarcoma; embryonal rhabdomyosarcoma; alveolar rhabdomyosarcoma; stromal sarcoma; mixed tumor, malignant; mullerian mixed tumor; nephroblastoma; hepatoblastoma; carcinosarcoma; mesenchymoma, malignant; brenner tumor, malignant; phyllodes tumor, malignant; synovial sarcoma; mesothelioma, malignant; dysgerminoma; embryonal carcinoma; teratoma, malignant; struma ovarii, malignant; choriocarcinoma; mesonephroma, malignant; hemangiosarcoma; hemangioendothelioma, malignant; kaposi's sarcoma; hemangiopericytoma, malignant; lymphangiosarcoma; osteosarcoma; juxtacortical osteosarcoma; chondrosarcoma; chondroblastoma, malignant; mesenchymal chondrosarcoma; giant cell tumor of bone; ewing's sarcoma; odontogenic tumor, malignant; ameloblastic odontosarcoma; ameloblastoma, malignant; ameloblastic fibrosarcoma; pinealoma, malignant; chordoma; glioma, malignant; ependymoma; astrocytoma; protoplasmic astrocytoma; fibrillary astrocytoma; astroblastoma; glioblastoma; oligodendroglioma; oligodendroblastoma; primitive neuroectodermal; cerebellar sarcoma; ganglioneuroblastoma; neuroblastoma; retinoblastoma; olfactory neurogenic tumor; meningioma, malignant; neurofibrosarcoma; neurilemmoma, malignant; granular cell tumor, malignant; malignant lymphoma; Hodgkin's disease; Hodgkin's lymphoma; paragranuloma; malignant lymphoma, small lymphocytic; malignant lymphoma, large cell, diffuse; malignant lymphoma, follicular; mycosis fungoides; other specified non-Hodgkin's lymphomas; malignant histiocytosis; multiple myeloma; mast cell sarcoma; immunoproliferative small intestinal disease; leukemia; lymphoid leukemia; plasma cell leukemia; erythroleukemia; lymphosarcoma cell leukemia; myeloid leukemia; basophilic leukemia; eosinophilic leukemia; monocytic leukemia; mast cell leukemia; megakaryoblastic leukemia; myeloid sarcoma; and hairy cell leukemia. [00361] It is contemplated that when used to treat various diseases, the compositions and methods can be combined with other therapeutic agents suitable for the same or similar diseases. Also, two or more embodiments described herein may be also co-administered to generate additive or synergistic effects. When co-administered with a second therapeutic
Attorney Docket No.250298.000682 agent, the embodiment described herein and the second therapeutic agent may be simultaneously or sequentially (in any order). Suitable therapeutically effective dosages for each agent may be lowered due to the additive action or synergy. [00362] In some embodiments, the compositions and methods disclosed herein are useful to enhance the efficacy of vaccines directed to HTLV-1 infection or HTLV-1- induced diseases. Thus, the compositions and methods described herein can be administered to a subject either simultaneously with or before (e.g., 1-30 days before) a reagent (including but not limited to small molecules, antibodies, or cellular reagents) that acts to elicit an immune response (e.g., to treat HTLV-1, HTLV-2, HTLV-3, and/or HTLV- 4 infection or cancer) is administered to the subject. [00363] The compositions and methods described herein can be also administered in combination with an anti-tumor antibody or an antibody directed at a pathogenic antigen (e.g., HTLV-1) or allergen. [00364] The compositions and methods described herein can be combined with other immunomodulatory treatments such as, e.g., therapeutic vaccines (including but not limited to GVAX, DC-based vaccines, etc.), checkpoint inhibitors (including but not limited to agents that block CTLA4, PD1, LAG3, TIM3, etc.) or activators (including but not limited to agents that enhance 41BB, OX40, etc.). The inhibitory treatments described herein can be also combined with other treatments that possess the ability to modulate NKT function or stability, including but not limited to CD1d, CD1d-fusion proteins, CD1d dimers or larger polymers of CD1d either unloaded or loaded with antigens, CD1d- chimeric antigen receptors (CD1d-CAR), or any other of the five known CD1 isomers existing in humans (CD1a, CD1b, CD1c, CD1e), in any of the aforementioned forms or formulations, alone or in combination with each other or other agents. [00365] Therapeutic methods described herein can be combined with additional immunotherapies and therapies. For example, when used for treating cancer, NKT cells described herein can be used in combination with cancer therapies, such as, e.g., surgery, radiotherapy, chemotherapy or combinations thereof, depending on type of the tumor, patient condition, other health issues, and a variety of factors. In certain aspects, other therapeutic agents useful for combination cancer therapy with the inhibitors described herein include anti-angiogenic agents. Many anti-angiogenic agents have been identified
Attorney Docket No.250298.000682 and are known in the art, including, e.g., TNP-470, platelet factor 4, thrombospondin-1, tissue inhibitors of metalloproteases (TIMP1 and TIMP2), prolactin (16-Kd fragment), angiostatin (38-Kd fragment of plasminogen), endostatin, bFGF soluble receptor, transforming growth factor β, interferon-α, soluble KDR and FLT-1 receptors, placental proliferin-related protein, as well as those listed by Carmeliet and Jain (2000). In some embodiments, the inhibitors described herein can be used in combination with a VEGF antagonist or a VEGF receptor antagonist such as anti-VEGF antibodies, VEGF variants, soluble VEGF receptor fragments, aptamers capable of blocking VEGF or VEGFR, neutralizing anti-VEGFR antibodies, inhibitors of VEGFR tyrosine kinases and any combinations thereof (e.g., anti-hVEGF antibody A4.6.1, bevacizumab or ranibizumab). [00366] Non-limiting examples of chemotherapeutic compounds which can be used in combination treatments include, for example, aminoglutethimide, amsacrine, anastrozole, asparaginase, bcg, bicalutamide, bleomycin, buserelin, busulfan, campothecin, capecitabine, carboplatin, carmustine, chlorambucil, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, estradiol, estramnustine, etoposide, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone, flutamide, gemcitabine, genistein, goserelin, hydroxyurea, idarubicin, ifosfamide, imatinib, interferon, irinotecan, ironotecan, letrozole, leucovorin, leuprolide, levamisole, lomustine, mechlorethamine, medroxyprogesterone, megestrol, melphalan, mercaptopurine, mesna, methotrexate, mitomycin, mitotane, mitoxantrone, nilutamide, nocodazole, octreotide, oxaliplatin, paclitaxel, pamidronate, pentostatin, plicamycin, porfimer, procarbazine, raltitrexed, rituximab, streptozocin, suramin, tamoxifen, temozolomide, teniposide, testosterone, thioguanine, thiotepa, titanocene dichloride, topotecan, trastuzumab, tretinoin, vinblastine, vincristine, vindesine, and vinorelbine. [00367] These chemotherapeutic compounds may be categorized by their mechanism of action into, for example, following groups: anti-metabolites/anti-cancer agents, such as pyrimidine analogs (5-fluorouracil, floxuridine, capecitabine, gemcitabine and cytarabine) and purine analogs, folate antagonists and related inhibitors (mercaptopurine, thioguanine, pentostatin and 2-chlorodeoxyadenosine (cladribine));
Attorney Docket No.250298.000682 antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinblastine, vincristine, and vinorelbine), microtubule disruptors such as taxane (paclitaxel, docetaxel), vincristin, vinblastin, nocodazole, epothilones and navelbine, epidipodophyllotoxins (etoposide, teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, busulfan, camptothecin, carboplatin, chlorambucil, cisplatin, cyclophosphamide, cytoxan, dactinomycin, daunorubicin, doxorubicin, epirubicin, hexamethyhnelamineoxaliplatin, iphosphamide, melphalan, merchlorehtamine, mitomycin, mitoxantrone, nitrosourea, plicamycin, procarbazine, taxol, taxotere, teniposide, triethylenethiophosphoramide and etoposide (VP16)); antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (adriamycin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin) and mitomycin; enzymes (L-asparaginase which systemically metabolizes L-asparagine and deprives cells which do not have the capacity to synthesize their own asparagine); antiplatelet agents; antiproliferative/antimitotic alkylating agents such as nitrogen mustards (mechlorethamine, cyclophosphamide and analogs, melphalan, chlorambucil), ethylenimines and methylmelamines (hexamethylmelamine and thiotepa), alkyl sulfonates-busulfan, nitrosoureas (carmustine (BCNU) and analogs, streptozocin), trazenes-dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes (cisplatin, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones, hormone analogs (estrogen, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole); anticoagulants (heparin, synthetic heparin salts and other inhibitors of thrombin); fibrinolytic agents (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidine, clopidogrel, abciximab; antimigratory agents; antisecretory agents (breveldin); immunosuppressives (cyclosporine, tacrolimus (FK-506), sirolimus (rapamycin), azathioprine, mycophenolate mofetil); anti- angiogenic compounds (e.g., TNP-470, genistein, bevacizumab) and growth factor inhibitors (e.g., fibroblast growth factor (FGF) inhibitors); angiotensin receptor blocker; nitric oxide donors; anti-sense oligonucleotides; antibodies (trastuzumab); cell cycle inhibitors and differentiation inducers (tretinoin); mTOR inhibitors, topoisomerase inhibitors (doxorubicin (adriamycin), amsacrine, camptothecin, daunorubicin,
Attorney Docket No.250298.000682 dactinomycin, eniposide, epirubicin, etoposide, idarubicin and mitoxantrone, topotecan, irinotecan), corticosteroids (cortisone, dexamethasone, hydrocortisone, methylpednisolone, prednisone, and prenisolone); growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers and caspase activators; and chromatin disruptors. [00368] For treatment of viral infections, combined therapy described herein can encompass co-administering compositions and methods described herein with an anti-viral drug. Non-limiting examples of useful anti-viral drugs include, adefovir and entecavir, telbivudine, immune system modulators such as interferon-α, -β or -γ, didanosine, lamivudine, zanamavir, lopanivir, nelfinavir, efavirenz, indinavir, valacyclovir, zidovudine, amantadine, rimantidine, ribavirin, ganciclovir, foscarnet, and acyclovir or any salts or variants thereof. See also Physician's Desk Reference, 59.sup.th edition, (2005), Thomson P D R, Montvale N.J.; Gennaro et al., Eds. Remington's The Science and Practice of Pharmacy 20.sup.th edition, (2000), Lippincott Williams and Wilkins, Baltimore Md.; Braunwald et al., Eds. Harrison's Principles of Internal Medicine, 15.sup.th edition, (2001), McGraw Hill, NY; Berkow et al., Eds. The Merck Manual of Diagnosis and Therapy, (1992), Merck Research Laboratories, Rahway N.J. Kits [00369] The present disclosure further comprises a kit which may comprise any of various compositions of the present disclosure, including the peptides, peptide-based molecules (such as complexes (e.g., peptide-MHC (pMHC) complexes), fusion proteins, or conjugates comprising the peptide(s)), nucleic acid molecules, vectors, cells, or binding moieties of the disclosure. [00370] In one aspect, the present disclosure may include a kit comprising, for example: (a) a container that contains a pharmaceutical composition disclosed herein, for example, a pharmaceutical composition in solution or in lyophilized form; (b) optionally, a second container containing a diluent or reconstituting solution for the lyophilized formulation; and/or (c) optionally, instructions for (i) use of the solution or (ii) reconstitution and/or use of the lyophilized formulation.
Attorney Docket No.250298.000682 [00371] In some embodiments, the kit may further comprise, for example, without limitation, one or more of (i) a buffer, (ii) a diluent, (iii) a filter, (iv) a needle, and/or (v) a syringe. As a non-limiting example, the container may be a bottle, a vial, a syringe or test tube. In some embodiments, the container may be a multi-use container. In some the pharmaceutical composition may be lyophilized. [00372] Kits of the present disclosure may comprise a lyophilized formulation of the present disclosure in a suitable container and instructions for its reconstitution and/or use. Suitable containers include, for example, bottles, vials (e.g. dual chamber vials), syringes (such as dual chamber syringes) and test tubes. The container may be formed from a variety of materials such as glass or plastic. The kit and/or container may contain instructions on or associated with the container that indicate directions for reconstitution of the lyophilized formulation and/or use of the kit. For example, the label may indicate that the lyophilized formulation is to be reconstituted to an appropriate peptide concentration. The label may indicate that the formulation is useful or intended for any route of administration disclosed herein, e.g., parenteral administration routes disclosed herein. [00373] The container holding the formulation may be a multi-use vial, which may allow for repeat administrations (e.g., from 2-6 administrations) of the reconstituted formulation. The kit may further comprise a second container comprising a suitable diluent (e.g., sodium bicarbonate solution). [00374] Upon mixing of the diluent and the lyophilized formulation, the final peptide concentration in the reconstituted formulation is reached. The kit may further include other materials desirable from a commercial and/or user standpoint, including, for example, without limitation, other buffers, diluents, filters, needles, syringes, and/or package inserts which may comprise, e.g., instructions for use. [00375] Kits of the present disclosure may have a single container that contains the formulation of the pharmaceutical compositions according to the present disclosure with or without other components (e.g., other compounds or pharmaceutical compositions of these other compounds) or may have a distinct container for each component. [00376] In some embodiments, kits of the disclosure may include a formulation of the disclosure packaged for use in combination with the coadministration of a second compound (such as adjuvants (e.g., GM-CSF, a chemotherapeutic agent, a natural product,
Attorney Docket No.250298.000682 a hormone or antagonist, an anti-angiogenesis agent or inhibitor, an apoptosis-inducing agent or a chelator) or a pharmaceutical composition thereof. The components of the kit may be pre-complexed or each component may be in a separate distinct container prior to administration to a patient. The components of the kit may be provided in one or more liquid solutions. A liquid solutions described herein may be an aqueous solution, for example, a sterile aqueous solution. The components of the kit may also be provided as solids, which may be converted into liquids such as by addition of suitable solvents, which may be provided in another distinct container. [00377] The container of a therapeutic kit may be a vial, test tube, flask, bottle, syringe, or any other means of enclosing a solid or liquid. When there is more than one component, the kit may contain a second vial or other container, which may allow for separate dosing. The kit may also contain another container for a pharmaceutically acceptable liquid. In some embodiment, a kit may contain an apparatus (e.g., one or more needles, syringes, eye droppers, pipettes, etc.), which may allow for administration of the agents of the disclosure that are components of the present kit. Method and System for Identifying an Immunogenic Virus-Derived Peptide [00378] The present disclosure further comprises a method and system for identifying an immunogenic virus-derived peptide. Some or all the steps of the method can be executed by a computational device. In some aspects, some or all of the steps of the method can be stored as computer-readable instructions within a memory such that the instructions can be executed by one or more processors to perform functions associated with the method. [00379] In one aspect, the present disclosure may include a method for identifying an immunogenic virus-derived peptide that includes the following steps: (a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus, wherein the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences and a plurality of infected-subject endogenous RNA contig sequences; (b) identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences; (c) assembling a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; (d) identifying a protein sequence based
Attorney Docket No.250298.000682 on the viral RNA sequence; and (e) identifying the immunogenic virus-derived peptide based at least in part on the identified protein sequence. [00380] In one aspect, a system configured to perform functions associated with the foregoing method can include a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device. The non-transitory computer-readable medium may include instructions thereon, that when executed by the processor(s), cause the computational device to: (a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences and a plurality of infected-subject endogenous RNA contig sequences, and wherein the infected subject is infected with a virus; (b) identify the plurality of virus- derived RNA contig sequences from within the plurality of RNA contig sequences; (c) assemble a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; (d) identify a protein sequence based on the viral RNA sequence; (e) identify an immunogenic virus-derived peptide based at least in part on the protein sequence; and (f) provide, as an output, the immunogenic virus-derived peptide. [00381] In some embodiments, the plurality of RNA contig sequences may be derived from one infected subject. [00382] In some embodiments, the infected subject may be a human. [00383] In some embodiments, the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject. [00384] In some embodiments, the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject. [00385] In some embodiments, identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences (step b) can further include: comparing at least a portion of contig sequences of the plurality of RNA contig sequences to a reference viral sequence; and identifying the plurality of virus-derived RNA contig sequences such that each contig sequence of the plurality of virus-derived RNA contig sequences comprises at least a portion that corresponds to the reference viral sequence.
Attorney Docket No.250298.000682 [00386] In some embodiments, each contig sequence of the plurality of virus- derived RNA contig may be distinct from the plurality of infected-subject endogenous RNA contig sequences. [00387] In some embodiments, each contig sequence of the plurality of virus- derived RNA contig sequences may lack infected-subject endogenous RNA contig sequences. [00388] In some embodiments, the reference viral sequence may include a reference genome. [00389] In some embodiments, the reference genome may include a HTLV-1 virus genome. [00390] In some embodiments, assembling the viral RNA sequence based on the plurality of virus-derived RNA contig sequences (step c) may include: overlapping common sequence portions at ends of at least a portion of the plurality of virus-derived RNA contig sequences such that the at least a portion of the plurality of virus-derived RNA contig sequences overlap linearly to assemble the viral RNA sequence. [00391] In some embodiments, identifying a protein sequence based on the viral RNA sequence such that the protein sequence includes a translation of the viral RNA sequence (step d) may include: identifying the protein sequence without requiring a comparison to a database of viral proteins. [00392] In some embodiments, identifying the protein sequence based on the viral RNA sequence such that the protein sequence includes a translation of the viral RNA sequence (step d) may include: identifying a plurality of protein sequences each based on the viral RNA sequence such that each of the plurality of protein sequences respectively include a translation of the viral RNA sequence, and identifying the protein sequence as a frequently occurring protein sequence within the plurality of protein sequences. [00393] In some embodiments, the protein sequence may be identified based on the viral RNA sequence associated with a single infected subject. [00394] In some embodiments, identifying the immunogenic virus-derived peptide based at least in part on the protein sequence (step 3) may include: identifying a MHC molecule associated with the single infected subject; identifying one or more peptides based at least in part on the protein sequence such that the one or more peptides each form
Attorney Docket No.250298.000682 a respective MHC-peptide complex with the MHC molecule; and identifying the immunogenic virus-derived peptide based on the one or more peptides. Method and System for Identifying an Integration Site of a Viral Gene within a Subject Gene [00395] The present disclosure further comprises a method and system for identifying an integration site of a viral gene within a subject gene. Some or all the steps of the method can be executed by a computational device. In some aspects, some or all of the steps of the method can be stored as computer-readable instructions within a memory such that the instructions can be executed by one or more processors to perform functions associated with the method. [00396] In one aspect, the present disclosure may include a method for identifying an integration site of a viral gene within a subject gene that includes the following steps: (a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus- derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and a plurality of hybrid RNA contig sequences comprising viral and infected- subject endogenous portions; (b) identifying the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; (c) comparing, for at least a portion of the plurality of hybrid RNA contig sequences, infected-subject endogenous portions to a subject reference genome; and (d) identifying, based at least in part on the comparison of infected-subject endogenous portions to the subject reference genome, an integration site comprising the subject gene. [00397] In one aspect, a system configured to perform functions associated with the foregoing method can include a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device. The non-transitory computer-readable medium may include instructions thereon, that when executed by the processor(s), cause the computational device to: (a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and plurality
Attorney Docket No.250298.000682 of hybrid RNA contig sequences comprising viral and infected-subject endogenous portions; (b) identify the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; (c) compare, for at least a portion of the plurality of hybrid RNA contig sequences, infected-subject endogenous portions to a subject reference genome; (d) identify, based at least in part on the comparison of infected-subject endogenous portions to the subject reference genome, an integration site comprising a subject gene; and (e) provide, as an output, the integration site. [00398] In some embodiments, the plurality of RNA contig sequences may be derived from one infected subject. [00399] In some embodiments, the infected subject may be a human. [00400] In some embodiments, the plurality of virus-derived RNA contig sequences may be derived from the virus infecting the infected subject. [00401] In some embodiments, the virus may include a HTLV-1 virus. [00402] In some embodiments, the plurality of infected-subject endogenous RNA contig sequences may be derived from RNA endogenous to the infected subject. [00403] In some embodiments, the subject reference genome can include the human genome. [00404] Certain embodiments and implementations of the disclosed technology are described above with reference to systems and methods and/or computer program products according to example embodiments or implementations of the disclosed technology. It will be understood that method steps can be implemented by computer-executable program instructions. Likewise, some method steps need not be performed in the order presented, may be repeated, or may not necessarily need to be performed at all, according to some embodiments or implementations of the disclosed technology. [00405] These computer-executable program instructions may be loaded onto a general-purpose computer, a special-purpose computer, a processor, or other programmable data processing apparatus to produce a particular machine, such that the instructions that execute on the computer, processor, or other programmable data processing apparatus create means for implementing one or more functions associated with method steps. These computer program instructions may also be stored in a computer- readable memory that can direct a computer or other programmable data processing
Attorney Docket No.250298.000682 apparatus to function in a particular manner, such that the instructions stored in the computer-readable memory produce an article of manufacture including instruction means that implement one or more functions associated with method steps. [00406] Non-transitory computer-readable media may include, but is not limited to, random access memory (RAM), read-only memory (ROM), electronically erasable programmable ROM (EEPROM), flash memory or other memory technology, compact disc ROM (CD-ROM), digital versatile disks (DVD) or other optical storage, magnetic cassettes, magnetic tape, magnetic disk storage or other magnetic storage devices, or any other tangible, physical medium which can be used to store computer readable information. [00407] Embodiments or implementations of the disclosed technology may provide for a computer program product, including a computer-usable medium having a computer- readable program code or program instructions embodied therein, said computer-readable program code adapted to be executed to implement one or more functions related to example methods presented herein. Likewise, the computer program instructions may be loaded onto a computer or other programmable data processing apparatus to cause a series of operational elements or steps to be performed on the computer or other programmable apparatus to produce a computer-implemented process such that the instructions that execute on the computer or other programmable apparatus provide elements or steps for implementing the functions related to example methods presented herein. [00408] Accordingly, illustrations and descriptions of methods support combinations of means for performing the specified functions, combinations of elements or steps for performing the specified functions, and program instruction means for performing the specified functions. It will also be understood that at least some method steps, and combinations of method steps, can be implemented by special-purpose, hardware-based computer systems that perform the specified functions, elements or steps, or combinations of special-purpose hardware and computer instructions. [00409] Certain implementations of the disclosed technology can be utilized with customer devices that may include mobile computing devices. Those skilled in the art will recognize that there are several categories of mobile devices, generally known as portable computing devices that can run on batteries but are not usually classified as laptops. For
Attorney Docket No.250298.000682 example, mobile devices can include, but are not limited to portable computers, tablet PCs, internet tablets, PDAs, ultra-mobile PCs (UMPCs), wearable devices, and smart phones. [00410] Certain implementations of the disclosed technology can be utilized with medical equipment, medical devices, and/or associated peripherals. Non-limiting Examples of Methods and Systems [00411] In various embodiments, the present disclosure provides a method for identifying an immunogenic virus-derived peptide, the method comprising: a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus, wherein the plurality of RNA contig sequences comprises a plurality of virus-derived RNA contig sequences and a plurality of infected-subject endogenous RNA contig sequences; b) identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences; c) assembling a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; d) identifying a protein sequence based on the viral RNA sequence; and e) identifying the immunogenic virus-derived peptide based at least in part on the identified protein sequence. [00412] In some embodiments, the plurality of RNA contig sequences are derived from one infected subject. [00413] In some embodiments, the infected subject is a human. [00414] In some embodiments, the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject. [00415] In some embodiments, the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject. [00416] In some embodiments, the identifying the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences comprises: comparing at least a portion of contig sequences of the plurality of RNA contig sequences to a reference viral sequence; and
Attorney Docket No.250298.000682 identifying the plurality of virus-derived RNA contig sequences such that each contig sequence of the plurality of virus-derived RNA contig sequences comprises at least a portion that corresponds to the reference viral sequence. [00417] In some embodiments, each contig sequence of the plurality of virus- derived RNA contig is distinct from the plurality of infected-subject endogenous RNA contig sequences. [00418] In some embodiments, each contig sequence of the plurality of virus- derived RNA contig sequences lacks infected-subject endogenous RNA contig sequences. [00419] In some embodiments, the reference viral sequence comprises a reference genome. [00420] In some embodiments, the reference genome comprises a HTLV-1 virus genome. [00421] In some embodiments, the assembling the viral RNA sequence based on the plurality of virus-derived RNA contig sequences comprises: overlapping common sequence portions at ends of at least a portion of the plurality of virus-derived RNA contig sequences such that the at least a portion of the plurality of virus-derived RNA contig sequences overlap linearly to assemble the viral RNA sequence. [00422] In some embodiments, the identifying a protein sequence based on the viral RNA sequence such that the identified protein sequence includes a translation of the viral RNA sequence comprises: identifying the protein sequence without requiring a comparison to a database of viral proteins. [00423] In some embodiments, the identifying the protein sequence based on the viral RNA sequence such that the identified protein sequence includes a translation of the viral RNA sequence further comprises: identifying a plurality of protein sequences each based on the viral RNA sequence such that each of the plurality of protein sequences respectively include a translation of the viral RNA sequence, and identifying the protein sequence as a frequently occurring protein sequence within the plurality of protein sequences.
Attorney Docket No.250298.000682 [00424] In some embodiments, the protein sequence identified based on the viral RNA sequence is associated with a single infected subject. [00425] In some embodiments, the identifying the immunogenic virus-derived peptide based at least in part on the protein sequence comprises: identifying an MHC molecule associated with the single infected subject; identifying one or more peptides based at least in part on the protein sequence such that the one or more peptides each form a respective MHC-peptide complex with the MHC molecule; and identifying the immunogenic virus-derived peptide based on the one or more peptides. [00426] In various embodiments, the present disclosure provides a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device, the non-transitory computer-readable medium including instructions thereon, that when executed by the processor(s), cause the computational device to: a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences and a plurality of infected- subject endogenous RNA contig sequences, and wherein the infected subject is infected with a virus; b) identify the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences; c) assemble a viral RNA sequence based on the plurality of virus-derived RNA contig sequences; d) identify a protein sequence based on the viral RNA sequence; e) identify an immunogenic virus-derived peptide based at least in part on the protein sequence; and f) provide, as an output, the immunogenic virus-derived peptide. [00427] In some embodiments, the plurality of RNA contig sequences are derived from only one infected subject. [00428] In some embodiments, the infected subject comprises a human.
Attorney Docket No.250298.000682 [00429] In some embodiments, the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject. [00430] In some embodiments, the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject. [00431] In some embodiments, the instructions which cause the computational device to identify the plurality of virus-derived RNA contig sequences from within the plurality of RNA contig sequences further comprise instructions, that when executed by the processor(s), cause the computational device to: compare at least a portion of contig sequences of the plurality of RNA contig sequences to a reference viral sequence; and identify the plurality of virus-derived RNA contig sequences such that each contig sequence of the plurality of virus-derived RNA contig sequences comprises at least a portion that corresponds to the reference viral sequence. [00432] In some embodiments, each contig sequence of the plurality of virus- derived RNA contig is distinct from the plurality of infected-subject endogenous RNA contig sequences. [00433] In some embodiments, each contig sequence of the plurality of virus- derived RNA contig sequences lacks portions of infected-subject endogenous RNA contig sequences. [00434] In some embodiments, the reference viral sequences comprises a reference genome. [00435] In some embodiments, the reference genome comprises a HTLV-1 genome. [00436] In some embodiments, the instructions which cause the computational device to assemble the viral RNA sequence based on the plurality of virus-derived RNA contig sequences further comprise instructions, that when executed by the processor(s), cause the computational device to: overlap common sequence portions at ends of at least a portion of the plurality of virus-derived RNA contig sequences such that the at least a portion of the plurality of virus- derived RNA contig sequences overlap linearly to assemble the viral RNA sequence. [00437] In some embodiments, the instructions which cause the computational device to identify a protein sequence based on the viral RNA sequence such that the protein
Attorney Docket No.250298.000682 sequence includes a translation of the viral RNA sequence further comprise instructions, that when executed by the processor, cause the computational device to: identify the protein sequence without requiring a comparison to a database of viral proteins. [00438] In some embodiments, the protein sequence identified based on the viral RNA sequence is a novel protein. [00439] In some embodiments, the protein sequence identified based on the viral RNA sequence is associated with a single infected subject. [00440] In some embodiments, the instructions which cause the computational device to identify a protein sequence based on the viral RNA sequence such that the protein sequence includes a translation of the viral RNA sequence further comprise instructions, that when executed by the processor(s), cause the computational device to: identify a plurality of protein sequences each based on the viral RNA sequence such that each of the plurality of protein sequences respectively include a translation of the viral RNA sequence; and identify the protein sequence as a frequently occurring protein sequence within the plurality of protein sequences. [00441] In some embodiments, the instructions which cause the computational device to identify the immunogenic virus-derived peptide based at least in part on the protein sequence further comprise instructions, that when executed by the processor(s), cause the computational device to: identify a major histocompatibility complex (MHC) molecule associated with the single infected subject; identify one or more peptides based at least in part on the protein sequence such that the one or more peptides are each capable of forming a respective MHC-peptide complex with the MHC molecule; and identify the immunogenic virus-derived peptide based on the one or more peptides. [00442] In some embodiments, the instructions, when executed by the processor(s), further cause the computational device to: store the protein sequence to a database such that the protein sequence is associated with the infected subject within the database.
Attorney Docket No.250298.000682 [00443] In various embodiments, the present disclosure provides a method for identifying an integration site of a viral gene within a subject gene, the method comprising: a) obtaining a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences comprise a plurality of virus-derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and a plurality of hybrid RNA contig sequences comprising viral and infected-subject endogenous portions; b) identifying the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; c) comparing, for at least a portion of the plurality of hybrid RNA contig sequences, infected-subject endogenous portions to a subject reference genome; and d) identifying, based at least in part on the comparison of infected-subject endogenous portions to the subject reference genome, an integration site comprising the subject gene. [00444] In some embodiments, the plurality of RNA contig sequences are derived from one infected subject. [00445] In some embodiments, the infected subject is a human. [00446] In some embodiments, the plurality of virus-derived RNA contig sequences are derived from the virus infecting the infected subject. [00447] In some embodiments, the virus is a HTLV-1. [00448] In some embodiments, the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject. [00449] In some embodiments, the subject reference genome comprises the human genome. [00450] In various embodiments, the present disclosure provides a non-transitory computer-readable medium configured to communicate with one or more processor(s) of a computational device, the non-transitory computer-readable medium including instructions thereon, that when executed by the processor(s), cause the computational device to: a) receive, as an input, a plurality of RNA contig sequences derived from an infected subject infected with a virus such that the plurality of RNA contig sequences
Attorney Docket No.250298.000682 comprise a plurality of virus-derived RNA contig sequences, a plurality of infected-subject endogenous RNA contig sequences, and plurality of hybrid RNA contig sequences comprising viral and infected-subject endogenous portions; b) identify the plurality of hybrid RNA contig sequences from within the plurality of RNA contig sequences; c) compare, for at least a portion of the plurality of hybrid RNA contig sequences, infected-subject endogenous portions to a subject reference genome; d) identify, based at least in part on the comparison of infected-subject endogenous portions to the subject reference genome, an integration site comprising a subject gene; and e) provide, as an output, the integration site. [00451] In some embodiments, the plurality of RNA contig sequences are derived from only one infected subject. [00452] In some embodiments, the infected subject comprises a human. [00453] In some embodiments, the plurality of virus-derived RNA contig sequences are derived from a virus infecting the infected subject. [00454] In some embodiments, the virus comprises a HTLV-1. [00455] In some embodiments, the plurality of infected-subject endogenous RNA contig sequences are derived from RNA endogenous to the infected subject. [00456] In some embodiments, the subject reference genome comprises the human genome. EXAMPLES [00457] The present disclosure is also described and demonstrated by way of the following examples. However, the use of these and other examples anywhere in the specification is illustrative only and in no way limits the scope and meaning of the disclosure or of any exemplified term. Likewise, the disclosure is not limited to any particular preferred embodiments described here. Indeed, many modifications and variations of the disclosure may be apparent to those skilled in the art upon reading this specification, and such variations can be made without departing from the disclosure in spirit or in scope. The disclosure is therefore to be limited only by the terms of the appended claims along with the full scope of equivalents to which those claims are entitled.
Attorney Docket No.250298.000682 [00458] Human T-lymphotropic Virus, type 1 (HTLV-1) is the etiological agent for Adult T-cell Leukemia/Lymphoma (ATL), HTLV-1 associated myelopathy (HAM), HTLV-1-associated infective dermatitis (IDH) and other inflammatory disorders like uveitis and arthritis. While HTLV-1 is the most well-studied of the four HTLV genotypes (HTLV-1, HTLV-2, HTLV-3 and HTLV-4) the immunopeptidome of HTLV-1-infected T-cells and cell lines have not yet been characterized. The surface presentation of HTLV- 1-specific peptides by Human Leukocyte Antigens (HLA) could be leveraged for developing immunotherapies targeting infected lymphocytes in ATL. In the Examples described below, HLA-I immunopeptidomics were performed on four model HTLV- transformed cell lines: MT-4, C8166, MT-2 and C5/MJ; primary cells and ATL patient samples. Example 1. Immunopeptidomics for HTLV-transformed cells. [00459] HLA-I complexes were purified from the 4 HTLV-transformed cell lines MT-4, C8166, MT-2 and C5/MJ, using anti-HLA-I W6/32 affinity purification (Figure 1A). Briefly, 100 million cells for each cell line were lysed in 1% NP-40 lysis buffer under mild conditions, and lysates were incubated with the W6/32-conjugated sepharose beads in column format. The beads were washed followed by an HLA elution using an acidic glycine solution (pH 2.7). The HLA-associated peptides were then separated from HLA molecules by 30% Acetonitrile/H2O selective elution from a C18-based sep-pak. The peptides were separated on a 25 cm column, and analyzed by liquid chromatography with tandem mass spectrometry (LC-MS/MS) using a Thermo Orbitrap Fusion Lumos connected online to nano-LC 1200 (Thermo). The mass spectrometry raw files were searched against a combined FASTA database containing both human (Uniprot) and HTLV-1 (Uniprot) protein sequences. More than 13,000 peptides were detected from the MT-4 cell line (Figure 1D) and between 8,000 to 10,000 peptides were detected from the C8166, MT-2, and C5/MJ cell lines (Figures 1B-1C and Figure 1E), with nonamers being the most abundant peptide length. [00460] Furthermore, a number of unique HTLV peptides were also identified. For example, 19 HTLV-1 peptides were detected from MT-4 cells, and 16 of the peptides overlapped between the three biological replicates (Figure 4 and Table 5). Similarly, 11 and 32 HTLV-1 specific peptides from C8166 and MT-2 cells were detected, respectively,
Attorney Docket No.250298.000682 with high reproducibility between replicates (Figure 2 and Table 3; Figure 3 and Table 4). For the C5/ MJ cells, the HTLV-1 peptides presented by HLA, with and without Interferon- ^ stimulation, were compared. No significant increase in number of HTLV peptides was observed after Interferon (IFN)- ^ stimulation of C5/ MJ cells (Figure 5 and Table 6). To increase confidence in the disclosed peptide identifications, HLA genotyping of these samples was performed, and the panNetMHC 4.0 algorithm was used to predict the binding affinity of the identified HTLV-1 peptides with the HLA alleles of the respective cell lines. Most of the HTLV-1 peptides identified from the four cell lines were strong (panNetMHC 4.0 Rank below 0.5) or weak (panNetMHC 4.0 Rank between 0.5 – 2.0) predicted binders to the HLA of described cell lines (Tables 3-6). Table 3. HTLV-1 Peptides Detected from C8166 Cells HLA- HLA- HLA- HLA- HLA- A01:01 B08:01 B44:02 C05:01 C07:01 nk 72 69 80 5 5 7 n =
engt Table 4. HTLV-1 Peptides Detected from MT-2 Cells HLA- HLA- HLA- HLA- HLA-
Attorney Docket No.250298.000682 Peptide Le Protein nM Ran nM Ran nM Ran nM Ran nM Ran n k k k k k 72 0 .26 .23 2 6 3 .87 2 .71 .02 .34 9 2 0 0
Attorney Docket No.250298.000682 VSSPCHNSL 9 ENV_HTL1 1325 7.01 2499 24.012217 7.48 81 0.32 530 1.42 (SEQ ID NO: M 4 1 4 46 .40 2 0 n =
Table 5. HTLV-1 Peptides Detected from MT-4 Cells HLA- HLA- HLA- HLA- HLA- A02:01 A11:01 A24:02 B07:02 B15:01 n 1 .25 .75 .34 .14 5 .04 .49 .91 .50 .72 .00 .59 .45
Attorney Docket No.250298.000682 LPAPHLTLP 9 ENV_HTL1 3401 51.133821 60.744133 52.693154 2.47 3207 56.66 (SEQ ID NO: 53) M 6 4 8 7 55 .17 .80 n =
Table 6. HTLV-1 Peptides Detected from C5/MJ Cells ± IFN-γ Treatment HLA- HLA- HLA- HLA- HLA- A3201 B3503 B5101 C0401 C1502 n 6 1 .7 0 3 .4 .8 .0 .1
Attorney Docket No.250298.000682 EALQTGITL 9 ENV_HTL1 1553 12.0 703 0.11 3947 0.69 2568 8.87 4484 3.81 (SEQ ID NO: 50) M 3 8 3 .2 n =
Example 2. Immunopeptidomics for ATL primary cells. [00461] Next, the HLA-I immunopeptidome of patient-derived ATL primary sample ED41214(-) was analyzed using the same method as discussed in Example 1. A slight increase in total peptide count with interferon- ^ treatment (Figure 6A and Figure 6C) was observed. More importantly, 16 and 21 unique HTLV-1 peptides from untreated and interferon- ^ treated ED41214(-) cells, respectively, were identified (Figure 6B, Figure 6D and Figure 6E). Only a few HTLV peptides were predicted binders to the common HLA alleles in the patient population (see, e.g., Table 7). Table 7. HTLV-1 Peptides Detected from C5/MJ Cells ± IFN-γ Treatment HLA- HLA- HLA- HLA- HLA- HLA-B54 A02:01 A11:01 A24:02 B07:02 B15:01 an 5.6 8.3 01 6.2 8.9 01
Attorney Docket No.250298.000682 SASPIPRPPR Gag 8-1710 337650.3706 2.11414053 286825.2276744.7320926.9 (SEQ ID NO: polyprot 1 9 2 0 1 7 6 2 5 i 20 7.4 01 4.2 0.6 00 06 2.9 5.8 26 59 06 4.9
Attorney Docket No.250298.000682 KNTSVLGAG Gag-Pro- 501- 11 431383.6349747.4468089.2447191.1366871.1470195.6 GQ (SEQ ID Pol 511 8 6 5 7 7 3 3 6 5 5 0 4 22 l 05 n =
[00462] Peptide distributions were obtained for all HTLV primary samples tested; however, HTLV peptides were detected only for ED-41214(-) (combined search against human UniProt + HTLV sequence) (Figure 6A and Figures 7A-7D). Example 3. Validation of HTLV peptides with heavy analogues in cell lines and calculation of copy number. [00463] A few HTLV peptides are also selected for validation using the synthetic heavy analogues. Briefly, synthetic peptides containing heavy Leucine (13C(6)15N(1)) (Thermo) are mixed with the HLA-eluted peptides from HTLV-transformed cell lines and are analyzed by LC-MS/MS. The co-elution and similarity in fragmentation profile of endogenous and heavy peptides are used to confirm the endogenous peptide identity. Example 4. Identification of HTLV-specific peptides from HLA-I immunopeptidomics of ATL patient samples. [00464] ATL (adult T-cell leukemia) patient samples
procured. Peripheral blood mononuclear cells (PBMCs) were suspended in lysis buffer containing 1% NP-40 and incubated at 4°C for 1 hour. HLA class I complexes were then purified using a pan class I anti-HLA W6/32 antibody covalently conjugated to sepharose beads, followed by subsequent washing of the beads and elution of the HLA complexes in acidic solution (pH 2.7). A C18-based solid-phase extraction was utilized to separate the HLA proteins from the associated peptides, which were then further analyzed by liquid chromatography-mass spectrometry (LC-MS). Out of 23 patient samples, 5 samples with cell counts in the range of 50-100 million cells were analyzed by a Thermo Orbitrap mass spectrometer, while the remaining 18 samples (10-20 million cells) were analyzed by a Bruker timsTOF SCP mass spectrometer. PEAKS online (Bioinformatics Solutions Inc.) was used to identify peptide sequences from the mass spectrometer raw files, by searching the raw spectrum against human Uniprot protein sequences. For identification of HLA-I-restricted HTLV-1
Attorney Docket No.250298.000682 peptides, FASTA-containing sequences derived from HTLV RNA reads from the patient samples were used in combination with human Uniprot. Patient DNAseq and RNAseq data were also used to identify HLA genotyping of individual patient samples. [00465] Analysis of all ATL patient samples revealed an abundance of nonamers representing peptides presented by HLA class I (Figures 8A-8D). While 800-2,300 nonamers were isolated from ATL1-5 with the Thermo Orbitrap mass spectrometer (Figures 8A-8B), much higher 9-mer peptide counts were recovered from the HLA-I peptidome analysis of ATL6-23 using the Bruker timsTOF SCP mass spectrometer (Figures 8C-8D). HLA genotyping of ATL patient samples highlighted the diversity of HLA-I in the patient population with a high proportion of patients with HLA-A02 (12 patient samples), HLA-A11 (6 patient samples) and HLA-A24 (9 patient samples) alleles (Table 8). Table 8. HLA Genotyping of ATL Patient Samples Lab ID HLA-A HLA-B HLA-C ATL1C *24:02 *26:03 *51:01 *54:01 *01:02 *14:02
Attorney Docket No.250298.000682 [00466] Using analysis from the Thermo Orbitrap mass spectrometer, peptide LPAPHLTLP (SEQ ID NO: 53) originating from the HTLV envelope protein and a predicted HLA-B51 and HLA-B54 binder, was identified from the ATL1C sample, and an 8-mer originating from the HTLV polymerase protein, ALPELQAL (SEQ ID NO: 72), was identified from the ATL1C and ATL2C samples (Table 9). However, the two unique HTLV peptides detected at the highest frequencies in the ATL patient samples were QSSSFIFHK (SEQ ID NO: 143) and EYTNIPISLL (SEQ ID NO: 144), as determined using analysis from the Bruker timsTOF SCP mass spectrometer (Table 10). Both peptides originated from the HTLV Tax protein and were detected in 2/6 HLA-A11 (33%) and 3/6 HLA-A24 (50%) of patient samples respectively. The HTLV peptides identified from the individual patient samples were matched with their respective HLA alleles using the panNetMHC 4.0 binding prediction algorithm to further enhance the confidence in peptide identifications.
Attorney Docket No.250298.000682 Table 9. HTLV Peptides Detected From ATL1-5 Samples Using Thermo Orbitrap Mass Spectrometer Sample Peptide Len Ion Uniprot ID % Rank_EL (aa sequence) Score (netMHC) 1) - -
er timsTOF SCP Mass Spectrometer Uniprot Peptide Len Ion Samples Sample Frequency %Rank_EL IDs (aa sequence) Scores Count by (netMHC) - - - -
Example 5. Generation of a Viral Sequence Database from HTLV-infected Patient Samples. [00467] An RNAseq-based approach was employed to generate a database of patient-specific HTLV genome sequences directly reconstituted from transcribed virus RNA sequences amplified from patient samples (Figure 9). Total human RNA reads were first converted into large contigs using a de novo sequencing approach, and contigs with
Attorney Docket No.250298.000682 loose homology to the HTLV reference genome (NCBI Accession No. NC_001436.1) were marked as HTLV-specific sequences. Fully-mapped contigs to the HTLV reference genome were re-arranged in a reference-free fashion to assemble patient-specific HTLV genomic sequences. Using this approach, seven patient-specific HTLV genome sequences were reconstructed. Genome reconstruction was partial, ranging from 13% to 42% of the HTLV reference genome. The patient-specific viral sequence was then used to extract and translate all HTLV coding sequences. Non-canonical protein sequences with start codons embedded within canonical protein sequences were also identified in each sequence. The RNA sequence read coverage of the patient-specific reconstructed genomes is depicted in Figure 10. [00468] A summary of the RNAseq data of the present Example, including patient- specific reconstructed genome length and percentage (%) of the HTLV reference genome, is provided in Table 11. Table 11. Summary of RNAseq data Reconstructed Lab ID Genome Reconstructed RNA Sequencing
[00469] Total RNA was extracted from human tissue using a MagMAX kit (ThermoFisher). Strand-specific RNA-seq libraries were prepared from 1 µg RNA using a KAPA stranded mRNA-Seq Kit (KAPA Biosystems). Twelve-cycle PCR was performed to amplify libraries. The amplified libraries were size-selected at 400-600 bp (base pairs) using PippinHT (Sage Science). Sequencing was performed on Illumina HiSeq®2500 (Illumina) by multiplexed paired-read run with 2X100 cycles.
Attorney Docket No.250298.000682 Mapping of Patient RNA Sequences to HTLV Reference Genome [00470] Bulk RNA-seq reads were aligned to the HTLV reference genome (NCBI Accession No. NC_001436.1) using minimap2 (v2.17) (see, e.g., Li 2018). Alignments were then sorted by coordinates and quality controlled by samtools flagstats (v1.9) (see, e.g., Li et al., 2009) and bedtools genomeCoverageBed (v2.17.0) (see, e.g., Quinlan et al., 2010) inspection. Duplicates were then marked and removed by the Picard toolkit (v2.18.2) (github.com/broadinstitute/picard). Workflow to Reconstruct HTLV Genomes from Patient RNAseq Data [00471] Paired-end Illumina RNA reads from each sample were de novo assembled into large contigs using megahit (see, e.g., Li et al., 2015) (options: --min-count 3 --k-min 27 --k-max 127 --prune-level 2) and mapped using BLAST to the HTLV reference genome (NCBI Accession No. NC_001436.1) to select HTLV-specific sequences. BLAST parameters for sequence comparisons included outfmt ‘7 std sgi stitle’; minimum E value = 0.001; cost to open a gap = 5; cost to extend a gap = 2; length of best perfect match = 11; reward for a nucleotide match = 2; reward for a nucleotide mis-match = -3. Contigs without BLAST matches were discarded, as well as BLAST results with E values greater than 0.001, percentage identity below 79%, or alignment length of less than 50 nucleotides. Overlapping contigs were merged using custom scripts and final sequences that covered the entire length or partial length of the reference genome sequence with the highest identity were selected. The final sequence was mapped against the reference genome to extract and translate all coding sequences. Non-redundant protein sequences were then added to customized databases for peptide identification by mass-spectrometry. References 1. Li, H., Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics, 2018.34(18): p.3094-3100. 2. Heng Li, B.H., Alec Wysoker, Tim Fennell, Jue Ruan, Nils Homer, Gabor Marth, Goncalo Abecasis, Richard Durbin, 1000 Genome Project Data Processing Subgroup, The Sequence Alignment/Map format and SAMtools. Bioinformatics, 2009. 25(16): p. 2078- 2079. 3. Aaron R Quinlan, I.M.H., BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics, 2010.26(6): p.841-842.
Attorney Docket No.250298.000682 4. Dinghua Li, C.-M.L., Ruibang Luo, Kunihiko Sadakane, Tak-Wah Lam, MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics, 2015.31(10): p.1674-1676. Example 6. Identification of HTLV-1 antigens or epitopes. [00472] The present Example relates to nucleic acid assay methods and compositions for identifying HTLV-1 antigens or epitopes. [00473] RNA or DNA is sequenced from a cancer patient’s tumor cell(s) and/or healthy tissue(s)/cell(s) to identify sequences which may contain HTLV-1-associated insertions in genes expressed in the tumor cell. [00474] Tumor material is assessed for HTLV-1-associated insertions by sequencing such as by RNA sequencing. Sequencing data are used to investigate HTLV- 1-associated insertions expressed in genes. Peptide stretches comprising any of the identified HTLV-1-associated insertions are created in silico and are filtered through the application of prediction algorithms or used to identify MHC-associated epitopes via mass spectrometry data. Where appropriate, suitable MHC-binding HTLV-1 epitope sets are used to identify physiologically relevant HTLV-1 epitope-specific T cell responses via functional assays and/or MHC multimer-based screens within both or either of CD8+ and/or CD4+ T cell populations. [00475] HTLV-1 epitopes are determined by sequencing the genome and/or exome of tumor tissue and/or healthy tissue from a cancer patient using next generation sequencing (NGS) approaches. Genes selected based on the presence of HTLV-1- associated insertions and ability to act as an antigen are sequenced using NGS technology. NGS applies to, without limitation, genome sequencing, genome resequencing, epigenome characterization, DNA-protein interactions (ChIP-sequencing), and transcriptome profiling (RNA-Seq). Similar to DNA-based assays using, e.g., NGS or massively parallel sequencing (MPS) approaches, RNA from tumors is analyzed by conversion to cDNA and generation of a library suitable for sequencing. [00476] Assays are employed to identify HTLV-1 epitopes in biological samples. Suitable assays used to identify HTLV-1 epitopes in biological samples include, but are not limited to, proteomics, NGS, solution hybridization, array hybridization nucleic amplification, polymerase chain reaction (PCR), RT-PCR, quantitative PCR, branched
Attorney Docket No.250298.000682 DNA (bDNA) assay, rolling circle amplification (RCA), in situ hybridization, Northern hybridization, hybridization protection assay (HPA), single molecule hybridization detection, Invader assay, and/or Oligo Ligation Assay (OLA), hybridization, and array analysis. [00477] The sequencing data derived from determining the presence of HTLV-1 epitopes in a cancer patient is analyzed to predict personal HTLV-1 peptides that can bind to HLA molecules of the individual. The data are analyzed using a computer. The sequence data are analyzed, in particular, for the presence of HTLV-1 antigens. [00478] HTLV-1 antigens are assessed by their affinity to MHC molecules. [00479] Neural network-based learning approaches with validated binding and non- binding peptides are used in prediction algorithms for the major HLA-A and -B alleles. Algorithms are used for predicting missense mutations that create strong binding peptides to a cancer patient’s cognate MHC molecules. A set of peptides representative of optimal HTLV-1 epitopes for each patient is identified and prioritized. HTLV-1 epitope prediction algorithms are used to predict binding of candidate peptides to MHC class I molecules or MHC class II molecules. [00480] In some cases, a peptide binding tool can be one of the following: Antibody Epitope Prediction, ANTIGENIC, BepiPred, CTLPred, DiscoTope, EPIPREDICT, Epitope Cluster Analysis, Epitope Conservancy Analysis, EUiPro, HLA Peptide Binding Predictions, HLABinding, MAPPP, MHCBench, MHC-I processing predictions, Mosaic Vaccine Tool Suite, NetChop, NetCTL, NetMHC, NetMHCII, NetMHCpan, nHLAPred- I, OptiTope, PAProC, POPI, PREDEP, Prediction of Antigenic Determinants, ProPred, ProPred-1, RankPep, SMM, SVMHC, TAPPred, VaxiJen, or combinations thereof. Additional exemplary programs are used such BIMAS or SYFPEITHI, Rankpep. [00481] In some cases, the Immune Epitope Database and Analysis Resource (IEDB) (Vita R, et al. Nucleic Acids Res.2015; 43 (Database issue):D405-D412) is used to identify a suitable tumor HTLV-1 antigen. Such algorithms predict peptide binding to different MHC class I variants based on artificial neural networks (ANN), providing predicted IC50 as an output. NetMHC (Lundegaard C, et al. Nucleic Acids Res. 2008;36 (Web Server issue):W509-W512.) is used. Programs such as SMMPMBEC (Kim Y, et al. BMC Bio informatics.2009; 10:394) and/or SMM (Peters B, et al. BMC Bio informatics.
Attorney Docket No.250298.000682 2005; 6: 132) are used. These programs use position-weight matrices to describe statistical preferences from peptide-MHC I binding data. This approach suppresses noise caused, for example, by a limited number of data points present in the training set and/or experimental error. [00482] In some instances, single nucleotide polymorphisms (SNPs) are removed from candidate HTLV-1 antigens or HTLV-1 epitopes. SNPs contain a range of molecular variation: (1) SNPs, (2) multinucleotide polymorphisms (MNPs), (3) short deletion and insertion polymorphisms (indels/DIPs), (4) micro satellite markers or short tandem repeats (STRs), (5) heterozygous sequences, and (6) named variants. [00483] Proteomic-based methods for identifying tumor specific HTLV-1 antigens such as direct protein sequencing are used. Protein sequencing of enzymatic digests using multidimensional MS techniques including tandem mass spectrometry (MS/MS) is used to identify HTLV-1 antigens. High-throughput methods for de novo sequencing of unknown proteins is used, for example, to analyze the proteome of a cancer patient's tumor to identify expressed HTLV-1 antigens. In some instances, meta-shotgun protein sequencing is used to identify expressed HTLV-1 antigens. [00484] Tumor specific HTLV-1 antigens are identified using MHC multimers to identify HTLV-1 antigen-specific T-cell responses. For example, high-throughput analysis of HTLV-1 antigen- specific T-cell responses in cancer patient samples may be performed using MHC tetramer-based screening approaches. Such tetramer-based screening approaches are used for the identification of tumor specific HTLV-1 antigens, or as a secondary screening protocol to assess HTLV-1 antigens to which a patient may have already been exposed, which may support the selection of candidate HTLV-1 antigens. Where appropriate, filters are applied to eliminate (1) epitopes with lower binding affinity than the corresponding wild-type sequences and/or (2) epitopes predicted to be poorly processed by the immunoproteasome. Candidate mutated peptides are synthesized and screened to identify T cell HTLV-1 antigens. [00485] Pulsing antigen presenting cells (APCs) with relatively long synthetic peptides that encompass minimal T cell epitopes is used to identify HTLV-1 epitopes. Nonsynonymous mutated epitopes are identified in tumors by evaluating the response of CD4+ tumor infiltrating lymphocytes (TILs) to autologous B cells that are pulsed with
Attorney Docket No.250298.000682 peptides encompassing individual mutations. Use of this approach results in the identification of mutated cell epitopes. A peptide screening assay is performed based on the combination of two peptide libraries: (1) overlapping long-peptides (2) peptides according to MHC-binding prediction. Screening leads to identification of mutated HTLV- 1-reactive T cells. [00486] A tandem minigene screening approach is used to identify HTLV-1 epitopes. A tandem minigene construct comprised, for example, without limitation 6 to 24 minigenes that encoded polypeptides comprising a mutated amino acid residue flanked on the N- and/or C-terminus by, e.g., 12 amino acids. Tandem minigene constructs are synthesized and used to transfect autologous APCs and/or cell lines co-expressing autologous HLA molecules. Using this approach, HTLV-1 epitopes are identified in cancer patients, e.g., patients with adult T-cell leukemia/lymphoma (ATL) or HTLV-1- myelopathy/tropical spastic paraparesis (HAM/TSP). [00487] HTLV-1 epitopes are identified using an approach combining whole- exome/transcriptome sequencing analysis, MHC binding prediction, and mass spectrometric technique to detect peptides eluted from HLA molecules. Predicted high- binding peptides are confirmed by mass spectrometry. Example 7. Determination of MHC binding capacity. [00488] Candidate peptides according to the present disclosure are tested for their MHC binding capacity (affinity). The individual peptide-MHC (pMHC) complexes are produced by UV-ligand exchange. A UV-sensitive peptide is cleaved upon UV-irradiation, and exchanged with the peptide of interest. Peptide candidates that effectively bind and stabilize the peptide-receptive MHC molecules prevent dissociation of the MHC complexes. To determine the yield of the exchange reaction, an ELISA is performed based on the detection of the light chain (β2m) of stabilized MHC complexes. Briefly, 96-well plates are coated with streptavidin, washed, and blocked. Refolded HLA-A monomers serve as standards, covering a pre-determined concentration range. Peptide-MHC monomers of the UV-exchange reaction are diluted in blocking buffer. Samples are incubated, washed, incubated with HRP conjugated anti- β2m, washed again and detected with a chromogenic substrate solution that is stopped per the manufacturer’s protocol. Absorption is measured, for example, at 450 nm. Candidate peptides that show a high
Attorney Docket No.250298.000682 exchange yield are preferred for generation and production of antibodies or fragments thereof, and/or T cell receptors or fragments thereof. Candidate peptides demonstrate avidity to the MHC molecules and prevent dissociation of the MHC complexes. Example 8. Preparation of peptide-MHC (pMHC) complexes. [00489] This example relates to a method for the preparation of soluble recombinant HLA loaded with an HTLV-1-derived peptide. [00490] Class I HLA molecules (HLA-heavy chain and HLA light-chain (β2m)) are expressed separately in E. coli as inclusion bodies using suitable expression vectors. HLA- heavy chain additionally comprises a C-terminal biotinylation tag which replaces, for example, the transmembrane and/or cytoplasmic domains. E. coli cells are lysed and inclusion bodies processed to approximately 80% purity. [00491] Inclusion bodies of β2m and heavy chain are denatured separately in denaturation buffer. Refolding buffer is prepared. Synthetic peptides are dissolved to a final concentration and added to the refold buffer. Then β2m followed by heavy chain are added. Refolding is performed to completion. [00492] The refold mixture is then dialyzed. The protein solution is subsequently filtered through a filter and loaded onto an exchange column (pre-equilibrated). Protein is eluted such as by way of a linear salt gradient using additional purifier. HLA-peptide complex is eluted, and peak fractions are collected. A cocktail of protease inhibitors is added and the fractions are chilled on ice. [00493] Biotin-tagged pHLA molecules are buffer exchanged into a buffer using a fast desalting column equilibrated in the same buffer. Upon elution, the protein-containing fractions are chilled on ice and protease inhibitor cocktail is added. Biotinylation reagents are then added. The mixture is then allowed to incubate. [00494] The biotinylated pHLA molecules are further purified, for example, by gel filtration chromatography using purifier with a column pre-equilibrated with filtered PBS. The biotinylated pHLA mixture is concentrated to a final volume, loaded onto the column and developed. Biotinylated pHLA molecules elute, e.g., as a single peak. Fractions containing protein are pooled, chilled on ice, and protease inhibitor cocktail is added. Protein concentration is determined and aliquots of biotinylated pHLA molecules are stored frozen.
Attorney Docket No.250298.000682 [00495] Such peptide-MHC (pMHC) complexes are used in soluble form or immobilized through their C-terminal biotin moiety on to a solid support, to be used for the detection of T cells and T cell receptors which bind the peptide-MHC complex. For example, such complexes are used in panning phage libraries, performing ELISA assays and/or preparing sensor chips for measurements of affinity and binding kinetics. Example 9. Identification of T cell receptors (TCRs) that bind to pMHC complexes. [00496] Antigen binding T cell receptors (TCRs) are obtained using peptides disclosed herein to pan a TCR phage library. The library is constructed using α- and β- chain sequences obtained from a natural repertoire. The random combination of these α- and β- chain sequences occurs during library creation, thereby producing a non-natural repertoire of α/β chain combinations. [00497] TCRs obtained from the library are assessed by enzyme-linked immunoassay (ELISA) to confirm specific antigen recognition. ELISA plates are coated with streptavidin and incubated with the biotinylated peptide-HLA complex. TCR-bearing phage clones are added to each well and detection is carried out using an HRP antibody conjugate. Bound antibody is detected using a peroxidase Substrate System. An absence of binding to alternative peptide-HLA complexes indicated that the TCR is not highly cross reactive. [00498] Further confirmation that TCRs can bind a peptide-HLA complex of the disclosure is obtained by surface plasmon resonance (SPR) using isolated TCRs. In this case α- and β- chain sequences are expressed in E. coli as soluble TCRs. Binding of the soluble TCRs to the complexes is analyzed by surface plasmon resonance. Biotinylated peptide-HLA monomers are prepared and immobilized on a streptavidin-coupled sensor chip. To measure affinity, serial dilutions of the soluble TCRs are flowed over the immobilized peptide-HLAs and the response values at equilibrium are determined for each concentration. Data are analyzed, for example, by plotting the specific equilibrium binding against protein concentration followed by a least squares fit to the Langmuir binding equation, assuming a 1:1 interaction. [00499] TCRs that specifically recognize peptide-HLA complexes of the disclosure are obtained from the library. Data generated according to the above-described experiments confirm that antigen specific TCRs can be isolated.
Attorney Docket No.250298.000682 Example 10. Characterization of binding to MHC and stability of pMHC complex. [00500] T2 cell-based peptide binding assay. T2 cells which do not express the transporter associated with antigen processing (TAP), and as such do not assemble stable MHC class I on the cell surface, are pulsed with different concentrations of peptides (controls or HTLV-1 peptide of interest [POI]), washed, detected with fluorescently-tagged antibody recognizing MHC class I (e.g., A2 allele), and run through a FACS Scan analyzer. The difference between the MFI (mean fluorescence intensity) corresponding to a given concentration of POI and the negative control (non-MHC binder) is a function of the number of stabilized pMHC complexes displayed on the cell surface. Therefore, at limiting concentrations of the peptide, it is largely a measurement of Kon, and at saturation levels of the peptide it is a measurement of both Kon and Koff. The binding is quantified by two related factors: relative affinity (1/RA) and half maximal binding (the peptide concentration responsible for 50% of the signal corresponding to saturation). Relative affinity, RA, is binding normalized to a reference (e.g., a wild-type peptide in instances where a mutant POI is being tested), e.g., the ratio between half maximal binding of control relative to POI. The higher the 1/RA index and the lower the half maximal binding, the higher the Kon of the interaction between the POI and the MHC. [00501] Characterization of binding and stability by ELISA. Avidin-coated microtiter plates containing class I monomer loaded with a placeholder peptide are used to evaluate peptide binding, affinity, and off-rate. The monomer-coated plates are supplied as part of a kit, e.g., the iTopia Epitope Discovery System Kit. Assay buffers, anti-MHC- FITC mAb and β2-microglobulin and control peptides are also supplied with the kits. [00502] Binding assay: POIs are first evaluated for their ability to bind each MHC molecule by binding assay. This assay measures the ability of individual peptides to bind HLA molecules under optimal standardized binding conditions. Monomer-coated plates are first stripped, releasing the placeholder peptide and leaving only the MHC heavy chain bound to the plate. Test peptides are then introduced under optimal folding conditions, along with the anti-MHC-FITC mAb. Plates were incubated. The anti-MHC-FITC mAb binds preferentially to a refolded MHC complex. Thus, the fluorescence intensity resultant from each peptide is related to the peptide's capacity to complex with MHC molecule. Each
Attorney Docket No.250298.000682 peptide's binding is evaluated relative to a positive control peptide, and the results are expressed, for example, as percent (%) binding. [00503] Affinity assay: For the affinity assay, after the initial stripping of the placeholder peptide, increasing concentrations of POI are added to a series of wells and incubated under the conditions described previously. Plates are read on the fluorometer. Dose response curves are generated. The amount of peptide required to achieve 50% of the maximum is recorded as ED50 value. [00504] Off-Rate assay: Plates are washed after incubation under conditions to remove excess peptide. The plates are then incubated on allele-specific monomer plates. The plates are measured at multiple time points (e.g., 0, 0.5, 1, 1.5, 2, 4, 6 and 8 hrs) for relative fluorescence intensity. The time required for 50% of the peptide to dissociate from the MHC monomer is defined as the T½ value (hrs). [00505] iScore calculation: an iScore is a multi-parameter calculation provided within the iTopia software. Its value is calculated based on the binding, affinity, and stability data. Example 11. Determination of responses against tumor cells. [00506] A suitable number of groups of mice are immunized with a plasmid expressing HTLV-1 peptides disclosed herein by direct inoculation. By way of a non- limiting example, mice are inoculated into lymph nodes, e.g., inguinal lymph nodes, with plasmids at an appropriate concentration at day 0, and at subsequent days over the time course of the experiment, e.g., at days 3, 14, and 17. In certain cases, this is followed by one or more additional peptide boost(s) on following days, e.g., days 28 and 31, using a negative control peptide and POI. Splenocytes are stimulated ex vivo with POI and tested against Chromium-51 (51Cr)--labeled tumor cells at various E:T ratios. Example 12. In vivo assessment of enhanced immunity against HTLV-1 peptides. [00507] A suitable number of groups of mice are immunized with a plasmid expressing HTLV-1 peptides disclosed herein by direct inoculation. By way of a non- limiting example, mice are inoculated into lymph nodes, e.g., inguinal lymph nodes, with plasmids at an appropriate concentration at day 0, and at subsequent days over the time course of the experiment, e.g., at days 3, 14, and 17. In certain cases, this is followed by
Attorney Docket No.250298.000682 one or more additional peptide boost(s) on following days, e.g., days 28 and 31, using a negative control peptide and POI. [00508] To evaluate the in vivo response against HTLV-1 peptides, splenocytes were isolated from littermate control mice and incubated with one or more appropriate concentrations of POI for a pre-determined period of time. These cells were then stained with CFSEhi fluorescence and intravenously co-injected into immunized mice with an equal number of control splenocytes stained with CFSElo fluorescence. After a pre- determined period of time, the specific elimination of target cells was measured by removing spleen and PBMCs from challenged animals and measuring CFSE fluorescence by flow cytometry. The relative depletion of the populations corresponding to peptide loaded splenocytes was calculated relative to the control (unloaded) population and expressed as percent (%) specific lysis. Example 13. Testing of POI’s increased immunogenicity and ability to overcome tolerization [00509] POIs are used in in vitro for immunization of blood to generate cytotoxic T lymphocytes (CTLs). [00510] PBMCs from normal donors are purified from buffy coats by centrifugation in standard sterile medium designed for isolating lymphocytes. Cultures are carried out using autologous plasma (AP). For in vitro generation of peptide-specific CTL, autologous dendritic cells (DCs) are used as antigen presenting cells (APCs). DCs are generated and CTLs are induced with DCs and peptides from PBMCs. Monocyte-enriched cell fractions are cultured to induce maturation. Specific numbers of CD8+-enriched T lymphocytes and peptide-pulsed DCs are co-cultured. Cultures are restimulated on various days with autologous irradiated peptide-pulsed DCs. Immunogenicity is assayed using in vitro cytotoxicity and cytokine production assays. Example 14. CD8+ T cell responses against peptides. [00511] Based on the predicted or experimentally verified HLA-binding peptides, whether T cells can be generated to recognize the tumor-specific peptides is determined. Peptides with appropriate binding scores are synthesized. To generate T cells of desired specificity, T cells are stimulated with peptide-pulsed (individual peptide or peptide pool) autologous APCs such as dendritic cells and/or CD40L-expanded autologous B cells on
Attorney Docket No.250298.000682 a predetermined schedule, for example, in the presence of IL-2 and IL-7. After a number of rounds of stimulation, the expanded CD8+ cells are tested on ELISpot for evidence of reactivity against the peptide based on IFNγ secretion. Example 15. Cytokine production assay. [00512] For cytokine (e.g., IL-2 and IFN ^) production assays, T cells are harvested after contact with the peptide-pulsed APC, centrifuged, and both cell pellets and supernatants are collected. Cytokine production is measured by ELISA from the supernatant. Example 16. In vivo activation of viral peptide-specific T cells as a method to limit viral production. [00513] For in vivo studies, the ability of the viral peptide in the context of MHC I molecules (e.g., HLA-A2, HLA-A24, HLA-A11) to enhance a CD8+ T cell response as a method to limit viral production following AAV-HTLV-1 challenge is tested in transgenic mice that express human HLA molecule(s): [00514] (i) MHC class I molecule(s) displaying the HTLV-1 viral peptide of interest (POI) or (ii) a control MHC class I molecule displaying the OVA257-264 peptide (OVA) are generated. [00515] A cohort of transgenic mice that express human HLA molecule(s) is initially injected with the MHC I molecule/peptide or MHC I molecule/OVA complexes and the level of effector and memory CD8+ T cells is monitored at different time points post- injection (from spleen and/or blood). Mock injected mice will serve as a negative control group in this experiment. A second cohort of mice are then injected with the MHC I molecule/POI complex and subsequently challenged using AAV-HTLV-1, at post- injection time points that will be determined according to the measurements of effector and memory CD8 + T cells levels from the first experiment. Control groups are constituted with (i) mice not injected with the MHC I molecule/POI complex but challenged with AAV-HTLV-1, (ii) mice injected with MHC I molecule/OVA complexes, and (iii) mice injected with MHC I molecule/POI complex but not challenged with AAV-HTLV-1. The viral burden is analyzed at different time points post-injection to determine MHC I molecule/POI limitation of viral infection from CD8+ T cells activation.
Attorney Docket No.250298.000682 Example 17. In vivo activation of antigen-specific T cells by the viral peptide as a method to decrease viral production. [00516] For in vivo studies, the ability of the HTLV-1 viral peptide of interest (POI) in the context of MHC I molecules (e.g., HLA-A2, HLA-A24, HLA-A11) to enhance a T cell response against HTLV-1 virus following AAV-HTLV-1 challenge is tested in transgenic mice that express human HLA molecule(s). More particularly, the in vivo activation of peptide-specific CD8+ T cells after (i) POI immunization, (ii) acute challenge with AAV-HTLV-1, and/or (iii) chronic challenge with AAV-HTLV-1, is compared. [00517] (i) MHC class I molecule(s) displaying the HTLV-1 viral POI or (ii) a control MHC class I molecule displaying the OVA257-262 peptide (OVA) are generated. [00518] Transgenic mice that express human HLA molecule(s) are first challenged with AAV-HTLV-1. The MHC class I molecule/peptide or MHC class I molecule/OVA complexes produced for this example are then injected 1 day and 7 days after infection with the acute challenge, and 1 day, 7 days, and 14 days after the chronic challenge. The viral burden is analyzed at different time points post-peptide injection by blood collection and post-mortem organs collection. At those time points, the levels of effector and memory CD8+ T cells are measured and compared to the control conditions to determine the capacity of MHC class I molecule/peptide to decrease viral production in response to an AAV-HTLV-1 challenge. Example 18. Enhancement of a peptide-specific CD8+ T cell immunity in a peptide- presenting tumor model. [00519] For in vivo studies, the ability of the HTLV-1 peptide of interest in the context of MHC I molecules to enhance a CD8+ T cell response against peptide-presenting tumors is tested in a general purpose strain mice, e.g., C57B1/6 mice, grafted with a peptide-expressing tumor cell line. [00520] The peptide-expressing tumor cell line is first grown in vitro, then injected (e.g., subcutaneously) into the mice. After tumor cell injection, when tumors are of palpable size, the HTLV-1 peptide, for example, in a specific groove of the MHC I or control peptide (e.g., ovalbumin [OVA] peptide) in an alternate groove of the MHC I molecule, are injected intratumorally. Tumor cell volume is measured. Tumor, blood, and/or spleen is collected, and homogenized in single cell suspensions, when applicable.
Attorney Docket No.250298.000682 The level of CD8+ T cells in the homogenized samples is determined at all suitable time points. [00521] Below are additional example methods that may be used in accordance with the disclosure. [00522] Splenocytes and antigen presenting cell primary cultures. Spleens of adult general purpose strain mice, e.g., C57Bl/6 mice, are excised and placed in cold buffer. Tissues are then homogenized to break apart the spleens. Dissociated cells are centrifuged. After centrifugation, the cell pellet is resuspended in a suitable volume of lysis buffer designed to remove red blood cells, e.g., ACK (Ammonium-Chloride-Potassium) lysis buffer, and incubated in the buffer. After incubation the cell suspension is added to buffer, and centrifuged. The cell pellet is then resuspended in buffer and the cell solution is filtered. The filtered cell suspension is centrifuged again, and the subsequent pellet is resuspended in a suitable volume of buffer. Total spleen cells are counted, and global antigen-presenting cells (APC) are sorted using anti-MHC class II Microbeads and appropriate sorting technology. After elution, the MHC class II-positive cell fraction is collected and resuspended at a given concentration in APC cell medium. [00523] Pulsing of APCs with peptides. APC are incubated with HTLV-1 or control peptides. Peptide-pulsed APC are harvested and washed prior to addition to any of various suitable T cell lines (e.g., J.RT3-T3.5 derived cells), after infection with the viral particles. The T cells are co-cultured with the pulsed APC and subsequently used in proliferation, luciferase and/or cytokines production assays. [00524] T cell activation post transduction. Alternatively, to co-culture with peptides-pulsed APCs, suitable T cell lines (e.g., J.RT3-T3.5-derived cell lines) are activated after transduction with either phytohemagglutinin (PHA,), phorbol 12-myristate 13-acetate (PMA), or a combination of PHA and PMA. [00525] Another alternative method to activate T cells after transduction is to use soluble or immobilized antibodies such as, but not limited to anti-CD3 and/or anti-CD28 monoclonal antibodies. Soluble antibodies are added at an appropriate concentration. In some experiments, plates pre-coated with anti-CD3 antibodies, for example, are used in combination with soluble anti-CD28 antibody.
Attorney Docket No.250298.000682 [00526] Another alternative method to activate T cell lines is activating beads coupled with anti-CD3/anti-CD28 antibodies. After transduction, infected cells are counted, and anti-CD3/anti-CD28 beads are added to the culture medium at a given ratio. [00527] Cells staining and FACS analysis. Fluorescence-activated cell sorting (FACS) is performed after the transduction. Transduced cells are counted and seeded into a cell culture plate. Cells are spun, washed, and then spun again any number of times. Cells are incubated with a dye for determining viability of cells, e.g., Live/DeadTM Fixable Near- IR stain, washed, and incubated with Fc block in buffer, e.g., FACS stain buffer. After being washed any number of times again with FACS stain buffer, cells are subsequently incubated with antibodies such as, but not limited to, fluorescent-labeled antibodies targeting TCR and control. Cells are washed with FACS stain buffer, fixed, washed again, and resuspended in FACS stain buffer. Samples are run for analysis with a FACS analyzer. [00528] Proliferation and cytokine production assays. To measure T cell proliferation, 3H-thymidine is added to assay cultures following contact with the peptide- pulsed APC. Following incubation, cultures are harvested onto filter bottom microplates. MicroScint 20 scintillation fluid or the like is added to each well, and plates are counted on a Scintillation Counter. [00529] For transcription factor activity analysis and cytokines production assay, cells are harvested after contact with the peptide-pulsed APC, centrifuged, and cell pellets and supernatants are collected. [00530] Cell pellets are processed for RNA extraction for transcriptomics analysis via qPCR. Cytokine production is measured by ELISA from the supernatant. Cell pellets are processed for luciferase detection assay to measure programmed cell death, e.g., AP1 activity. [00531] Splenocytes isolation and CD8+T cell culture. Spleens of adult general purpose strain mice, e.g., C57Bl/6 mice, are excised and placed in cold buffer. Tissues are then homogenized to break apart the spleens. Dissociated cells are centrifuged. After centrifugation, the cell pellet is resuspended in a suitable volume of lysis buffer designed to remove red blood cells, e.g., ACK (Ammonium-Chloride-Potassium) lysis buffer, and incubated in the buffer. After incubation, the cell suspension is added to buffer, and centrifuged. The cell pellet is then resuspended in buffer and the cell solution is filtered.
Attorney Docket No.250298.000682 The filtered cell suspension is centrifuged again and the subsequent pellet is resuspended in a suitable volume of buffer. CD8+ T cells are then isolated such as, for example, by using a CD8a+ T cell Isolation Kit. [00532] Peptide immunization of mice. For immunization, a suitable amount of HTLV-1 peptide or control peptide is diluted in buffer and emulsified at a given ratio with an adjuvant such as, but not limited to, complete Freund’s adjuvant (CFA) or incomplete Freund’s adjuvant (IFA) using, for example, a double syringes system, to reach a final desired volume of peptide/adjuvant emulsion. A measured volumed (e.g., 200 µl) of the peptide/adjuvant emulsion is then injected by a given route of administration, for example, subcutaneously, into mice (e.g., C57Bl/6 mice) under appropriate anesthesia in appropriate location(s). [00533] Quantification of cytotoxicity activity by flow cytometry. [00534] Preparation of target cells: For preparation of target cells (e.g., autologous B cells), the target cells are isolated from splenocytes of OT-1 or P14 mice by using a Mouse B cell Kit (or the like, according to the manufacturer's instruction), and are subsequently stimulated for a predetermined duration in the presence of a set concentration of IFN-γ. Targets cells are then counted, divided into tubes, and washed in buffer. A proportion of the target cells are stained with a high concentration (e.g., 0.2 μM) of CFSE (CFSEHigh) and a separate proportion with a low concentration (e.g, 0.02 μM) of CFSE (CFSELow) in buffer. After the incubation, cells are pelleted and resuspended in an appropriate media to quench the labeling reaction. Target cells stained with the low concentration of CFSE are pulsed by adding the HTLV-1 peptides or control OVA peptides at a suitable final concentration under appropriate culture conditions. Both target cells stained with different concentration of CFSE are then washed, resuspended at a measured concentration in media and mixed with an appropriate ratio, such as a 1:1 ratio (CFSEHigh:CFSELow). [00535] Effector cells: Total CD8+ T cells containing the effector cells are enriched from splenocytes of P14 and OT-1 mice using a Negative Selection Human CD8 T cell isolation Kit. CD8+ T cells are counted, resuspended in complete T cell medium, and serially diluted volume:volume in a given volume of complete T cell medium. From each dilution, a given volume of cells are seeded in duplicate to multi-well cell culture plate.
Attorney Docket No.250298.000682 The mixed target cells are added to each dilution of effector cells. To measure basal apoptosis, a number of wells are seeded with target cells alone. Cell mixtures are incubated under standard cell culture conditions. [00536] Flow cytometry staining and acquisition: Cells are transferred to multi-well cell culture plate, washed in FACS staining buffer and stained with, for example, iTag Tetramer/APC-H-2Kb OVA9 ^ (MBL), iTag Tetramer/APC-H-2Db HTLV-1 ^Alexa Fluor ^ and Live/Dead ^ Fixable Near-IR stain (ThermoFisher) under appropriate conditions. Cells are then washed in FACS stain buffer before staining with, for example, Fluorescently-labeled monoclonal antibody that specifically binds to CD8 alpha such as, but not limited to, BV421 αCD8a. Cells are washed with FACS stain buffer and resuspended fixative. Acquisition is performed and all cells are acquired. Post-acquisition data analysis performed. [00537] Preparation of vectors encoding peptides. A therapeutic DNA or RNA vaccine comprising polynucleotides or vectors encoding polynucleotides to be used is prepared by GMP manufacturing of the plasmid vaccine according to regulatory authorities' guidelines. The vaccine is appropriately formulated, for example, by dissolving in a saline solution, at a suitable concentration. The vaccine may be administered either intradermal or intramuscular with or without following electroporation or alternatively with a jet injector. * * * [00538] The claimed subject matter is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the claimed subject matter in addition to those described herein will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims. [00539] All patents, applications, publications, test methods, literature, and other materials cited herein are hereby incorporated by reference in their entirety as if physically present in this specification.
Claims
Attorney Docket No.250298.000682 CLAIMS 1. An isolated peptide comprising an amino acid sequence that is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof, wherein the isolated peptide is 5-20 amino acids in length. 2. The isolated peptide of claim 1, wherein the isolated peptide comprises an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. 3. The isolated peptide of claim 1 or 2, wherein the isolated peptide consists essentially of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. 4. The isolated peptide of any one of claims 1-3, wherein the isolated peptide consists of an amino acid sequence of any one of SEQ ID NOs: 1-89 and 143-146. 5. An isolated peptide comprising two or more amino acid sequences selected from any one of SEQ ID NO: 1-89 and 143-146, or a pharmaceutically acceptable salt thereof, or a fragment or derivative thereof. 6. The isolated peptide of any one of claims 1-5, wherein the isolated peptide comprises one or more reverse peptide bonds, one or more non-peptide bonds, one or more D- isomers of amino acids, one or more chemical modifications, or any combination thereof. 7. The isolated peptide of any one of claims 1-6, wherein the isolated peptide is produced by expression in a heterologous host cell. 8. The isolated peptide of any one of claims 1-6, wherein the isolated peptide is produced synthetically. 9. The isolated peptide of any one of claims 1-8, wherein the isolated peptide, or pharmaceutically acceptable salt thereof, or fragment or derivative thereof induces a
Attorney Docket No.250298.000682 Human T-Lymphotropic Virus Type-1 (HTLV-1)-specific immune response in a subject when presented in a complex with a major histocompatibility complex (MHC) molecule on the surface of an antigen presenting cell (APC). 10. A fusion protein comprising one or more isolated peptides of any one of claims 1-9 fused to one or more heterologous molecules. 11. The fusion protein of claim 10, wherein the one or more heterologous molecules enhance a peptide-specific immune response in a subject. 12. The fusion protein of claim 10, wherein the one or more heterologous molecules mediate peptide delivery to a specific site within a subject. 13. The fusion protein of any one of claims 10-12, wherein the one or more heterologous molecules are a MHC molecule, or a fragment or derivative thereof. 14. A conjugate comprising one or more isolated peptides of any one of claims 1-9 conjugated to one or more heterologous molecules. 15. The conjugate of claim 14, wherein the one or more heterologous molecules enhance a peptide-specific immune response in a subject. 16. The conjugate of claim 14, wherein the one or more heterologous molecules mediate peptide delivery to a specific site within a subject. 17. The conjugate of any one of claims 14-16, wherein the one or more heterologous molecules are an MHC molecule, or a fragment or derivative thereof. 18. The conjugate of any one of claims 14-16, wherein the one or more peptides are conjugated to a particle. 19. An oligomeric complex comprising two or more isolated peptides of any one of claims 1-9.
Attorney Docket No.250298.000682 20. A non-covalent complex comprising the isolated peptide of any one of claims 1-9 and an MHC molecule, or a fragment or derivative thereof. 21. The non-covalent complex of claim 20, wherein the MHC molecule, or the fragment thereof, is a class I MHC molecule. 22. The non-covalent complex of claim 21, wherein the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule. 23. The non-covalent complex of claim 20, wherein the MHC molecule, or the fragment thereof, is a class II MHC molecule. 24. The non-covalent complex of claim 23, wherein the class II MHC molecule is a class II HLA molecule. 25. A fusion protein comprising the isolated peptide of any one of claims 1-9 and an MHC molecule, or a fragment or derivative thereof. 26. The fusion protein of claim 25, wherein the MHC molecule, or the fragment thereof, is a class I MHC molecule. 27. The fusion protein of claim 26, wherein the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule. 28. The fusion protein of claim 25, wherein the MHC molecule, or the fragment thereof, is a class II MHC molecule. 29. The fusion protein of claim 28, wherein the class II MHC molecule is a class II HLA molecule. 30. A conjugate comprising the isolated peptide of any one of claims 1-9 and a MHC molecule, or a fragment or derivative thereof.
Attorney Docket No.250298.000682 31. The conjugate of claim 30, wherein the MHC molecule, or the fragment thereof, is a class I MHC molecule. 32. The conjugate of claim 31, wherein the class I MHC molecule is a class I human leukocyte antigen (HLA) molecule. 33. The conjugate of claim 30, wherein the MHC molecule, or the fragment thereof, is a class II MHC molecule. 34. The conjugate of claim 33, wherein the class II MHC molecule is a class II HLA molecule. 35. A pharmaceutical composition comprising (i) one or more isolated peptides of any one of claims 1-9, one or more fusion proteins of any one of claims 10-13 and 25-29, one or more conjugates of any one of claims 14-18 and 30-34, one or more oligomeric complexes of claim 19, or one or more non-covalent complexes of any one of claims 20-24, or any combination thereof; and (ii) a pharmaceutically acceptable carrier or excipient. 36. The pharmaceutical composition of claim 35, further comprising an adjuvant. 37. An isolated molecule that binds the isolated peptide of any one of claims 1-9, the fusion protein of any one of claims 10-13 and 25-29, the conjugate of any one of claims 14-18 and 30-34, the oligomeric complex of claim 19, or the non-covalent complex of any one of claims 20-24. 38. The isolated molecule of claim 37, wherein the molecule is an antibody or an antigen- binding fragment thereof. 39. The isolated molecule of claim 38, wherein the antibody is a bispecific antibody.
Attorney Docket No.250298.000682 40. The isolated molecule of claim 37, wherein the molecule is an alternative scaffold. 41. The isolated molecule of claim 37, wherein the molecule is a chimeric antigen receptor (CAR). 42. The isolated molecule of claim 37, wherein the molecule is a T cell receptor (TCR). 43. An isolated cell comprising the CAR of claim 41. 44. The isolated cell of claim 43, wherein the isolated cell is an immune cell. 45. The isolated cell of claim 44, wherein the immune cell is a T cell, an NK cell, or a macrophage. 46. An isolated cell comprising the TCR of claim 42. 47. The isolated cell of claim 46, wherein the isolated cell is an immune cell. 48. The isolated cell of claim 47, wherein the immune cell is a T cell, an NK cell, or a macrophage. 49. A pharmaceutical composition comprising (i) the isolated molecule of any one of claims 38-43, or the isolated cell of any one of claims 43-48; and (ii) a pharmaceutically acceptable carrier or excipient. 50. An isolated polynucleotide comprising a nucleotide sequence encoding one or more isolated peptides of any one of claims 1-9 or the fusion protein of any one of claims 10-13 and 25-29. 51. The isolated polynucleotide of claim 50, wherein the nucleotide sequence is operably linked to a promoter.
Attorney Docket No.250298.000682 52. The isolated polynucleotide of claim 50 or claim 51, wherein the isolated polynucleotide comprises DNA. 53. The isolated polynucleotide of claim 50 or claim 51, wherein the isolated polynucleotide comprises RNA. 54. The isolated polynucleotide of claim 53, wherein the RNA is mRNA. 55. The isolated polynucleotide of claim 53, wherein the RNA is self-replicating RNA. 56. A vector comprising the isolated polynucleotide of any one of claims 50-55. 57. The vector of claim 56, wherein the vector is an expression vector. 58. The vector of claim 56 or claim 57, wherein the vector is a viral vector. 59. A host cell comprising the isolated polynucleotide of any one of claims 50-55 or the vector of any one of claims 56-58. 60. The host cell of claim 59, wherein the host cell is a prokaryotic cell. 61. The host cell of claim 59, wherein the host cell is a eukaryotic cell. 62. The host cell of claim 61, wherein the host cell is an APC. 63. A pharmaceutical composition comprising (i) the isolated polynucleotide of any one of claims 50-55, or the vector of any one of claims 56-58; and (ii) a pharmaceutically acceptable carrier or excipient. 64. The pharmaceutical composition of claim 63, wherein the pharmaceutically acceptable carrier is a lipid nanoparticle carrier.
Attorney Docket No.250298.000682 65. A method of inducing an immune response against a HTLV-1infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of: a) one or more isolated peptides of any one of claims 1-9; b) the fusion protein of any one of claims 10-13 and 25-29; c) the conjugate of any one of claims 14-18 and 30-34; d) the oligomeric complex of claim 19; e) the non-covalent complex of any one of claims 20-24; f) the pharmaceutical composition of any one of claims 35, 36, 49, 63, and 64; g) the molecule of any one of claims 37-42; h) the isolated cell of any one of claims 43-48 and 59-62; i) the isolated polynucleotide of any one of claims 50-55; or j) the vector of any one of claims 56-58. 66. A method of inducing an immune response against a HTLV-1 infection in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of one or more isolated peptides of any one of claims 1-9. 67. A method of inducing an immune response against a HTLV-1 infection in a subject in need thereof, the method comprising administering to the subject an activated T cell that is produced by contacting a T cell with an APC that presents the isolated peptide of any one of claims 1-9 in complex with an MHC molecule. 68. A method of treating a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of: a) one or more isolated peptides of any one of claims 1-9; b) the fusion protein of any one of claims 10-13 and 25-29; c) the conjugate of any one of claims 14-18 and 30-34; d) the oligomeric complex of claim 19; e) the non-covalent complex of any one of claims 20-24;
Attorney Docket No.250298.000682 f) the pharmaceutical composition of any one of claims 35, 36, 49, 63, and 64; g) the molecule of any one of claims 37-42; h) the isolated cell of any one of claims 43-48 and 59-62; i) the isolated polynucleotide of any one of claims 50-55; or j) the vector of any one of claims 56-58. 69. A method of preventing or reducing the likelihood of a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of: a) one or more isolated peptides of any one of claims 1-9; b) the fusion protein of any one of claims 10-13 and 25-29; c) the conjugate of any one of claims 14-18 and 30-34; d) the oligomeric complex of claim 19; e) the non-covalent complex of any one of claims 20-24; f) the pharmaceutical composition of any one of claims 35, 36, 49, 63, and 64; g) the molecule of any one of claims 37-42; h) the isolated cell of any one of claims 43-48 and 59-62; i) the isolated polynucleotide of any one of claims 50-55; or j) the vector of any one of claims 56-58. 70. A method of treating a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of one or more isolated peptides of any one of claims 1-9. 71. A method of preventing or reducing the likelihood of a HTLV-1-induced disease or disorder in a subject in need thereof, the method comprising administering to the subject an effective amount of one or more isolated peptides of any one of claims 1-9. 72. The method of any one of claims 69-71, wherein the HTLV-1-induced disease or disorder is adult T-cell leukemia/lymphoma (ATL) or HTLV-1-myelopathy/tropical spastic paraparesis (HAM/TSP).
Attorney Docket No.250298.000682 73. A kit comprising: (i) a) one or more isolated peptides of any one of claims 1-9; b) the fusion protein of any one of claims 10-13 and 25-29; c) the conjugate of any one of claims 14-18 and 30-34; d) the oligomeric complex of claim 19; e) the non-covalent complex of any one of claims 20-24; f) the pharmaceutical composition of any one of claims 35, 36, 49, 63, and 64; g) the molecule of any one of claims 37-42; h) the isolated cell of any one of claims 43-48 and 59-62; i) the isolated polynucleotide of any one of claims 50-55; or j) the vector of any one of claims 56-58; and (ii) packaging and/or instructions for use for the same.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202363583834P | 2023-09-19 | 2023-09-19 | |
US63/583,834 | 2023-09-19 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2025064470A1 true WO2025064470A1 (en) | 2025-03-27 |
Family
ID=93010712
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2024/047158 WO2025064470A1 (en) | 2023-09-19 | 2024-09-18 | Viral peptides and uses thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2025064470A1 (en) |
-
2024
- 2024-09-18 WO PCT/US2024/047158 patent/WO2025064470A1/en unknown
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7525442B2 (en) | Novel peptides and peptide combinations for use in immunotherapy against lung cancer, including NSCLC, and other cancers - Patent Application 20070123333 | |
US20230382997A1 (en) | Antigen-binding proteins targeting shared antigens | |
JP2024028750A (en) | Antigen-binding proteins that target shared antigens | |
JP2024028730A (en) | Novel peptides and peptide-scaffold combinations for use in immunotherapy against renal cell carcinoma (RCC) and other cancers | |
JP2023518821A (en) | Coronavirus vaccines and how to use them | |
JP2021040639A (en) | Novel peptides and peptide combinations for use in immunotherapy against prostate cancer and other cancers | |
JP2022058349A (en) | Novel peptides and peptide combinations for use in immunotherapy for CLL and other cancers | |
KR20180012865A (en) | Combinations of novel cell epitopes and cell epitopes for use in immunotherapy of myeloma and other cancers | |
CN115175934A (en) | Antigen binding proteins targeting consensus neoantigens | |
KR20180030506A (en) | Combinations of novel peptides and peptides for use in immunotherapy for epithelial ovarian cancer and other cancers | |
TW201841937A (en) | Novel peptides and combination of peptides for use in immunotherapy against leukemias and other cancers | |
JP5292550B2 (en) | T cell receptor β chain gene and α chain gene | |
CN111315769A (en) | Dual-function receptor for signaling and antigen presentation (SABR) | |
JP7026970B2 (en) | Peptides and combinations of peptides for use in immunotherapy for leukemia and other cancers | |
JP2017537642A (en) | Chimeric antigen receptor and method of use thereof | |
TW202043284A (en) | Multispecific binding proteins | |
US20220033460A1 (en) | Identification and use of t cell epitopes in designing diagnostic and therapeutic approaches for covid-19 | |
JP2021533785A (en) | Antigen-binding protein that targets shared antigens | |
JP2024535896A (en) | Coronavirus vaccines and how to use them | |
CN107002073A (en) | Tumor antigen peptides | |
WO2023240085A1 (en) | Viral peptides and uses thereof | |
WO2025064470A1 (en) | Viral peptides and uses thereof | |
TW201309727A (en) | Two-component antigen presenting composition and use thereof | |
US12209137B2 (en) | Antigen binding proteins specifically binding CT45 | |
NL2034657B1 (en) | RCN1-derived TEIPP neoantigens and uses thereof |